1
|
Taroc EZM, Amato E, Semon A, Dolphin N, Beck B, Belin S, Poitelon Y, Forni PE. Shared Lineage, Distinct Outcomes: Yap and Taz Loss Differentially Impact Schwann and Olfactory Ensheathing Cell Development Without Disrupting GnRH-1 Migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638196. [PMID: 40027653 PMCID: PMC11870449 DOI: 10.1101/2025.02.13.638196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Olfactory Ensheathing Cells (OECs) are glial cells originating from the neural crest, critical for bundling olfactory axons to the brain. Their development is crucial for the migration of Gonadotropin-Releasing Hormone-1 (GnRH-1) neurons, which are essential for puberty and fertility. OECs have garnered interest as potential therapeutic targets for central nervous system lesions, although their development is not fully understood. Our single-cell RNA sequencing of mouse embryonic nasal tissues suggests that OECs and Schwann cells share a common origin from Schwann cell precursors yet exhibit significant genetic differences. The transcription factors Yap and Taz have previously been shown to play a crucial role in Schwann cell development. We used Sox10 -Cre mice to conditionally ablate Yap and Taz in migrating the neural crest and its derivatives. Our analyses showed reduced Sox10+ glial cells along nerves in the nasal region, altered gene expression of SCs, melanocytes, and OECs, and a significant reduction in olfactory sensory neurons and vascularization in the vomeronasal organ. However, despite these changes, GnRH-1 neuronal migration remained unaffected. Our findings highlight the importance of the Hippo pathway in OEC development and how changes in cranial neural crest derivatives indirectly impact the development of olfactory epithelia.
Collapse
|
2
|
Ajongbolo AO, Langhans SA. YAP/TAZ-associated cell signaling - at the crossroads of cancer and neurodevelopmental disorders. Front Cell Dev Biol 2025; 13:1522705. [PMID: 39936032 PMCID: PMC11810912 DOI: 10.3389/fcell.2025.1522705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
YAP/TAZ (Yes-associated protein/paralog transcriptional co-activator with PDZ-binding domain) are transcriptional cofactors that are the key and major downstream effectors of the Hippo signaling pathway. Both are known to play a crucial role in defining cellular outcomes, including cell differentiation, cell proliferation, and apoptosis. Aside from the canonical Hippo signaling cascade with the key components MST1/2 (mammalian STE20-like kinase 1/2), SAV1 (Salvador homologue 1), MOB1A/B (Mps one binder kinase activator 1A/B) and LATS1/2 (large tumor suppressor kinase 1/2) upstream of YAP/TAZ, YAP/TAZ activation is also influenced by numerous other signaling pathways. Such non-canonical regulation of YAP/TAZ includes well-known growth factor signaling pathways such as the epidermal growth factor receptor (EGFR)/ErbB family, Notch, and Wnt signaling as well as cell-cell adhesion, cell-matrix interactions and mechanical cues from a cell's microenvironment. This puts YAP/TAZ at the center of a complex signaling network capable of regulating developmental processes and tissue regeneration. On the other hand, dysregulation of YAP/TAZ signaling has been implicated in numerous diseases including various cancers and neurodevelopmental disorders. Indeed, in recent years, parallels between cancer development and neurodevelopmental disorders have become apparent with YAP/TAZ signaling being one of these pathways. This review discusses the role of YAP/TAZ in brain development, cancer and neurodevelopmental disorders with a special focus on the interconnection in the role of YAP/TAZ in these different conditions.
Collapse
Affiliation(s)
- Aderonke O. Ajongbolo
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
- Biological Sciences Graduate Program, University of Delaware, Newark, DE, United States
| | - Sigrid A. Langhans
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
| |
Collapse
|
3
|
Kalanithy JC, Mingardo E, Stegmann JD, Dhakar R, Dakal TC, Rosenfeld JA, Tan WH, Coury SA, Woerner AC, Sebastian J, Levy PA, Fleming LR, Waffenschmidt L, Lindenberg TT, Yilmaz Ö, Channab K, Babra BK, Christ A, Eiberger B, Hölzel S, Vidic C, Häberlein F, Ishorst N, Rodriguez-Gatica JE, Pezeshkpoor B, Kupczyk PA, Vanakker OM, Loddo S, Novelli A, Dentici ML, Becker A, Thiele H, Posey JE, Lupski JR, Hilger AC, Reutter HM, Merz WM, Dworschak GC, Odermatt B. TFAP2E is implicated in central nervous system, orofacial and maxillofacial anomalies. J Med Genet 2025; 62:126-137. [PMID: 39715634 PMCID: PMC11777392 DOI: 10.1136/jmg-2023-109799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/02/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Previous studies in mouse, Xenopus and zebrafish embryos show strong tfap2e expression in progenitor cells of neuronal and neural crest tissues suggesting its involvement in neural crest specification. However, the role of human transcription factor activator protein 2 (TFAP2E) in human embryonic central nervous system (CNS), orofacial and maxillofacial development is unknown. METHODS Through a collaborative work, exome survey was performed in families with congenital CNS, orofacial and maxillofacial anomalies. Exome variant prioritisation prompted TFAP2E gene for functional analysis in zebrafish embryos. Embryonic morphology and development were assessed after antisense morpholino (MO) knockdown (KD), CRISPR/Cas9 knockout and overexpression of tfap2e in fluorescent zebrafish reporter lines using in vivo microscopy. Computational structural protein modelling of the identified human variants was performed. RESULTS In total, exome survey identified novel or ultra-rare heterozygous missense variants in TFAP2E in seven individuals from five independent families with predominantly CNS, orofacial and maxillofacial anomalies. One variant was found de novo and another variant segregated in an affected multiplex family. Protein modelling of the identified variants indicated potential distortion of TFAP2E in the transactivation or dimerisation domain. MO KD and CRISPR/Cas9 knockout of tfap2e in zebrafish revealed hydrocephalus and a significant reduction of brain volume, consistent with a microencephaly phenotype. Furthermore, mRNA overexpression of TFAP2E indicates dosage-sensitive phenotype expression. In addition, zebrafish showed orofacial and maxillofacial anomalies following tfap2e KD, recapitulating the human phenotype. CONCLUSION Our human genetic data and analysis of Tfap2e manipulation in zebrafish indicate a potential role of TFAP2E in human CNS, orofacial and maxillofacial anomalies.
Collapse
Affiliation(s)
- Jeshurun C Kalanithy
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Enrico Mingardo
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jil D Stegmann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Ramgopal Dhakar
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Jill A Rosenfeld
- Baylor Genetics Laboratories, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Wen-Hann Tan
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Stephanie A Coury
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Genomes2People Research Program, Division of Genetics, Department of Medicine, Mass General Brigham Inc, Boston, Massachusetts, USA
| | - Audrey C Woerner
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jessica Sebastian
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Paul A Levy
- Department of Pediatrics, Children's Hospital at Montefiore, New York, New York, USA
| | - Leah R Fleming
- Genetics and Metabolic Clinic, St Luke's Health System, Boise, Idaho, USA
| | - Lea Waffenschmidt
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Tobias T Lindenberg
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
| | - Öznur Yilmaz
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
| | - Khadija Channab
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Bimaljeet K Babra
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andrea Christ
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Britta Eiberger
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Selina Hölzel
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Clara Vidic
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Felix Häberlein
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Pharmaceutical Biology, Molecular, Cellular, and Pharmacobiology Section, University of Bonn, Bonn, Germany
| | - Nina Ishorst
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | | | - Behnaz Pezeshkpoor
- Institute for Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
- Center for Rare Diseases Bonn, University Hospital Bonn, Bonn, Germany
| | - Patrick A Kupczyk
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | | | - Sara Loddo
- Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria L Dentici
- Medical Genetics Unit, Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Albert Becker
- Institute for Cellular Neurosciences II, University Hospital Bonn, Bonn, Germany
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - James R Lupski
- Molecular and Human Genetics; Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - Alina C Hilger
- Research Center On Rare Kidney Diseases (RECORD), Erlangen University Hospital, Erlangen, Germany
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Heiko M Reutter
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Pediatric and Adolescent Medicine, Division Neonatology and Pediatric Intensive Care, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Institute of Human Genetics, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Waltraut M Merz
- Department of Obstetrics and Prenatal Medicine, University Hospital Bonn, Bonn, Germany
| | - Gabriel C Dworschak
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Neuropediatrics, University Hospital Bonn, Bonn, Germany
| | - Benjamin Odermatt
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
4
|
Huang Y, Flentke GR, Smith SM. Alcohol induces p53-mediated apoptosis in neural crest by stimulating an AMPK-mediated suppression of TORC1, S6K, and ribosomal biogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601754. [PMID: 39005448 PMCID: PMC11244973 DOI: 10.1101/2024.07.02.601754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Prenatal alcohol exposure is a leading cause of permanent neurodevelopmental disability and can feature distinctive craniofacial deficits that partly originate from the apoptotic deletion of craniofacial progenitors, a stem cell lineage called the neural crest (NC). We recently demonstrated that alcohol causes nucleolar stress in NC through its suppression of ribosome biogenesis (RBG) and this suppression is causative in their p53/MDM2-mediated apoptosis. Here, we show that this nucleolar stress originates from alcohol's activation of AMPK, which suppresses TORC1 and the p70/S6K-mediated stimulation of RBG. Alcohol-exposed cells of the pluripotent, primary cranial NC line O9-1 were evaluated with respect to their S6K, TORC1, and AMPK activity. The functional impact of these signals with respect to RBG, p53, and apoptosis were assessed using gain-of-function constructs and small molecule mediators. Alcohol rapidly (<2hr) increased pAMPK, pTSC2, and pRaptor, and reduced both total and pS6K in NC cells. These changes persisted for at least 12hr to 18hr following alcohol exposure. Attenuation of these signals via gain- or loss-of-function approaches that targeted AMPK, S6K, or TORC1 prevented alcohol's suppression of rRNA synthesis and the induction of p53-stimulated apoptosis. We conclude that alcohol induces ribosome dysbiogenesis and activates their p53/MDM2-mediated apoptosis via its activation of pAMPK, which in turn activates TSC2 and Raptor to suppress the TORC1/S6K-mediated promotion of ribosome biogenesis. This represents a novel mechanism underlying alcohol's neurotoxicity and is consistent with findings that TORC1/S6K networks are critical for cranial NC survival.
Collapse
|
5
|
Li Z, Lin J, Wu J, Suo J, Wang Z. The Hippo signalling pathway in bone homeostasis: Under the regulation of mechanics and aging. Cell Prolif 2024; 57:e13652. [PMID: 38700015 PMCID: PMC11471399 DOI: 10.1111/cpr.13652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The Hippo signalling pathway is a conserved kinase cascade that orchestrates diverse cellular processes, such as proliferation, apoptosis, lineage commitment and stemness. With the onset of society ages, research on skeletal aging-mechanics-bone homeostasis has exploded. In recent years, aging and mechanical force in the skeletal system have gained groundbreaking research progress. Under the regulation of mechanics and aging, the Hippo signalling pathway has a crucial role in the development and homeostasis of bone. We synthesize the current knowledge on the role of the Hippo signalling pathway, particularly its downstream effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), in bone homeostasis. We discuss the regulation of the lineage specification and function of different skeletal cell types by the Hippo signalling pathway. The interactions of the Hippo signalling pathway with other pathways, such as Wnt, transforming growth factor beta and nuclear factor kappa-B, are also mentioned because of their importance for modulating bone homeostasis. Furthermore, YAP/TAZ have been extensively studied as mechanotransducers. Due to space limitations, we focus on reviewing how mechanical forces and aging influence cell fate, communications and homeostasis through a dysregulated Hippo signalling pathway.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Junqing Lin
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Jing Wu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| |
Collapse
|
6
|
Gao S, Thillaikumaran T, Dominguez MH, Giang W, Hayes K, Chen X, Pace J, Bockman J, Jathan D, Sung D, Narayan S, Frankfurter M, Mericko-Ishizuka P, Yang J, Castro M, Potente M, Kahn ML. YAP/TAZ signaling in allantois-derived cells is required for placental vascularization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613151. [PMID: 39345443 PMCID: PMC11429833 DOI: 10.1101/2024.09.15.613151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Normal placental development and angiogenesis are crucial for fetal growth and maternal health during pregnancy. However, molecular regulation of placental angiogenesis has been difficult to study due to a lack of specific genetic tools that isolate the placenta from the embryo and yolk sac. To address this gap in knowledge we recently developed Hoxa13 Cre mice in which Cre is expressed in allantois-derived cells, including placental endothelial and stromal cells. Mice lacking the transcriptional regulators Yes-associated protein (YAP) and PDZ-binding motif (TAZ) in allantois-derived cells exhibit embryonic lethality at midgestation with compromised placental vasculature. snRNA-seq analysis revealed transcriptional changes in placental stromal cells and endothelial cells. YAP/TAZ mutants exhibited significantly reduced placental stromal cells prior to the endothelial architectural change, highlighting the role of these cells in placental vascular growth. These results reveal a central role for YAP/TAZ signaling during placental vascular growth and implicate Hoxa13 -derived placental stromal cells as a critical component of placental vascularization.
Collapse
|
7
|
Alexander BE, Zhao H, Astrof S. SMAD4: A Critical Regulator of Cardiac Neural Crest Cell Fate and Vascular Smooth Muscle Differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532676. [PMID: 36993156 PMCID: PMC10055180 DOI: 10.1101/2023.03.14.532676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Background The pharyngeal arch arteries (PAAs) are precursor vessels which remodel into the aortic arch arteries (AAAs) during embryonic cardiovascular development. Cardiac neural crest cells (NCs) populate the PAAs and differentiate into vascular smooth muscle cells (vSMCs), which is critical for successful PAA-to-AAA remodeling. SMAD4, the central mediator of canonical TGFβ signaling, has been implicated in NC-to-vSMC differentiation; however, its distinct roles in vSMC differentiation and NC survival are unclear. Results Here, we investigated the role of SMAD4 in cardiac NC differentiation to vSMCs using lineage-specific inducible mouse strains in an attempt to avoid early embryonic lethality and NC cell death. We found that with global SMAD4 loss, its role in smooth muscle differentiation could be uncoupled from its role in the survival of the cardiac NC in vivo . Moreover, we found that SMAD4 may regulate the induction of fibronectin, a known mediator of NC-to-vSMC differentiation. Finally, we found that SMAD4 is required in NCs cell-autonomously for NC-to-vSMC differentiation and for NC contribution to and persistence in the pharyngeal arch mesenchyme. Conclusions Overall, this study demonstrates the critical role of SMAD4 in the survival of cardiac NCs, their differentiation to vSMCs, and their contribution to the developing pharyngeal arches.
Collapse
|
8
|
Huang JC, Yue ZP, Yu HF, Yang ZQ, Wang YS, Guo B. TAZ ameliorates the microglia-mediated inflammatory response via the Nrf2-ROS-NF-κB pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:435-449. [PMID: 35505966 PMCID: PMC9043866 DOI: 10.1016/j.omtn.2022.03.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 03/27/2022] [Indexed: 01/03/2023]
Abstract
Transcriptional co-activator with PDZ-binding motif (TAZ), one of core modules of the Hippo pathway, involves inflammatory cell infiltration in the liver, but little information is available regarding its physiological function in the microglia-mediated inflammatory response. Here we revealed that activation of TAZ prevented microglia production of proinflammatory cytokines, indicating TAZ’s importance in anti-inflammation. After translocation into the nucleus, TAZ interacted with transcriptional enhanced associate domain (TEAD) and bound to the promoter of nuclear factor erythroid 2-related factor 2 (Nrf2), whose blockage caused inability of TAZ to improve inflammation, implying that Nrf2 is a direct target of TAZ. Further analysis showed that TAZ induced Nrf2 nuclear translocation to enhance antioxidant capacity with attenuation of oxidative stress and the inflammatory response. Under inflammatory conditions, TAZ impeded mitochondrial dysfunction, as indicated by amelioration of ATP levels, mtDNA copy numbers, and mitochondrial membrane potential with an obvious reduction in mitochondrial superoxide, but this impediment was neutralized by blockage of Nrf2. TAZ hindered opening of the mitochondrial permeability transition pore, restrained release of cytochrome c from mitochondria into the cytosol, and was sufficient to rescue microglia from apoptosis dependent on Nrf2. Nrf2 acted as a downstream target of TAZ to repress NF-κB activation by enhancing antioxidant capacity. Collectively, TAZ might ameliorate the microglia-mediated inflammatory response through the Nrf2-reactive oxygen species (ROS)-nuclear factor κB (NF-κB) pathway.
Collapse
Affiliation(s)
- Ji-Cheng Huang
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Zhan-Peng Yue
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Hai-Fan Yu
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Zhan-Qing Yang
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Yu-Si Wang
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| | - Bin Guo
- College of Veterinary Medicine, Jilin University, Changchun, P.R. China
| |
Collapse
|
9
|
Terry BK, Kim S. The Role of Hippo-YAP/TAZ Signaling in Brain Development. Dev Dyn 2022; 251:1644-1665. [PMID: 35651313 DOI: 10.1002/dvdy.504] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/08/2022] Open
Abstract
In order for our complex nervous system to develop normally, both precise spatial and temporal regulation of a number of different signaling pathways is critical. During both early embryogenesis and in organ development, one pathway that has been repeatedly implicated is the Hippo-YAP/TAZ signaling pathway. The paralogs YAP and TAZ are transcriptional co-activators that play an important role in cell proliferation, cell differentiation, and organ growth. Regulation of these proteins by the Hippo kinase cascade is therefore important for normal development. In this article, we review the growing field of research surrounding the role of Hippo-YAP/TAZ signaling in normal and atypical brain development. Starting from the development of the neural tube to the development and refinement of the cerebral cortex, cerebellum, and ventricular system, we address the typical role of these transcriptional co-activators, the functional consequences that manipulation of YAP/TAZ and their upstream regulators have on brain development, and where further research may be of benefit. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bethany K Terry
- Shriners Hospitals Pediatrics Research Center, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA.,Biomedical Sciences Graduate Program, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Seonhee Kim
- Shriners Hospitals Pediatrics Research Center, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA
| |
Collapse
|
10
|
Liao J, Huang Y, Wang Q, Chen S, Zhang C, Wang D, Lv Z, Zhang X, Wu M, Chen G. Gene regulatory network from cranial neural crest cells to osteoblast differentiation and calvarial bone development. Cell Mol Life Sci 2022; 79:158. [PMID: 35220463 PMCID: PMC11072871 DOI: 10.1007/s00018-022-04208-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/02/2022] [Accepted: 02/14/2022] [Indexed: 11/03/2022]
Abstract
Calvarial bone is one of the most complex sequences of developmental events in embryology, featuring a uniquely transient, pluripotent stem cell-like population known as the cranial neural crest (CNC). The skull is formed through intramembranous ossification with distinct tissue lineages (e.g. neural crest derived frontal bone and mesoderm derived parietal bone). Due to CNC's vast cell fate potential, in response to a series of inductive secreted cues including BMP/TGF-β, Wnt, FGF, Notch, Hedgehog, Hippo and PDGF signaling, CNC enables generations of a diverse spectrum of differentiated cell types in vivo such as osteoblasts and chondrocytes at the craniofacial level. In recent years, since the studies from a genetic mouse model and single-cell sequencing, new discoveries are uncovered upon CNC patterning, differentiation, and the contribution to the development of cranial bones. In this review, we summarized the differences upon the potential gene regulatory network to regulate CNC derived osteogenic potential in mouse and human, and highlighted specific functions of genetic molecules from multiple signaling pathways and the crosstalk, transcription factors and epigenetic factors in orchestrating CNC commitment and differentiation into osteogenic mesenchyme and bone formation. Disorders in gene regulatory network in CNC patterning indicate highly close relevance to clinical birth defects and diseases, providing valuable transgenic mouse models for subsequent discoveries in delineating the underlying molecular mechanisms. We also emphasized the potential regenerative alternative through scientific discoveries from CNC patterning and genetic molecules in interfering with or alleviating clinical disorders or diseases, which will be beneficial for the molecular targets to be integrated for novel therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Junguang Liao
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yuping Huang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qiang Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Sisi Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Chenyang Zhang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Dan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zhengbing Lv
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Mengrui Wu
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
11
|
Zarka M, Haÿ E, Cohen-Solal M. YAP/TAZ in Bone and Cartilage Biology. Front Cell Dev Biol 2022; 9:788773. [PMID: 35059398 PMCID: PMC8764375 DOI: 10.3389/fcell.2021.788773] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
YAP and TAZ were initially described as the main regulators of organ growth during development and more recently implicated in bone biology. YAP and TAZ are regulated by mechanical and cytoskeletal cues that lead to the control of cell fate in response to the cellular microenvironment. The mechanical component represents a major signal for bone tissue adaptation and remodelling, so YAP/TAZ contributes significantly in bone and cartilage homeostasis. Recently, mice and cellular models have been developed to investigate the precise roles of YAP/TAZ in bone and cartilage cells, and which appear to be crucial. This review provides an overview of YAP/TAZ regulation and function, notably providing new insights into the role of YAP/TAZ in bone biology.
Collapse
Affiliation(s)
- Mylène Zarka
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| | - Eric Haÿ
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| | - Martine Cohen-Solal
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| |
Collapse
|
12
|
Molina E, Hong L, Chefetz I. NUAK Kinases: Brain-Ovary Axis. Cells 2021; 10:cells10102760. [PMID: 34685740 PMCID: PMC8535158 DOI: 10.3390/cells10102760] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Liver kinase B (LKB1) and adenosine monophosphate (AMP)-activated protein kinase (AMPK) are two major kinases that regulate cellular metabolism by acting as adenosine triphosphate (ATP) sensors. During starvation conditions, LKB1 and AMPK activate different downstream pathways to increase ATP production, while decreasing ATP consumption, which abrogates cellular proliferation and cell death. Initially, LKB1 was considered to be a tumor suppressor due to its loss of expression in various tumor types. Additional studies revealed amplifications in LKB1 and AMPK kinases in several cancers, suggesting a role in tumor progression. The AMPK-related proteins were described almost 20 years ago as a group of key kinases involved in the regulation of cellular metabolism. As LKB1-downstream targets, AMPK-related proteins were also initially considered to function as tumor suppressors. However, further research demonstrated that AMPK-related kinases play a major role not only in cellular physiology but also in tumor development. Furthermore, aside from their role as regulators of metabolism, additional functions have been described for these proteins, including roles in the cell cycle, cell migration, and cell death. In this review, we aim to highlight the major role of AMPK-related proteins beyond their functions in cellular metabolism, focusing on cancer progression based on their role in cell migration, invasion, and cell survival. Additionally, we describe two main AMPK-related kinases, Novel (nua) kinase family 1 (NUAK1) and 2 (NUAK2), which have been understudied, but play a major role in cellular physiology and tumor development.
Collapse
Affiliation(s)
- Ester Molina
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA;
| | - Linda Hong
- School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Ilana Chefetz
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA;
- Masonic Cancer Center, Minneapolis, MN 55455, USA
- Stem Cell Institute, Minneapolis, MN 55455, USA
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence: ; Tel.: +1-507-437-9624
| |
Collapse
|
13
|
Engel-Pizcueta C, Pujades C. Interplay Between Notch and YAP/TAZ Pathways in the Regulation of Cell Fate During Embryo Development. Front Cell Dev Biol 2021; 9:711531. [PMID: 34490262 PMCID: PMC8417249 DOI: 10.3389/fcell.2021.711531] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/02/2021] [Indexed: 12/23/2022] Open
Abstract
Cells in growing tissues receive both biochemical and physical cues from their microenvironment. Growing evidence has shown that mechanical signals are fundamental regulators of cell behavior. However, how physical properties of the microenvironment are transduced into critical cell behaviors, such as proliferation, progenitor maintenance, or differentiation during development, is still poorly understood. The transcriptional co-activators YAP/TAZ shuttle between the cytoplasm and the nucleus in response to multiple inputs and have emerged as important regulators of tissue growth and regeneration. YAP/TAZ sense and transduce physical cues, such as those from the extracellular matrix or the actomyosin cytoskeleton, to regulate gene expression, thus allowing them to function as gatekeepers of progenitor behavior in several developmental contexts. The Notch pathway is a key signaling pathway that controls binary cell fate decisions through cell-cell communication in a context-dependent manner. Recent reports now suggest that the crosstalk between these two pathways is critical for maintaining the balance between progenitor maintenance and cell differentiation in different tissues. How this crosstalk integrates with morphogenesis and changes in tissue architecture during development is still an open question. Here, we discuss how progenitor cell proliferation, specification, and differentiation are coordinated with morphogenesis to construct a functional organ. We will pay special attention to the interplay between YAP/TAZ and Notch signaling pathways in determining cell fate decisions and discuss whether this represents a general mechanism of regulating cell fate during development. We will focus on research carried out in vertebrate embryos that demonstrate the important roles of mechanical cues in stem cell biology and discuss future challenges.
Collapse
Affiliation(s)
- Carolyn Engel-Pizcueta
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
14
|
Kinoshita A, Ohyama K, Tanimura S, Matsuda K, Kishino T, Negishi Y, Asahina N, Shiraishi H, Hosoki K, Tomiwa K, Ishihara N, Mishima H, Mori R, Nakashima M, Saitoh S, Yoshiura KI. Itpr1 regulates the formation of anterior eye segment tissues derived from neural crest cells. Development 2021; 148:271160. [PMID: 34338282 DOI: 10.1242/dev.188755] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/19/2021] [Indexed: 01/23/2023]
Abstract
Mutations in ITPR1 cause ataxia and aniridia in individuals with Gillespie syndrome (GLSP). However, the pathogenic mechanisms underlying aniridia remain unclear. We identified a de novo GLSP mutation hotspot in the 3'-region of ITPR1 in five individuals with GLSP. Furthermore, RNA-sequencing and immunoblotting revealed an eye-specific transcript of Itpr1, encoding a 218amino acid isoform. This isoform is localized not only in the endoplasmic reticulum, but also in the nuclear and cytoplasmic membranes. Ocular-specific transcription was repressed by SOX9 and induced by MAF in the anterior eye segment (AES) tissues. Mice lacking seven base pairs of the last Itpr1 exon exhibited ataxia and aniridia, in which the iris lymphatic vessels, sphincter and dilator muscles, corneal endothelium and stroma were disrupted, but the neural crest cells persisted after completion of AES formation. Our analyses revealed that the 218-amino acid isoform regulated the directionality of actin fibers and the intensity of focal adhesion. The isoform might control the nuclear entry of transcriptional regulators, such as YAP. It is also possible that ITPR1 regulates both AES differentiation and muscle contraction in the iris.
Collapse
Affiliation(s)
- Akira Kinoshita
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Kaname Ohyama
- Department of Pharmacy Practice, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-3131, Japan
| | - Susumu Tanimura
- Department of Cell Regulation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-3131, Japan
| | - Katsuya Matsuda
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Tatsuya Kishino
- Gene Research Center, Center for Frontier Life Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Yutaka Negishi
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8602, Japan
| | - Naoko Asahina
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Hideaki Shiraishi
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Kana Hosoki
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka 594-1101, Japan
| | - Kiyotaka Tomiwa
- Department of Pediatrics, Todaiji Ryoiku Hospital for Children, Nara 630-8211, Japan
| | - Naoko Ishihara
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Hiroyuki Mishima
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Ryoichi Mori
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Masahiro Nakashima
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8602, Japan
| | - Koh-Ichiro Yoshiura
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
15
|
The Cardiac Neural Crest Cells in Heart Development and Congenital Heart Defects. J Cardiovasc Dev Dis 2021; 8:jcdd8080089. [PMID: 34436231 PMCID: PMC8397082 DOI: 10.3390/jcdd8080089] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/22/2022] Open
Abstract
The neural crest (NC) is a multipotent and temporarily migratory cell population stemming from the dorsal neural tube during vertebrate embryogenesis. Cardiac neural crest cells (NCCs), a specified subpopulation of the NC, are vital for normal cardiovascular development, as they significantly contribute to the pharyngeal arch arteries, the developing cardiac outflow tract (OFT), cardiac valves, and interventricular septum. Various signaling pathways are shown to orchestrate the proper migration, compaction, and differentiation of cardiac NCCs during cardiovascular development. Any loss or dysregulation of signaling pathways in cardiac NCCs can lead to abnormal cardiovascular development during embryogenesis, resulting in abnormalities categorized as congenital heart defects (CHDs). This review focuses on the contributions of cardiac NCCs to cardiovascular formation, discusses cardiac defects caused by a disruption of various regulatory factors, and summarizes the role of multiple signaling pathways during embryonic development. A better understanding of the cardiac NC and its vast regulatory network will provide a deeper insight into the mechanisms of the associated abnormalities, leading to potential therapeutic advancements.
Collapse
|
16
|
Zhao X, Le TP, Erhardt S, Findley TO, Wang J. Hippo-Yap Pathway Orchestrates Neural Crest Ontogenesis. Front Cell Dev Biol 2021; 9:706623. [PMID: 34307386 PMCID: PMC8298320 DOI: 10.3389/fcell.2021.706623] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
Neural crest (NC) cells are a migratory stem cell population in vertebrate embryogenesis that can give rise to multiple cell types, including osteoblasts, chondrocytes, smooth muscle cells, neurons, glia, and melanocytes, greatly contributing to the development of different tissues and organs. Defects in NC development are implicated in many human diseases, such as numerous syndromes, craniofacial aberration and congenital heart defects. Research on NC development has gained intense interest and made significant progress. Recent studies showed that the Hippo-Yap pathway, a conserved fundamental pathway with key roles in regulation of cell proliferation, survival, and differentiation, is indispensable for normal NC development. However, the roles and mechanisms of the Hippo-Yap pathway in NC development remain largely unknown. In this review, we summarize the key functions of the Hippo-Yap pathway indicated in NC induction, migration, proliferation, survival, and differentiation, as well as the diseases caused by its dysfunction in NC cells. We also discuss emerging current and future studies in the investigation of the Hippo-Yap pathway in NC development.
Collapse
Affiliation(s)
| | | | | | | | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
17
|
Dieterle MP, Husari A, Steinberg T, Wang X, Ramminger I, Tomakidi P. From the Matrix to the Nucleus and Back: Mechanobiology in the Light of Health, Pathologies, and Regeneration of Oral Periodontal Tissues. Biomolecules 2021; 11:824. [PMID: 34073044 PMCID: PMC8228498 DOI: 10.3390/biom11060824] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Among oral tissues, the periodontium is permanently subjected to mechanical forces resulting from chewing, mastication, or orthodontic appliances. Molecularly, these movements induce a series of subsequent signaling processes, which are embedded in the biological concept of cellular mechanotransduction (MT). Cell and tissue structures, ranging from the extracellular matrix (ECM) to the plasma membrane, the cytosol and the nucleus, are involved in MT. Dysregulation of the diverse, fine-tuned interaction of molecular players responsible for transmitting biophysical environmental information into the cell's inner milieu can lead to and promote serious diseases, such as periodontitis or oral squamous cell carcinoma (OSCC). Therefore, periodontal integrity and regeneration is highly dependent on the proper integration and regulation of mechanobiological signals in the context of cell behavior. Recent experimental findings have increased the understanding of classical cellular mechanosensing mechanisms by both integrating exogenic factors such as bacterial gingipain proteases and newly discovered cell-inherent functions of mechanoresponsive co-transcriptional regulators such as the Yes-associated protein 1 (YAP1) or the nuclear cytoskeleton. Regarding periodontal MT research, this review offers insights into the current trends and open aspects. Concerning oral regenerative medicine or weakening of periodontal tissue diseases, perspectives on future applications of mechanobiological principles are discussed.
Collapse
Affiliation(s)
- Martin Philipp Dieterle
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Ayman Husari
- Center for Dental Medicine, Department of Orthodontics, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany;
- Faculty of Engineering, University of Freiburg, Georges-Köhler-Allee 101, 79110 Freiburg, Germany
| | - Thorsten Steinberg
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Xiaoling Wang
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Imke Ramminger
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| | - Pascal Tomakidi
- Center for Dental Medicine, Division of Oral Biotechnology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (M.P.D.); (X.W.); (I.R.); (P.T.)
| |
Collapse
|
18
|
Young JI, Slifer S, Hecht JT, Blanton SH. DNA Methylation Variation Is Identified in Monozygotic Twins Discordant for Non-syndromic Cleft Lip and Palate. Front Cell Dev Biol 2021; 9:656865. [PMID: 34055787 PMCID: PMC8149607 DOI: 10.3389/fcell.2021.656865] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Non-syndromic cleft lip with or without cleft palate (NSCLP) is the most common craniofacial birth defect. The etiology of NSCLP is complex with multiple genes and environmental factors playing causal roles. Although studies have identified numerous genetic markers associated with NSCLP, the role of epigenetic variation remains relatively unexplored. Because of their identical DNA sequences, monozygotic (MZ) twins discordant for NSCLP are an ideal model for examining the potential contribution of DNA methylation to non-syndromic orofacial clefting. In this study, we compared the patterns of whole genome DNA methylation in six MZ twin pairs discordant for NSCLP. Differentially methylated positions (DMPs) and regions (DMRs) were identified in NSCLP candidate genes, including differential methylation in MAFB and ZEB2 in two independent MZ twin pairs. In addition to DNA methylation differences in NSCLP candidate genes, we found common differential methylation in genes belonging to the Hippo signaling pathway, implicating this mechanosensory pathway in the etiology of NSCLP. The results of this novel approach using MZ twins discordant for NSCLP suggests that differential methylation is one mechanism contributing to NSCLP, meriting future studies on the role of DNA methylation in familial and sporadic NSCLP.
Collapse
Affiliation(s)
- Juan I. Young
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Susan Slifer
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Jacqueline T. Hecht
- McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Susan H. Blanton
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
19
|
Jeong MG, Kim HK, Hwang ES. The essential role of TAZ in normal tissue homeostasis. Arch Pharm Res 2021; 44:253-262. [PMID: 33770379 PMCID: PMC8009801 DOI: 10.1007/s12272-021-01322-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/14/2021] [Indexed: 02/06/2023]
Abstract
Transcriptional coactivator with PDZ-binding motif (TAZ) has been extensively characterized in organ development, tissue regeneration, and tumor progression. In particular, TAZ functions as a Hippo mediator that regulates organ size, tumor growth and migration. It is highly expressed in various types of human cancer, and has been reported to be associated with tumor metastasis and poor outcomes in cancer patients, suggesting that TAZ is an oncogenic regulator. Yes-associated protein (YAP) has 60% similarity in amino acid sequence to TAZ and plays redundant roles with TAZ in the regulation of cell proliferation and migration of cancer cells. Therefore, TAZ and YAP, which are encoded by paralogous genes, are referred to as TAZ/YAP and are suggested to be functionally equivalent. Despite its similarity to YAP, TAZ can be clearly distinguished from YAP based on its genetic, structural, and functional aspects. In addition, targeting superabundant TAZ can be a promising therapeutic strategy for cancer treatment; however, persistent TAZ inactivation may cause failure of tissue homeostatic control. This review focuses primarily on TAZ, not YAP, discusses its structural features and physiological functions in the regulation of tissue homeostasis, and provides new insights into the drug development targeting TAZ to control reproductive and musculoskeletal disorders.
Collapse
Affiliation(s)
- Mi Gyeong Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, C206 Science building, 52 Ewhayeodae-Gil, Seodaemun-Gu, Seoul, 03760, Korea
| | - Hyo Kyeong Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, C206 Science building, 52 Ewhayeodae-Gil, Seodaemun-Gu, Seoul, 03760, Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, C206 Science building, 52 Ewhayeodae-Gil, Seodaemun-Gu, Seoul, 03760, Korea.
| |
Collapse
|
20
|
Bi-Lin KW, Seshachalam PV, Tuoc T, Stoykova A, Ghosh S, Singh MK. Critical role of the BAF chromatin remodeling complex during murine neural crest development. PLoS Genet 2021; 17:e1009446. [PMID: 33750945 PMCID: PMC8016319 DOI: 10.1371/journal.pgen.1009446] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/01/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
The BAF complex plays an important role in the development of a wide range of tissues by modulating gene expression programs at the chromatin level. However, its role in neural crest development has remained unclear. To determine the role of the BAF complex, we deleted BAF155/BAF170, the core subunits required for the assembly, stability, and functions of the BAF complex in neural crest cells (NCCs). Neural crest-specific deletion of BAF155/BAF170 leads to embryonic lethality due to a wide range of developmental defects including craniofacial, pharyngeal arch artery, and OFT defects. RNAseq and transcription factor enrichment analysis revealed that the BAF complex modulates the expression of multiple signaling pathway genes including Hippo and Notch, essential for the migration, proliferation, and differentiation of the NCCs. Furthermore, we demonstrated that the BAF complex is essential for the Brg1-Yap-Tead-dependent transcription of target genes in NCCs. Together, our results demonstrate an important role of the BAF complex in modulating the gene regulatory network essential for neural crest development. Neural crest cells (NCCs) are a multipotent and migratory cell population that is induced at the neural plate border during neurulation and contributes to the formation of a wide range of tissues. Defects in the development, differentiation, or migration of NCCs lead to various birth defects including craniofacial and heart anomalies. Here, by genetically deleting BAF155/BAF170, the core subunits required for the assembly, stability, and functions of the BAF chromatin remodeling complex, we demonstrate that the BAF complex is essential for the proliferation, survival, and differentiation of the NCCs. Neural crest-specific deletion of BAF155/BAF170 leads to embryonic lethality due to a wide range of developmental defects including craniofacial and cardiovascular defects. By performing RNAseq and transcription factor enrichment analysis we show that the BAF complex modulates the expression of multiple signaling pathway genes including Hippo and Notch, essential for the development of the NCCs. Furthermore, the BAF complex component physically interacts with the Hippo signaling components in NCCs to regulate gene expression. We demonstrated that the BAF complex is essential for the Brg1-Yap-Tead-dependent transcription of target genes in NCCs. Together, our results demonstrate a critical role of the BAF complex in modulating the gene regulatory network essential for the proper development of neural crest and neural crest-derived tissues.
Collapse
Affiliation(s)
- Kathleen Wung Bi-Lin
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | | | - Tran Tuoc
- Department of Human Genetics, Ruhr University of Bochum, Bochum, Germany
- Institute of Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | | | - Sujoy Ghosh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Manvendra K. Singh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Center Singapore, Singapore
- * E-mail:
| |
Collapse
|
21
|
Bonnard C, Navaratnam N, Ghosh K, Chan PW, Tan TT, Pomp O, Ng AYJ, Tohari S, Changede R, Carling D, Venkatesh B, Altunoglu U, Kayserili H, Reversade B. A loss-of-function NUAK2 mutation in humans causes anencephaly due to impaired Hippo-YAP signaling. J Exp Med 2021; 217:152044. [PMID: 32845958 PMCID: PMC7953732 DOI: 10.1084/jem.20191561] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/21/2020] [Accepted: 05/19/2020] [Indexed: 01/18/2023] Open
Abstract
Failure of neural tube closure during embryonic development can result in anencephaly, one of the most common birth defects in humans. A family with recurrent anencephalic fetuses was investigated to understand its etiology and pathogenesis. Exome sequencing revealed a recessive germline 21-bp in-frame deletion in NUAK2 segregating with the disease. In vitro kinase assays demonstrated that the 7–amino acid truncation in NUAK2, a serine/threonine kinase, completely abrogated its catalytic activity. Patient-derived disease models including neural progenitor cells and cerebral organoids showed that loss of NUAK2 activity led to decreased Hippo signaling via cytoplasmic YAP retention. In neural tube–like structures, endogenous NUAK2 colocalized apically with the actomyosin network, which was disrupted in patient cells, causing impaired nucleokinesis and apical constriction. Our results establish NUAK2 as an indispensable kinase for brain development in humans and suggest that a NUAK2-Hippo signaling axis regulates cytoskeletal processes that govern cell shape during neural tube closure.
Collapse
Affiliation(s)
- Carine Bonnard
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Naveenan Navaratnam
- Medical Research Council London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kakaly Ghosh
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Puck Wee Chan
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Thong Teck Tan
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Oz Pomp
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Alvin Yu Jin Ng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Sumanty Tohari
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Rishita Changede
- Mechanobiology Institute, National University of Singapore, Singapore
| | - David Carling
- Medical Research Council London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Byrappa Venkatesh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Paediatrics, National University of Singapore, Singapore
| | - Umut Altunoglu
- Medical Genetics Department, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey.,Medical Genetics Department, Koç University School of Medicine, Istanbul, Turkey
| | - Hülya Kayserili
- Medical Genetics Department, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey.,Medical Genetics Department, Koç University School of Medicine, Istanbul, Turkey
| | - Bruno Reversade
- Human Genetics and Embryology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Paediatrics, National University of Singapore, Singapore.,Medical Genetics Department, Koç University School of Medicine, Istanbul, Turkey
| |
Collapse
|
22
|
Hong L, Sun H, Amendt BA. MicroRNA function in craniofacial bone formation, regeneration and repair. Bone 2021; 144:115789. [PMID: 33309989 PMCID: PMC7869528 DOI: 10.1016/j.bone.2020.115789] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
Bone formation in the craniofacial complex is regulated by cranial neural crest (CNC) and mesoderm-derived cells. Different elements of the developing skull, face, mandible, maxilla (jaws) and nasal bones are regulated by an array of transcription factors, signaling molecules and microRNAs (miRs). miRs are molecular modulators of these factors and act to restrict their expression in a temporal-spatial mechanism. miRs control the different genetic pathways that form the craniofacial complex. By understanding how miRs function in vivo during development they can be adapted to regenerate and repair craniofacial genetic anomalies as well as bone diseases and defects due to traumatic injuries. This review will highlight some of the new miR technologies and functions that form new bone or inhibit bone regeneration.
Collapse
Affiliation(s)
- Liu Hong
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, USA
| | - Hongli Sun
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, USA
| | - Brad A Amendt
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, USA; The University of Iowa, Department of Anatomy and Cell Biology, Iowa City, IA, USA; Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
23
|
LeBlanc L, Ramirez N, Kim J. Context-dependent roles of YAP/TAZ in stem cell fates and cancer. Cell Mol Life Sci 2021; 78:4201-4219. [PMID: 33582842 PMCID: PMC8164607 DOI: 10.1007/s00018-021-03781-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/30/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
Hippo effectors YAP and TAZ control cell fate and survival through various mechanisms, including transcriptional regulation of key genes. However, much of this research has been marked by conflicting results, as well as controversy over whether YAP and TAZ are redundant. A substantial portion of the discordance stems from their contradictory roles in stem cell self-renewal vs. differentiation and cancer cell survival vs. apoptosis. In this review, we present an overview of the multiple context-dependent functions of YAP and TAZ in regulating cell fate decisions in stem cells and organoids, as well as their mechanisms of controlling programmed cell death pathways in cancer.
Collapse
Affiliation(s)
- Lucy LeBlanc
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Nereida Ramirez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.,Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA. .,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
24
|
Du W, Bhojwani A, Hu JK. FACEts of mechanical regulation in the morphogenesis of craniofacial structures. Int J Oral Sci 2021; 13:4. [PMID: 33547271 PMCID: PMC7865003 DOI: 10.1038/s41368-020-00110-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
During embryonic development, organs undergo distinct and programmed morphological changes as they develop into their functional forms. While genetics and biochemical signals are well recognized regulators of morphogenesis, mechanical forces and the physical properties of tissues are now emerging as integral parts of this process as well. These physical factors drive coordinated cell movements and reorganizations, shape and size changes, proliferation and differentiation, as well as gene expression changes, and ultimately sculpt any developing structure by guiding correct cellular architectures and compositions. In this review we focus on several craniofacial structures, including the tooth, the mandible, the palate, and the cranium. We discuss the spatiotemporal regulation of different mechanical cues at both the cellular and tissue scales during craniofacial development and examine how tissue mechanics control various aspects of cell biology and signaling to shape a developing craniofacial organ.
Collapse
Affiliation(s)
- Wei Du
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Arshia Bhojwani
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Jimmy K Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Feltri ML, Weaver MR, Belin S, Poitelon Y. The Hippo pathway: Horizons for innovative treatments of peripheral nerve diseases. J Peripher Nerv Syst 2021; 26:4-16. [PMID: 33449435 DOI: 10.1111/jns.12431] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022]
Abstract
Initially identified in Drosophila, the Hippo signaling pathway regulates how cells respond to their environment by controlling proliferation, migration and differentiation. Many recent studies have focused on characterizing Hippo pathway function and regulation in mammalian cells. Here, we present a brief overview of the major components of the Hippo pathway, as well as their regulation and function. We comprehensively review the studies that have contributed to our understanding of the Hippo pathway in the function of the peripheral nervous system and in peripheral nerve diseases. Finally, we discuss innovative approaches that aim to modulate Hippo pathway components in diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- M Laura Feltri
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
26
|
Vanyai HK, Prin F, Guillermin O, Marzook B, Boeing S, Howson A, Saunders RE, Snoeks T, Howell M, Mohun TJ, Thompson B. Control of skeletal morphogenesis by the Hippo-YAP/TAZ pathway. Development 2020; 147:dev187187. [PMID: 32994166 PMCID: PMC7673359 DOI: 10.1242/dev.187187] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 09/07/2020] [Indexed: 12/12/2022]
Abstract
The Hippo-YAP/TAZ pathway is an important regulator of tissue growth, but can also control cell fate or tissue morphogenesis. Here, we investigate the function of the Hippo pathway during the development of cartilage, which forms the majority of the skeleton. Previously, YAP was proposed to inhibit skeletal size by repressing chondrocyte proliferation and differentiation. We find that, in vitro, Yap/Taz double knockout impairs murine chondrocyte proliferation, whereas constitutively nuclear nls-YAP5SA accelerates proliferation, in line with the canonical role of this pathway in most tissues. However, in vivo, cartilage-specific knockout of Yap/Taz does not prevent chondrocyte proliferation, differentiation or skeletal growth, but rather results in various skeletal deformities including cleft palate. Cartilage-specific expression of nls-YAP5SA or knockout of Lats1/2 do not increase cartilage growth, but instead lead to catastrophic malformations resembling chondrodysplasia or achondrogenesis. Physiological YAP target genes in cartilage include Ctgf, Cyr61 and several matrix remodelling enzymes. Thus, YAP/TAZ activity controls chondrocyte proliferation in vitro, possibly reflecting a regenerative response, but is dispensable for chondrocyte proliferation in vivo, and instead functions to control cartilage morphogenesis via regulation of the extracellular matrix.
Collapse
Affiliation(s)
- Hannah K Vanyai
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Fabrice Prin
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Oriane Guillermin
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Bishara Marzook
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Stefan Boeing
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Alexander Howson
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Rebecca E Saunders
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Thomas Snoeks
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Michael Howell
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Timothy J Mohun
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
| | - Barry Thompson
- The Francis Crick Institute, 1 Midland Rd, St Pancras, NW1 1AT London, UK
- EMBL Australia, Department of Cancer Biology & Therapeutics, The John Curtin School of Medical Research, The Australian National University, 131 Garran Rd, Acton, 2601, Canberra, Australia
| |
Collapse
|
27
|
Sun R, Wang Z, Claus Henn B, Su L, Lu Q, Lin X, Wright RO, Bellinger DC, Kile M, Mazumdar M, Tellez-Rojo MM, Schnaas L, Christiani DC. Identification of novel loci associated with infant cognitive ability. Mol Psychiatry 2020; 25:3010-3019. [PMID: 30120420 PMCID: PMC6378130 DOI: 10.1038/s41380-018-0205-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 12/02/2022]
Abstract
It is believed that genetic factors play a large role in the development of many cognitive and neurological processes; however, epidemiological evidence for the genetic basis of childhood neurodevelopment is very limited. Identification of the genetic polymorphisms associated with early-stage neurodevelopment will help elucidate biological mechanisms involved in neuro-behavior and provide a better understanding of the developing brain. To search for such variants, we performed a genome-wide association study (GWAS) for infant mental and motor ability at two years of age with mothers and children recruited from cohorts in Bangladesh and Mexico. Infant ability was assessed using mental and motor composite scores calculated with country-specific versions of the Bayley Scales of Infant Development. A missense variant (rs1055153) located in the gene WWTR1 reached genome-wide significance in association with mental composite score (meta-analysis effect size of minor allele βmeta = -6.04; 95% CI: -8.13 to -3.94; P = 1.56×10-8). Infants carrying the minor allele reported substantially lower cognitive scores in both cohorts, and this variant is predicted to be in the top 0.3% of most deleterious substitutions in the human genome. Fine mapping and region-based association testing provided additional suggestive evidence that both WWTR1 and a second gene, LRP1B, were associated with infant cognitive ability. Comparisons with recently conducted GWAS in intelligence and educational attainment indicate that our phenotypes do not possess a high genetic correlation with either adolescent or adult cognitive traits, suggesting that infant neurological assessments should be treated as an independent outcome of interest. Additional functional studies and replication efforts in other cohorts may help uncover new biological pathways and genetic architectures that are crucial to the developing brain.
Collapse
Affiliation(s)
- Ryan Sun
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| | - Zhaoxi Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Birgit Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Robert O Wright
- Department of Preventive Medicine, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - David C Bellinger
- Department of Psychiatry, Harvard Medical School and Boston Children's Hospital, Boston, MA, 02115, USA
| | - Molly Kile
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Maitreyi Mazumdar
- Department of Neurology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Martha Maria Tellez-Rojo
- Center of Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, 62100, Mexico
| | - Lourdes Schnaas
- Center of Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, 62100, Mexico
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| |
Collapse
|
28
|
Ray H, Chang C. The transcription factor Hypermethylated in Cancer 1 (Hic1) regulates neural crest migration via interaction with Wnt signaling. Dev Biol 2020; 463:169-181. [PMID: 32502469 DOI: 10.1016/j.ydbio.2020.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 01/20/2023]
Abstract
The transcription factor Hypermethylated in Cancer 1 (HIC1) is associated with both tumorigenesis and the complex human developmental disorder Miller-Dieker Syndrome. While many studies have characterized HIC1 as a tumor suppressor, HIC1 function in development is less understood. Loss-of-function mouse alleles show embryonic lethality accompanied with developmental defects, including craniofacial abnormalities that are reminiscent of human Miller-Dieker Syndrome patients. However, the tissue origin of the defects has not been reported. In this study, we use the power of the Xenopus laevis model system to explore Hic1 function in early development. We show that hic1 mRNA is expressed throughout early Xenopus development and has a spatial distribution within the neural plate border and in migrating neural crest cells in branchial arches. Targeted manipulation of hic1 levels in the dorsal ectoderm that gives rise to neural and neural crest tissues reveals that both overexpression and knockdown of hic1 result in craniofacial defects with malformations of the craniofacial cartilages. Neural crest specification is not affected by altered hic1 levels, but migration of the cranial neural crest is impaired both in vivo and in tissue explants. Mechanistically, we find that Hic1 regulates cadherin expression profiles and canonical Wnt signaling. Taken together, these results identify Hic1 as a novel regulator of the canonical Wnt pathway during neural crest migration.
Collapse
Affiliation(s)
- Heather Ray
- Dept. of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, MCLM 338, 1918 University Dr. Birmingham, AL, 35294, USA.
| | - Chenbei Chang
- Dept. of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, MCLM 338, 1918 University Dr. Birmingham, AL, 35294, USA
| |
Collapse
|
29
|
Goodwin AF, Chen CP, Vo NT, Bush JO, Klein OD. YAP/TAZ Regulate Elevation and Bone Formation of the Mouse Secondary Palate. J Dent Res 2020; 99:1387-1396. [PMID: 32623954 DOI: 10.1177/0022034520935372] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clefting of the secondary palate is one of the most common congenital anomalies, and the multiple corrective surgeries that individuals with isolated cleft palate undergo are associated with major costs and morbidities. Secondary palate development is a complex, multistep process that includes the elevation of the palatal shelves from a vertical to horizontal position, a process that is not well understood. The Hippo signaling cascade is a mechanosensory pathway that regulates morphogenesis, homeostasis, and regeneration by controlling cell proliferation, apoptosis, and differentiation, primarily via negative regulation of the downstream effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). We deleted Yap/Taz throughout the palatal shelf mesenchyme as well as specifically in the posterior palatal shelf mesenchyme, using the Osr2Cre and Col2Cre drivers, respectively, which resulted in palatal shelf elevation delay and clefting of the secondary palate. In addition, the deletion resulted in undersized bones of the secondary palate. We next determined downstream targets of YAP/TAZ in the posterior palatal shelves, which included Ibsp and Phex, genes involved in mineralization, and Loxl4, which encodes a lysyl oxidase that catalyzes collagen crosslinking. Ibsp, Phex, and Loxl4 were expressed at decreased levels in the ossification region in the posterior palatal shelf mesenchyme upon deletion of Yap/Taz. Furthermore, collagen levels were decreased specifically in the same region prior to elevation. Thus, our data suggest that YAP/TAZ may regulate collagen crosslinking in the palatal shelf mesenchyme, thus controlling palatal shelf elevation, as well as mineralization of the bones of the secondary palate.
Collapse
Affiliation(s)
- A F Goodwin
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - C P Chen
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - N T Vo
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - J O Bush
- Program in Craniofacial Biology, University of California, San Francisco, CA, USA.,Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.,Institute of Human Genetics, University of California, San Francisco, CA, USA
| | - O D Klein
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Program in Craniofacial Biology, University of California, San Francisco, CA, USA.,Institute of Human Genetics, University of California, San Francisco, CA, USA.,Department of Pediatrics, University of California, San Francisco, CA, USA
| |
Collapse
|
30
|
The Hippo Pathway as a Driver of Select Human Cancers. Trends Cancer 2020; 6:781-796. [PMID: 32446746 DOI: 10.1016/j.trecan.2020.04.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022]
Abstract
The Hippo pathway regulates myriad biological processes in diverse species and is a key cancer signaling network in humans. Although Hippo has been linked to multiple aspects of cancer, its role in this disease is incompletely understood. Large-scale pan-cancer analyses of core Hippo pathway genes reveal that the pathway is mutated at a high frequency only in select human cancers, including malignant mesothelioma and meningioma. Hippo pathway deregulation is also enriched in squamous epithelial cancers. We discuss cancer-related functions of the Hippo pathway and potential explanations for the cancer-restricted mutation profile of core Hippo pathway genes. Greater understanding of Hippo pathway deregulation in cancers will be essential to guide the imminent use of Hippo-targeted therapies.
Collapse
|
31
|
Ouyang N, Li H, Wang M, Shen H, Si J, Shen G. The Transcription Factor Foxc1 Promotes Osteogenesis by Directly Regulating Runx2 in Response of Intermittent Parathyroid Hormone (1-34) Treatment. Front Pharmacol 2020; 11:592. [PMID: 32431614 PMCID: PMC7216818 DOI: 10.3389/fphar.2020.00592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/17/2020] [Indexed: 01/23/2023] Open
Abstract
Parathyroid hormone (PTH) is crucial for bone remodeling. Intermittent PTH (1–34) administration stimulates osteogenesis and promotes bone formation; however, the possible targets and underlying mechanisms still remain unclear. In this study, functional links between PTH and Foxc1, a transcription factor reported to be predominant in skeletal development and formation, were indicated. We determined the impacts of Foxc1 on in vitro osteogenic differentiation and in vivo bone regeneration under intermittent PTH induction, and further explored its possible targets. We found that the expression level of Foxc1 was upregulated during osteogenic induction by intermittent PTH treatment, and the elevated expression of Foxc1 induced by PTH was inhibited by PTH1R silencing, while rescued by intermittent PTH supplement. By gain- and loss-of-function strategies targeting Foxc1 in MC3T3-E1 cells, we demonstrated that Foxc1 could promote in vitro osteogenic differentiation by intermittent PTH induction. Moreover, immunofluorescence analysis indicated the nuclear co-localization of Foxc1 with Runx2. Luciferase-reporter and chromatin immunoprecipitation analysis further confirmed that Foxc1 could bind to the P1 promoter region of Runx2 directly, which plays an indispensable part in osteogenic differentiation and bone mineralization. Meanwhile, we also revealed that Foxc1 could promote bone regeneration induced by intermittent PTH treatment in vivo. Taken together, this study revealed the role and mechanism of Foxc1 on in vitro osteogenic differentiation and in vivo bone regeneration in response of intermittent PTH treatment.
Collapse
Affiliation(s)
- Ningjuan Ouyang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hongliang Li
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Minjiao Wang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hongzhou Shen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jiawen Si
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Guofang Shen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
32
|
Bhattacharya D, Azambuja AP, Simoes-Costa M. Metabolic Reprogramming Promotes Neural Crest Migration via Yap/Tead Signaling. Dev Cell 2020; 53:199-211.e6. [PMID: 32243782 PMCID: PMC7236757 DOI: 10.1016/j.devcel.2020.03.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/05/2020] [Accepted: 03/04/2020] [Indexed: 02/04/2023]
Abstract
The Warburg effect is one of the metabolic hallmarks of cancer cells, characterized by enhanced glycolysis even under aerobic conditions. This physiological adaptation is associated with metastasis , but we still have a superficial understanding of how it affects cellular processes during embryonic development. Here we report that the neural crest, a migratory stem cell population in vertebrate embryos, undergoes an extensive metabolic remodeling to engage in aerobic glycolysis prior to delamination. This increase in glycolytic flux promotes Yap/Tead signaling, which activates the expression of a set of transcription factors to drive epithelial-to-mesenchymal transition. Our results demonstrate how shifts in carbon metabolism can trigger the gene regulatory circuits that control complex cell behaviors. These findings support the hypothesis that the Warburg effect is a precisely regulated developmental mechanism that is anomalously reactivated during tumorigenesis and metastasis.
Collapse
Affiliation(s)
| | - Ana Paula Azambuja
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850, USA
| | - Marcos Simoes-Costa
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
33
|
Liu W, Wang K, Lv X, Wang Q, Li X, Yang Z, Liu X, Yan L, Fu X, Xiao R. Up-regulation of RNA Binding Proteins Contributes to Folate Deficiency-Induced Neural Crest Cells Dysfunction. Int J Biol Sci 2020; 16:85-98. [PMID: 31892848 PMCID: PMC6930370 DOI: 10.7150/ijbs.33976] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 09/09/2019] [Indexed: 12/21/2022] Open
Abstract
Folate deficiency has long been associated with the abnormal development of the neural crest cells (NCCs) and neural tube defects (NTDs). RNA binding proteins (RBPs) also play important roles in the normal neural crest development and neural tube formation. Nevertheless, the causative mechanism by which folate status influences human NCCs development and the RBPs functions remains unknown. In this study, we differentiated H9 human embryonic stem cells into neural crest cells (H9-NCCs) and then constructed three folic acid (FA) deficiency (FAD) H9-NCCs models in vitro. Decreased viability, impaired migration and promoted apoptosis of H9-NCCs were observed in three FAD H9-NCCs models. In addition, we showed that three RBPs, namely, hnRNPC, LARP6 and RCAN2, were up-regulated both in the FAD H9-NCC models in vitro and in the FAD mouse model in vivo. Knocking down of these three RBPs increased the H9-NCC viability and RCAN2 knockdown further promoted H9-NCC migration under FAD conditions. In normal culture condition, overexpression of RCAN2 and HnRNPC did not affect viabilities and migration of H9-NCCs while overexpression of LARP6 reduced the H9-NCC viability. Our findings demonstrate important regulatory effects of RBPs underlying FAD-induced impaired function of NCCs.
Collapse
Affiliation(s)
- Wenbo Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Kang Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Xiaoyan Lv
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Xiu Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing, 100144, People's Republic of China
| |
Collapse
|
34
|
Kakiuchi-Kiyota S, Schutten MM, Zhong Y, Crawford JJ, Dey A. Safety Considerations in the Development of Hippo Pathway Inhibitors in Cancers. Front Cell Dev Biol 2019; 7:156. [PMID: 31475147 PMCID: PMC6707765 DOI: 10.3389/fcell.2019.00156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/25/2019] [Indexed: 01/04/2023] Open
Abstract
The Hippo pathway is a critical regulator of cell and organ growth and has emerged as a target for therapeutic intervention in cancers. Its signaling is thought to play an important role in various physiological processes including homeostasis and tissue regeneration. To date there has been limited information about potential pharmacology-related (on-target) safety liabilities of Hippo pathway inhibitors in the context of cancer indications. Herein, we review data from human genetic disorders and genetically engineered rodent models to gain insight into safety liabilities that may emerge from the inhibition of Hippo pathway. Germline systemic deletion of murine Hippo pathway effectors (Yap, Taz, and Teads) resulted in embryonic lethality or developmental phenotypes. Mouse models with tissue-specific deletion (or mutant overexpression) of the key effectors in Hippo pathways have indicated that, at least in some tissues, Hippo signaling may be dispensable for physiological homeostasis; and appears to be critical for regeneration upon tissue damage, indicating that patients with underlying comorbidities and/or insults caused by therapeutic agents and/or comedications may have a higher risk. Caution should be taken in interpreting phenotypes from tissue-specific transgenic animal models since some tissue-specific promoters are turned on during development. In addition, therapeutic agents may result in systemic effects not well-predicted by animal models with tissue-specific gene deletion. Therefore, the development of models that allows for systemic deletion of Yap and/or Taz in adult animals will be key in evaluating the potential safety liabilities of Hippo pathway modulation. In this review, we focus on potential challenges and strategies for targeting the Hippo pathway in cancers.
Collapse
Affiliation(s)
- Satoko Kakiuchi-Kiyota
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, United States
| | - Melissa M Schutten
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, United States
| | - Yu Zhong
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, United States
| | - James J Crawford
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, CA, United States
| | - Anwesha Dey
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
35
|
Kumar D, Nitzan E, Kalcheim C. YAP promotes neural crest emigration through interactions with BMP and Wnt activities. Cell Commun Signal 2019; 17:69. [PMID: 31228951 PMCID: PMC6589182 DOI: 10.1186/s12964-019-0383-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background Premigratory neural crest progenitors undergo an epithelial-to-mesenchymal transition and leave the neural tube as motile cells. Previously, we showed that BMP generates trunk neural crest emigration through canonical Wnt signaling which in turn stimulates G1/S transition. The molecular network underlying this process is, however, not yet completely deciphered. Yes-associated-protein (YAP), an effector of the Hippo pathway, controls various aspects of development including cell proliferation, migration, survival and differentiation. In this study, we examined the possible involvement of YAP in neural crest emigration and its relationship with BMP and Wnt. Methods We implemented avian embryos in which levels of YAP gene activity were either reduced or upregulated by in ovo plasmid electroporation, and monitored effects on neural crest emigration, survival and proliferation. Neural crest-derived sensory neuron and melanocyte development were assessed upon gain of YAP function. Imunohistochemistry was used to assess YAP expression. In addition, the activity of specific signaling pathways including YAP, BMP and Wnt was monitored with specific reporters. Results We find that the Hippo pathway transcriptional co-activator YAP is expressed and is active in premigratory crest of avian embryos. Gain of YAP function stimulates neural crest emigration in vivo, and attenuating YAP inhibits cell exit. This is associated with an accumulation of FoxD3-expressing cells in the dorsal neural tube, with reduced proliferation, and enhanced apoptosis. Furthermore, gain of YAP function inhibits differentiation of Islet-1-positive sensory neurons and augments the number of EdnrB2-positive melanocytes. Using specific in vivo reporters, we show that loss of YAP function in the dorsal neural tube inhibits BMP and Wnt activities whereas gain of YAP function stimulates these pathways. Reciprocally, inhibition of BMP and Wnt signaling by noggin or Xdd1, respectively, downregulates YAP activity. In addition, YAP-dependent stimulation of neural crest emigration is compromised upon inhibition of either BMP or Wnt activities. Together, our results suggest a positive bidirectional cross talk between these pathways. Conclusions Our data show that YAP is necessary for emigration of neural crest progenitors. In addition, they incorporate YAP signaling into a BMP/Wnt-dependent molecular network responsible for emigration of trunk-level neural crest.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Erez Nitzan
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel.
| |
Collapse
|
36
|
Shen H, Lu C, Shi J, Li H, Si J, Shen G. Satb2 expression in Foxc1-promoted osteogenic differentiation of MC3T3-E1 cells is negatively regulated by microRNA-103-3p. Acta Biochim Biophys Sin (Shanghai) 2019; 51:588-597. [PMID: 31089719 DOI: 10.1093/abbs/gmz037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Indexed: 12/12/2022] Open
Abstract
The forkhead transcription factor C1 (Foxc1) is a cell-fate-determining factor that controls cranial bone development and osteogenic differentiation. Previously, it was demonstrated that various microRNAs (miRNAs) play important roles in osteogenesis and regulate the complex process of osteogenic differentiation. However, it remains unclear how miRNA expression changes during Foxc1-promoted osteogenic differentiation. In this study, we successfully overexpressed the Foxc1 gene in MC3T3-E1 cells and investigated the alterations in the miRNA expression profile on day 3 after osteogenic induction by using a miRNA microarray. Nine downregulated miRNAs and eight upregulated miRNAs were found to be differentially expressed. Among these miRNAs, miR-103-3p was consistently downregulated in the Foxc1-overexpressing MC3T3-E1 cells and was identified as a negative regulator of osteogenic differentiation by using a gain- and lose-of-function assay. The special AT-rich sequence-binding protein 2 (Satb2), a pivotal osteogenic transcription factor, was identified as the miR-103-3p targeting gene and was verified by real-time polymerase chain reaction, western blot analysis, and luciferase assay. Overexpression of miR-103-3p markedly inhibited the expression of Satb2 and attenuated Foxc1-promoted osteogenic differentiation. Taken together, our results elucidated the miRNA expression profiles of MC3T3-E1 cells in the early stage of Foxc1-promoted osteogenic differentiation and suggested that miR-103-3p acts as a negative regulator of the osteogenic differentiation of MC3T3-E1 cells by directly targeting Satb2.
Collapse
Affiliation(s)
- Hongzhou Shen
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chenpei Lu
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jun Shi
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Hongliang Li
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiawen Si
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Guofang Shen
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
37
|
Dooley CM, Wali N, Sealy IM, White RJ, Stemple DL, Collins JE, Busch-Nentwich EM. The gene regulatory basis of genetic compensation during neural crest induction. PLoS Genet 2019; 15:e1008213. [PMID: 31199790 PMCID: PMC6594659 DOI: 10.1371/journal.pgen.1008213] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/26/2019] [Accepted: 05/27/2019] [Indexed: 12/15/2022] Open
Abstract
The neural crest (NC) is a vertebrate-specific cell type that contributes to a wide range of different tissues across all three germ layers. The gene regulatory network (GRN) responsible for the formation of neural crest is conserved across vertebrates. Central to the induction of the NC GRN are AP-2 and SoxE transcription factors. NC induction robustness is ensured through the ability of some of these transcription factors to compensate loss of function of gene family members. However the gene regulatory events underlying compensation are poorly understood. We have used gene knockout and RNA sequencing strategies to dissect NC induction and compensation in zebrafish. We genetically ablate the NC using double mutants of tfap2a;tfap2c or remove specific subsets of the NC with sox10 and mitfa knockouts and characterise genome-wide gene expression levels across multiple time points. We find that compensation through a single wild-type allele of tfap2c is capable of maintaining early NC induction and differentiation in the absence of tfap2a function, but many target genes have abnormal expression levels and therefore show sensitivity to the reduced tfap2 dosage. This separation of morphological and molecular phenotypes identifies a core set of genes required for early NC development. We also identify the 15 somites stage as the peak of the molecular phenotype which strongly diminishes at 24 hpf even as the morphological phenotype becomes more apparent. Using gene knockouts, we associate previously uncharacterised genes with pigment cell development and establish a role for maternal Hippo signalling in melanocyte differentiation. This work extends and refines the NC GRN while also uncovering the transcriptional basis of genetic compensation via paralogues. Embryonic development is an intricate process that requires genes to be active at the right time and place. Organisms have evolved mechanisms that ensure faithful execution of developmental programmes even if genes fail to function. For example, in a process called genetic compensation, one or more genes become activated in response to loss of function of another. In this work we use the zebrafish model to investigate how two related genes, tfap2a and tfap2c, interact to ensure establishment of the neural crest, a vertebrate-specific cell type that contributes to many different tissues. Losing tfap2a activity causes mild morphological defects and losing tfap2c has no visible effect. Yet when both are inactive, embryos are severely abnormal due to lack of neural crest-derived tissues. Here we show that loss of tfap2a triggers upregulation of tfap2c which prevents the loss of neural crest tissue. However, the genes normally regulated by tfap2a respond differently to tfap2c allowing us to identify the first tier of the Ap2 network and new players in neural crest biology. Our work demonstrates that the expression signature of partial, but morphologically sufficient, genetic compensation provides an opportunity to dissect gene regulatory networks.
Collapse
Affiliation(s)
| | - Neha Wali
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Ian M. Sealy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Richard J. White
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Derek L. Stemple
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - John E. Collins
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Elisabeth M. Busch-Nentwich
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Kim S, Thomasy SM, Raghunathan VK, Teixeira LB, Moshiri A, FitzGerald P, Murphy CJ. Ocular phenotypic consequences of a single copy deletion of the Yap1 gene ( Yap1 +/-) in mice. Mol Vis 2019; 25:129-142. [PMID: 30820148 PMCID: PMC6382475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 02/15/2019] [Indexed: 11/29/2022] Open
Abstract
PURPOSE To identify the effects of a single copy deletion of Yap1 (Yap1 +/-) in the mouse eye, the ocular phenotypic consequences of Yap1 +/- were determined in detail. METHODS Complete ophthalmic examinations, as well as corneal esthesiometry, the phenol red thread test, intraocular pressure, and Fourier-domain optical coherence tomography were performed on Yap1 +/- and age-matched wild-type (WT) mice between eyelid opening (2 weeks after birth) and adulthood (2 months and 1 year after birth). Following euthanasia, enucleated eyes were characterized histologically. RESULTS Microphthalmia with small palpebral fissures, corneal fibrosis, and reduced corneal sensation were common findings in the Yap1 +/- mice. Generalized corneal fibrosis precluded clinical examination of the posterior structures. Histologically, thinning and keratinization of the corneal epithelium were observed in the Yap1 +/- mice in comparison with the WT mice. Distorted collagen fiber arrangement and hypercellularity of keratocytes were observed in the stroma. Descemet's membrane was extremely thin and lacked an endothelial layer in the Yap1 +/- mice. The iris was adherent to the posterior cornea along most of its surface creating a distorted contour. Most of the Yap1 +/- eyes were microphakic with swollen fibers and bladder cells. The retinas of the Yap1 +/- mice were normal at 2 weeks and 2 months of age, but the presence of retinal abnormalities, including retinoschisis and detachment, was markedly increased in the Yap1 +/- mice at 1 year of age. CONCLUSIONS The results show that the heterozygous deletion of the Yap1 gene in mice leads to complex ocular abnormalities, including microphthalmia, corneal fibrosis, anterior segment dysgenesis, and cataract.
Collapse
Affiliation(s)
- Soohyun Kim
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA
| | - Sara M. Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA,Department of Ophthalmology & Vision Science, School of Medicine, School of Medicine, University of California Davis, Davis, CA
| | - Vijay Krishna Raghunathan
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX,The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX,Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX
| | - Leandro B.C. Teixeira
- Comparative Ocular Pathology Laboratory of Wisconsin, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, School of Medicine, University of California Davis, Davis, CA
| | - Paul FitzGerald
- Department of Ophthalmology & Vision Science, School of Medicine, School of Medicine, University of California Davis, Davis, CA,Department of Cell Biology and Human Anatomy, School of Medicine, School of Medicine, University of California Davis, Davis, CA
| | - Christopher J. Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA,Department of Ophthalmology & Vision Science, School of Medicine, School of Medicine, University of California Davis, Davis, CA
| |
Collapse
|
39
|
Cho H, Kim J, Ahn JH, Hong YK, Mäkinen T, Lim DS, Koh GY. YAP and TAZ Negatively Regulate Prox1 During Developmental and Pathologic Lymphangiogenesis. Circ Res 2019; 124:225-242. [DOI: 10.1161/circresaha.118.313707] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Hyunsoo Cho
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
| | - Jaeryung Kim
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea (J.K., G.Y.K.)
| | - Ji Hoon Ahn
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
| | - Young-Kwon Hong
- Department of Surgery (Y.-K.H.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Biology (Y.-K.H.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Taija Mäkinen
- Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (T.M.)
| | - Dae-Sik Lim
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
| | - Gou Young Koh
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea (J.K., G.Y.K.)
| |
Collapse
|
40
|
Cox AG, Tsomides A, Yimlamai D, Hwang KL, Miesfeld J, Galli GG, Fowl BH, Fort M, Ma KY, Sullivan MR, Hosios AM, Snay E, Yuan M, Brown KK, Lien EC, Chhangawala S, Steinhauser ML, Asara JM, Houvras Y, Link B, Vander Heiden MG, Camargo FD, Goessling W. Yap regulates glucose utilization and sustains nucleotide synthesis to enable organ growth. EMBO J 2018; 37:embj.2018100294. [PMID: 30348863 DOI: 10.15252/embj.2018100294] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/14/2022] Open
Abstract
The Hippo pathway and its nuclear effector Yap regulate organ size and cancer formation. While many modulators of Hippo activity have been identified, little is known about the Yap target genes that mediate these growth effects. Here, we show that yap -/- mutant zebrafish exhibit defects in hepatic progenitor potential and liver growth due to impaired glucose transport and nucleotide biosynthesis. Transcriptomic and metabolomic analyses reveal that Yap regulates expression of glucose transporter glut1, causing decreased glucose uptake and use for nucleotide biosynthesis in yap -/- mutants, and impaired glucose tolerance in adults. Nucleotide supplementation improves Yap deficiency phenotypes, indicating functional importance of glucose-fueled nucleotide biosynthesis. Yap-regulated glut1 expression and glucose uptake are conserved in mammals, suggesting that stimulation of anabolic glucose metabolism is an evolutionarily conserved mechanism by which the Hippo pathway controls organ growth. Together, our results reveal a central role for Hippo signaling in glucose metabolic homeostasis.
Collapse
Affiliation(s)
- Andrew G Cox
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Allison Tsomides
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dean Yimlamai
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Katie L Hwang
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | | | - Giorgio G Galli
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brendan H Fowl
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Fort
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kimberly Y Ma
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark R Sullivan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aaron M Hosios
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Erin Snay
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Min Yuan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kristin K Brown
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Evan C Lien
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sagar Chhangawala
- Weill Cornell Medical College and New York Presbyterian Hospital, New York, NY, USA
| | - Matthew L Steinhauser
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John M Asara
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yariv Houvras
- Weill Cornell Medical College and New York Presbyterian Hospital, New York, NY, USA
| | - Brian Link
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Fernando D Camargo
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Wolfram Goessling
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA .,Harvard Stem Cell Institute, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Boston, MA, USA
| |
Collapse
|
41
|
Nguyen BH, Ishii M, Maxson RE, Wang J. Culturing and Manipulation of O9-1 Neural Crest Cells. J Vis Exp 2018. [PMID: 30371662 DOI: 10.3791/58346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Neural crest cells (NCCs) are migrating multipotent stem cells that can differentiate into different cell types and give rise to multiple tissues and organs. The O9-1 cell line is derived from the endogenous mouse embryonic NCCs and maintains its multipotency. However, under specific culture conditions, O9-1 cells can differentiate into different cell types and be utilized in a wide range of research applications. Recently, with the combination of mouse studies and O9-1 cell studies, we have shown that the Hippo signaling pathway effectors Yap and Taz play important roles in neural crest-derived craniofacial development. Although the culturing process for O9-1 cells is more complicated than that used for other cell lines, the O9-1 cell line is a powerful model for investigating NCCs in vitro. Here, we present a protocol for culturing the O9-1 cell line to maintain its stemness, as well as protocols for differentiating O9-1 cells into different cell types, such as smooth muscle cells and osteoblasts. In addition, protocols are described for performing gene loss-of-function studies in O9-1 cells by using CRISPR-Cas9 deletion and small interfering RNA-mediated knockdown.
Collapse
Affiliation(s)
- Bao H Nguyen
- Molecular Physiology and Biophysics, Baylor College of Medicine
| | - Mamoru Ishii
- Department of Biochemistry and Molecular Biology, University of Southern California
| | - Robert E Maxson
- Department of Biochemistry and Molecular Biology, University of Southern California;
| | - Jun Wang
- Molecular Physiology and Biophysics, Baylor College of Medicine;
| |
Collapse
|
42
|
Sun Z, da Fontoura CSG, Moreno M, Holton NE, Sweat M, Sweat Y, Lee MK, Arbon J, Bidlack FB, Thedens DR, Nopoulos P, Cao H, Eliason S, Weinberg SM, Martin JF, Moreno-Uribe L, Amendt BA. FoxO6 regulates Hippo signaling and growth of the craniofacial complex. PLoS Genet 2018; 14:e1007675. [PMID: 30286078 PMCID: PMC6197693 DOI: 10.1371/journal.pgen.1007675] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/22/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
The mechanisms that regulate post-natal growth of the craniofacial complex and that ultimately determine the size and shape of our faces are not well understood. Hippo signaling is a general mechanism to control tissue growth and organ size, and although it is known that Hippo signaling functions in neural crest specification and patterning during embryogenesis and before birth, its specific role in postnatal craniofacial growth remains elusive. We have identified the transcription factor FoxO6 as an activator of Hippo signaling regulating neonatal growth of the face. During late stages of mouse development, FoxO6 is expressed specifically in craniofacial tissues and FoxO6-/- mice undergo expansion of the face, frontal cortex, olfactory component and skull. Enlargement of the mandible and maxilla and lengthening of the incisors in FoxO6-/- mice are associated with increases in cell proliferation. In vitro and in vivo studies demonstrated that FoxO6 activates Lats1 expression, thereby increasing Yap phosphorylation and activation of Hippo signaling. FoxO6-/- mice have significantly reduced Hippo Signaling caused by a decrease in Lats1 expression and decreases in Shh and Runx2 expression, suggesting that Shh and Runx2 are also linked to Hippo signaling. In vitro, FoxO6 activates Hippo reporter constructs and regulates cell proliferation. Furthermore PITX2, a regulator of Hippo signaling is associated with Axenfeld-Rieger Syndrome causing a flattened midface and we show that PITX2 activates FoxO6 expression. Craniofacial specific expression of FoxO6 postnatally regulates Hippo signaling and cell proliferation. Together, these results identify a FoxO6-Hippo regulatory pathway that controls skull growth, odontogenesis and face morphology.
Collapse
Affiliation(s)
- Zhao Sun
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
| | - Clarissa S. G. da Fontoura
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA, United States of America
| | - Myriam Moreno
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
| | - Nathan E. Holton
- Department of Orthodontics, College of Dentistry, The University of Iowa, Iowa City, IA, United States of America
| | - Mason Sweat
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
| | - Yan Sweat
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
| | - Myoung Keun Lee
- Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh PA, United States of America
| | - Jed Arbon
- Private practice, Cary, North Carolina United States of America
| | | | - Daniel R. Thedens
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
| | - Peggy Nopoulos
- Department of Psychiatry, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
| | - Huojun Cao
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA, United States of America
| | - Steven Eliason
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
| | - Seth M. Weinberg
- Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh PA, United States of America
| | - James F. Martin
- Department of Physiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Lina Moreno-Uribe
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA, United States of America
- Department of Orthodontics, College of Dentistry, The University of Iowa, Iowa City, IA, United States of America
| | - Brad A. Amendt
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States of America
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA, United States of America
- Department of Orthodontics, College of Dentistry, The University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
43
|
Oristian KM, Crose LES, Kuprasertkul N, Bentley RC, Lin YT, Williams N, Kirsch DG, Linardic CM. Loss of MST/Hippo Signaling in a Genetically Engineered Mouse Model of Fusion-Positive Rhabdomyosarcoma Accelerates Tumorigenesis. Cancer Res 2018; 78:5513-5520. [PMID: 30093562 DOI: 10.1158/0008-5472.can-17-3912] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/26/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022]
Abstract
A hallmark of fusion-positive alveolar rhabdomyosarcoma (aRMS) is the presence of a chromosomal translocation encoding the PAX3-FOXO1 fusion oncogene. Primary cell-based modeling experiments have shown that PAX3-FOXO1 is necessary, but not sufficient for aRMS tumorigenesis, indicating additional molecular alterations are required to initiate and sustain tumor growth. Previously, we showed that PAX3-FOXO1-positive aRMS is promoted by dysregulated Hippo pathway signaling, as demonstrated by increased YAP1 expression and decreased MST activity. We hypothesized that ablating MST/Hippo signaling in a genetically engineered mouse model (GEMM) of aRMS would accelerate tumorigenesis. To this end, MST1/2-floxed (Stk3F/F;Stk4F/F ) mice were crossed with a previously established aRMS GEMM driven by conditional expression of Pax3:Foxo1 from the endogenous Pax3 locus and conditional loss of Cdkn2a in Myf6 (myogenic factor 6)-expressing cells. Compared with Pax3PF/PF;Cdkn2aF/F;Myf6ICN/+ controls, Stk3F/F;Stk4F/F;Pax3PF/PF;Cdkn2aF/F;Myf6ICN/+ animals displayed accelerated tumorigenesis (P < 0.0001) and increased tumor penetrance (88% vs. 27%). GEMM tumors were histologically consistent with aRMS. GEMM tumor-derived cell lines showed increased proliferation and invasion and decreased senescence and myogenic differentiation. These data suggest that loss of MST/Hippo signaling acts with Pax3:Foxo1 expression and Cdkn2a loss to promote tumorigenesis. The rapid onset and increased penetrance of tumorigenesis in this model provide a powerful tool for interrogating aRMS biology and screening novel therapeutics.Significance: A novel mouse model sheds light on the critical role of Hippo/MST downregulation in PAX3-FOXO1-positive rhabdomyosarcoma tumorigenesis. Cancer Res; 78(19); 5513-20. ©2018 AACR.
Collapse
Affiliation(s)
- Kristianne M Oristian
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina.,Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Lisa E S Crose
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | | | - Rex C Bentley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Yi-Tzu Lin
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Nerissa Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - David G Kirsch
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina.,Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Corinne M Linardic
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina. .,Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
44
|
Sittewelle M, Monsoro-Burq AH. AKT signaling displays multifaceted functions in neural crest development. Dev Biol 2018; 444 Suppl 1:S144-S155. [PMID: 29859890 DOI: 10.1016/j.ydbio.2018.05.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/23/2022]
Abstract
AKT signaling is an essential intracellular pathway controlling cell homeostasis, cell proliferation and survival, as well as cell migration and differentiation in adults. Alterations impacting the AKT pathway are involved in many pathological conditions in human disease. Similarly, during development, multiple transmembrane molecules, such as FGF receptors, PDGF receptors or integrins, activate AKT to control embryonic cell proliferation, migration, differentiation, and also cell fate decisions. While many studies in mouse embryos have clearly implicated AKT signaling in the differentiation of several neural crest derivatives, information on AKT functions during the earliest steps of neural crest development had remained relatively scarce until recently. However, recent studies on known and novel regulators of AKT signaling demonstrate that this pathway plays critical roles throughout the development of neural crest progenitors. Non-mammalian models such as fish and frog embryos have been instrumental to our understanding of AKT functions in neural crest development, both in neural crest progenitors and in the neighboring tissues. This review combines current knowledge acquired from all these different vertebrate animal models to describe the various roles of AKT signaling related to neural crest development in vivo. We first describe the importance of AKT signaling in patterning the tissues involved in neural crest induction, namely the dorsal mesoderm and the ectoderm. We then focus on AKT signaling functions in neural crest migration and differentiation.
Collapse
Affiliation(s)
- Méghane Sittewelle
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France
| | - Anne H Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Universitaire de France, F-75005 Paris, France.
| |
Collapse
|
45
|
Xiang L, Yu H, Zhang X, Wang B, Yuan Y, Zhang Q, Ye R, Gong P, Wu Y. The versatile hippo pathway in oral-maxillofacial development and bone remodeling. Dev Biol 2018; 440:53-63. [PMID: 29792855 DOI: 10.1016/j.ydbio.2018.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/20/2018] [Accepted: 05/20/2018] [Indexed: 02/05/2023]
Abstract
The Hippo signaling pathway is implicated in key aspects of cell proliferation, control of organ size, stem cell functions and tumor suppression. Its functions are primarily mediated either through direct effects on transcription factors to influence target gene expression or through crosstalk with other signaling pathways that regulate multiple physiological activities. Studies are revealing Hippo pathway involvement in diverse functions including renewal of intestinal epithelium, promotion of myocardial cell proliferation, cancer suppression, etc. In this review we discuss Hippo pathway signaling in oral-maxillofacial development and bone remodeling under normal and pathological conditions and highlight promising future research directions.
Collapse
Affiliation(s)
- Lin Xiang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyuan Zhang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Wang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying Yuan
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qin Zhang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Ye
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Yingying Wu
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
46
|
Fu V, Plouffe SW, Guan KL. The Hippo pathway in organ development, homeostasis, and regeneration. Curr Opin Cell Biol 2018; 49:99-107. [PMID: 29316535 DOI: 10.1016/j.ceb.2017.12.012] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/05/2017] [Accepted: 12/16/2017] [Indexed: 12/12/2022]
Abstract
The Hippo pathway is a universal governor of organ size, tissue homeostasis, and regeneration. A growing body of work has advanced our understanding of Hippo pathway regulation of cell proliferation, differentiation, and spatial patterning not only in organ development but also upon injury-induced regeneration. The pathway's central role in stem cell biology thus implicates its potential for therapeutic manipulation in mammalian organ regeneration. In this review, we survey recent literature linking the Hippo pathway to the development, homeostasis, and regeneration of various organs, including Hippo-independent roles for YAP, defined here as YAP functions that are not regulated by the Hippo pathway kinases LATS1/2.
Collapse
Affiliation(s)
- Vivian Fu
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, United States
| | - Steven W Plouffe
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, United States
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
47
|
Hippo pathway coactivators Yap and Taz are required to coordinate mammalian liver regeneration. Exp Mol Med 2018; 50:e423. [PMID: 29303509 PMCID: PMC5992983 DOI: 10.1038/emm.2017.205] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 05/11/2017] [Indexed: 12/15/2022] Open
Abstract
The mammalian liver has a remarkable capacity for repair following injury. Removal of up to two-third of liver mass results in a series of events that include extracellular matrix remodeling, coordinated hepatic cell cycle re-entry, restoration of liver mass and tissue remodeling to return the damaged liver to its normal state. Although there has been considerable advancement of our knowledge concerning the regenerative capacity of the mammalian liver, many outstanding questions remaining, such as: how does the regenerating liver stop proliferating when appropriate mass is restored and how do these mechanisms relate to normal regulation of organ size during development? Hippo pathway has been proposed to be central in mediating both events: organ size control during development and following regeneration. In this report, we examined the role of Yap and Taz, key components of the Hippo pathway in liver organ size regulation, both in the context of development and homeostasis. Our studies reveal that contrary to the current paradigms that Yap/Taz are not required for developmental regulation of liver size but are required for proper liver regeneration. In livers depleted of Yap and Taz, liver mass is elevated in neonates and adults. However, Yap/Taz-depleted livers exhibit profound defects in liver regeneration, including an inability to restore liver mass and to properly coordinate cell cycle entry. Taken together, our results highlight requirements for the Hippo pathway during liver regeneration and indicate that there are additional pathways that cooperate with Hippo signaling to control liver size during development and in the adult.
Collapse
|
48
|
Okuno H, Renault Mihara F, Ohta S, Fukuda K, Kurosawa K, Akamatsu W, Sanosaka T, Kohyama J, Hayashi K, Nakajima K, Takahashi T, Wysocka J, Kosaki K, Okano H. CHARGE syndrome modeling using patient-iPSCs reveals defective migration of neural crest cells harboring CHD7 mutations. eLife 2017; 6. [PMID: 29179815 PMCID: PMC5705211 DOI: 10.7554/elife.21114] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/22/2017] [Indexed: 12/11/2022] Open
Abstract
CHARGE syndrome is caused by heterozygous mutations in the chromatin remodeler, CHD7, and is characterized by a set of malformations that, on clinical grounds, were historically postulated to arise from defects in neural crest formation during embryogenesis. To better delineate neural crest defects in CHARGE syndrome, we generated induced pluripotent stem cells (iPSCs) from two patients with typical syndrome manifestations, and characterized neural crest cells differentiated in vitro from these iPSCs (iPSC-NCCs). We found that expression of genes associated with cell migration was altered in CHARGE iPSC-NCCs compared to control iPSC-NCCs. Consistently, CHARGE iPSC-NCCs showed defective delamination, migration and motility in vitro, and their transplantation in ovo revealed overall defective migratory activity in the chick embryo. These results support the historical inference that CHARGE syndrome patients exhibit defects in neural crest migration, and provide the first successful application of patient-derived iPSCs in modeling craniofacial disorders. CHARGE syndrome is a disease in which organs including the heart, eyes and ears may not develop properly. The cells that form the tissues affected by CHARGE syndrome develop in embryos from precursor cells called neural crest cells. Individuals with CHARGE syndrome also have mutations in a gene called CHD7. However, it is difficult to examine how CHD7 mutations affect neural crest cells in embryos. In recent years, cell reprogramming techniques have made it possible to create induced pluripotent stem cells (iPSCs) from the specialized somatic cells found in the human body. These iPSCs can be developed into many different cell types, including neural crest cells. Okuno et al. created iPSCs from the skin cells of people with CHARGE syndrome, developed these cells into neural crest cells, and compared them with neural crest cells that were developed from the skin cells of people without CHARGE syndrome. The neural crest cells developed from people with CHARGE syndrome showed multiple abnormalities. For example, they were not able to move around correctly. This is an important observation because neural crest cells must move through tissues to form the various organs affected by CHARGE syndrome. Okuno et al. also observed changes in the activity of many genes other than CHD7 in the neural crest cells developed from CHARGE patients. Further research is now needed to find out which genes are the most important for restoring the normal activity of neural crest cells.
Collapse
Affiliation(s)
- Hironobu Okuno
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | | | - Shigeki Ohta
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Kimiko Fukuda
- Department of Biological Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Kenji Kurosawa
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Wado Akamatsu
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Tsukasa Sanosaka
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Jun Kohyama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Kanehiro Hayashi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Takao Takahashi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Joanna Wysocka
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, United States.,Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, United States
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
Davis JA, Saunders SJ, Mann M, Backofen R. Combinatorial ensemble miRNA target prediction of co-regulation networks with non-prediction data. Nucleic Acids Res 2017; 45:8745-8757. [PMID: 28911111 PMCID: PMC5587804 DOI: 10.1093/nar/gkx605] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are key regulators of cell-fate decisions in development and disease with a vast array of target interactions that can be investigated using computational approaches. For this study, we developed metaMIR, a combinatorial approach to identify miRNAs that co-regulate identified subsets of genes from a user-supplied list. We based metaMIR predictions on an improved dataset of human miRNA–target interactions, compiled using a machine-learning-based meta-analysis of established algorithms. Simultaneously, the inverse dataset of negative interactions not likely to occur was extracted to increase classifier performance, as measured using an expansive set of experimentally validated interactions from a variety of sources. In a second differential mode, candidate miRNAs are predicted by indicating genes to be targeted and others to be avoided to potentially increase specificity of results. As an example, we investigate the neural crest, a transient structure in vertebrate development where miRNAs play a pivotal role. Patterns of metaMIR-predicted miRNA regulation alone partially recapitulated functional relationships among genes, and separate differential analysis revealed miRNA candidates that would downregulate components implicated in cancer progression while not targeting tumour suppressors. Such an approach could aid in therapeutic application of miRNAs to reduce unintended effects. The utility is available at http://rna.informatik.uni-freiburg.de/metaMIR/.
Collapse
Affiliation(s)
- Jason A Davis
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Sita J Saunders
- Bioinformatics Group, Department of Computer Science, Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 106, 79110 Freiburg, Germany
| | - Martin Mann
- Bioinformatics Group, Department of Computer Science, Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 106, 79110 Freiburg, Germany
| | - Rolf Backofen
- Bioinformatics Group, Department of Computer Science, Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 106, 79110 Freiburg, Germany.,ZBSA Centre for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstr. 49, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Cluster of Excellence, Albert-Ludwigs-University Freiburg, Germany.,Centre for non-coding RNA in Technology and Health, University of Copenhagen, Gr⊘nnegårdsvej 3, DK-1870 Frederiksberg C, Denmark
| |
Collapse
|
50
|
Jeong MG, Song H, Shin JH, Jeong H, Kim HK, Hwang ES. Transcriptional coactivator with PDZ-binding motif is required to sustain testicular function on aging. Aging Cell 2017; 16:1035-1042. [PMID: 28613007 PMCID: PMC5595677 DOI: 10.1111/acel.12631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2017] [Indexed: 12/15/2022] Open
Abstract
Transcriptional coactivator with PDZ‐binding motif (TAZ) directly interacts with transcription factors and regulates their transcriptional activity. Extensive functional studies have shown that TAZ plays critical regulatory roles in stem cell proliferation, differentiation, and survival and also modulates the development of organs such as the lung, kidney, heart, and bone. Despite the importance of TAZ in stem cell maintenance, TAZ function has not yet been evaluated in spermatogenic stem cells of the male reproductive system. Here, we investigated the expression and functions of TAZ in mouse testis. TAZ was expressed in spermatogenic stem cells; however, its deficiency caused significant structural abnormalities, including atrophied tubules, widened interstitial space, and abnormal Leydig cell expansion, thereby resulting in lowered sperm counts and impaired fertility. Furthermore, TAZ deficiency increased the level of apoptosis and senescence in spermatogenic cells and Leydig cells upon aging. The expression of senescence‐associated β‐galactosidase (SA‐βgal), secretory phenotypes, and cyclin‐dependent kinase inhibitors (p16, p19, and p21) significantly increased in the absence of TAZ. TAZ downregulation in testicular cells further increased SA‐βgal and p21 expression induced by oxidative stress, whereas TAZ overexpression decreased p21 induction and prevented senescence. Mechanistic studies showed that TAZ suppressed DNA‐binding activity of p53 through a direct interaction and thus attenuated p53‐induced p21 gene transcription. Our results suggested that TAZ may suppress apoptosis and premature senescence in spermatogenic cells by inhibiting the p53‐p21 signaling pathway, thus playing important roles in the maintenance and control of reproductive function.
Collapse
Affiliation(s)
- Mi Gyeong Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Hyuna Song
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Ji Hyun Shin
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Hana Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Hyo Kyeong Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences; Ewha Womans University; Seoul 03760 Korea
| |
Collapse
|