1
|
Dawson LE, Sekar A, Fulford AD, Lambert RI, Burgess HS, Ribeiro PS. The deubiquitylating enzyme Fat facets promotes Fat signalling and restricts tissue growth. Nat Commun 2025; 16:1938. [PMID: 39994229 PMCID: PMC11850632 DOI: 10.1038/s41467-025-57164-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Tissue growth is regulated by many signals, including polarity cues. The Hippo signalling pathway restricts tissue growth and receives inputs from the planar cell polarity-controlling Fat signalling pathway. The atypical cadherin Fat restricts growth via several mechanisms that ultimately control the activity of the pro-growth transcriptional co-activator Yorkie. Fat signalling activates the Yorkie inhibitory kinase Warts, and modulates the function of the FERM protein Expanded, which promotes Hippo signalling and also directly inhibits Yorkie. Although several Fat pathway activity modulators are known to be involved in ubiquitylation, the role of this post-translational modification in the pathway remains unclear. Moreover, no deubiquitylating enzymes have been described in this pathway. Here, using in vivo RNAi screening, we identify the deubiquitylating enzyme Fat facets as a positive regulator of Fat signalling with roles in tissue growth control. Fat facets interacts genetically and physically with Fat signalling components and regulates Yorkie target gene expression. Thus, we uncover a role for reversible ubiquitylation in the control of Fat signalling and tissue growth regulation.
Collapse
Affiliation(s)
- Lauren E Dawson
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, Fulham Road, London, UK
| | - Aashika Sekar
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, UK
| | - Alexander D Fulford
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel I Lambert
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Hannah S Burgess
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Paulo S Ribeiro
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK.
| |
Collapse
|
2
|
Gallego-Zazo N, Tenorio-Castano J, Parra A, Nevado J, Cazalla M, Lucas-Castro E, Heath KE, Palomares M, Soengas E, Lledín MD, Larrea E, Olveira A, Morte B, Carracedo Á, Lapunzina P. AMOTL1 -Associated Multiple Congenital Anomalies (Craniofaciocardiohepatic Syndrome, CFCHS): A Novel Clinical Spectrum Including Craniofacial, Heart and Liver Abnormalities. Clin Genet 2025; 107:234-236. [PMID: 39538375 DOI: 10.1111/cge.14644] [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: 09/13/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
We identified an AMOTL1 variant in a patient that adds evidence supporting the clinical and molecular overlap between AMOTL1-related disorders and other syndromes affecting craniofacial, cardiac, and hepatic development. As more cases are identified, we propose naming this entity as AMOTL1-associated multiple congenital anomalies or craniofaciocardiohepatic syndrome (CFCHS).
Collapse
Affiliation(s)
- Natalia Gallego-Zazo
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ERN ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and Other Neuro-Developmental Disorders, Paris, France
| | - Jair Tenorio-Castano
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ERN ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and Other Neuro-Developmental Disorders, Paris, France
| | - Alejandro Parra
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ERN ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and Other Neuro-Developmental Disorders, Paris, France
| | - Julián Nevado
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ERN ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and Other Neuro-Developmental Disorders, Paris, France
| | - Mario Cazalla
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ERN ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and Other Neuro-Developmental Disorders, Paris, France
| | - Elsa Lucas-Castro
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
| | - Karen E Heath
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
| | - María Palomares
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ERN ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and Other Neuro-Developmental Disorders, Paris, France
| | - Emma Soengas
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - M Dolores Lledín
- Department of Pediatric Hepatology and Liver Transplant, Hospital Universitario La Paz, Madrid, Spain
| | - Emily Larrea
- Department of Gastrology and Hepatology, Hospital Universitario La Paz, Madrid, Spain
| | - Antonio Olveira
- Department of Gastrology and Hepatology, Hospital Universitario La Paz, Madrid, Spain
| | - Beatriz Morte
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Ángel Carracedo
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- Department of Genetics, Fundación de Medicina Xenómica, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica (SERGAS), IDIS, Santiago de Compostela, Spain
- Grupo de Medicina Genómica, CIMUS, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Pablo Lapunzina
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- INGEMM-Idipaz, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ERN ITHACA, European Reference Network on Rare Malformations Syndromes, Intellectual and Other Neuro-Developmental Disorders, Paris, France
| |
Collapse
|
3
|
Shewale B, Ebrahim T, Samal A, Dubois N. Molecular Regulation of Cardiomyocyte Maturation. Curr Cardiol Rep 2025; 27:32. [PMID: 39836238 DOI: 10.1007/s11886-024-02189-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE OF THE REVIEW This review aims to discuss the process of cardiomyocyte maturation, with a focus on the underlying molecular mechanisms required to form a fully functional heart. We examine both long-standing concepts associated with cardiac maturation and recent developments, and the overall complexity of molecularly integrating all the processes that lead to a mature heart. RECENT FINDINGS Cardiac maturation, defined here as the sequential changes that occurring before the heart reaches full maturity, has been a subject of investigation for decades. Recently, there has been a renewed, highly focused interest in this process, driven by clinically motivated research areas where enhancing maturation may lead to improved therapeutic opportunities. These include using pluripotent stem cell models for cell therapy and disease modeling, as well as recent advancements in adult cardiac regeneration approaches. We highlight key processes underlying maturation of the heart, including cellular and organ growth, and electrophysiological, metabolic, and contractile maturation. We further discuss how these processes integrate and interact to contribute to the overall complexity of the developing heart. Finally, we emphasize the transformative potential for translating relevant maturation concepts to emerging models of heart disease and regeneration.
Collapse
Affiliation(s)
- Bhavana Shewale
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tasneem Ebrahim
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arushi Samal
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Liu S, Deshmukh V, Meng F, Wang Y, Morikawa Y, Steimle JD, Li RG, Wang J, Martin JF. Microtubules Sequester Acetylated YAP in the Cytoplasm and Inhibit Heart Regeneration. Circulation 2025; 151:59-75. [PMID: 39185559 PMCID: PMC11671299 DOI: 10.1161/circulationaha.123.067646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The Hippo pathway effector YAP (Yes-associated protein) plays an essential role in cardiomyocyte proliferation and heart regeneration. In response to physiological changes, YAP moves in and out of the nucleus. The pathophysiological mechanisms regulating YAP subcellular localization after myocardial infarction remain poorly defined. METHODS We identified YAP acetylation at site K265 by in vitro acetylation followed by mass spectrometry analysis. We used adeno-associated virus to express YAP-containing mutations that either abolished acetylation (YAP-K265R) or mimicked acetylation (YAP-K265Q) and studied how acetylation regulates YAP subcellular localization in mouse hearts. We generated a cell line with YAP-K265R mutation and investigated the protein-protein interactors by YAP immunoprecipitation followed by mass spectrometry, then validated the YAP interaction in neonatal rat ventricular myocytes. We examined colocalization of YAP and TUBA4A (tubulin α 4A) by superresolution imaging. Furthermore, we developed YAP-K265R and αMHC-MerCreMer (MCM); Yap-loxP/K265R mutant mice to examine the pathophysiological role of YAP acetylation in cardiomyocytes during cardiac regeneration. RESULTS We found that YAP is acetylated at K265 by CBP (CREB-binding protein)/P300 (E1A-binding protein P300) and is deacetylated by nicotinamide phosphoribosyltransferase/nicotinamide adenine dinucleotide/sirtuins axis in cardiomyocytes. After myocardial infarction, YAP acetylation is increased, which promotes YAP cytoplasmic localization. Compared with controls, mice that were genetically engineered to express a K265R mutation that prevents YAP K265 acetylation showed improved cardiac regenerative ability and increased YAP nuclear localization. Mechanistically, YAP acetylation facilitates its interaction with TUBA4A, a component of the microtubule network that sequesters acetylated YAP in the cytoplasm. After myocardial infarction, the microtubule network increased in cardiomyocytes, resulting in the accumulation of YAP in the cytoplasm. CONCLUSIONS After myocardial infarction, decreased sirtuin activity enriches YAP acetylation at K265. The growing TUBA4A network sequesters acetylated YAP within the cytoplasm, which is detrimental to cardiac regeneration.
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (S.L.)
| | - Vaibhav Deshmukh
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
- Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, MO (V.D.)
| | - Fansen Meng
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| | | | - Yuka Morikawa
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - Jeffrey D Steimle
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| | - Rich Gang Li
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - Jun Wang
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| |
Collapse
|
5
|
Guo Y, Dupart M, Irondelle M, Peraldi P, Bost F, Mazure NM. YAP1 modulation of primary cilia-mediated ciliogenesis in 2D and 3D prostate cancer models. FEBS Lett 2024; 598:3071-3086. [PMID: 39424416 DOI: 10.1002/1873-3468.15029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
The primary cilium, a non-motile organelle present in most human cells, plays a crucial role in detecting microenvironmental changes and regulating intracellular signaling. Its dysfunction is linked to various diseases, including cancer. We explored the role of ciliated cells in prostate cancer by using Gefitinib and Jasplakinolide compounds to induce ciliated cells in both normal and tumor-like prostate cell lines. We assessed GLI1 and IFT20 expression and investigated YAP1 protein's role, which is implicated in primary cilium regulation. Finally, we examined these compounds in 3D cell models, aiming to simulate in vivo conditions. Our study highlights YAP1 as a potential target for novel genetic models to understand the primary cilium's role in mediating resistance to anticancer treatments.
Collapse
Affiliation(s)
- Yingbo Guo
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Mathilde Dupart
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
- IRCAN, Université Côte d'Azur, Nice Cedex 02, France
| | - Marie Irondelle
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
| | - Pascal Peraldi
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Frederic Bost
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Nathalie M Mazure
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| |
Collapse
|
6
|
Li P, Que Y, Wong C, Lin Y, Qiu J, Gao B, Zhou H, Hu W, Shi H, Peng Y, Huang D, Gao W, Qiu X, Liang A. IL-32 aggravates metabolic disturbance in human nucleus pulposus cells by activating FAT4-mediated Hippo/YAP signaling. Int Immunopharmacol 2024; 141:112966. [PMID: 39178518 DOI: 10.1016/j.intimp.2024.112966] [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: 02/07/2024] [Revised: 07/21/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
Extracellular matrix (ECM) metabolism disorders in the inflammatory microenvironment play a key role in the pathogenesis of intervertebral disc degeneration (IDD). Interleukin-32 (IL-32) has been reported to be involved in the progression of various inflammatory diseases; however, it remains unclear whether it participates in the matrix metabolism of nucleus pulposus (NP) cells. Therefore, this study aimed to investigate the mechanism of IL-32 on regulating the ECM metabolism in the inflammatory microenvironment. RNA-seq was used to identify aberrantly expressed genes in NP cells in the inflammatory microenvironment. Western blotting, real-time quantitative PCR, immunohistochemistry and immunofluorescence analysis were performed to measure the expression of IL-32 and metabolic markers in human NP tissues or NP cells treated with or without tumor necrosis factor-α (TNF-α). In vivo, an adeno-associated virus overexpressing IL-32 was injected into the caudal intervertebral discs of rats to assess its effect on IDD. Proteins interacting with IL-32 were identified via immunoprecipitation and mass spectrometry. Lentivirus overexpressing IL-32 or knocking down Fat atypical cadherin 4 (FAT4), yes-associated protein (YAP) inhibitor-Verteporfin (VP) were used to treat human NP cells, to explore the pathogenesis of IL-32. Hippo/YAP signaling activity was verified in human NP tissues. IL-32 expression was significantly upregulated in degenerative NP tissues, as indicated in the clinical samples. Furthermore, IL-32 was remarkably overexpressed in TNF-α-induced degenerative NP cells. IL-32 overexpression induced IDD progression in the rat model. Mechanistically, the elevation of IL-32 in the inflammatory microenvironment enhanced its interactions with FAT4 and mammalian sterile 20-like kinase1/2 (MST1/2) proteins, prompting MST1/2 phosphorylation, and activating the Hippo/YAP signaling pathway, causing matrix metabolism disorder in NP cells. Our results suggest that IL-32 mediates matrix metabolism disorders in NP cells in the inflammatory micro-environment via the FAT4/MST/YAP axis, providing a theoretical basis for the precise treatment of IDD.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yichen Que
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Orthopedic Surgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical Universit, Qingyuan, Guangdong, China
| | - Chipiu Wong
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Youxi Lin
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jincheng Qiu
- Department of Minimally Invasive Spine Surgery, Panyu Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Bo Gao
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hang Zhou
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenjun Hu
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huihong Shi
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Peng
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dongsheng Huang
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenjie Gao
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xianjian Qiu
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Anjing Liang
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Meijuan C, Fang M, Qian W. Dachsous cadherin related 1 (DCHS1) is a novel biomarker for immune infiltration and epithelial-mesenchymal transition in endometrial cancer via pan-cancer analysis. J Ovarian Res 2024; 17:162. [PMID: 39123216 PMCID: PMC11312386 DOI: 10.1186/s13048-024-01478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 07/15/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Dachsous cadherin related 1 (DCHS1) is one of calcium-dependent adhesion membrane proteins and is mainly involved in the development of mammalian tissues. There is a lack of more detailed research on the biological function of DCHS1 in pan-cancer. MATERIALS AND METHODS We evaluated the expression, the prognostic value, the diagnostic value and genomic alterations of DCHS1 by using the databases, including TCGA, UALCAN, HPA, GEPIA2.0 and GSCA. We employed the databases of UCSC, TIMER2.0, TISIDB, GSCA to analyze the association between DCHS1 expression and the immune microenvironment, stemness, TMB, MSI and anticancer drug sensitivity. BioGRID, STRING and GEPIA2.0 were used to perform protein interaction and functional enrichment analysis. Real-time quantitative PCR, CCK8, Transwell assay and Western blot were performed to determine the function of DCHS1 in UCEC. RESULTS DCHS1 is differentially expressed in many cancers and its expression is significantly associated with tumor prognosis and diagnosis. DCHS1 expression was significantly correlated with the infiltration of cancer-associated fibroblasts (CAFs), Endothelial cell (ECs), and Hematopoietic stem cell in most cancers. In addition, DCHS1 was significantly associated with sensitivity to many antitumor drugs. Functional enrichment analysis revealed that DCHS1-related proteins were involved in Focal adhesion, Endometrial cancer and Wnt signaling pathway. GSEA results showed that DCHS1 was related to epithelial-mesenchymal transition (EMT) in many cancers. In vitro experiments in UCEC showed that DCHS1 regulated cell proliferation, migration and EMT. CONCLUSIONS Our findings indicated that DCHS1 might be a novel prognostic and diagnostic biomarker and immunotherapy target, and plays an important role in the proliferation, migration and EMT in UCEC.
Collapse
Affiliation(s)
- Cai Meijuan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), No.758 Hefei Road, Qingdao, 266035, Shandong, China
| | - Min Fang
- Department of Gynecology, Qingdao Women's and Children's Hospital, Qingdao University, Qingdao, Shandong, China
| | - Wang Qian
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), No.758 Hefei Road, Qingdao, 266035, Shandong, China.
| |
Collapse
|
8
|
Wang XX, Zhao YM, Zhang QY, Zhao JX, Yin DH, Zhang ZZ, Jin XY, Li SN, Ji HY, Chen HY, Guo XF, Yu Y, Ma WY, Yan H, Li H, Ou-Yang QM, Pan ZW, Liang HH, Wang N, Chen W, Cai BZ, Liu Y. Acetylcytidine modification of Amotl1 by N-acetyltransferase 10 contributes to cardiac fibrotic expansion in mice after myocardial infarction. Acta Pharmacol Sin 2024; 45:1425-1437. [PMID: 38839936 PMCID: PMC11192918 DOI: 10.1038/s41401-024-01306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024]
Abstract
Cardiac fibrosis is a pathological scarring process that impairs cardiac function. N-acetyltransferase 10 (Nat10) is recently identified as the key enzyme for the N4-acetylcytidine (ac4C) modification of mRNAs. In this study, we investigated the role of Nat10 in cardiac fibrosis following myocardial infarction (MI) and the related mechanisms. MI was induced in mice by ligation of the left anterior descending coronary artery; cardiac function was assessed with echocardiography. We showed that both the mRNA and protein expression levels of Nat10 were significantly increased in the infarct zone and border zone 4 weeks post-MI, and the expression of Nat10 in cardiac fibroblasts was significantly higher compared with that in cardiomyocytes after MI. Fibroblast-specific overexpression of Nat10 promoted collagen deposition and induced cardiac systolic dysfunction post-MI in mice. Conversely, fibroblast-specific knockout of Nat10 markedly relieved cardiac function impairment and extracellular matrix remodeling following MI. We then conducted ac4C-RNA binding protein immunoprecipitation-sequencing (RIP-seq) in cardiac fibroblasts transfected with Nat10 siRNA, and revealed that angiomotin-like 1 (Amotl1), an upstream regulator of the Hippo signaling pathway, was the target gene of Nat10. We demonstrated that Nat10-mediated ac4C modification of Amotl1 increased its mRNA stability and translation in neonatal cardiac fibroblasts, thereby increasing the interaction of Amotl1 with yes-associated protein 1 (Yap) and facilitating Yap translocation into the nucleus. Intriguingly, silencing of Amotl1 or Yap, as well as treatment with verteporfin, a selective and potent Yap inhibitor, attenuated the Nat10 overexpression-induced proliferation of cardiac fibroblasts and prevented their differentiation into myofibroblasts in vitro. In conclusion, this study highlights Nat10 as a crucial regulator of myocardial fibrosis following MI injury through ac4C modification of upstream activators within the Hippo/Yap signaling pathway.
Collapse
Affiliation(s)
- Xiu-Xiu Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Yi-Ming Zhao
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Qian-Yun Zhang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Jing-Xuan Zhao
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Dao-Hong Yin
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Zi-Zhen Zhang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Xiao-Yan Jin
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Shuai-Nan Li
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Hao-Yu Ji
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Hong-Yang Chen
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Xiao-Fei Guo
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Yang Yu
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Wen-Ya Ma
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Hong Yan
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Han Li
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Qi-Meng Ou-Yang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Zhen-Wei Pan
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Hai-Hai Liang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Ning Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China.
| | - Wei Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Ben-Zhi Cai
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China.
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China.
- NHC Key Laboratory of Cell Transplantation, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University, Harbin, 150001, China.
| | - Yu Liu
- Department of Clinical Laboratory at the Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
9
|
Zhao Y, Sun B, Fu X, Zuo Z, Qin H, Yao K. YAP in development and disease: Navigating the regulatory landscape from retina to brain. Biomed Pharmacother 2024; 175:116703. [PMID: 38713948 DOI: 10.1016/j.biopha.2024.116703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
The distinctive role of Yes-associated protein (YAP) in the nervous system has attracted widespread attention. This comprehensive review strategically uses the retina as a vantage point, embarking on an extensive exploration of YAP's multifaceted impact from the retina to the brain in development and pathology. Initially, we explore the crucial roles of YAP in embryonic and cerebral development. Our focus then shifts to retinal development, examining in detail YAP's regulatory influence on the development of retinal pigment epithelium (RPE) and retinal progenitor cells (RPCs), and its significant effects on the hierarchical structure and functionality of the retina. We also investigate the essential contributions of YAP in maintaining retinal homeostasis, highlighting its precise regulation of retinal cell proliferation and survival. In terms of retinal-related diseases, we explore the epigenetic connections and pathophysiological regulation of YAP in diabetic retinopathy (DR), glaucoma, and proliferative vitreoretinopathy (PVR). Lastly, we broaden our exploration from the retina to the brain, emphasizing the research paradigm of "retina: a window to the brain." Special focus is given to the emerging studies on YAP in brain disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD), underlining its potential therapeutic value in neurodegenerative disorders and neuroinflammation.
Collapse
Affiliation(s)
- Yaqin Zhao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Bin Sun
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xuefei Fu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhuan Zuo
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
10
|
Wang H, Ye M, Jin X. Role of angiomotin family members in human diseases (Review). Exp Ther Med 2024; 27:258. [PMID: 38766307 PMCID: PMC11099588 DOI: 10.3892/etm.2024.12546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/23/2023] [Indexed: 05/22/2024] Open
Abstract
Angiomotin (Amot) family members, including Amot, Amot-like protein 1 (Amotl1) and Amot-like protein 2 (Amotl2), have been found to interact with angiostatins. In addition, Amot family members are involved in various physiological and pathological functions such as embryonic development, angiogenesis and tumorigenesis. Some studies have also demonstrated its regulation in signaling pathways such as the Hippo signaling pathway, AMPK signaling pathway and mTOR signaling pathways. Amot family members play an important role in neural stem cell differentiation, dendritic formation and synaptic maturation. In addition, an increasing number of studies have focused on their function in promoting and/or suppressing cancer, but the underlying mechanisms remain to be elucidated. The present review integrated relevant studies on upstream regulation and downstream signals of Amot family members, as well as the latest progress in physiological and pathological functions and clinical applications, hoping to offer important ideas for further research.
Collapse
Affiliation(s)
- Haoyun Wang
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
11
|
Song M, Wang H, Liu C, Jin S, Liu B, Sun W. Non-coding RNAs as regulators of the Hippo pathway in cardiac development and cardiovascular disease. Front Pharmacol 2024; 15:1348280. [PMID: 38698813 PMCID: PMC11063341 DOI: 10.3389/fphar.2024.1348280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Cardiovascular diseases pose a serious threat to human health. The onset of cardiovascular diseases involves the comprehensive effects of multiple genes and environmental factors, and multiple signaling pathways are involved in regulating the occurrence and development of cardiovascular diseases. The Hippo pathway is a highly conserved signaling pathway involved in the regulation of cell proliferation, apoptosis, and differentiation. Recently, it has been widely studied in the fields of cardiovascular disease, cancer, and cell regeneration. Non-coding RNA (ncRNAs), which are important small molecules for the regulation of gene expression in cells, can directly target genes and have diverse regulatory functions. Recent studies have found that ncRNAs interact with Hippo pathway components to regulate myocardial fibrosis, cardiomyocyte proliferation, apoptosis, and hypertrophy and play an important role in cardiovascular disease. In this review, we describe the mode of action of ncRNAs in regulating the Hippo pathway, provide new ideas for further research, and identify molecules involved in the mechanism of action of ncRNAs and the Hippo pathway as potential therapeutic targets, with the aim of finding new modes of action for the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Mengyang Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Caixia Liu
- Department of Neurology, The Liaoning Province People’s Hospital, Shenyang, China
| | - Sijie Jin
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Noël ES. Cardiac construction-Recent advances in morphological and transcriptional modeling of early heart development. Curr Top Dev Biol 2024; 156:121-156. [PMID: 38556421 DOI: 10.1016/bs.ctdb.2024.02.005] [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] [Indexed: 04/02/2024]
Abstract
During human embryonic development the early establishment of a functional heart is vital to support the growing fetus. However, forming the embryonic heart is an extremely complex process, requiring spatiotemporally controlled cell specification and differentiation, tissue organization, and coordination of cardiac function. These complexities, in concert with the early and rapid development of the embryonic heart, mean that understanding the intricate interplay between these processes that help shape the early heart remains highly challenging. In this review I focus on recent insights from animal models that have shed new light on the earliest stages of heart development. This includes specification and organization of cardiac progenitors, cell and tissue movements that make and shape the early heart tube, and the initiation of the first beat in the developing heart. In addition I highlight relevant in vitro models that could support translation of findings from animal models to human heart development. Finally I discuss challenges that are being addressed in the field, along with future considerations that together may help move us towards a deeper understanding of how our hearts are made.
Collapse
Affiliation(s)
- Emily S Noël
- School of Biosciences and Bateson Centre, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
13
|
Parrish AG, Arora S, Thirimanne HN, Rudoy D, Schmid S, Sievers P, Sahm F, Holland EC, Szulzewsky F. Aggressive high-grade NF2 mutant meningiomas downregulate oncogenic YAP signaling via the upregulation of VGLL4 and FAT3/4. Neurooncol Adv 2024; 6:vdae148. [PMID: 39380691 PMCID: PMC11459063 DOI: 10.1093/noajnl/vdae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Background Meningiomas are the most common primary central nervous system tumors in adults. Although generally benign, a subset is of higher grade and ultimately fatal. Around half of all meningiomas harbor inactivating mutations in NF2, leading to deregulation of oncogenic YAP1 activity. While benign NF2 mutant meningiomas exhibit few genetic events in addition to NF2 inactivation, aggressive high-grade NF2 mutant meningiomas frequently harbor a highly aberrant genome. It is unclear if NF2 mutant meningiomas of different grades are equally reliant on YAP activity. Methods We analyzed bulk and single-cell RNA-Seq data from a large cohort of human meningiomas for the expression of YAP1 target genes and Hippo effectors as well as in vitro cell line experiments. Results Aggressive NF2 mutant meningiomas harbor decreased expression levels of YAP1 target genes and increased expression levels of the YAP1 antagonist VGLL4 and the upstream regulators FAT3/4 compared to their benign counterparts. Decreased expression of YAP1 target genes as well as high expression of VGLL4 and FAT3/4 is significantly associated with an increased risk of recurrence. In vitro, overexpression of VGLL4 resulted in the downregulation of YAP activity in benign NF2 mutant meningioma cells, confirming the direct link between VGLL4 expression and decreased levels of YAP activity observed in aggressive NF2 mutant meningiomas. Conclusions Our results shed new insight into the biology of benign and aggressive NF2 mutant meningiomas and may have important implications for the efficacy of therapies targeting oncogenic YAP1 activity in NF2 mutant meningiomas.
Collapse
Affiliation(s)
- Abigail G Parrish
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sonali Arora
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Dmytro Rudoy
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sebastian Schmid
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Philipp Sievers
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Sahm
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Eric C Holland
- Seattle Translational Tumor Research Center, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Frank Szulzewsky
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
14
|
Harvey DC, Verma R, Sedaghat B, Hjelm BE, Morton SU, Seidman JG, Kumar SR. Mutations in genes related to myocyte contraction and ventricular septum development in non-syndromic tetralogy of Fallot. Front Cardiovasc Med 2023; 10:1249605. [PMID: 37840956 PMCID: PMC10569225 DOI: 10.3389/fcvm.2023.1249605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Objective Eighty percent of patients with a diagnosis of tetralogy of Fallot (TOF) do not have a known genetic etiology or syndrome. We sought to identify key molecular pathways and biological processes that are enriched in non-syndromic TOF, the most common form of cyanotic congenital heart disease, rather than single driver genes to elucidate the pathogenesis of this disease. Methods We undertook exome sequencing of 362 probands with non-syndromic TOF and their parents within the Pediatric Cardiac Genomics Consortium (PCGC). We identified rare (minor allele frequency <1 × 10-4), de novo variants to ascertain pathways and processes affected in this population to better understand TOF pathogenesis. Pathways and biological processes enriched in the PCGC TOF cohort were compared to 317 controls without heart defects (and their parents) from the Simons Foundation Autism Research Initiative (SFARI). Results A total of 120 variants in 117 genes were identified as most likely to be deleterious, with CHD7, CLUH, UNC13C, and WASHC5 identified in two probands each. Gene ontology analyses of these variants using multiple bioinformatic tools demonstrated significant enrichment in processes including cell cycle progression, chromatin remodeling, myocyte contraction and calcium transport, and development of the ventricular septum and ventricle. There was also a significant enrichment of target genes of SOX9, which is critical in second heart field development and whose loss results in membranous ventricular septal defects related to disruption of the proximal outlet septum. None of these processes was significantly enriched in the SFARI control cohort. Conclusion Innate molecular defects in cardiac progenitor cells and genes related to their viability and contractile function appear central to non-syndromic TOF pathogenesis. Future research utilizing our results is likely to have significant implications in stratification of TOF patients and delivery of personalized clinical care.
Collapse
Affiliation(s)
- Drayton C. Harvey
- Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Riya Verma
- Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Brandon Sedaghat
- Department of Medicine, Rosalind Franklin University School of Medicine and Science, Chicago, IL, United States
| | - Brooke E. Hjelm
- Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Sarah U. Morton
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Jon G. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - S. Ram Kumar
- Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
15
|
Xu XC, Jiang JX, Zhou YQ, He S, Liu Y, Li YQ, Wei PP, Bei JX, Sun J, Luo CL. SRSF3/AMOTL1 splicing axis promotes the tumorigenesis of nasopharyngeal carcinoma through regulating the nucleus translocation of YAP1. Cell Death Dis 2023; 14:511. [PMID: 37558679 PMCID: PMC10412622 DOI: 10.1038/s41419-023-06034-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Dysregulation of serine/arginine splicing factors (SRSFs) and abnormal alternative splicing (AS) have been widely implicated in various cancers but scarcely investigated in nasopharyngeal carcinoma (NPC). Here we examine the expression of 12 classical SRSFs between 87 NPC and 10 control samples, revealing a significant upregulation of SRSF3 and its association with worse prognosis in NPC. Functional assays demonstrate that SRSF3 exerts an oncogenic function in NPC progression. Transcriptome analysis reveals 1,934 SRSF3-regulated AS events in genes related to cell cycle and mRNA metabolism. Among these events, we verify the generation of a long isoform of AMOTL1 (AMOTL1-L) through a direct bond of the SRSF3 RRM domain with the exon 12 of AMOTL1 to promote exon inclusion. Functional studies also reveal that AMOTL1-L promotes the proliferation and migration of NPC cells, while AMOTL1-S does not. Furthermore, overexpression of AMOTL1-L, but not -S, significantly rescues the inhibitory effects of SRSF3 knockdown. Additionally, compared with AMOTL1-S, AMOTL1-L has a localization preference in the intracellular than the cell membrane, leading to a more robust interaction with YAP1 to promote nucleus translocation. Our findings identify SRSF3/AMOTL1 as a novel alternative splicing axis with pivotal roles in NPC development, which could serve as promising prognostic biomarkers and therapeutic targets for NPC.
Collapse
Affiliation(s)
- Xiao-Chen Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jia-Xin Jiang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
| | - Ya-Qing Zhou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
| | - Shuai He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
| | - Yang Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
| | - Yi-Qi Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
| | - Pan-Pan Wei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
| | - Jin-Xin Bei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Medical Oncology, National Cancer Centre of Singapore, Singapore, Singapore
| | - Jian Sun
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China.
| | - Chun-Ling Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, P. R. China.
| |
Collapse
|
16
|
Xin JW, Chai ZX, Jiang H, Cao HW, Chen XY, Zhang CF, Zhu Y, Zhang Q, Ji QM. Genome-wide comparison of DNA methylation patterns between yak and three cattle strains and their potential association with mRNA transcription. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2023; 340:316-328. [PMID: 36148637 DOI: 10.1002/jez.b.23174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 07/29/2022] [Accepted: 09/01/2022] [Indexed: 05/11/2023]
Abstract
Yak has evolved specific adaptative mechanisms to high-altitude environment. Up to date, only a few studies reported the DNA methylation in yak. In the present study, genome-wide DNA methylome and transcriptome profiles in lung, mammary, and biceps brachii muscle tissues were compared between yak and three cattle breeds (Tibetan cattle, Sanjiang cattle, and Holstein cattle). The association between differentially expressed genes (DEGs) and differentially methylated regions (DMRs) was analyzed, and the biological functions of DEGs potentially driven by DMRs were explored by KEGG enrichment analysis. Finally, we found that yak-specific DMRs-driven DEGs were mainly involved in neuromodulation, respiration, lung development, blood pressure regulation, cardiovascular protection, energy metabolism, DNA repair, and immune functions. The higher levels of the key genes associated with these functions were observed in yak than in cattle, suggesting that DNA methylation might regulate these genes. Overall, the present study contributes basic data at the DNA methylation level to further understand the physiological metabolism in yak.
Collapse
Affiliation(s)
- Jin-Wei Xin
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| | - Zhi-Xin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hui Jiang
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| | - Han-Wen Cao
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| | - Xiao-Ying Chen
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| | - Cheng-Fu Zhang
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| | - Yong Zhu
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| | - Qiang Zhang
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa, Tibet, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| | - Qiu-Mei Ji
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, Tibet, China
| |
Collapse
|
17
|
Tyser RCV. Formation of the Heart: Defining Cardiomyocyte Progenitors at Single-Cell Resolution. Curr Cardiol Rep 2023; 25:495-503. [PMID: 37119451 PMCID: PMC10188409 DOI: 10.1007/s11886-023-01880-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE OF REVIEW Formation of the heart requires the coordinated addition of multiple progenitor sources which have undergone different pathways of specification and differentiation. In this review, I aim to put into context how recent studies defining cardiac progenitor heterogeneity build on our understanding of early heart development and also discuss the questions raised by this new insight. RECENT FINDINGS With the development of sequencing technologies and imaging approaches, it has been possible to define, at high temporal resolution, the molecular profile and anatomical location of cardiac progenitors at the single-cell level, during the formation of the mammalian heart. Given the recent progress in our understanding of early heart development and technical advances in high-resolution time-lapse imaging and lineage analysis, we are now in a position of great potential, allowing us to resolve heart formation at previously impossible levels of detail. Understanding how this essential organ forms not only addresses questions of fundamental biological significance but also provides a blueprint for strategies to both treat and model heart disease.
Collapse
Affiliation(s)
- Richard C V Tyser
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, CB2 0AW, UK.
| |
Collapse
|
18
|
Strong A, Rao S, von Hardenberg S, Li D, Cox LL, Lee PC, Zhang LQ, Awotoye W, Diamond T, Gold J, Gooch C, Gowans LJJ, Hakonarson H, Hing A, Loomes K, Martin N, Marazita ML, Mononen T, Piccoli D, Pfundt R, Raskin S, Scherer SW, Sobriera N, Vaccaro C, Wang X, Watson D, Weksberg R, Bhoj E, Murray JC, Lidral AC, Butali A, Buckley MF, Roscioli T, Koolen DA, Seaver LH, Prows CA, Stottmann RW, Cox TC. A mutational hotspot in AMOTL1 defines a new syndrome of orofacial clefting, cardiac anomalies, and tall stature. Am J Med Genet A 2023; 191:1227-1239. [PMID: 36751037 PMCID: PMC10081944 DOI: 10.1002/ajmg.a.63130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/03/2023] [Accepted: 01/17/2023] [Indexed: 02/09/2023]
Abstract
AMOTL1 encodes angiomotin-like protein 1, an actin-binding protein that regulates cell polarity, adhesion, and migration. The role of AMOTL1 in human disease is equivocal. We report a large cohort of individuals harboring heterozygous AMOTL1 variants and define a core phenotype of orofacial clefting, congenital heart disease, tall stature, auricular anomalies, and gastrointestinal manifestations in individuals with variants in AMOTL1 affecting amino acids 157-161, a functionally undefined but highly conserved region. Three individuals with AMOTL1 variants outside this region are also described who had variable presentations with orofacial clefting and multi-organ disease. Our case cohort suggests that heterozygous missense variants in AMOTL1, most commonly affecting amino acid residues 157-161, define a new orofacial clefting syndrome, and indicates an important functional role for this undefined region.
Collapse
Affiliation(s)
- Alanna Strong
- The Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Soumya Rao
- Department of Oral & Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City Kansas City, Missouri
| | | | - Dong Li
- The Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Liza L. Cox
- Department of Oral & Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City Kansas City, Missouri
| | - Paul C. Lee
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Li Q. Zhang
- Department of Oral & Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City Kansas City, Missouri
| | - Waheed Awotoye
- Department of Orthodontics, College of Dentistry, University of Iowa, Iowa City, Iowa
| | - Tamir Diamond
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Gastroenterology, Hepatology and Nutrition. Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Jessica Gold
- The Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Catherine Gooch
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Lord Jephthah Joojo Gowans
- Department of Biochemistry and Biotechnology, Kwame Nkurumah University of Science and Technology, Kumasi, Ghana
| | - Hakon Hakonarson
- The Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Pulmonary Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Anne Hing
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington, Seattle, Washington
| | - Kathleen Loomes
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Gastroenterology, Hepatology and Nutrition. Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Nicole Martin
- Division of Clinical & Metabolic Genetics and Department of Genetic Counselling, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mary L. Marazita
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics School of Dental Medicine, Pittsburgh, Pennsylvania
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tarja Mononen
- Department of Clinical Genetics, Kuopio University Hospital, Kuopio, Finland
| | - David Piccoli
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Gastroenterology, Hepatology and Nutrition. Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Rolph Pfundt
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Salmo Raskin
- Assistance Center for Cleft Lip and Palate (CAIF), Curitiba-PR, Brazil
| | - Stephen W. Scherer
- The Centre for Applied Genomics and Department of Genetics & Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- McLaughlin Centre and Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Nara Sobriera
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Courtney Vaccaro
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Xiang Wang
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Deborah Watson
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rosanna Weksberg
- Institute of Medical Sciences and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Clinical & Metabolic Genetics, Department of Pediatrics, and Genetics and Genome Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elizabeth Bhoj
- The Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Genomic Diagnostics and Department of Pathology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | | | - Azeez Butali
- Departments of Oral Pathology, Radiology and Medicine, College of Dentistry & Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Michael F. Buckley
- NSW Health Pathology Genomics Laboratory, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Tony Roscioli
- NSW Health Pathology Genomics Laboratory, Prince of Wales Hospital, Randwick, NSW, Australia
- Centre for Clinical Genetics, Sydney Children’s Hospital, Randwick, NSW, Australia
- Neuroscience Research Australia and Prince of Wales Clinical School, University of New South Wales, Kensington, NSW, Australia
| | - David A. Koolen
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Laurie H. Seaver
- Spectrum Health Helen DeVos Children’s Hospital, Grand Rapids, Michigan
- Department of Pediatrics and Human Development, Michigan State University College of Human Medicine, Grand Rapids, Michigan
| | - Cynthia A. Prows
- Divisions of Human Genetics and Patient Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Rolf W. Stottmann
- Divisions of Human Genetics and Patient Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Steve & Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University School of Medicine, Columbus, Ohio, USA
| | - Timothy C. Cox
- Department of Oral & Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City Kansas City, Missouri
- Department of Pediatrics, School of Medicine, University of Missouri-Kansas City Kansas City, Missouri, 64108, USA
| |
Collapse
|
19
|
Fulford AD, Enderle L, Rusch J, Hodzic D, Holder MV, Earl A, Oh RH, Tapon N, McNeill H. Expanded directly binds conserved regions of Fat to restrain growth via the Hippo pathway. J Cell Biol 2023; 222:e202204059. [PMID: 37071483 PMCID: PMC10120405 DOI: 10.1083/jcb.202204059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/26/2022] [Accepted: 02/09/2023] [Indexed: 04/19/2023] Open
Abstract
The Hippo pathway is a conserved and critical regulator of tissue growth. The FERM protein Expanded is a key signaling hub that promotes activation of the Hippo pathway, thereby inhibiting the transcriptional co-activator Yorkie. Previous work identified the polarity determinant Crumbs as a primary regulator of Expanded. Here, we show that the giant cadherin Fat also regulates Expanded directly and independently of Crumbs. We show that direct binding between Expanded and a highly conserved region of the Fat cytoplasmic domain recruits Expanded to the apicolateral junctional zone and stabilizes Expanded. In vivo deletion of Expanded binding regions in Fat causes loss of apical Expanded and promotes tissue overgrowth. Unexpectedly, we find Fat can bind its ligand Dachsous via interactions of their cytoplasmic domains, in addition to the known extracellular interactions. Importantly, Expanded is stabilized by Fat independently of Dachsous binding. These data provide new mechanistic insights into how Fat regulates Expanded, and how Hippo signaling is regulated during organ growth.
Collapse
Affiliation(s)
- Alexander D. Fulford
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, USA
| | - Leonie Enderle
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Jannette Rusch
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, USA
| | - Didier Hodzic
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, USA
| | - Maxine V. Holder
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, London, UK
| | - Alex Earl
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, USA
| | - Robin Hyunseo Oh
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Nicolas Tapon
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, London, UK
| | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, USA
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
20
|
Structure of the planar cell polarity cadherins Fat4 and Dachsous1. Nat Commun 2023; 14:891. [PMID: 36797229 PMCID: PMC9935876 DOI: 10.1038/s41467-023-36435-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
The atypical cadherins Fat and Dachsous are key regulators of cell growth and animal development. In contrast to classical cadherins, which form homophilic interactions to segregate cells, Fat and Dachsous cadherins form heterophilic interactions to induce cell polarity within tissues. Here, we determine the co-crystal structure of the human homologs Fat4 and Dachsous1 (Dchs1) to establish the molecular basis for Fat-Dachsous interactions. The binding domains of Fat4 and Dchs1 form an extended interface along extracellular cadherin (EC) domains 1-4 of each protein. Biophysical measurements indicate that Fat4-Dchs1 affinity is among the highest reported for cadherin superfamily members, which is attributed to an extensive network of salt bridges not present in structurally similar protocadherin homodimers. Furthermore, modeling suggests that unusual extracellular phosphorylation modifications directly modulate Fat-Dachsous binding by introducing charged contacts across the interface. Collectively, our analyses reveal how the molecular architecture of Fat4-Dchs1 enables them to form long-range, high-affinity interactions to maintain planar cell polarity.
Collapse
|
21
|
CircAMOTL1 RNA and AMOTL1 Protein: Complex Functions of AMOTL1 Gene Products. Int J Mol Sci 2023; 24:ijms24032103. [PMID: 36768425 PMCID: PMC9916871 DOI: 10.3390/ijms24032103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
The complexity of the cellular proteome facilitates the control of a wide range of cellular processes. Non-coding RNAs, including microRNAs and long non-coding RNAs, greatly contribute to the repertoire of tools used by cells to orchestrate various functions. Circular RNAs (circRNAs) constitute a specific class of non-coding RNAs that have recently emerged as a widely generated class of molecules produced from many eukaryotic genes that play essential roles in regulating cellular processes in health and disease. This review summarizes current knowledge about circRNAs and focuses on the functions of AMOTL1 circRNAs and AMOTL1 protein. Both products from the AMOTL1 gene have well-known functions in physiology, cancer, and other disorders. Using AMOTL1 as an example, we illustrate how focusing on both circRNAs and proteins produced from the same gene contributes to a better understanding of gene functions.
Collapse
|
22
|
Kasiah J, McNeill H. Fat and Dachsous cadherins in mammalian development. Curr Top Dev Biol 2023; 154:223-244. [PMID: 37100519 DOI: 10.1016/bs.ctdb.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Cell growth and patterning are critical for tissue development. Here we discuss the evolutionarily conserved cadherins, Fat and Dachsous, and the roles they play during mammalian tissue development and disease. In Drosophila, Fat and Dachsous regulate tissue growth via the Hippo pathway and planar cell polarity (PCP). The Drosophila wing has been an ideal tissue to observe how mutations in these cadherins affect tissue development. In mammals, there are multiple Fat and Dachsous cadherins, which are expressed in many tissues, but mutations in these cadherins that affect growth and tissue organization are context dependent. Here we examine how mutations in the Fat and Dachsous mammalian genes affect development in mammals and contribute to human disease.
Collapse
Affiliation(s)
- Jennysue Kasiah
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States.
| |
Collapse
|
23
|
Langa P, Wolska BM, Solaro RJ. The Hippo Signaling Pathway as a Drug Target in Familial Dilated Cardiomyopathy. INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2022; 1:4. [PMID: 38818406 PMCID: PMC11139043 DOI: 10.53941/ijddp.v1i1.189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
We focus here on the Hippo pathway in the hierarchical sensing and modulation of the mechanical state of the adult heart in health and disease. The Hippo pathway interrogates the micro-environment of cardiac myocytes providing surveillance of the mechanical state with engagement of signaling pathways critical to homeostasis of cardiac development, remodeling, and function and vulnerable to pathologies. Our discussion centers on Hippo signaling in the altered mechanical state instigated by variants of genes expressing mutant sarcomere proteins that trigger a progression to dilated cardiomyopathy (familial DCM). There is an unmet need for therapies in DCM. Recent progress in the discovery of small molecules that target Hippo signaling and are intended for use in cardiac disorders provides leads for modifying Hippo in DCM. As we emphasize, identifying useful targets in DCM requires in depth understanding of cell specific Hippo signaling in the cardiac micro-environment.
Collapse
Affiliation(s)
- Paulina Langa
- Department of Physiology and Biophysics and the Center for Cardiovascular Research,University of Illinois at Chicago, Chicago, IL,USA
| | - Beata M. Wolska
- Department of Physiology and Biophysics and the Center for Cardiovascular Research,University of Illinois at Chicago, Chicago, IL,USA
- Department of Medicine, Division of Cardiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - R. John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research,University of Illinois at Chicago, Chicago, IL,USA
| |
Collapse
|
24
|
Zhang CH, Gao Y, Hung HH, Zhuo Z, Grodzinsky AJ, Lassar AB. Creb5 coordinates synovial joint formation with the genesis of articular cartilage. Nat Commun 2022; 13:7295. [PMID: 36435829 PMCID: PMC9701237 DOI: 10.1038/s41467-022-35010-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/15/2022] [Indexed: 11/28/2022] Open
Abstract
While prior work has established that articular cartilage arises from Prg4-expressing perichondrial cells, it is not clear how this process is specifically restricted to the perichondrium of synovial joints. We document that the transcription factor Creb5 is necessary to initiate the expression of signaling molecules that both direct the formation of synovial joints and guide perichondrial tissue to form articular cartilage instead of bone. Creb5 promotes the generation of articular chondrocytes from perichondrial precursors in part by inducing expression of signaling molecules that block a Wnt5a autoregulatory loop in the perichondrium. Postnatal deletion of Creb5 in the articular cartilage leads to loss of both flat superficial zone articular chondrocytes coupled with a loss of both Prg4 and Wif1 expression in the articular cartilage; and a non-cell autonomous up-regulation of Ctgf. Our findings indicate that Creb5 promotes joint formation and the subsequent development of articular chondrocytes by driving the expression of signaling molecules that both specify the joint interzone and simultaneously inhibit a Wnt5a positive-feedback loop in the perichondrium.
Collapse
Affiliation(s)
- Cheng-Hai Zhang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA.
| | - Yao Gao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Han-Hwa Hung
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhu Zhuo
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Andrew B Lassar
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA.
| |
Collapse
|
25
|
Domingo-Relloso A, Makhani K, Riffo-Campos AL, Tellez-Plaza M, Klein KO, Subedi P, Zhao J, Moon KA, Bozack AK, Haack K, Goessler W, Umans JG, Best LG, Zhang Y, Herreros-Martinez M, Glabonjat RA, Schilling K, Galvez-Fernandez M, Kent JW, Sanchez TR, Taylor KD, Craig Johnson W, Durda P, Tracy RP, Rotter JI, Rich SS, Berg DVD, Kasela S, Lappalainen T, Vasan RS, Joehanes R, Howard BV, Levy D, Lohman K, Liu Y, Daniele Fallin M, Cole SA, Mann KK, Navas-Acien A. Arsenic Exposure, Blood DNA Methylation, and Cardiovascular Disease. Circ Res 2022; 131:e51-e69. [PMID: 35658476 PMCID: PMC10203287 DOI: 10.1161/circresaha.122.320991] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/18/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Epigenetic dysregulation has been proposed as a key mechanism for arsenic-related cardiovascular disease (CVD). We evaluated differentially methylated positions (DMPs) as potential mediators on the association between arsenic and CVD. METHODS Blood DNA methylation was measured in 2321 participants (mean age 56.2, 58.6% women) of the Strong Heart Study, a prospective cohort of American Indians. Urinary arsenic species were measured using high-performance liquid chromatography coupled to inductively coupled plasma mass spectrometry. We identified DMPs that are potential mediators between arsenic and CVD. In a cross-species analysis, we compared those DMPs with differential liver DNA methylation following early-life arsenic exposure in the apoE knockout (apoE-/-) mouse model of atherosclerosis. RESULTS A total of 20 and 13 DMPs were potential mediators for CVD incidence and mortality, respectively, several of them annotated to genes related to diabetes. Eleven of these DMPs were similarly associated with incident CVD in 3 diverse prospective cohorts (Framingham Heart Study, Women's Health Initiative, and Multi-Ethnic Study of Atherosclerosis). In the mouse model, differentially methylated regions in 20 of those genes and DMPs in 10 genes were associated with arsenic. CONCLUSIONS Differential DNA methylation might be part of the biological link between arsenic and CVD. The gene functions suggest that diabetes might represent a relevant mechanism for arsenic-related cardiovascular risk in populations with a high burden of diabetes.
Collapse
Affiliation(s)
- Arce Domingo-Relloso
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
- Department of Statistics and Operations Research, University of Valencia, Spain
| | - Kiran Makhani
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Angela L. Riffo-Campos
- Millennium Nucleus on Sociomedicine (SocioMed) and Vicerrectoría Académica, Universidad de La Frontera, Temuco, Chile
- Department of Computer Science, ETSE, University of Valencia, Valencia, Spain
| | - Maria Tellez-Plaza
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Kathleen Oros Klein
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Pooja Subedi
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jinying Zhao
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Katherine A. Moon
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anne K. Bozack
- Department of Environmental Health Sciences, School of Public Health, University of California, Berkeley, USA
| | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Walter Goessler
- Institute of Chemistry - Analytical Chemistry for Health and Environment, University of Graz, Austria
| | | | - Lyle G. Best
- Missouri Breaks Industries and Research Inc., Eagle Butte, SD, USA
| | - Ying Zhang
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, OK, USA
| | | | - Ronald A. Glabonjat
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Kathrin Schilling
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Marta Galvez-Fernandez
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Jack W. Kent
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Tiffany R Sanchez
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Peter Durda
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Russell P. Tracy
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - David Van Den Berg
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Silva Kasela
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Tuuli Lappalainen
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Ramachandran S Vasan
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA; Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine, Department of Medicine, department of Epidemiology, Boston University Schools of medicine and Public health, Boston, MA, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | | | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
- Framingham Heart Study, Framingham, MA
| | - Kurt Lohman
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Yongmei Liu
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - M Daniele Fallin
- Departments of Mental Health and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Shelley A. Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Koren K. Mann
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
26
|
Zhang X, Chen N, Chen H, Lei C, Sun T. Comparative analyses of copy number variations between swamp and river buffalo. Gene X 2022; 830:146509. [PMID: 35460806 DOI: 10.1016/j.gene.2022.146509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 11/30/2022] Open
Abstract
Domestic buffalo is an important livestock in the tropical and sub-tropical region, including two types: swamp and river buffalo. The swamp buffalo is mainly used as draft animal, while the river buffalo is raised for milk production. In this study, based on the new high-quality buffalo reference genome UOA_WB_1, we firstly investigated the copy number variants in buffalo using whole-genome Illumina sequencing. A total of 3,734 CNV regions (CNVRs) were detected in 106 buffalo population with a total length of 23,429,066 bp, corresponding to ∼ 0.88% of the water buffalo genome (UOA_WB_1). Our results revealed a clear population differentiation in CNV between swamp and river buffalo. In addition, a total of 667 highly differentiated CNVRs (covering 886 genes) were detected between river and swamp buffalo population. We detected a set of CNVR-overlapping genes associated with exercise, immunity, nerve, and milk trait which exhibited different copy numbers between swamp and river buffalo population. This study provides valuable genome variation resources for buffalo and would contribute to understanding the genetic differences between swamp and river buffalo.
Collapse
Affiliation(s)
- Xianfu Zhang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University, Hangzhou, Zhejiang 311300, China.
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ting Sun
- College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China.
| |
Collapse
|
27
|
Liu S, Li RG, Martin JF. The cell-autonomous and non–cell-autonomous roles of the Hippo pathway in heart regeneration. J Mol Cell Cardiol 2022; 168:98-106. [PMID: 35526477 DOI: 10.1016/j.yjmcc.2022.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/04/2022] [Accepted: 04/23/2022] [Indexed: 10/18/2022]
|
28
|
Zani ALS, Gouveia MH, Aquino MM, Quevedo R, Menezes RL, Rotimi C, Lwande GO, Ouma C, Mekonnen E, Fagundes NJR. Genetic differentiation in East African ethnicities and its relationship with endurance running success. PLoS One 2022; 17:e0265625. [PMID: 35588128 PMCID: PMC9119534 DOI: 10.1371/journal.pone.0265625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/04/2022] [Indexed: 12/02/2022] Open
Abstract
Since the 1960s, East African athletes, mainly from Kenya and Ethiopia, have dominated long-distance running events in both the male and female categories. Further demographic studies have shown that two ethnic groups are overrepresented among elite endurance runners in each of these countries: the Kalenjin, from Kenya, and the Oromo, from Ethiopia, raising the possibility that this dominance results from genetic or/and cultural factors. However, looking at the life history of these athletes or at loci previously associated with endurance athletic performance, no compelling explanation has emerged. Here, we used a population approach to identify peaks of genetic differentiation for these two ethnicities and compared the list of genes close to these regions with a list, manually curated by us, of genes that have been associated with traits possibly relevant to endurance running in GWAS studies, and found a significant enrichment in both populations (Kalenjin, P = 0.048, and Oromo, P = 1.6x10-5). Those traits are mainly related to anthropometry, circulatory and respiratory systems, energy metabolism, and calcium homeostasis. Our results reinforce the notion that endurance running is a systemic activity with a complex genetic architecture, and indicate new candidate genes for future studies. Finally, we argue that a deterministic relationship between genetics and sports must be avoided, as it is both scientifically incorrect and prone to reinforcing population (racial) stereotyping.
Collapse
Affiliation(s)
- André L. S. Zani
- Postgraduate Program in Genetics and Molecular Biology, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Mateus H. Gouveia
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Marla M. Aquino
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo Quevedo
- School of Physical Education, Physical Therapy and Dance, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rodrigo L. Menezes
- School of Physical Education, Physical Therapy and Dance, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Charles Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gerald O. Lwande
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya
| | - Collins Ouma
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya
| | - Ephrem Mekonnen
- Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Nelson J. R. Fagundes
- Postgraduate Program in Genetics and Molecular Biology, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Postgraduate Program in Animal Biology, Institute of Biosciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- * E-mail:
| |
Collapse
|
29
|
Vogel A, Crawford A, Nyarko A. Multivalent Angiomotin-like 1 and Yes-associated protein form a dynamic complex. Protein Sci 2022; 31:e4295. [PMID: 35481651 PMCID: PMC8994507 DOI: 10.1002/pro.4295] [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: 11/10/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/11/2022]
Abstract
Multivalent complexes formed between the cancer-promoting transcriptional co-activator, Yes-associated protein (YAP), and proteins containing short linear motifs of type PPxY modulate cell proliferation and are attractive therapeutic targets. However, challenges producing PPxY polypeptides containing the full binding domain has limited understanding of the assembly process. Here, we successfully produced a polypeptide containing the complete set of three PPxY binding sites of Angiomotin-like 1 (AMOTL1), a scaffolding protein that regulates the nucleo-cytoplasmic shuttling of YAP via WW-PPxY interactions. Using an array of biophysical techniques including isothermal titration calorimetry, size-exclusion chromatography coupled to multi-angle light scattering, and solution nuclear magnetic resonance spectroscopy, we show that the AMOTL1 polypeptide is partially disordered, and binds the YAP WW domains to form an ensemble of complexes of varying stabilities. The binding process is initiated by the binding of one YAP WW domain to one AMOTL1 PPxY motif and is completed by transient interactions of the second YAP WW domain with a second AMOTL1 PPxY motif to form an equilibrating mixture composed of various species having two YAP sites bound to two conjugate AMOTL1 sites. We rationalize that the transient interactions fine-tune the stability of the complex for rapid assembly and disassembly in response to changes in the local cellular environment.
Collapse
Affiliation(s)
- Amber Vogel
- Department of Biochemistry & BiophysicsOregon State UniversityCorvallisOregonUSA
| | - Alexandra Crawford
- Department of Biochemistry & BiophysicsOregon State UniversityCorvallisOregonUSA
| | - Afua Nyarko
- Department of Biochemistry & BiophysicsOregon State UniversityCorvallisOregonUSA
| |
Collapse
|
30
|
Gridnev A, Misra JR. Emerging Mechanisms of Growth and Patterning Regulation by Dachsous and Fat Protocadherins. Front Cell Dev Biol 2022; 10:842593. [PMID: 35372364 PMCID: PMC8967653 DOI: 10.3389/fcell.2022.842593] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/08/2022] [Indexed: 01/14/2023] Open
Abstract
Dachsous (Ds) and Fat are evolutionarily conserved cell adhesion molecules that play a critical role in development of multiple organ systems, where they coordinate tissue growth and morphogenesis. Much of our understanding of Ds-Fat signaling pathway comes from studies in Drosophila, where they initiate a signaling pathway that regulate growth by influencing Hippo signaling and morphogenesis by regulating Planar Cell Polarity (PCP). In this review, we discuss recent advances in our understanding of the mechanisms by which Ds-Fat signaling pathway regulates these critical developmental processes. Further, we discuss the progress in our understanding about how they function in mammals.
Collapse
|
31
|
Xu G, Seng Z, Zhang M, Qu J. Angiomotin-like 1 plays a tumor-promoting role in glioma by enhancing the activation of YAP1 signaling. ENVIRONMENTAL TOXICOLOGY 2021; 36:2500-2511. [PMID: 34480788 DOI: 10.1002/tox.23363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 06/13/2023]
Abstract
Angiomotin-like 1 (AMOTL1) is reportedly a pivotal tumor-associated protein that is strongly associated with the tumorigenesis of multiple malignant tumors. However, the issue of whether AMOTL1 plays a role in the tumorigenesis of glioma remains unclear. The aim of this work was to explore the possible relationship between AMOTL1 and glioma progression. Results demonstrated that high levels of AMOTL1 in glioma tissues were associated with a reduced survival rate in patients with glioma. Cellular functional assays revealed that silencing of AMOTL1 in glioma cell lines substantially decreased cell proliferation and invasion and increased cell apoptosis. Further investigation revealed that silencing of AMOTL1 inhibited the activation of yes-associated protein 1 (YAP1) and decreased the expression of YAP1 target genes. Reactivation of YAP1 reversed AMOTL1-silencing-induced antitumor effects, whereas inhibition of YAP1 abolished AMOTL1-overexpression-induced tumor-promoting effects in glioma cells. Silencing of AMOTL1 also retarded the growth of glioma cell-derived xenograft tumors in vivo. In conclusion, these findings suggested that AMOTL1 may exert a tumor-promoting function in glioma by enhancing the activation of YAP1 signaling. This work suggested AMOTL1 as a potential target for the development of antiglioma therapy.
Collapse
Affiliation(s)
- Gang Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhiyuan Seng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianqiang Qu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
32
|
Bhatt B, Prakhar P, Lohia GK, Rajmani RS, Balaji KN. Pre-existing mycobacterial infection modulates Candida albicans-driven pyroptosis. FEBS J 2021; 289:1536-1551. [PMID: 34670010 DOI: 10.1111/febs.16243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/26/2021] [Accepted: 10/20/2021] [Indexed: 11/26/2022]
Abstract
Active tuberculosis patients are at high risk of coinfection with opportunistic fungal pathogen Candida albicans. However, the molecular mechanisms that orchestrate pathogenesis of Mycobacterium tuberculosis (Mtb)-C. albicans coinfection remain elusive. In the current study, we utilize a mouse model to demonstrate that Mtb promotes a macrophage environment that is conducive for C. albicans survival. Mtb-dependent protein kinase Cζ-WNT signalling axis induces expression of an E3 ubiquitin ligase, constitutive photomorphogenesis protein 1 (COP1). A secondary infection of C. albicans in such Mtb-infected macrophages causes COP1 to mediate the proteasomal degradation of interferon regulatory factor 9 (IRF9), a cardinal factor that we identified to arbitrate an inflammatory programmed cell death, pyroptosis. In vivo experiments mimicking a pre-existing Mtb infection demonstrate that inhibition of pyroptosis in mice results in increased C. albicans burden and aberrant lung tissue architecture, leading to increased host mortality. Together, our study reveals the crucial role of pyroptosis regulation for manifesting a successful C. albicans-Mtb coinfection.
Collapse
Affiliation(s)
- Bharat Bhatt
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Praveen Prakhar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Gaurav Kumar Lohia
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Raju S Rajmani
- Centre of Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | | |
Collapse
|
33
|
Meng F, Xie B, Martin JF. Targeting the Hippo pathway in heart repair. Cardiovasc Res 2021; 118:2402-2414. [PMID: 34528077 DOI: 10.1093/cvr/cvab291] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo pathway is an evolutionarily and functionally conserved signaling pathway that controls organ size by regulating cell proliferation, apoptosis, and differentiation. Emerging evidence has shown that the Hippo pathway plays critical roles in cardiac development, homeostasis, disease, and regeneration. Targeting the Hippo pathway has tremendous potential as a therapeutic strategy for treating intractable cardiovascular diseases such as heart failure. In this review, we summarize the function of the Hippo pathway in the heart. Particularly, we highlight the posttranslational modification of Hippo pathway components, including the core kinases LATS1/2 and their downstream effectors YAP/TAZ, in different contexts, which has provided new insights and avenues in cardiac research.
Collapse
Affiliation(s)
- Fansen Meng
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Bing Xie
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030.,Texas Heart Institute, Houston, Texas, 77030
| |
Collapse
|
34
|
Matos-Nieves A, Manivannan S, Majumdar U, McBride KL, White P, Garg V. A Multi-Omics Approach Using a Mouse Model of Cardiac Malformations for Prioritization of Human Congenital Heart Disease Contributing Genes. Front Cardiovasc Med 2021; 8:683074. [PMID: 34504875 PMCID: PMC8421733 DOI: 10.3389/fcvm.2021.683074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023] Open
Abstract
Congenital heart disease (CHD) is the most common type of birth defect, affecting ~1% of all live births. Malformations of the cardiac outflow tract (OFT) account for ~30% of all CHD and include a range of CHDs from bicuspid aortic valve (BAV) to tetralogy of Fallot (TOF). We hypothesized that transcriptomic profiling of a mouse model of CHD would highlight disease-contributing genes implicated in congenital cardiac malformations in humans. To test this hypothesis, we utilized global transcriptional profiling differences from a mouse model of OFT malformations to prioritize damaging, de novo variants identified from exome sequencing datasets from published cohorts of CHD patients. Notch1 +/- ; Nos3 -/- mice display a spectrum of cardiac OFT malformations ranging from BAV, semilunar valve (SLV) stenosis to TOF. Global transcriptional profiling of the E13.5 Notch1 +/- ; Nos3 -/- mutant mouse OFTs and wildtype controls was performed by RNA sequencing (RNA-Seq). Analysis of the RNA-Seq dataset demonstrated genes belonging to the Hif1α, Tgf-β, Hippo, and Wnt signaling pathways were differentially expressed in the mutant OFT. Mouse to human comparative analysis was then performed to determine if patients with TOF and SLV stenosis display an increased burden of damaging, genetic variants in gene homologs that were dysregulated in Notch1 +/- ; Nos3 -/- OFT. We found an enrichment of de novo variants in the TOF population among the 1,352 significantly differentially expressed genes in Notch1 +/- ; Nos3 -/- mouse OFT but not the SLV population. This association was not significant when comparing only highly expressed genes in the murine OFT to de novo variants in the TOF population. These results suggest that transcriptomic datasets generated from the appropriate temporal, anatomic and cellular tissues from murine models of CHD may provide a novel approach for the prioritization of disease-contributing genes in patients with CHD.
Collapse
Affiliation(s)
- Adrianna Matos-Nieves
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Uddalak Majumdar
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Kim L. McBride
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
| | - Peter White
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Vidu Garg
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, Ohio State University, Columbus, OH, United States
| |
Collapse
|
35
|
Strepkos D, Markouli M, Papavassiliou KA, Papavassiliou AG, Piperi C. Emerging roles for the YAP/TAZ transcriptional regulators in brain tumour pathology and targeting options. Neuropathol Appl Neurobiol 2021; 48:e12762. [PMID: 34409639 DOI: 10.1111/nan.12762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
The transcriptional co-activators Yes-associated protein 1/transcriptional co-activator with PDZ-binding motif (YAP/TAZ) have emerged as significant regulators of a wide variety of cellular and organ functions with impact in early embryonic development, especially during the expansion of the neural progenitor cell pool. YAP/TAZ signalling regulates organ size development, tissue homeostasis, wound healing and angiogenesis by participating in a complex network of various pathways. However, recent evidence suggests an association of these physiologic regulatory effects of YAP/TAZ with pro-oncogenic activities. Herein, we discuss the physiological functions of YAP/TAZ as well as the extensive network of signalling pathways that control their expression and activity, leading to brain tumour development and progression. Furthermore, we describe current targeting approaches and drug options including direct YAP/TAZ and YAP-TEA domain transcription factor (TEAD) interaction inhibitors, G-protein coupled receptors (GPCR) signalling modulators and kinase inhibitors, which may be used to successfully attack YAP/TAZ-dependent tumours.
Collapse
Affiliation(s)
- Dimitrios Strepkos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Mariam Markouli
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas A Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
36
|
Xie J, Wang Y, Ai D, Yao L, Jiang H. The role of the Hippo pathway in heart disease. FEBS J 2021; 289:5819-5833. [PMID: 34174031 DOI: 10.1111/febs.16092] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/18/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022]
Abstract
Heart disease, including coronary artery disease, myocardial infarction, heart failure, cardiac hypertrophy, and cardiomyopathies, is the leading causes of death worldwide. The Hippo pathway is a central controller for organ size and tissue growth, which plays a pivotal role in determining cardiomyocytes and nonmyocytes proliferation, regeneration, differentiation, and apoptosis. In this review, we summarize the effects of the Hippo pathway on heart disease and propose potential intervention targets. Especially, we discuss the molecular mechanisms of the Hippo pathway involved in maintaining cardiac homeostasis by regulating cardiomyocytes and nonmyocytes function in the heart. Based on this, we conclude that the Hippo pathway is a promising therapeutic target for cardiovascular therapy, which will bring new perspectives for their treatments.
Collapse
Affiliation(s)
- Jiahong Xie
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yuxin Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ding Ai
- Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, China
| | - Liu Yao
- Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, China
| | - Hongfeng Jiang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Peralta M, Ortiz Lopez L, Jerabkova K, Lucchesi T, Vitre B, Han D, Guillemot L, Dingare C, Sumara I, Mercader N, Lecaudey V, Delaval B, Meilhac SM, Vermot J. Intraflagellar Transport Complex B Proteins Regulate the Hippo Effector Yap1 during Cardiogenesis. Cell Rep 2021; 32:107932. [PMID: 32698004 DOI: 10.1016/j.celrep.2020.107932] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/30/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023] Open
Abstract
Cilia and the intraflagellar transport (IFT) proteins involved in ciliogenesis are associated with congenital heart diseases (CHDs). However, the molecular links between cilia, IFT proteins, and cardiogenesis are yet to be established. Using a combination of biochemistry, genetics, and live-imaging methods, we show that IFT complex B proteins (Ift88, Ift54, and Ift20) modulate the Hippo pathway effector YAP1 in zebrafish and mouse. We demonstrate that this interaction is key to restrict the formation of the proepicardium and the myocardium. In cellulo experiments suggest that IFT88 and IFT20 interact with YAP1 in the cytoplasm and functionally modulate its activity, identifying a molecular link between cilia-related proteins and the Hippo pathway. Taken together, our results highlight a noncanonical role for IFT complex B proteins during cardiogenesis and shed light on a mechanism of action for ciliary proteins in YAP1 regulation.
Collapse
Affiliation(s)
- Marina Peralta
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Laia Ortiz Lopez
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Katerina Jerabkova
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Tommaso Lucchesi
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France; Sorbonne Université, Collège Doctoral, F-75005, Paris, France
| | - Benjamin Vitre
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), CNRS, Université de Montpellier, Montpellier, France
| | - Dong Han
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France
| | - Laurent Guillemot
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France
| | - Chaitanya Dingare
- Institute for Cell Biology and Neurosciences, Goethe University of Frankfurt, Frankfurt, Germany
| | - Izabela Sumara
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Virginie Lecaudey
- Institute for Cell Biology and Neurosciences, Goethe University of Frankfurt, Frankfurt, Germany
| | - Benedicte Delaval
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), CNRS, Université de Montpellier, Montpellier, France
| | - Sigolène M Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France; Sorbonne Université, Collège Doctoral, F-75005, Paris, France; Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
38
|
New insights into the Hippo/YAP pathway in idiopathic pulmonary fibrosis. Pharmacol Res 2021; 169:105635. [PMID: 33930530 DOI: 10.1016/j.phrs.2021.105635] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterised by an inexorable decline in lung function. The development of IPF involves multiple positive feedback loops; and a strong support role of the Hippo/YAP signalling pathway, which is essential for regulating cell proliferation and organ size, in IPF pathogenesis has been unveiled recently in cell and animal models. YAP/TAZ contributes to both pulmonary fibrosis and alveolar regeneration via the conventional Hippo/YAP signalling pathway, G protein-coupled receptor signalling, and mechanotransduction. Selectively inhibiting YAP/TAZ in lung fibroblasts may inhibit fibroblast proliferation and extracellular matrix deposition, while activating YAP/TAZ in alveolar epithelial cells may promote alveolar regeneration. In this review, we explore, for the first time, the bidirectional and cell-specific regulation of the Hippo/YAP pathway in IPF pathogenesis and discuss recent research progress and future prospects of IPF treatment based on Hippo/YAP signalling, thus providing a basis for the development of new therapeutic strategies to alleviate or even reverse IPF.
Collapse
|
39
|
YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming. Metabolites 2021; 11:metabo11030154. [PMID: 33800464 PMCID: PMC7999074 DOI: 10.3390/metabo11030154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
Cell reprogramming can either refer to a direct conversion of a specialized cell into another or to a reversal of a somatic cell into an induced pluripotent stem cell (iPSC). It implies a peculiar modification of the epigenetic asset and gene regulatory networks needed for a new cell, to better fit the new phenotype of the incoming cell type. Cellular reprogramming also implies a metabolic rearrangement, similar to that observed upon tumorigenesis, with a transition from oxidative phosphorylation to aerobic glycolysis. The induction of a reprogramming process requires a nexus of signaling pathways, mixing a range of local and systemic information, and accumulating evidence points to the crucial role exerted by the Hippo pathway components Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ). In this review, we will first provide a synopsis of the Hippo pathway and its function during reprogramming and tissue regeneration, then we introduce the latest knowledge on the interplay between YAP/TAZ and metabolism and, finally, we discuss the possible role of YAP/TAZ in the orchestration of the metabolic switch upon cellular reprogramming.
Collapse
|
40
|
Wang P, Wang M, Hu Y, Chen J, Cao Y, Liu C, Wu Z, Shen J, Lu J, Liu P. Isorhapontigenin protects against doxorubicin-induced cardiotoxicity via increasing YAP1 expression. Acta Pharm Sin B 2021; 11:680-693. [PMID: 33777675 PMCID: PMC7982427 DOI: 10.1016/j.apsb.2020.10.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
As an effective anticancer drug, the clinical limitation of doxorubicin (Dox) is the time- and dose-dependent cardiotoxicity. Yes-associated protein 1 (YAP1) interacts with transcription factor TEA domain 1 (TEAD1) and plays an important role in cell proliferation and survival. However, the role of YAP1 in Dox-induced cardiomyopathy has not been reported. In this study, the expression of YAP1 was reduced in clinical human failing hearts with dilated cardiomyopathy and Dox-induced in vivo and in vitro cardiotoxic model. Ectopic expression of Yap1 significantly blocked Dox-induced cardiomyocytes apoptosis in TEAD1 dependent manner. Isorhapontigenin (Isor) is a new derivative of stilbene and responsible for a wide range of biological processes. Here, we found that Isor effectively relieved Dox-induced cardiomyocytes apoptosis in a dose-dependent manner in vitro. Administration with Isor (30 mg/kg/day, intraperitoneally, 3 weeks) significantly protected against Dox-induced cardiotoxicity in mice. Interestingly, Isor increased Dox-caused repression in YAP1 and the expression of its target genes in vivo and in vitro. Knockout or inhibition of Yap1 blocked the protective effects of Isor on Dox-induced cardiotoxicity. In conclusion, YAP1 may be a novel target for Dox-induced cardiotoxicity and Isor might be a new compound to fight against Dox-induced cardiotoxicity by increasing YAP1 expression.
Collapse
Key Words
- AMPK, AMP-activated protein kinase
- AP-1, anti-microbial protein
- AREG, amphiregulin
- AUC/Dose, dose-normalized plasma exposures
- Amphiregulin
- Ang II, angiotensin II
- CO, cardiac output
- CTGF, connective tissue growth factor
- Cardiomyocytes apoptosis
- Cardiotoxicity
- Cmax/Dose, dose-normalized maximal plasma concentrations
- Connective tissue growth factor
- DAB, 3,3′-diaminobenzidine
- DMEM, Dulbecco's modified Eagle's medium
- Dob, dobutamine
- Dox, doxorubicin
- Doxorubicin
- EMT, epithelial mesenchymal transformation
- FOXO1, forkhead box class O1
- FS, fractional shortening
- HE, hematoxylin–eosin
- ISO, isoproterenol
- Isor, isorhapontigenin
- Isorhapontigenin
- LVAW;d, left ventricular end-diastolic anterior wall thickness
- LVAW;s, left ventricular end-systolic anterior wall thickness
- LVEF, left ventricular ejection fraction
- LVID;d, left ventricular end-diastolic internal diameter
- LVID;s, left ventricular end-systolic internal diameter
- LVPW;d, left ventricular end-diastolic posterior wall thickness
- LVPW;s, left ventricular end-systolic posterior wall thickness
- MAPK, mitogen-activated protein kinase
- MI, myocardial infarction
- NF-κB, nuclear factor kappa-B
- NRCMs, neonatal rat cardiomyocytes
- P2Y12 receptor, ADP receptor
- PGC-1α, peroxisome proliferator-activated receptor γ coactivator-1α
- PMSF, phenylmethanesulfonyl fluoride
- PVDF, polyvinylidene fluoride
- ROS, reactive oxygen species
- SD, Sprague–Dawley
- SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- SESN2, sestrin2
- TCF4, T-cell factor 4
- TEAD, TEA domain transcription factor proteins
- TEAD1
- TUNEL, TdT-mediated dUTP nick end labeling
- WGA, wheat germ agglutinin
- YAP1
- YAP1, Yes-associated protein 1
- qRT-PCR, quantitative real-time polymerase chain reaction
- sgRNAs, sequence guiding RNAs
- Δψm, mitochondrial membrane potential
Collapse
Affiliation(s)
- Panxia Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Minghui Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuehuai Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jianxing Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yanjun Cao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Cui Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Corresponding authors.
| | - Jing Lu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Corresponding authors.
| | - Peiqing Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Engineering Laboratory of Druggability and New Drugs Evaluation, Guangzhou 510006, China
- Corresponding authors.
| |
Collapse
|
41
|
Betterman KL, Sutton DL, Secker GA, Kazenwadel J, Oszmiana A, Lim L, Miura N, Sorokin L, Hogan BM, Kahn ML, McNeill H, Harvey NL. Atypical cadherin FAT4 orchestrates lymphatic endothelial cell polarity in response to flow. J Clin Invest 2021; 130:3315-3328. [PMID: 32182215 DOI: 10.1172/jci99027] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/05/2020] [Indexed: 01/07/2023] Open
Abstract
The atypical cadherin FAT4 has established roles in the regulation of planar cell polarity and Hippo pathway signaling that are cell context dependent. The recent identification of FAT4 mutations in Hennekam syndrome, features of which include lymphedema, lymphangiectasia, and mental retardation, uncovered an important role for FAT4 in the lymphatic vasculature. Hennekam syndrome is also caused by mutations in collagen and calcium binding EGF domains 1 (CCBE1) and ADAM metallopeptidase with thrombospondin type 1 motif 3 (ADAMTS3), encoding a matrix protein and protease, respectively, that regulate activity of the key prolymphangiogenic VEGF-C/VEGFR3 signaling axis by facilitating the proteolytic cleavage and activation of VEGF-C. The fact that FAT4, CCBE1, and ADAMTS3 mutations underlie Hennekam syndrome suggested that all 3 genes might function in a common pathway. We identified FAT4 as a target gene of GATA-binding protein 2 (GATA2), a key transcriptional regulator of lymphatic vascular development and, in particular, lymphatic vessel valve development. Here, we demonstrate that FAT4 functions in a lymphatic endothelial cell-autonomous manner to control cell polarity in response to flow and is required for lymphatic vessel morphogenesis throughout development. Our data reveal a crucial role for FAT4 in lymphangiogenesis and shed light on the mechanistic basis by which FAT4 mutations underlie a human lymphedema syndrome.
Collapse
Affiliation(s)
- Kelly L Betterman
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Drew L Sutton
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Genevieve A Secker
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Jan Kazenwadel
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Anna Oszmiana
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| | - Lillian Lim
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, University of Queensland, Saint Lucia, Queensland, Australia.,Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Helen McNeill
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,SA Pathology, Adelaide, South Australia, Australia
| |
Collapse
|
42
|
Szulzewsky F, Holland EC, Vasioukhin V. YAP1 and its fusion proteins in cancer initiation, progression and therapeutic resistance. Dev Biol 2021; 475:205-221. [PMID: 33428889 DOI: 10.1016/j.ydbio.2020.12.018] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/14/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
YAP1 is a transcriptional co-activator whose activity is controlled by the Hippo signaling pathway. In addition to important functions in normal tissue homeostasis and regeneration, YAP1 has also prominent functions in cancer initiation, aggressiveness, metastasis, and therapy resistance. In this review we are discussing the molecular functions of YAP1 and its roles in cancer, with a focus on the different mechanisms of de-regulation of YAP1 activity in human cancers, including inactivation of upstream Hippo pathway tumor suppressors, regulation by intersecting pathways, miRNAs, and viral oncogenes. We are also discussing new findings on the function and biology of the recently identified family of YAP1 gene fusions, that constitute a new type of activating mutation of YAP1 and that are the likely oncogenic drivers in several subtypes of human cancers. Lastly, we also discuss different strategies of therapeutic inhibition of YAP1 functions.
Collapse
Affiliation(s)
- Frank Szulzewsky
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Eric C Holland
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA; Seattle Tumor Translational Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Valeri Vasioukhin
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| |
Collapse
|
43
|
Lodge EJ, Xekouki P, Silva TS, Kochi C, Longui CA, Faucz FR, Santambrogio A, Mills JL, Pankratz N, Lane J, Sosnowska D, Hodgson T, Patist AL, Francis-West P, Helmbacher F, Stratakis CA, Andoniadou CL. Requirement of FAT and DCHS protocadherins during hypothalamic-pituitary development. JCI Insight 2020; 5. [PMID: 33108146 PMCID: PMC7714405 DOI: 10.1172/jci.insight.134310] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Pituitary developmental defects lead to partial or complete hormone deficiency and significant health problems. The majority of cases are sporadic and of unknown cause. We screened 28 patients with pituitary stalk interruption syndrome (PSIS) for mutations in the FAT/DCHS family of protocadherins that have high functional redundancy. We identified seven variants, four of which putatively damaging, in FAT2 and DCHS2 in six patients with pituitary developmental defects recruited through a cohort of patients with mostly ectopic posterior pituitary gland and/or pituitary stalk interruption. All patients had growth hormone deficiency and two presented with multiple hormone deficiencies and small glands. FAT2 and DCHS2 were strongly expressed in the mesenchyme surrounding the normal developing human pituitary. We analyzed Dchs2-/- mouse mutants and identified anterior pituitary hypoplasia and partially penetrant infundibular defects. Overlapping infundibular abnormalities and distinct anterior pituitary morphogenesis defects were observed in Fat4-/- and Dchs1-/- mouse mutants but all animal models displayed normal commitment to the anterior pituitary cell type. Together our data implicate FAT/DCHS protocadherins in normal hypothalamic-pituitary development and identify FAT2 and DCHS2 as candidates underlying pituitary gland developmental defects such as ectopic pituitary gland and/or pituitary stalk interruption.
Collapse
Affiliation(s)
- Emily J. Lodge
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Paraskevi Xekouki
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Tatiane S. Silva
- Pediatric Endocrinology Unit, Irmandade da Santa Casa de Misericórdia de São Paulo, São Paulo, Brazil
| | - Cristiane Kochi
- Pediatric Endocrinology Unit, Irmandade da Santa Casa de Misericórdia de São Paulo, São Paulo, Brazil
| | - Carlos A. Longui
- Pediatric Endocrinology Unit, Irmandade da Santa Casa de Misericórdia de São Paulo, São Paulo, Brazil
| | - Fabio R. Faucz
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Alice Santambrogio
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - James L. Mills
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - John Lane
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Dominika Sosnowska
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Tina Hodgson
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Amanda L. Patist
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Philippa Francis-West
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | | | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Cynthia L. Andoniadou
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
44
|
Rao-Bhatia A, Zhu M, Yin WC, Coquenlorge S, Zhang X, Woo J, Sun Y, Dean CH, Liu A, Hui CC, Shivdasani RA, McNeill H, Hopyan S, Kim TH. Hedgehog-Activated Fat4 and PCP Pathways Mediate Mesenchymal Cell Clustering and Villus Formation in Gut Development. Dev Cell 2020; 52:647-658.e6. [PMID: 32155439 DOI: 10.1016/j.devcel.2020.02.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 11/06/2019] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
During development, intestinal epithelia undergo dramatic morphogenesis mediated by mesenchymal signaling to form villi, which are required for efficient nutrient absorption and host defense. Although both smooth-muscle-induced physical forces and mesenchymal cell clustering beneath emerging villi are implicated in epithelial folding, the underlying cellular mechanisms are unclear. Hedgehog (Hh) signaling can mediate both processes. We therefore analyzed its direct targetome and revealed GLI2 transcriptional activation of atypical cadherin and planar cell polarity (PCP) genes. By examining Fat4 and Dchs1 knockout mice, we demonstrate their critical roles in villus formation. Analyses of PCP-mutant mice and genetic interaction studies show that the Fat4-Dchs1 axis acts in parallel to the core-Vangl2 PCP axis to control mesenchymal cell clustering. Moreover, live light-sheet fluorescence microscopy and cultured PDGFRα+ cells reveal a requirement for PCP in their oriented cell migration guided by WNT5A. Therefore, mesenchymal PCP induced by Hh signaling drives cell clustering and subsequent epithelial remodeling.
Collapse
Affiliation(s)
- Abilasha Rao-Bhatia
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Min Zhu
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Wen-Chi Yin
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sabrina Coquenlorge
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiaoyun Zhang
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Janghee Woo
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Charlotte H Dean
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Aimin Liu
- Department of Biology, Eberly College of Science, Centers for Cellular Dynamics and Molecular Investigation of Neurological Disorders, Huck Institute of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Helen McNeill
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sevan Hopyan
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
45
|
ERBB2 drives YAP activation and EMT-like processes during cardiac regeneration. Nat Cell Biol 2020; 22:1346-1356. [PMID: 33046882 DOI: 10.1038/s41556-020-00588-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 09/01/2020] [Indexed: 01/06/2023]
Abstract
Cardiomyocyte loss after injury results in adverse remodelling and fibrosis, inevitably leading to heart failure. The ERBB2-Neuregulin and Hippo-YAP signalling pathways are key mediators of heart regeneration, yet the crosstalk between them is unclear. We demonstrate that transient overexpression of activated ERBB2 in cardiomyocytes (OE CMs) promotes cardiac regeneration in a heart failure model. OE CMs present an epithelial-mesenchymal transition (EMT)-like regenerative response manifested by cytoskeletal remodelling, junction dissolution, migration and extracellular matrix turnover. We identified YAP as a critical mediator of ERBB2 signalling. In OE CMs, YAP interacts with nuclear-envelope and cytoskeletal components, reflecting an altered mechanical state elicited by ERBB2. We identified two YAP-activating phosphorylations on S352 and S274 in OE CMs, which peak during metaphase, that are ERK dependent and Hippo independent. Viral overexpression of YAP phospho-mutants dampened the proliferative competence of OE CMs. Together, we reveal a potent ERBB2-mediated YAP mechanotransduction signalling, involving EMT-like characteristics, resulting in robust heart regeneration.
Collapse
|
46
|
Flinn MA, Otten C, Brandt ZJ, Bostrom JR, Kenarsary A, Wan TC, Auchampach JA, Abdelilah-Seyfried S, O'Meara CC, Link BA. Llgl1 regulates zebrafish cardiac development by mediating Yap stability in cardiomyocytes. Development 2020; 147:147/16/dev193581. [PMID: 32843528 DOI: 10.1242/dev.193581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/10/2020] [Indexed: 01/19/2023]
Abstract
The Hippo-Yap pathway regulates multiple cellular processes in response to mechanical and other stimuli. In Drosophila, the polarity protein Lethal (2) giant larvae [L(2)gl], negatively regulates Hippo-mediated transcriptional output. However, in vertebrates, little is known about its homolog Llgl1. Here, we define a novel role for vertebrate Llgl1 in regulating Yap stability in cardiomyocytes, which impacts heart development. In contrast to the role of Drosophila L(2)gl, Llgl1 depletion in cultured rat cardiomyocytes decreased Yap protein levels and blunted target gene transcription without affecting Yap transcript abundance. Llgl1 depletion in zebrafish resulted in larger and dysmorphic cardiomyocytes, pericardial effusion, impaired blood flow and aberrant valvulogenesis. Cardiomyocyte Yap protein levels were decreased in llgl1 morphants, whereas Notch, which is regulated by hemodynamic forces and participates in valvulogenesis, was more broadly activated. Consistent with the role of Llgl1 in regulating Yap stability, cardiomyocyte-specific overexpression of Yap in Llgl1-depleted embryos ameliorated pericardial effusion and restored blood flow velocity. Altogether, our data reveal that vertebrate Llgl1 is crucial for Yap stability in cardiomyocytes and its absence impairs cardiac development.
Collapse
Affiliation(s)
- Michael A Flinn
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cécile Otten
- Institute for Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
| | - Zachary J Brandt
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jonathan R Bostrom
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aria Kenarsary
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tina C Wan
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Pharmacology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John A Auchampach
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Pharmacology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Salim Abdelilah-Seyfried
- Institute for Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany.,Institute for Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Caitlin C O'Meara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian A Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA .,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
47
|
Heng BC, Zhang X, Aubel D, Bai Y, Li X, Wei Y, Fussenegger M, Deng X. Role of YAP/TAZ in Cell Lineage Fate Determination and Related Signaling Pathways. Front Cell Dev Biol 2020; 8:735. [PMID: 32850847 PMCID: PMC7406690 DOI: 10.3389/fcell.2020.00735] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
The penultimate effectors of the Hippo signaling pathways YAP and TAZ, are transcriptional co-activator proteins that play key roles in many diverse biological processes, ranging from cell proliferation, tumorigenesis, mechanosensing and cell lineage fate determination, to wound healing and regeneration. In this review, we discuss the regulatory mechanisms by which YAP/TAZ control stem/progenitor cell differentiation into the various major lineages that are of interest to tissue engineering and regenerative medicine applications. Of particular interest is the key role of YAP/TAZ in maintaining the delicate balance between quiescence, self-renewal, proliferation and differentiation of endogenous adult stem cells within various tissues/organs during early development, normal homeostasis and regeneration/healing. Finally, we will consider how increasing knowledge of YAP/TAZ signaling might influence the trajectory of future progress in regenerative medicine.
Collapse
Affiliation(s)
- Boon C. Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Faculty of Science and Technology, Sunway University, Subang Jaya, Malaysia
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
| | - Dominique Aubel
- IUTA Department Genie Biologique, Universite Claude Bernard Lyon 1, Villeurbanne, France
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH-Zürich, Basel, Switzerland
| | - Xuliang Deng
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
48
|
Zheng M, Jacob J, Hung SH, Wang J. The Hippo Pathway in Cardiac Regeneration and Homeostasis: New Perspectives for Cell-Free Therapy in the Injured Heart. Biomolecules 2020; 10:biom10071024. [PMID: 32664346 PMCID: PMC7407108 DOI: 10.3390/biom10071024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Intractable cardiovascular diseases are leading causes of mortality around the world. Adult mammalian hearts have poor regenerative capacity and are not capable of self-repair after injury. Recent studies of cell-free therapeutics such as those designed to stimulate endogenous cardiac regeneration have uncovered new feasible therapeutic avenues for cardiac repair. The Hippo pathway, a fundamental pathway with pivotal roles in cell proliferation, survival and differentiation, has tremendous potential for therapeutic manipulation in cardiac regeneration. In this review, we summarize the most recent studies that have revealed the function of the Hippo pathway in heart regeneration and homeostasis. In particular, we discuss the molecular mechanisms of how the Hippo pathway maintains cardiac homeostasis by directing cardiomyocyte chromatin remodeling and regulating the cell-cell communication between cardiomyocytes and non-cardiomyocytes in the heart.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA;
| | - Joan Jacob
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston, TX 77030, USA; (J.J.); (S.-H.H.)
| | - Shao-Hsi Hung
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston, TX 77030, USA; (J.J.); (S.-H.H.)
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA;
- Correspondence: ; Tel.: +1-7135-005-723
| |
Collapse
|
49
|
Helmbacher F, Stricker S. Tissue cross talks governing limb muscle development and regeneration. Semin Cell Dev Biol 2020; 104:14-30. [PMID: 32517852 DOI: 10.1016/j.semcdb.2020.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022]
Abstract
For decades, limb development has been a paradigm of three-dimensional patterning. Moreover, as the limb muscles and the other tissues of the limb's musculoskeletal system arise from distinct developmental sources, it has been a prime example of integrative morphogenesis and cross-tissue communication. As the limbs grow, all components of the musculoskeletal system (muscles, tendons, connective tissue, nerves) coordinate their growth and differentiation, ultimately giving rise to a functional unit capable of executing elaborate movement. While the molecular mechanisms governing global three-dimensional patterning and formation of the skeletal structures of the limbs has been a matter of intense research, patterning of the soft tissues is less understood. Here, we review the development of limb muscles with an emphasis on their interaction with other tissue types and the instructive roles these tissues play. Furthermore, we discuss the role of adult correlates of these embryonic accessory tissues in muscle regeneration.
Collapse
Affiliation(s)
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany.
| |
Collapse
|
50
|
Yang JX, Xie P, Li YS, Wen T, Yang XC. Osteoclast-derived miR-23a-5p-containing exosomes inhibit osteogenic differentiation by regulating Runx2. Cell Signal 2020; 70:109504. [PMID: 31857240 DOI: 10.1016/j.cellsig.2019.109504] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/10/2019] [Accepted: 12/15/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Some microRNAs (miRNAs) are involved in osteogenic differentiation. In recent years, increasing evidences have revealed that exosomes contain specific miRNAs. However, the effect and mechanism of miR-23a-5p-containing exosomes in osteoblast remain largely unclear. METHODS We extracted exosomes from RANKL-induced RAW 264.7 cells, and identified exosomes via transmission electron microscopy, western blot and flow cytometry analysis. In addition, exosome secretion was inhibited by GW4869 and Rab27a siRNAs. miR-23a-5p expression was analyzed by qRT-PCR, and the related protein levels were examined by western blot assay. Furthermore, the number and distribution of osteoclasts were detected by TRAP staining, and early osteogenesis was evaluated by ALP staining. Combination of YAP1 and Runx2 was verified by Co-IP assay, and the regulation of miR-23a-5p and Runx2 was measured by dual luciferase reporter assay. RESULTS We successfully extracted exosomes from RANKL-induced RAW 264.7 cells, and successfully verified exosomes morphology. We also indicated that miR-23a-5p was highly expressed in exosomes from RANKL-induced RAW 264.7 cells, and osteoclast-derived miR-23a-5p-containing exosomes inhibited osteoblast activity, while its inhibition weakened osteoclasts. In mechanism, we demonstrated that Runx2 was a target gene of miR-23a-5p, YAP interacted with Runx2, and YAP or Runx2 inhibited MT1DP expression. In addition, we proved that knockdown of MT1DP facilitated osteogenic differentiation by regulating FoxA1 and Runx2. CONCLUSIONS We demonstrated that osteoclast-derived miR-23a-5p-containing exosomes could efficiently suppress osteogenic differentiation by inhibiting Runx2 and promoting YAP1-mediated MT1DP. Therefore, we suggested miR-23a-5p in exosomes might provide a novel mechanism for osteoblast function.
Collapse
Affiliation(s)
- Jun-Xiao Yang
- Department of orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, PR China
| | - Peng Xie
- Department of orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, PR China
| | - Yu-Sheng Li
- Department of orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, PR China
| | - Ting Wen
- Department of orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, PR China
| | - Xu-Cheng Yang
- Department of orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, PR China.
| |
Collapse
|