1
|
Zhao P, Zhao Y, Ma Y, Liang C, Yuan Q, Gao Y, Liu X, Zhu X, Hao X, Liang G, Fan H, Wang D. Gestational and lactational exposure to DEHP triggers ACSL4/TFR-mediated hippocampal neuronal ferroptosis via YAP activation: Implication for the neurocognitive disorders in male offspring. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138081. [PMID: 40187248 DOI: 10.1016/j.jhazmat.2025.138081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is one of the most extensively used phthalate and poses a public health concern. Perinatal exposure to DEHP has been shown to cause neurodevelopmental abnormalities and neurobehavioral disorders in offspring. However, the precise molecular mechanism has not yet been fully elucidated. In this study, pregnant C57BL/6 mice were exposed to DEHP from gestation to weaning. By RNA sequencing and animal experiments, ferroptosis has been identified as the key pathologic process contributing to DEHP-induced hippocampal injury in adult male offspring. In vitro results also showed that Ferrostatin-1 (Fer-1) effectively ameliorated Mono-(2-ethylhexyl) phthalate (MEHP) -induced cell survival via the inhibiting ferroptosis in HT22 cells. Consistently, we found that the expression of ACSL4 and TFR was significantly up-regulated in offspring hippocampi and MEHP-exposed HT22 neurons. However, silencing ACSL4 or knockdown TFR relieved MEHP-induced generation of lipid ROS and cellular iron accumulation, thereby blocking ferroptosis. Mechanistically, ACSL4/TFR-mediated ferroptosis seemed to be a Yes-associated protein (YAP) dependent via TEA domain transcription factor 4 in HT22 neurons. Importantly, treatment with Fer-1, rosiglitazone, and Deferoxamine effectively rescued DEHP-evoked cognitive decline in adult male offspring. Our findings certified that gestational and lactational exposure to DEHP provoked ACSL4/TFR-mediated hippocampal neuronal ferroptosis via YAP activation.
Collapse
Affiliation(s)
- Pu Zhao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Yuhang Zhao
- Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yilu Ma
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Chen Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Quan Yuan
- Henan Province Rongkang Hospital, Luoyang, China
| | - Yufei Gao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoli Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoying Zhu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xueqin Hao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China.
| | - Dongmei Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
2
|
Ahmad ST, Li Y, Garcia-Lopez J, Gudenas BL, Hadley J, Paul L, Wu SC, Refaat A, Kojic M, Batts M, Soliman T, Pitre A, Arnskötter F, Zindy F, Jones A, Twarog NR, Mayasundari A, Bianski B, Tinkle C, Shirinifard A, Janke L, Lu M, Lewis SA, Onar-Thomas A, Pfister SM, Gajjar A, Baker SJ, Roussel MF, Rankovic Z, Robinson GW, Orr BA, Wainwright B, Shelat AA, Waszak SM, Kutscher LM, Lin H, Northcott PA. Genetic modeling of ELP1-associated Sonic hedgehog medulloblastoma identifies MDM2 as a selective therapeutic target. Cancer Cell 2025:S1535-6108(25)00173-4. [PMID: 40378836 DOI: 10.1016/j.ccell.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 02/23/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025]
Abstract
Germline loss-of-function (LOF) variants in Elongator acetyltransferase complex subunit 1 (ELP1) are the most prevalent predisposing genetic events in childhood medulloblastoma (MB), accounting for ∼30% of the Sonic hedgehog (SHH) 3 subtype. The mechanism(s) by which germline ELP1 deficiency provokes SHH-MB pathogenesis remain unknown. Genetically engineered mice mimicking heterozygous Elp1 LOF (Elp1HET) seen in affected germline carriers exhibit hallmark features of premalignancy in cerebellar granule neuron progenitors (GNPs), including increased DNA replication stress, genomic instability, accelerated cell cycle, and stalled differentiation. Orthotopic transplantation of Elp1HET GNPs harboring somatic Ptch1 inactivation yields SHH-MB-like tumors with compromised p53 signaling, providing a plausible explanation for the exclusivity of ELP1-associated MBs in the SHH-3 subtype. Preclinical treatment of ELP1-mutant patient-derived xenografts with an FDA-approved MDM2 inhibitor reactivates p53-dependent apoptosis and extends survival. Our findings functionally substantiate the role of ELP1 deficiency in SHH-MB predisposition and nominate therapeutics targeting MDM2 as a rational treatment option.
Collapse
Affiliation(s)
- Shiekh Tanveer Ahmad
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yiran Li
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jesus Garcia-Lopez
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brian L Gudenas
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer Hadley
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Leena Paul
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephanie C Wu
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alaa Refaat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marija Kojic
- Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Melissa Batts
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Taha Soliman
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Aaron Pitre
- Cell and Tissue Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Frederik Arnskötter
- Hopp Children's Cancer Center Heidelberg (KiTZ), JRG Developmental Origins of Pediatric Cancers, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Frederique Zindy
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alun Jones
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Nathaniel R Twarog
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anand Mayasundari
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brandon Bianski
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher Tinkle
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Laura Janke
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Meifen Lu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sara A Lewis
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Division Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Amar Gajjar
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Suzanne J Baker
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Martine F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Giles W Robinson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brandon Wainwright
- Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sebastian M Waszak
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland; Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Lena M Kutscher
- Hopp Children's Cancer Center Heidelberg (KiTZ), JRG Developmental Origins of Pediatric Cancers, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hong Lin
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Paul A Northcott
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
3
|
Kim YK, Ramalho-Santos M. 20 years of stemness: From stem cells to hypertranscription and back. Stem Cell Reports 2025; 20:102406. [PMID: 39919752 PMCID: PMC11960510 DOI: 10.1016/j.stemcr.2025.102406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
Transcriptional profiling of stem cells came of age at the beginning of the century with the use of microarrays to analyze cell populations in bulk. Since then, stem cell transcriptomics has become increasingly sophisticated, notably with the recent widespread use of single-cell RNA sequencing. Here, we provide a perspective on how an early signature of genes upregulated in embryonic and adult stem cells, identified using microarrays over 20 years ago, serendipitously led to the recent discovery that stem/progenitor cells across organs are in a state of hypertranscription, a global elevation of the transcriptome. Looking back, we find that the 2002 stemness signature is a robust marker of stem cell hypertranscription, even though it was developed well before it was known what hypertranscription meant or how to detect it. We anticipate that studies of stem cell hypertranscription will be rich in novel insights in physiological and disease contexts for years to come.
Collapse
Affiliation(s)
- Yun-Kyo Kim
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto ON M5G 1X5, Canada.
| | - Miguel Ramalho-Santos
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto ON M5T 3L9, Canada; Department of Molecular Genetics, University of Toronto, Toronto ON M5G 1X5, Canada.
| |
Collapse
|
4
|
Guo D, Yao B, Shao W, Zuo J, Chang Z, Shi J, Hu N, Bao S, Chen M, Fan X, Li X. The Critical Role of YAP/BMP/ID1 Axis on Simulated Microgravity-Induced Neural Tube Defects in Human Brain Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410188. [PMID: 39656892 PMCID: PMC11792043 DOI: 10.1002/advs.202410188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/05/2024] [Indexed: 12/17/2024]
Abstract
Integrated biochemical and biophysical signals regulate embryonic development. Correct neural tube formation is critical for the development of central nervous system. However, the role of microgravity in neurodevelopment and its underlying molecular mechanisms remain unclear. In this study, the effects of stimulated microgravity (SMG) on the development of human brain organoids are investigated. SMG impairs N-cadherin-based adherens junction formation, leading to neural tube defects associated with dysregulated self-renewal capacity and neuroepithelial disorganization in human brain organoids. Bulk gene expression analyses reveal that SMG alters Hippo and BMP signaling in brain organoids. The neuropathological deficits in SMG-treated organoids can be rescued by regulating YAP/BMP/ID1 axis. Furthermore, sing-cell RNA sequencing data show that SMG results in perturbations in the number and function of neural stem and progenitor cell subpopulations. One of these subpopulations senses SMG cues and transmits BMP signals to the subpopulation responsible for tube morphogenesis, ultimately affecting the proliferating cell population. Finally, SMG intervention leads to persistent neurologic damage even after returning to normal gravity conditions. Collectively, this study reveals molecular and cellular abnormalities associated with SMG during human brain development, providing opportunities for countermeasures to maintain normal neurodevelopment in space.
Collapse
Affiliation(s)
- Di Guo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Wen‐Wei Shao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jia‐Chen Zuo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Zhe‐Han Chang
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jian‐Xin Shi
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Nan Hu
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Shuang‐Qing Bao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Meng‐Meng Chen
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiu Fan
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiao‐Hong Li
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| |
Collapse
|
5
|
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
|
6
|
Li C, Chen W, Cui Y, Zhang D, Yuan Q, Yu X, He Z. Essential Regulation of YAP1 in Fate Determinations of Spermatogonial Stem Cells and Male Fertility by Interacting with RAD21 and Targeting NEDD4 in Humans and Mice. RESEARCH (WASHINGTON, D.C.) 2024; 7:0544. [PMID: 39659446 PMCID: PMC11628678 DOI: 10.34133/research.0544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
Spermatogenesis is a sophisticated biological process by which spermatogonial stem cells (SSCs) undergo self-renewal and differentiation into spermatozoa. Molecular mechanisms underlying fate determinations of human SSCs by key genes and signaling pathways remain elusive. Here, we report for the first time that Yes1-associated transcriptional regulator (YAP1) is required for fate determinations of SSCs and male fertility by interacting with RAD21 and targeting NEDD4 in humans and mice. YAP1 was mainly located at cell nuclei of human SSCs. YAP1 silencing resulted in the decreases in proliferation and DNA synthesis as well as an enhancement in apoptosis of human SSCs both in vivo and in vitro. RNA sequencing and real-time polymerase chain reaction assays identified NEDD4 as a target of YAP1, and NEDD4 knockdown inhibited the proliferation of human SSCs and increased their apoptosis. Furthermore, YAP1 interacted with RAD21 to regulate NEDD4 transcription in human SSCs. Importantly, YAP1 abnormalities were found to be associated with non-obstructive azoospermia (NOA) as manifested as lower expression level of YAP1 in testicular tissues of NOA patients and YAP1 single-nucleotide variants (SNVs) in 777 NOA patients. Finally, Yap1 germline conditional knockout (cKO) mice assumed mitotic arrest, low sperm count, and motility. Collectively, these results highlight a critical role of YAP1 in determining the fate determinations of human SSCs and male infertility through the YAP1/RAD21/NEDD4 pathway. This study provides new insights into the genetic regulatory mechanisms underlying human spermatogenesis and the pathogenesis of NOA, and it offers new targets for gene therapy of male infertility.
Collapse
Affiliation(s)
- Chunyun Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Wei Chen
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Dong Zhang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Qingqing Yuan
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200135, China
| | - Xing Yu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| |
Collapse
|
7
|
Kim YK, Collignon E, Martin SB, Ramalho-Santos M. Hypertranscription: the invisible hand in stem cell biology. Trends Genet 2024; 40:1032-1046. [PMID: 39271397 DOI: 10.1016/j.tig.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024]
Abstract
Stem cells are the fundamental drivers of growth during development and adult organ homeostasis. The properties that define stem cells - self-renewal and differentiation - are highly biosynthetically demanding. In order to fuel this demand, stem and progenitor cells engage in hypertranscription, a global amplification of the transcriptome. While standard normalization methods in transcriptomics typically mask hypertranscription, new approaches are beginning to reveal a remarkable range in global transcriptional output in stem and progenitor cells. We discuss technological advancements to probe global transcriptional shifts, review recent findings that contribute to defining hallmarks of stem cell hypertranscription, and propose future directions in this field.
Collapse
Affiliation(s)
- Yun-Kyo Kim
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada.
| | - Evelyne Collignon
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Centre (U-CRC) and Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - S Bryn Martin
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada.
| | - Miguel Ramalho-Santos
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada.
| |
Collapse
|
8
|
Li XH, Guo D, Chen LQ, Chang ZH, Shi JX, Hu N, Chen C, Zhang XW, Bao SQ, Chen MM, Ming D. Low-intensity ultrasound ameliorates brain organoid integration and rescues microcephaly deficits. Brain 2024; 147:3817-3833. [PMID: 38739753 DOI: 10.1093/brain/awae150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Human brain organoids represent a remarkable platform for modelling neurological disorders and a promising brain repair approach. However, the effects of physical stimulation on their development and integration remain unclear. Here, we report that low-intensity ultrasound significantly increases neural progenitor cell proliferation and neuronal maturation in cortical organoids. Histological assays and single-cell gene expression analyses revealed that low-intensity ultrasound improves the neural development in cortical organoids. Following organoid grafts transplantation into the injured somatosensory cortices of adult mice, longitudinal electrophysiological recordings and histological assays revealed that ultrasound-treated organoid grafts undergo advanced maturation. They also exhibit enhanced pain-related gamma-band activity and more disseminated projections into the host brain than the untreated groups. Finally, low-intensity ultrasound ameliorates neuropathological deficits in a microcephaly brain organoid model. Hence, low-intensity ultrasound stimulation advances the development and integration of brain organoids, providing a strategy for treating neurodevelopmental disorders and repairing cortical damage.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Di Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Li-Qun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Jian-Xin Shi
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Shuang-Qing Bao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Meng-Meng Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
9
|
Wang Y, Yu FX. Angiomotin family proteins in the Hippo signaling pathway. Bioessays 2024; 46:e2400076. [PMID: 38760875 DOI: 10.1002/bies.202400076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
The Motin family proteins (Motins) are a class of scaffolding proteins consisting of Angiomotin (AMOT), AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). Motins play a pivotal role in angiogenesis, tumorigenesis, and neurogenesis by modulating multiple cellular signaling pathways. Recent findings indicate that Motins are components of the Hippo pathway, a signaling cascade involved in development and cancer. This review discusses how Motins are integrated into the Hippo signaling network, as either upstream regulators or downstream effectors, to modulate cell proliferation and migration. The repression of YAP/TAZ by Motins contributes to growth inhibition, whereas subcellular localization of Motins and their interactions with actin fibers are critical in regulating cell migration. The net effect of Motins on cell proliferation and migration may contribute to their diverse biological functions.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Andrade P, Alves JM, Pereira P, Rubin CJ, Silva E, Sprehn CG, Enbody E, Afonso S, Faria R, Zhang Y, Bonino N, Duckworth JA, Garreau H, Letnic M, Strive T, Thulin CG, Queney G, Villafuerte R, Jiggins FM, Ferrand N, Andersson L, Carneiro M. Selection against domestication alleles in introduced rabbit populations. Nat Ecol Evol 2024; 8:1543-1555. [PMID: 38907020 DOI: 10.1038/s41559-024-02443-3] [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: 10/16/2023] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Humans have moved domestic animals around the globe for thousands of years. These have occasionally established feral populations in nature, often with devastating ecological consequences. To understand how natural selection shapes re-adaptation into the wild, we investigated one of the most successful colonizers in history, the European rabbit. By sequencing the genomes of 297 rabbits across three continents, we show that introduced populations exhibit a mixed wild-domestic ancestry. We show that alleles that increased in frequency during domestication were preferentially selected against in novel natural environments. Interestingly, causative mutations for common domestication traits sometimes segregate at considerable frequencies if associated with less drastic phenotypes (for example, coat colour dilution), whereas mutations that are probably strongly maladaptive in nature are absent. Whereas natural selection largely targeted different genomic regions in each introduced population, some of the strongest signals of parallelism overlap genes associated with neuronal or brain function. This limited parallelism is probably explained by extensive standing genetic variation resulting from domestication together with the complex mixed ancestry of introduced populations. Our findings shed light on the selective and molecular mechanisms that enable domestic animals to re-adapt to the wild and provide important insights for the mitigation and management of invasive populations.
Collapse
Affiliation(s)
- Pedro Andrade
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal.
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal.
| | - Joel M Alves
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
- Palaeogenomics and Bio-Archaeology Research Network Research Laboratory for Archaeology and History of Art, University of Oxford, Oxford, UK
| | - Paulo Pereira
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Carl-Johan Rubin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Institute of Marine Research, Bergen, Norway
| | - Eugénio Silva
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - C Grace Sprehn
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Erik Enbody
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA, USA
| | - Sandra Afonso
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
| | - Rui Faria
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
| | - Yexin Zhang
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Never Bonino
- Estación Experimental Bariloche, Instituto Nacional de Tecnología Agropecuaria, Casilla de Correo Bariloche, Argentina
| | - Janine A Duckworth
- Wildlife Ecology and Management Group, Manaaki Whenua - Landcare Research, Lincoln, New Zealand
- Invasive Animals Cooperative Research Centre, University of Canberra, Bruce, Australian Capital Territory, Australia
| | - Hervé Garreau
- GenPhySE, Université de Toulouse, Castanet-Tolosan, France
| | - Mike Letnic
- Centre for Ecosystem Science, School of BEES, University of New South Wales, Sydney, New South Wales, Australia
- Evolution and Ecology Research Centre, School of BEES, University of New South Wales, Sydney, New South Wales, Australia
| | - Tanja Strive
- Centre for Invasive Species Solutions, University of Canberra, Bruce, Australian Capital Territory, Australia
- Commonwealth Scientific and Industrial Research Organisation, Canberra, Australian Capital Territory, Australia
| | - Carl-Gustaf Thulin
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Guillaume Queney
- ANTAGENE, Wildlife Genetics Laboratory, La Tour de Salvagny, France
| | | | | | - Nuno Ferrand
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Department of Zoology, Faculty of Sciences, University of Johannesburg, Auckland Park, South Africa
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA.
| | - Miguel Carneiro
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal.
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal.
| |
Collapse
|
11
|
Kiang KM, Ahad L, Zhong X, Lu QR. Biomolecular condensates: hubs of Hippo-YAP/TAZ signaling in cancer. Trends Cell Biol 2024; 34:566-577. [PMID: 38806345 DOI: 10.1016/j.tcb.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
Biomolecular condensates, the membraneless cellular compartments formed by liquid-liquid phase separation (LLPS), represent an important mechanism for physiological and tumorigenic processes. Recent studies have advanced our understanding of how these condensates formed in the cytoplasm or nucleus regulate Hippo signaling, a central player in organogenesis and tumorigenesis. Here, we review recent findings on the dynamic formation and function of biomolecular condensates in regulating the Hippo-yes-associated protein (YAP)/transcription coactivator with PDZ-binding motif (TAZ) signaling pathway under physiological and pathological processes. We further discuss how the nuclear condensates of YAP- or TAZ-fusion oncoproteins compartmentalize crucial transcriptional co-activators and alter chromatin architecture to promote oncogenic programs. Finally, we highlight key questions regarding how these findings may pave the way for novel therapeutics to target cancer.
Collapse
Affiliation(s)
- Karrie M Kiang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Leena Ahad
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
12
|
Zhao Y, Sun B, Fu X, Zuo Z, Qin H, Yao K. YAP in development and disease: Navigating the regulatory landscape from retina to brain. Biomed Pharmacother 2024; 175:116703. [PMID: 38713948 DOI: 10.1016/j.biopha.2024.116703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
The distinctive role of Yes-associated protein (YAP) in the nervous system has attracted widespread attention. This comprehensive review strategically uses the retina as a vantage point, embarking on an extensive exploration of YAP's multifaceted impact from the retina to the brain in development and pathology. Initially, we explore the crucial roles of YAP in embryonic and cerebral development. Our focus then shifts to retinal development, examining in detail YAP's regulatory influence on the development of retinal pigment epithelium (RPE) and retinal progenitor cells (RPCs), and its significant effects on the hierarchical structure and functionality of the retina. We also investigate the essential contributions of YAP in maintaining retinal homeostasis, highlighting its precise regulation of retinal cell proliferation and survival. In terms of retinal-related diseases, we explore the epigenetic connections and pathophysiological regulation of YAP in diabetic retinopathy (DR), glaucoma, and proliferative vitreoretinopathy (PVR). Lastly, we broaden our exploration from the retina to the brain, emphasizing the research paradigm of "retina: a window to the brain." Special focus is given to the emerging studies on YAP in brain disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD), underlining its potential therapeutic value in neurodegenerative disorders and neuroinflammation.
Collapse
Affiliation(s)
- Yaqin Zhao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Bin Sun
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xuefei Fu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhuan Zuo
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
13
|
Wang H, Ye M, Jin X. Role of angiomotin family members in human diseases (Review). Exp Ther Med 2024; 27:258. [PMID: 38766307 PMCID: PMC11099588 DOI: 10.3892/etm.2024.12546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/23/2023] [Indexed: 05/22/2024] Open
Abstract
Angiomotin (Amot) family members, including Amot, Amot-like protein 1 (Amotl1) and Amot-like protein 2 (Amotl2), have been found to interact with angiostatins. In addition, Amot family members are involved in various physiological and pathological functions such as embryonic development, angiogenesis and tumorigenesis. Some studies have also demonstrated its regulation in signaling pathways such as the Hippo signaling pathway, AMPK signaling pathway and mTOR signaling pathways. Amot family members play an important role in neural stem cell differentiation, dendritic formation and synaptic maturation. In addition, an increasing number of studies have focused on their function in promoting and/or suppressing cancer, but the underlying mechanisms remain to be elucidated. The present review integrated relevant studies on upstream regulation and downstream signals of Amot family members, as well as the latest progress in physiological and pathological functions and clinical applications, hoping to offer important ideas for further research.
Collapse
Affiliation(s)
- Haoyun Wang
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
14
|
Ryu HJ, Kim C, Jang H, Kim SI, Shin SJ, Chung KY, Torres-Cabala C, Kim SK. Nuclear Localization of Yes-Associated Protein Is Associated With Tumor Progression in Cutaneous Melanoma. J Transl Med 2024; 104:102048. [PMID: 38490470 DOI: 10.1016/j.labinv.2024.102048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024] Open
Abstract
Yes-associated protein (YAP), an effector molecule of the Hippo signaling pathway, is expressed at high levels in cutaneous melanoma. However, the role of YAP in melanoma progression according to cellular localization is poorly understood. Tissues from 140 patients with invasive melanoma were evaluated by immunohistochemistry. Flow cytometry, western blotting, viability assays, wound healing assays, verteporfin treatment, and xenograft assays were conducted using melanoma cell lines B16F1 and B16F10 subjected to YapS127A transfection and siYap knockdown. Nuclear YAP localization was identified in 63 tumors (45.0%) and was more frequent than cytoplasmic YAP in acral lentiginous and nodular subtypes (P = .007). Compared with cytoplasmic YAP melanomas, melanomas with nuclear YAP had higher mitotic activity (P = .016), deeper invasion (P < .001), and more frequently metastasized to lymph nodes (P < .001) and distant organs (P < .001). Patients with nuclear YAP melanomas had poorer disease-free survival (P < .001) and overall survival (P < .001). Nuclear YAP was an independent risk factor for distant metastasis (hazard ratio: 3.206; 95% CI, 1.032-9.961; P = .044). Proliferative ability was decreased in siYapB16F1 (P < .001) and siYapB16F10 (P = .001) cells and increased in YapS127AB16F1 (P = .003) and YapS127AB16F10 (P = .002) cells. Cell cycle analysis demonstrated relative G1 retention in siYapB16F1 (P < .001) and siYapB16F10 (P < .001) cells and S retention in YapS127AB16F1 cells (P = .008). Wound healing assays showed that Yap knockdown inhibited cell invasion (siYapB16F1, P = .001; siYapB16F10, P < .001), whereas nuclear YAP promoted it (YapS127AB16F, P < .001; YapS127AB16F1, P = .017). Verteporfin, a direct YAP inhibitor, reduced cellular proliferation in B16F1 (P = .003) and B16F10 (P < .001) cells. Proliferative effects of nuclear YAP were confirmed in xenograft mice (P < .001). In conclusion, nuclear YAP in human melanomas showed subtype specificity and correlated with proliferative activity and proinvasiveness. It is expected that YAP becomes a useful prognostic marker, and its inhibition may be a potential therapy for melanoma patients.
Collapse
Affiliation(s)
- Hyang Joo Ryu
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chayeon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyenguk Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Il Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Joon Shin
- Department of Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Yang Chung
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Carlos Torres-Cabala
- Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas.
| | - Sang Kyum Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
15
|
Zhong Z, Jiao Z, Yu FX. The Hippo signaling pathway in development and regeneration. Cell Rep 2024; 43:113926. [PMID: 38457338 DOI: 10.1016/j.celrep.2024.113926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/10/2024] Open
Abstract
The Hippo signaling pathway is a central growth control mechanism in multicellular organisms. By integrating diverse mechanical, biochemical, and stress cues, the Hippo pathway orchestrates proliferation, survival, differentiation, and mechanics of cells, which in turn regulate organ development, homeostasis, and regeneration. A deep understanding of the regulation and function of the Hippo pathway therefore holds great promise for developing novel therapeutics in regenerative medicine. Here, we provide updates on the molecular organization of the mammalian Hippo signaling network, review the regulatory signals and functional outputs of the pathway, and discuss the roles of Hippo signaling in development and regeneration.
Collapse
Affiliation(s)
- Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhihan Jiao
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
16
|
Li MY, Yang XL, Chung CC, Lai YJ, Tsai JC, Kuo YL, Yu JY, Wang TW. TRIP6 promotes neural stem cell maintenance through YAP-mediated Sonic Hedgehog activation. FASEB J 2024; 38:e23501. [PMID: 38411462 DOI: 10.1096/fj.202301805rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
In the adult mammalian brain, new neurons are continuously generated from neural stem cells (NSCs) in the subventricular zone (SVZ)-olfactory bulb (OB) pathway. YAP, a transcriptional co-activator of the Hippo pathway, promotes cell proliferation and inhibits differentiation in embryonic neural progenitors. However, the role of YAP in postnatal NSCs remains unclear. Here, we showed that YAP was present in NSCs of the postnatal mouse SVZ. Forced expression of Yap promoted NSC maintenance and inhibited differentiation, whereas depletion of Yap by RNA interference or conditional knockout led to the decline of NSC maintenance, premature neuronal differentiation, and collapse of neurogenesis. For the molecular mechanism, thyroid hormone receptor-interacting protein 6 (TRIP6) recruited protein phosphatase PP1A to dephosphorylate LATS1/2, therefore inducing YAP nuclear localization and activation. Moreover, TRIP6 promoted NSC maintenance, cell proliferation, and inhibited differentiation through YAP. In addition, YAP regulated the expression of the Sonic Hedgehog (SHH) pathway effector Gli2 and Gli1/2 mediated the effect of YAP on NSC maintenance. Together, our findings demonstrate a novel TRIP6-YAP-SHH axis, which is critical for regulating postnatal neurogenesis in the SVZ-OB pathway.
Collapse
Affiliation(s)
- Ming-Yang Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Xiu-Li Yang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chia-Chi Chung
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Yun-Ju Lai
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jui-Cheng Tsai
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ya-Lin Kuo
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jenn-Yah Yu
- Department of Life Sciences, Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tsu-Wei Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
17
|
Kwon Y. YAP/TAZ as Molecular Targets in Skeletal Muscle Atrophy and Osteoporosis. Aging Dis 2024; 16:AD.2024.0306. [PMID: 38502585 PMCID: PMC11745433 DOI: 10.14336/ad.2024.0306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024] Open
Abstract
Skeletal muscles and bones are closely connected anatomically and functionally. Age-related degeneration in these tissues is associated with physical disability in the elderly and significantly impacts their quality of life. Understanding the mechanisms of age-related musculoskeletal tissue degeneration is crucial for identifying molecular targets for therapeutic interventions for skeletal muscle atrophy and osteoporosis. The Hippo pathway is a recently identified signaling pathway that plays critical roles in development, tissue homeostasis, and regeneration. The Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are key downstream effectors of the mammalian Hippo signaling pathway. This review highlights the fundamental roles of YAP and TAZ in the homeostatic maintenance and regeneration of skeletal muscles and bones. YAP/TAZ play a significant role in stem cell function by relaying various environmental signals to stem cells. Skeletal muscle atrophy and osteoporosis are related to stem cell dysfunction or senescence triggered by YAP/TAZ dysregulation resulting from reduced mechanosensing and mitochondrial function in stem cells. In contrast, the maintenance of YAP/TAZ activation can suppress stem cell senescence and tissue dysfunction and may be used as a basis for the development of potential therapeutic strategies. Thus, targeting YAP/TAZ holds significant therapeutic potential for alleviating age-related muscle and bone dysfunction and improving the quality of life in the elderly.
Collapse
Affiliation(s)
- Youngjoo Kwon
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Collignon E. Unveiling the role of cellular dormancy in cancer progression and recurrence. Curr Opin Oncol 2024; 36:74-81. [PMID: 38193374 DOI: 10.1097/cco.0000000000001013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
PURPOSE OF REVIEW Cellular dormancy is a major contributor to cancer progression and recurrence. This review explores recent findings on the molecular mechanisms implicated in cancer dormancy and investigates potential strategies to improve therapeutic interventions. RECENT FINDINGS Research on cancer dormancy reveals a complex and multifaceted phenomenon. Providing a latent reservoir of tumor cells with reduced proliferation and enhanced drug-tolerance, dormant cancer cells emerge from a clonally diverse population after therapy or at metastatic sites. These cells exhibit distinct transcriptional and epigenetic profiles, involving the downregulation of Myc and mechanistic target of rapamycin (mTOR) pathways, and the induction of autophagy. Senescence traits, under the control of factors such as p53, also contribute significantly. The tumor microenvironment can either promote or prevent dormancy establishment, notably through the involvement of T and NK cells within the dormant tumor niche. Strategies to combat dormancy-related relapse include direct elimination of dormant tumor cells, sustaining dormancy to prolong survival, or awakening dormant cells to re-sensitize them to antiproliferative drugs. SUMMARY Improving our understanding of cancer dormancy at primary and secondary sites provides valuable insights into patient care and relapse prevention.
Collapse
Affiliation(s)
- Evelyne Collignon
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Centre (U-CRC) and Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
19
|
Hong J, Kirkland JM, Acheta J, Marziali LN, Beck B, Jeanette H, Bhatia U, Davis G, Herron J, Roué C, Abi-Ghanem C, Feltri ML, Zuloaga K, Bechler ME, Poitelon Y, Belin S. YAP and TAZ regulate remyelination in the central nervous system. Glia 2024; 72:156-166. [PMID: 37724047 PMCID: PMC10659087 DOI: 10.1002/glia.24467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/20/2023]
Abstract
Myelinating cells are sensitive to mechanical stimuli from their extracellular matrix. Ablation of YAP and TAZ mechanotransducers in Schwann cells abolishes the axon-Schwann cell recognition, myelination, and remyelination in the peripheral nervous system. It was unknown if YAP and TAZ are also required for myelination and remyelination in the central nervous system. Here we define the importance of oligodendrocyte (OL) YAP and TAZ in vivo, by specific deletion in oligodendroglial cells in adult OLs during myelin repair. Blocking YAP and TAZ expression in OL lineage cells did not affect animal viability or any major defects on OL maturation and myelination. However, using a mouse model of demyelination/remyelination, we demonstrate that YAP and TAZ modulate the capacity of OLs to remyelinate axons, particularly during the early stage of the repair process, when OL proliferation is most important. These results indicate that YAP and TAZ signaling is necessary for effective remyelination of the mouse brain.
Collapse
Affiliation(s)
- Jiayue Hong
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Jules M Kirkland
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Jenica Acheta
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Leandro N Marziali
- Institute for Myelin and Glia Exploration, Dept. Biochemistry, University at Buffalo, Buffalo, NY, 14203, USA
| | - Brianna Beck
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Haley Jeanette
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Urja Bhatia
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Grace Davis
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Jacob Herron
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Clémence Roué
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Dept. Biochemistry, University at Buffalo, Buffalo, NY, 14203, USA
- Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Kristen Zuloaga
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Marie E Bechler
- Department of Cell and Developmental Biology, and Department of Neuroscience and Physiology State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Yannick Poitelon
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Sophie Belin
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| |
Collapse
|
20
|
Lau MS, Hu Z, Zhao X, Tan YS, Liu J, Huang H, Yeo CJ, Leong HF, Grinchuk OV, Chan JK, Yan J, Tee WW. Transcriptional repression by a secondary DNA binding surface of DNA topoisomerase I safeguards against hypertranscription. Nat Commun 2023; 14:6464. [PMID: 37833256 PMCID: PMC10576097 DOI: 10.1038/s41467-023-42078-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Regulation of global transcription output is important for normal development and disease, but little is known about the mechanisms involved. DNA topoisomerase I (TOP1) is an enzyme well-known for its role in relieving DNA supercoils for enabling transcription. Here, we report a non-enzymatic function of TOP1 that downregulates RNA synthesis. This function is dependent on specific DNA-interacting residues located on a conserved protein surface. A loss-of-function knock-in mutation on this surface, R548Q, is sufficient to cause hypertranscription and alter differentiation outcomes in mouse embryonic stem cells (mESCs). Hypertranscription in mESCs is accompanied by reduced TOP1 chromatin binding and change in genomic supercoiling. Notably, the mutation does not impact TOP1 enzymatic activity; rather, it diminishes TOP1-DNA binding and formation of compact protein-DNA structures. Thus, TOP1 exhibits opposing influences on transcription through distinct activities which are likely to be coordinated. This highlights TOP1 as a safeguard of appropriate total transcription levels in cells.
Collapse
Affiliation(s)
- Mei Sheng Lau
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
| | - Zhenhua Hu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaodan Zhao
- Department of Physics, National University of Singapore, Singapore, 117551, Singapore
- Centre for Bioimaging Sciences, National University of Singapore, Singapore, 117557, Singapore
| | - Yaw Sing Tan
- Bioinformatics Institute (BII), A*STAR, 30 Biopolis Street, Matrix, Singapore, 138671, Singapore
| | - Jinyue Liu
- Genome Institute of Singapore (GIS), A*STAR, 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Hua Huang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Electrophysiology Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Clarisse Jingyi Yeo
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Hwei Fen Leong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Oleg V Grinchuk
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Justin Kaixuan Chan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Jie Yan
- Department of Physics, National University of Singapore, Singapore, 117551, Singapore.
- Centre for Bioimaging Sciences, National University of Singapore, Singapore, 117557, Singapore.
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore.
| | - Wee-Wei Tee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
21
|
Azzi A, Tao Z, Sun Y, Erb H, Guarino C, Wu X. The circadian clock protein Cryptochrome 1 is a direct target and feedback regulator of the Hippo pathway. iScience 2023; 26:107449. [PMID: 37593458 PMCID: PMC10428131 DOI: 10.1016/j.isci.2023.107449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Circadian clock controls daily behavior and physiology. The activity of various signaling pathways affects clock gene expression. Here, we show that the core circadian clock gene CRY1 is a direct target of the Hippo pathway effector YAP. YAP binds to TEADs and occupies the proximal promoter regions of CRY1, positively regulating its transcription. Interestingly, we further identified that CRY1 acts in a feedback loop to fine-tune Hippo pathway activation by modulating the expression of YAP and MOB1. Indeed, loss of CRY1 results in enhanced YAP activation. Consistently, we found that YAP levels and activity control clock gene expression and oscillation in synchronized cells. Furthermore, in breast cancer cells, CRY1 downregulation causes YAP/TAZ hyperactivation and enhanced DNA damage. Together, our findings provide a direct mechanistic link between the Hippo pathway and the circadian clock, where CRY1 and Hippo components form an orchestrated signaling network that influences cell growth and circadian rhythm.
Collapse
Affiliation(s)
- Abdelhalim Azzi
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yang Sun
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hannah Erb
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Carla Guarino
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
22
|
Fan W, Jurado‐Arjona J, Alanis‐Lobato G, Péron S, Berger C, Andrade‐Navarro MA, Falk S, Berninger B. The transcriptional co-activator Yap1 promotes adult hippocampal neural stem cell activation. EMBO J 2023; 42:e110384. [PMID: 37083045 PMCID: PMC10233373 DOI: 10.15252/embj.2021110384] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
Most adult hippocampal neural stem cells (NSCs) remain quiescent, with only a minor portion undergoing active proliferation and neurogenesis. The molecular mechanisms that trigger the transition from quiescence to activation are still poorly understood. Here, we found the activity of the transcriptional co-activator Yap1 to be enriched in active NSCs. Genetic deletion of Yap1 led to a significant reduction in the relative proportion of active NSCs, supporting a physiological role of Yap1 in regulating the transition from quiescence to activation. Overexpression of wild-type Yap1 in adult NSCs did not induce NSC activation, suggesting tight upstream control mechanisms, but overexpression of a gain-of-function mutant (Yap1-5SA) elicited cell cycle entry in NSCs and hilar astrocytes. Consistent with a role of Yap1 in NSC activation, single cell RNA sequencing revealed a partial induction of an activated NSC gene expression program. Furthermore, Yap1-5SA expression also induced expression of Taz and other key components of the Yap/Taz regulon that were previously identified in glioblastoma stem cell-like cells. Consequently, dysregulated Yap1 activity led to repression of hippocampal neurogenesis, aberrant cell differentiation, and partial acquisition of a glioblastoma stem cell-like signature.
Collapse
Affiliation(s)
- Wenqiang Fan
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Present address:
Neuroscience Therapeutic Area, New MedicinesUCB Biopharma SPRLBraine‐l'AlleudBelgium
| | - Jerónimo Jurado‐Arjona
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Gregorio Alanis‐Lobato
- Faculty of BiologyJohannes Gutenberg University MainzMainzGermany
- Present address:
Global Computational Biology and Data SciencesBoehringer Ingelheim Pharma GmbH & Co. KGBiberach an der RissGermany
| | - Sophie Péron
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Christian Berger
- Institute of GeneticsJohannes Gutenberg University MainzMainzGermany
| | | | - Sven Falk
- Institute of BiochemistryFriedrich‐Alexander‐Universität Nürnberg‐ErlangenErlangenGermany
| | - Benedikt Berninger
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
- The Francis Crick InstituteLondonUK
- Focus Program Translational NeuroscienceJohannes Gutenberg University MainzMainzGermany
| |
Collapse
|
23
|
Wang J, Chen H, Hou W, Han Q, Wang Z. Hippo Pathway in Schwann Cells and Regeneration of Peripheral Nervous System. Dev Neurosci 2023; 45:276-289. [PMID: 37080186 DOI: 10.1159/000530621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
Hippo pathway is an evolutionarily conserved signaling pathway comprising a series of MST/LATS kinase complexes. Its key transcriptional coactivators YAP and TAZ regulate transcription factors such as TEAD family to direct gene expression. The regulation of Hippo pathway, especially the nuclear level change of YAP and TAZ, significantly influences the cell fate switching from proliferation to differentiation, regeneration, and postinjury repair. This review outlines the main findings of Hippo pathway in peripheral nerve development, regeneration, and tumorigenesis, especially the studies in Schwann cells. We also summarize other roles of Hippo pathway in damage repair of the peripheral nerve system and discuss the potential future research which probably contributes to novel therapeutic strategies.
Collapse
Affiliation(s)
- Jingyuan Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Jing'an District Central Hospital of Shanghai, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haofeng Chen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wulei Hou
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Jing'an District Central Hospital of Shanghai, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingjian Han
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Huashan Hospital, Fudan University, Shanghai, China
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Jing'an District Central Hospital of Shanghai, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Messelodi D, Strocchi S, Bertuccio SN, Baden P, Indio V, Giorgi FM, Taddia A, Serravalle S, Valente S, di Fonzo A, Frattini E, Bernardoni R, Pession A, Grifoni D, Deleidi M, Astolfi A, Pession A. Neuronopathic Gaucher disease models reveal defects in cell growth promoted by Hippo pathway activation. Commun Biol 2023; 6:431. [PMID: 37076591 PMCID: PMC10115838 DOI: 10.1038/s42003-023-04813-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/05/2023] [Indexed: 04/21/2023] Open
Abstract
Gaucher Disease (GD), the most common lysosomal disorder, arises from mutations in the GBA1 gene and is characterized by a wide spectrum of phenotypes, ranging from mild hematological and visceral involvement to severe neurological disease. Neuronopathic patients display dramatic neuronal loss and increased neuroinflammation, whose molecular basis are still unclear. Using a combination of Drosophila dGBA1b loss-of-function models and GD patient-derived iPSCs differentiated towards neuronal precursors and mature neurons we showed that different GD- tissues and neuronal cells display an impairment of growth mechanisms with an increased cell death and reduced proliferation. These phenotypes are coupled with the downregulation of several Hippo transcriptional targets, mainly involved in cells and tissue growth, and YAP exclusion from nuclei. Interestingly, Hippo knock-down in the GBA-KO flies rescues the proliferative defect, suggesting that targeting the Hippo pathway can be a promising therapeutic approach to neuronopathic GD.
Collapse
Affiliation(s)
- Daria Messelodi
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Silvia Strocchi
- Laboratory of Translational Research, USL-IRCCS of Reggio Emilia, 42123, Reggio Emilia, Italy
| | | | - Pascale Baden
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Hertie Institut for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Valentina Indio
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia (BO), Italy
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Alberto Taddia
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Salvatore Serravalle
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Sabrina Valente
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Alessio di Fonzo
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Emanuele Frattini
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Roberto Bernardoni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | | | - Daniela Grifoni
- Department of Life, Health and Environmental Sciences (MeSVA), University of L'Aquila, 67100, L'Aquila, Italy.
| | - Michela Deleidi
- Hertie Institut for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
- Institut Imagine, INSERM UMR1163, 75015, Paris, France
| | - Annalisa Astolfi
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Andrea Pession
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| |
Collapse
|
25
|
Capuz A, Osien S, Karnoub MA, Aboulouard S, Laurent E, Coyaud E, Raffo-Romero A, Duhamel M, Bonnefond A, Derhourhi M, Trerotola M, El Yazidi-Belkoura I, Devos D, Zilkova M, Kobeissy F, Vanden Abeele F, Fournier I, Cizkova D, Rodet F, Salzet M. Astrocytes express aberrant immunoglobulins as putative gatekeeper of astrocytes to neuronal progenitor conversion. Cell Death Dis 2023; 14:237. [PMID: 37015912 PMCID: PMC10073301 DOI: 10.1038/s41419-023-05737-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 04/06/2023]
Abstract
Using multi-omics analyses including RNAseq, RT-PCR, RACE-PCR, and shotgun proteomic with enrichment strategies, we demonstrated that newborn rat astrocytes produce neural immunoglobulin constant and variable heavy chains as well as light chains. However, their edification is different from the ones found in B cells and they resemble aberrant immunoglobulins observed in several cancers. Moreover, the complete enzymatic V(D)J recombination complex has also been identified in astrocytes. In addition, the constant heavy chain is also present in adult rat astrocytes, whereas in primary astrocytes from human fetus we identified constant and variable kappa chains as well as the substitution lambda chains known to be involved in pre-B cells. To gather insights into the function of these neural IgGs, CRISPR-Cas9 of IgG2B constant heavy chain encoding gene (Igh6), IgG2B overexpression, proximal labeling of rat astrocytes IgG2B and targets identification through 2D gels were performed. In Igh6 KO astrocytes, overrepresentation of factors involved in hematopoietic cells, neural stem cells, and the regulation of neuritogenesis have been identified. Moreover, overexpression of IgG2B in astrocytes induces the CRTC1-CREB-BDNF signaling pathway known to be involved in gliogenesis, whereas Igh6 KO triggers the BMP/YAP1/TEAD3 pathway activated in astrocytes dedifferentiation into neural progenitors. Proximal labeling experiments revealed that IgG2B is N-glycosylated by the OST complex, addressed to vesicle membranes containing the ATPase complex, and behaves partially like CD98hc through its association with LAT1. These experiments also suggest that proximal IgG2B-LAT1 interaction occurs concomitantly with MACO-1 and C2CD2L, at the heart of a potentially novel cell signaling platform. Finally, we demonstrated that these chains are synthesized individually and associated to recognize specific targets. Indeed, intermediate filaments Eif4a2 and Pdia6 involved in astrocyte fate constitute targets for these neural IgGs. Taken together, we hypothese that neural aberrant IgG chains may act as gatekeepers of astrocytes' fate.
Collapse
Affiliation(s)
- Alice Capuz
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Sylvain Osien
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Mélodie Anne Karnoub
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Soulaimane Aboulouard
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Estelle Laurent
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Etienne Coyaud
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Antonella Raffo-Romero
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Marie Duhamel
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Amélie Bonnefond
- Univ. Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, CHU de Lille, 1 place de Verdun, 59000, Lille, France
| | - Mehdi Derhourhi
- Univ. Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, CHU de Lille, 1 place de Verdun, 59000, Lille, France
| | - Marco Trerotola
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), University 'G. D'Annunzio', Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, University 'G. D'Annunzio', Chieti, Italy
| | - Ikram El Yazidi-Belkoura
- Université de Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59655, Villeneuve d'Ascq, France
| | - David Devos
- Université de Lille, INSERM, U1172, CHU-Lille, Lille Neuroscience Cognition Research Centre, 1 place de Verdun, 59000, Lille, France
| | - Monika Zilkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 84510, Bratislava, Slovakia
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fabien Vanden Abeele
- Université de Lille, INSERM U1003, Laboratory of Cell Physiology, 59655, Villeneuve d'Ascq, France
| | - Isabelle Fournier
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
- Institut Universitaire de France, 75005, Paris, France
| | - Dasa Cizkova
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 84510, Bratislava, Slovakia
- Centre for Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovakia
| | - Franck Rodet
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France.
| | - Michel Salzet
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France.
- Institut Universitaire de France, 75005, Paris, France.
| |
Collapse
|
26
|
Leonov S, Inyang O, Achkasov K, Bogdan E, Kontareva E, Chen Y, Fu Y, Osipov AN, Pustovalova M, Merkher Y. Proteomic Markers for Mechanobiological Properties of Metastatic Cancer Cells. Int J Mol Sci 2023; 24:ijms24054773. [PMID: 36902201 PMCID: PMC10003476 DOI: 10.3390/ijms24054773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/26/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
The major cause (more than 90%) of all cancer-related deaths is metastasis, thus its prediction can critically affect the survival rate. Metastases are currently predicted by lymph-node status, tumor size, histopathology and genetic testing; however, all these are not infallible, and obtaining results may require weeks. The identification of new potential prognostic factors will be an important source of risk information for the practicing oncologist, potentially leading to enhanced patient care through the proactive optimization of treatment strategies. Recently, the new mechanobiology-related techniques, independent of genetics, based on the mechanical invasiveness of cancer cells (microfluidic, gel indentation assays, migration assays etc.), demonstrated a high success rate for the detection of tumor cell metastasis propensity. However, they are still far away from clinical implementation due to complexity. Hence, the exploration of novel markers related to the mechanobiological properties of tumor cells may have a direct impact on the prognosis of metastasis. Our concise review deepens our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incites further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit. It may open a new clinical dimension that will improve cancer prognosis and increase the effectiveness of tumor therapies.
Collapse
Affiliation(s)
- Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Olumide Inyang
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Konstantin Achkasov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Elizaveta Bogdan
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Elizaveta Kontareva
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ying Fu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Andreyan N. Osipov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical-Biological Agency, 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
- Correspondence:
| | - Margarita Pustovalova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical-Biological Agency, 123098 Moscow, Russia
| | - Yulia Merkher
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
27
|
Kim YK, Cho B, Cook DP, Trcka D, Wrana JL, Ramalho-Santos M. Absolute scaling of single-cell transcriptomes identifies pervasive hypertranscription in adult stem and progenitor cells. Cell Rep 2023; 42:111978. [PMID: 36640358 DOI: 10.1016/j.celrep.2022.111978] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 10/27/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023] Open
Abstract
Hypertranscription supports biosynthetically demanding cellular states through global transcriptome upregulation. Despite its potential widespread relevance, documented examples of hypertranscription remain few and limited to early development. Here, we demonstrate that absolute scaling of single-cell RNA-sequencing data enables the estimation of total transcript abundances per cell. We validate absolute scaling in known cases of developmental hypertranscription and apply it to adult cell types, revealing a remarkable dynamic range in transcriptional output. In adult organs, hypertranscription marks activated stem/progenitor cells with multilineage potential and is redeployed in conditions of tissue injury, where it precedes bursts of proliferation during regeneration. Our analyses identify a common set of molecular pathways associated with both adult and embryonic hypertranscription, including chromatin remodeling, DNA repair, ribosome biogenesis, and translation. These shared features across diverse cell contexts support hypertranscription as a general and dynamic cellular program that is pervasively employed during development, organ maintenance, and regeneration.
Collapse
Affiliation(s)
- Yun-Kyo Kim
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada.
| | - Brandon Cho
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - David P Cook
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada
| | - Dan Trcka
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada
| | - Jeffrey L Wrana
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Miguel Ramalho-Santos
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
28
|
TAZ promotes osteogenic differentiation of mesenchymal stem cells line C3H10T1/2, murine multi-lineage cells lines C2C12, and MEFs induced by BMP9. Cell Death Dis 2022; 8:499. [PMID: 36575168 PMCID: PMC9794779 DOI: 10.1038/s41420-022-01292-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
Bone morphogenetic protein 9 (BMP9), also named as growth differentiation factor 2 (GDF-2), is the strongest cytokine that promotes osteogenic differentiation in the BMP family, and has broad clinical application value. Nevertheless, the mechanism of BMP9 promotes osteogenic differentiation remain unclear. TAZ, a transcriptional co-activator, has great effects on cell proliferation, differentiation, and stem cell self-renewal. In this research, we investigated the effects of TAZ in BMP9-induced osteogenic differentiation of mesenchymal stem cell line C3H10T1/2 (MSCs) and murine multi-lineage cell lines C2C12 and MEFs (MMCs) and explored its possible mechanisms. This study has found that BMP9 induces the expression of TAZ and promotes its nuclear translocation. Meanwhile, our study found that Ad-TAZ and TM-25659, a TAZ agonist, can enhance the osteogenic differentiation of MSCs and MMCs induced by BMP9. Conversely, Ad-si-TAZ and verteporfin, an inhibitor of TAZ, have the contradictory effect. Likewise, the promotion of TAZ to the BMP9-induced ectopic bone formation in vivo was confirmed by the subcutaneous transplantation of MSCs in nude mice. Furthermore, we have detected that TAZ might increase the levels of the phosphorylation of Smad1/5/8, p38, ERK1/2, and JNK induced by BMP9. Additionally, we also found that TAZ increased the total protein level of β-catenin induced by BMP9. In summary, our results strongly indicated that TAZ will promote the osteogenic differentiation in MSCs and MMCs induced by BMP9 through multiple signal pathways.
Collapse
|
29
|
Howard A, Bojko J, Flynn B, Bowen S, Jungwirth U, Walko G. Targeting the Hippo/YAP/TAZ signalling pathway: Novel opportunities for therapeutic interventions into skin cancers. Exp Dermatol 2022; 31:1477-1499. [PMID: 35913427 PMCID: PMC9804452 DOI: 10.1111/exd.14655] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 01/05/2023]
Abstract
Skin cancers are by far the most frequently diagnosed human cancers. The closely related transcriptional co-regulator proteins YAP and TAZ (WWTR1) have emerged as important drivers of tumour initiation, progression and metastasis in melanoma and non-melanoma skin cancers. YAP/TAZ serve as an essential signalling hub by integrating signals from multiple upstream pathways. In this review, we summarize the roles of YAP/TAZ in skin physiology and tumorigenesis and discuss recent efforts of therapeutic interventions that target YAP/TAZ in in both preclinical and clinical settings, as well as their prospects for use as skin cancer treatments.
Collapse
Affiliation(s)
| | - Jodie Bojko
- Department of Life SciencesUniversity of BathBathUK
| | | | - Sophie Bowen
- Department of Life SciencesUniversity of BathBathUK
| | - Ute Jungwirth
- Department of Life SciencesUniversity of BathBathUK,Centre for Therapeutic InnovationUniversity of BathBathUK
| | - Gernot Walko
- Department of Life SciencesUniversity of BathBathUK,Centre for Therapeutic InnovationUniversity of BathBathUK
| |
Collapse
|
30
|
A kinase-independent function of cyclin-dependent kinase 6 promotes outer radial glia expansion and neocortical folding. Proc Natl Acad Sci U S A 2022; 119:e2206147119. [PMID: 36095192 PMCID: PMC9499540 DOI: 10.1073/pnas.2206147119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The neocortex, the center for higher brain function, first emerged in mammals and has become massively expanded and folded in humans, constituting almost half the volume of the human brain. Primary microcephaly, a developmental disorder in which the brain is smaller than normal at birth, results mainly from there being fewer neurons in the neocortex because of defects in neural progenitor cells (NPCs). Outer radial glia (oRGs), NPCs that are abundant in gyrencephalic species but rare in lissencephalic species, are thought to play key roles in the expansion and folding of the neocortex. However, how oRGs expand, whether they are necessary for neocortical folding, and whether defects in oRGs cause microcephaly remain important questions in the study of brain development, evolution, and disease. Here, we show that oRG expansion in mice, ferrets, and human cerebral organoids requires cyclin-dependent kinase 6 (CDK6), the mutation of which causes primary microcephaly via an unknown mechanism. In a mouse model in which increased Hedgehog signaling expands oRGs and intermediate progenitor cells and induces neocortical folding, CDK6 loss selectively decreased oRGs and abolished neocortical folding. Remarkably, this function of CDK6 in oRG expansion did not require its kinase activity, was not shared by the highly similar CDK4 and CDK2, and was disrupted by the mutation causing microcephaly. Therefore, our results indicate that CDK6 is conserved to promote oRG expansion, that oRGs are necessary for neocortical folding, and that defects in oRG expansion may cause primary microcephaly.
Collapse
|
31
|
Kelenis DP, Rodarte KE, Kollipara RK, Pozo K, Choudhuri SP, Spainhower KB, Wait SJ, Stastny V, Oliver TG, Johnson JE. Inhibition of Karyopherin β1-Mediated Nuclear Import Disrupts Oncogenic Lineage-Defining Transcription Factor Activity in Small Cell Lung Cancer. Cancer Res 2022; 82:3058-3073. [PMID: 35748745 PMCID: PMC9444950 DOI: 10.1158/0008-5472.can-21-3713] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/29/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022]
Abstract
Genomic studies support the classification of small cell lung cancer (SCLC) into subtypes based on the expression of lineage-defining transcription factors ASCL1 and NEUROD1, which together are expressed in ∼86% of SCLC. ASCL1 and NEUROD1 activate SCLC oncogene expression, drive distinct transcriptional programs, and maintain the in vitro growth and oncogenic properties of ASCL1 or NEUROD1-expressing SCLC. ASCL1 is also required for tumor formation in SCLC mouse models. A strategy to inhibit the activity of these oncogenic drivers may therefore provide both a targeted therapy for the predominant SCLC subtypes and a tool to investigate the underlying lineage plasticity of established SCLC tumors. However, there are no known agents that inhibit ASCL1 or NEUROD1 function. In this study, we identify a novel strategy to pharmacologically target ASCL1 and NEUROD1 activity in SCLC by exploiting the nuclear localization required for the function of these transcription factors. Karyopherin β1 (KPNB1) was identified as a nuclear import receptor for both ASCL1 and NEUROD1 in SCLC, and inhibition of KPNB1 led to impaired ASCL1 and NEUROD1 nuclear accumulation and transcriptional activity. Pharmacologic targeting of KPNB1 preferentially disrupted the growth of ASCL1+ and NEUROD1+ SCLC cells in vitro and suppressed ASCL1+ tumor growth in vivo, an effect mediated by a combination of impaired ASCL1 downstream target expression, cell-cycle activity, and proteostasis. These findings broaden the support for targeting nuclear transport as an anticancer therapeutic strategy and have implications for targeting lineage-transcription factors in tumors beyond SCLC. SIGNIFICANCE The identification of KPNB1 as a nuclear import receptor for lineage-defining transcription factors in SCLC reveals a viable therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Demetra P. Kelenis
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kathia E. Rodarte
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rahul K. Kollipara
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Karine Pozo
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA,Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Kyle B. Spainhower
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sarah J. Wait
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Victor Stastny
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Trudy G. Oliver
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Jane E. Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
32
|
Zhang SF, Dai SK, Du HZ, Wang H, Li XG, Tang Y, Liu CM. The epigenetic state of EED-Gli3-Gli1 regulatory axis controls embryonic cortical neurogenesis. Stem Cell Reports 2022; 17:2064-2080. [PMID: 35931079 PMCID: PMC9481917 DOI: 10.1016/j.stemcr.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in the embryonic ectoderm development (EED) cause Weaver syndrome, but whether and how EED affects embryonic brain development remains elusive. Here, we generated a mouse model in which Eed was deleted in the forebrain to investigate the role of EED. We found that deletion of Eed decreased the number of upper-layer neurons but not deeper-layer neurons starting at E16.5. Transcriptomic and genomic occupancy analyses revealed that the epigenetic states of a group of cortical neurogenesis-related genes were altered in Eed knockout forebrains, followed by a decrease of H3K27me3 and an increase of H3K27ac marks within the promoter regions. The switching of H3K27me3 to H3K27ac modification promoted the recruitment of RNA-Pol2, thereby enhancing its expression level. The small molecule activator SAG or Ptch1 knockout for activating Hedgehog signaling can partially rescue aberrant cortical neurogenesis. Taken together, we proposed a novel EED-Gli3-Gli1 regulatory axis that is critical for embryonic brain development.
Collapse
Affiliation(s)
- Shuang-Feng Zhang
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Shang-Kun Dai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hong-Zhen Du
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hui Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Xing-Guo Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Yi Tang
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|
33
|
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
|
34
|
Diaz JR, Martá-Ariza M, Khodadadi-Jamayran A, Heguy A, Tsirigos A, Pankiewicz JE, Sullivan PM, Sadowski MJ. Apolipoprotein E4 Effects a Distinct Transcriptomic Profile and Dendritic Arbor Characteristics in Hippocampal Neurons Cultured in vitro. Front Aging Neurosci 2022; 14:845291. [PMID: 35572125 PMCID: PMC9099260 DOI: 10.3389/fnagi.2022.845291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
The APOE gene is diversified by three alleles ε2, ε3, and ε4 encoding corresponding apolipoprotein (apo) E isoforms. Possession of the ε4 allele is signified by increased risks of age-related cognitive decline, Alzheimer's disease (AD), and the rate of AD dementia progression. ApoE is secreted by astrocytes as high-density lipoprotein-like particles and these are internalized by neurons upon binding to neuron-expressed apoE receptors. ApoE isoforms differentially engage neuronal plasticity through poorly understood mechanisms. We examined here the effects of native apoE lipoproteins produced by immortalized astrocytes homozygous for ε2, ε3, and ε4 alleles on the maturation and the transcriptomic profile of primary hippocampal neurons. Control neurons were grown in the presence of conditioned media from Apoe -/- astrocytes. ApoE2 and apoE3 significantly increase the dendritic arbor branching, the combined neurite length, and the total arbor surface of the hippocampal neurons, while apoE4 fails to produce similar effects and even significantly reduces the combined neurite length compared to the control. ApoE lipoproteins show no systemic effect on dendritic spine density, yet apoE2 and apoE3 increase the mature spines fraction, while apoE4 increases the immature spine fraction. This is associated with opposing effects of apoE2 or apoE3 and apoE4 on the expression of NR1 NMDA receptor subunit and PSD95. There are 1,062 genes differentially expressed across neurons cultured in the presence of apoE lipoproteins compared to the control. KEGG enrichment and gene ontology analyses show apoE2 and apoE3 commonly activate expression of genes involved in neurite branching, and synaptic signaling. In contrast, apoE4 cultured neurons show upregulation of genes related to the glycolipid metabolism, which are involved in dendritic spine turnover, and those which are usually silent in neurons and are related to cell cycle and DNA repair. In conclusion, our work reveals that lipoprotein particles comprised of various apoE isoforms differentially regulate various neuronal arbor characteristics through interaction with neuronal transcriptome. ApoE4 produces a functionally distinct transcriptomic profile, which is associated with attenuated neuronal development. Differential regulation of neuronal transcriptome by apoE isoforms is a newly identified biological mechanism, which has both implication in the development and aging of the CNS.
Collapse
Affiliation(s)
- Jenny R. Diaz
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Mitchell Martá-Ariza
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | | | - Adriana Heguy
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Joanna E. Pankiewicz
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Patrick M. Sullivan
- Department of Medicine (Geriatrics), Duke University School of Medicine, Durham, NC, United States
- Durham VA Medical Center’s, Geriatric Research Education and Clinical Center, Durham, NC, United States
| | - Martin J. Sadowski
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
35
|
Piccolo FM, Kastan NR, Haremaki T, Tian Q, Laundos TL, De Santis R, Beaudoin AJ, Carroll TS, Luo JD, Gnedeva K, Etoc F, Hudspeth AJ, Brivanlou AH. Role of YAP in early ectodermal specification and a Huntington's Disease model of human neurulation. eLife 2022; 11:e73075. [PMID: 35451959 PMCID: PMC9033270 DOI: 10.7554/elife.73075] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
The Hippo pathway, a highly conserved signaling cascade that functions as an integrator of molecular signals and biophysical states, ultimately impinges upon the transcription coactivator Yes-associated protein 1 (YAP). Hippo-YAP signaling has been shown to play key roles both at the early embryonic stages of implantation and gastrulation, and later during neurogenesis. To explore YAP's potential role in neurulation, we used self-organizing neuruloids grown from human embryonic stem cells on micropatterned substrates. We identified YAP activation as a key lineage determinant, first between neuronal ectoderm and nonneuronal ectoderm, and later between epidermis and neural crest, indicating that YAP activity can enhance the effect of BMP4 stimulation and therefore affect ectodermal specification at this developmental stage. Because aberrant Hippo-YAP signaling has been implicated in the pathology of Huntington's Disease (HD), we used isogenic mutant neuruloids to explore the relationship between signaling and the disease. We found that HD neuruloids demonstrate ectopic activation of gene targets of YAP and that pharmacological reduction of YAP's transcriptional activity can partially rescue the HD phenotype.
Collapse
Affiliation(s)
- Francesco M Piccolo
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Nathaniel R Kastan
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
- Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew YorkUnited States
| | - Tomomi Haremaki
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Qingyun Tian
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Tiago L Laundos
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do PortoPortoPortugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do PortoPortoPortugal
- INEB - Instituto de Engenharia Biomédica, Universidade do PortoPortoPortugal
| | - Riccardo De Santis
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Andrew J Beaudoin
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Ksenia Gnedeva
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
- Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew YorkUnited States
| | - Fred Etoc
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - AJ Hudspeth
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
- Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew YorkUnited States
| | - Ali H Brivanlou
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
36
|
Vainshelbaum NM, Salmina K, Gerashchenko BI, Lazovska M, Zayakin P, Cragg MS, Pjanova D, Erenpreisa J. Role of the Circadian Clock "Death-Loop" in the DNA Damage Response Underpinning Cancer Treatment Resistance. Cells 2022; 11:880. [PMID: 35269502 PMCID: PMC8909334 DOI: 10.3390/cells11050880] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/14/2022] [Accepted: 03/01/2022] [Indexed: 12/11/2022] Open
Abstract
Here, we review the role of the circadian clock (CC) in the resistance of cancer cells to genotoxic treatments in relation to whole-genome duplication (WGD) and telomere-length regulation. The CC drives the normal cell cycle, tissue differentiation, and reciprocally regulates telomere elongation. However, it is deregulated in embryonic stem cells (ESCs), the early embryo, and cancer. Here, we review the DNA damage response of cancer cells and a similar impact on the cell cycle to that found in ESCs—overcoming G1/S, adapting DNA damage checkpoints, tolerating DNA damage, coupling telomere erosion to accelerated cell senescence, and favouring transition by mitotic slippage into the ploidy cycle (reversible polyploidy). Polyploidy decelerates the CC. We report an intriguing positive correlation between cancer WGD and the deregulation of the CC assessed by bioinformatics on 11 primary cancer datasets (rho = 0.83; p < 0.01). As previously shown, the cancer cells undergoing mitotic slippage cast off telomere fragments with TERT, restore the telomeres by ALT-recombination, and return their depolyploidised offspring to telomerase-dependent regulation. By reversing this polyploidy and the CC “death loop”, the mitotic cycle and Hayflick limit count are thus again renewed. Our review and proposed mechanism support a life-cycle concept of cancer and highlight the perspective of cancer treatment by differentiation.
Collapse
Affiliation(s)
- Ninel Miriam Vainshelbaum
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
- Faculty of Biology, University of Latvia, LV-1050 Riga, Latvia
| | - Kristine Salmina
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Bogdan I. Gerashchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, 03022 Kyiv, Ukraine;
| | - Marija Lazovska
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Pawel Zayakin
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Mark Steven Cragg
- Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
| | - Dace Pjanova
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| | - Jekaterina Erenpreisa
- Cancer Research Division, Latvian Biomedicine Research and Study Centre, LV-1067 Riga, Latvia; (N.M.V.); Latvia; (K.S.); (M.L.); (P.Z.); (D.P.)
| |
Collapse
|
37
|
Terry BK, Park R, Cho SH, Crino PB, Kim S. Abnormal activation of Yap/Taz contributes to the pathogenesis of tuberous sclerosis complex. Hum Mol Genet 2022; 31:1979-1996. [PMID: 34999833 PMCID: PMC9239747 DOI: 10.1093/hmg/ddab374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/31/2021] [Accepted: 12/27/2021] [Indexed: 01/09/2023] Open
Abstract
The multi-systemic genetic disorder tuberous sclerosis complex (TSC) impacts multiple neurodevelopmental processes including neuronal morphogenesis, neuronal migration, myelination and gliogenesis. These alterations contribute to the development of cerebral cortex abnormalities and malformations. Although TSC is caused by mTORC1 hyperactivation, cognitive and behavioral impairments are not improved through mTORC1 targeting, making the study of the downstream effectors of this complex important for understanding the mechanisms underlying TSC. As mTORC1 has been shown to promote the activity of the transcriptional co-activator Yap, we hypothesized that altered Yap/Taz signaling contributes to the pathogenesis of TSC. We first observed that the levels of Yap/Taz are increased in human cortical tuber samples and in embryonic cortices of Tsc2 conditional knockout (cKO) mice. Next, to determine how abnormal upregulation of Yap/Taz impacts the neuropathology of TSC, we deleted Yap/Taz in Tsc2 cKO mice. Importantly, Yap/Taz/Tsc2 triple conditional knockout (tcKO) animals show reduced cortical thickness and cortical neuron cell size, despite the persistence of high mTORC1 activity, suggesting that Yap/Taz play a downstream role in cytomegaly. Furthermore, Yap/Taz/Tsc2 tcKO significantly restored cortical and hippocampal lamination defects and reduced hippocampal heterotopia formation. Finally, the loss of Yap/Taz increased the distribution of myelin basic protein in Tsc2 cKO animals, consistent with an improvement in myelination. Overall, our results indicate that targeting Yap/Taz lessens the severity of neuropathology in a TSC animal model. This study is the first to implicate Yap/Taz as contributors to cortical pathogenesis in TSC and therefore as potential novel targets in the treatment of this disorder.
Collapse
Affiliation(s)
- Bethany K Terry
- Department of Neural Sciences, Lewis Katz School of Medicine, Shriners Hospitals Pediatrics Research Center, Temple University, Philadelphia, PA 19140, USA,Biomedical Sciences Graduate Program, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Raehee Park
- Department of Neural Sciences, Lewis Katz School of Medicine, Shriners Hospitals Pediatrics Research Center, Temple University, Philadelphia, PA 19140, USA
| | - Seo-Hee Cho
- Department of Medicine, Sidney Kimmel Medical College, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Seonhee Kim
- To whom correspondence should be addressed. Tel: 215-926-9360; Fax: 215-926-9325;
| |
Collapse
|
38
|
Su YC, Hung TH, Wang TF, Lee YH, Wang TW, Yu JY. YAP maintains the production of intermediate progenitor cells and upper-layer projection neurons in the mouse cerebral cortex. Dev Dyn 2021; 251:846-863. [PMID: 34931379 DOI: 10.1002/dvdy.448] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The Hippo pathway is conserved through evolution and plays critical roles in development, tissue homeostasis and tumorigenesis. Yes-associated protein (YAP) is a transcriptional coactivator downstream of the Hippo pathway. Previous studies have demonstrated that activation of YAP promotes proliferation in the developing brain. Whether YAP is required for the production of neural progenitor cells or neurons in vivo remains unclear. RESULTS We demonstrated that SATB homeobox 2 (SATB2)-positive projection neurons (PNs) in upper layers, but not T-box brain transcription factor 1-positive and Coup-TF interacting protein 2-positive PNs in deep layers, were decreased in the neonatal cerebral cortex of Yap conditional knockout (cKO) mice driven by Nestin-Cre. Cell proliferation was reduced in the developing cerebral cortex of Yap-cKO. SATB2-positive PNs are largely generated from intermediate progenitor cells (IPCs), which are derived from radial glial cells (RGCs) during cortical development. Among these progenitor cells, IPCs but not RGCs were decreased in Yap-cKO. We further demonstrated that cell cycle re-entry was reduced in progenitor cells of Yap-cKO, suggesting that fewer IPCs were generated in Yap-cKO. CONCLUSION YAP is required for the production of IPCs and upper-layer SATB2-positive PNs during development of the cerebral cortex in mice.
Collapse
Affiliation(s)
- Yi-Ching Su
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Heng Hung
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Fang Wang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Hsuan Lee
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tsu-Wei Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jenn-Yah Yu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
39
|
Xiao Y, Dong J. The Hippo Signaling Pathway in Cancer: A Cell Cycle Perspective. Cancers (Basel) 2021; 13:cancers13246214. [PMID: 34944834 PMCID: PMC8699626 DOI: 10.3390/cancers13246214] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/25/2023] Open
Abstract
Simple Summary Cancer is increasingly viewed as a cell cycle disease in that the dysregulation of the cell cycle machinery is a common feature in cancer. The Hippo signaling pathway consists of a core kinase cascade as well as extended regulators, which together control organ size and tissue homeostasis. The aberrant expression of cell cycle regulators and/or Hippo pathway components contributes to cancer development, and for this reason, we specifically focus on delineating the roles of the Hippo pathway in the cell cycle. Improving our understanding of the Hippo pathway from a cell cycle perspective could be used as a powerful weapon in the cancer battlefield. Abstract Cell cycle progression is an elaborate process that requires stringent control for normal cellular function. Defects in cell cycle control, however, contribute to genomic instability and have become a characteristic phenomenon in cancers. Over the years, advancement in the understanding of disrupted cell cycle regulation in tumors has led to the development of powerful anti-cancer drugs. Therefore, an in-depth exploration of cell cycle dysregulation in cancers could provide therapeutic avenues for cancer treatment. The Hippo pathway is an evolutionarily conserved regulator network that controls organ size, and its dysregulation is implicated in various types of cancers. Although the role of the Hippo pathway in oncogenesis has been widely investigated, its role in cell cycle regulation has not been comprehensively scrutinized. Here, we specifically focus on delineating the involvement of the Hippo pathway in cell cycle regulation. To that end, we first compare the structural as well as functional conservation of the core Hippo pathway in yeasts, flies, and mammals. Then, we detail the multi-faceted aspects in which the core components of the mammalian Hippo pathway and their regulators affect the cell cycle, particularly with regard to the regulation of E2F activity, the G1 tetraploidy checkpoint, DNA synthesis, DNA damage checkpoint, centrosome dynamics, and mitosis. Finally, we briefly discuss how a collective understanding of cell cycle regulation and the Hippo pathway could be weaponized in combating cancer.
Collapse
Affiliation(s)
| | - Jixin Dong
- Correspondence: ; Tel.: +402-559-5596; Fax: +402-559-4651
| |
Collapse
|
40
|
Sun H, Cao X, Gong A, Huang Y, Xu Y, Zhang J, Sun J, Lv B, Li Z, Guan S, Lu L, Yin G. Extracellular vesicles derived from astrocytes facilitated neurite elongation by activating the Hippo pathway. Exp Cell Res 2021; 411:112937. [PMID: 34863709 DOI: 10.1016/j.yexcr.2021.112937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 01/01/2023]
Abstract
Spinal cord injury (SCI) often causes severe neurological dysfunction, and facilitating neurite elongation is particularly important in its treatment. Astrocytes (AS) play an important role in the central nervous system (CNS), and their high plasticity and versatility provide a feasible entry point for relevant research. Our purpose was to explore whether extracellular vesicles (EVs) from astrocytes (AS-EVs) and lipopolysaccharide (LPS)-preactivated astrocytes (LPAS-EVs) facilitate neurite elongation, to explore the underlying mechanism, and to verify whether these EVs promote locomotor recovery in rats. We used LPS to preactivate astrocytes and cocultured them with PC12 cells to observe neurite changes, then extracted and identified AS-EVs and LPAS-EVs and the role and mechanism of these EVs in facilitating neurite elongation was examined in vivo and vitro. We demonstrated that AS-EVs and LPAS-EVs facilitated the elongation of neurites and the recovery of rats with SCI. LPAS-EVs had a stronger effect than AS-EVs, by activating the Hippo pathway, promoting monopole spindle binding protein 1 (MOB1) expression, and reducing Yes-associated protein (YAP) levels. The data also suggest a feedback regulation between MOB1 and p-YAP/YAP. In sum, AS-EVs and LPAS-EVs can play an active role in facilitating neurite elongation by activating the Hippo pathway. These findings provide a new strategy for treating SCI and other CNS-related injuries.
Collapse
Affiliation(s)
- Haitao Sun
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xingbing Cao
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Aihua Gong
- Medical College, Jiangsu University, Zhenjiang City, Jiangsu, China
| | - Yonghui Huang
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yi Xu
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jinglong Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jifu Sun
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bin Lv
- Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhen Li
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shihao Guan
- Department of Spinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ling Lu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Guoyong Yin
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
41
|
Shim J, Goldsmith KC. A New Player in Neuroblastoma: YAP and Its Role in the Neuroblastoma Microenvironment. Cancers (Basel) 2021; 13:cancers13184650. [PMID: 34572875 PMCID: PMC8472533 DOI: 10.3390/cancers13184650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma is the most common extra-cranial pediatric solid tumor that accounts for more than 15% of childhood cancer-related deaths. High risk neuroblastomas that recur during or after intense multimodal therapy have a <5% chance at a second sustained remission or cure. The solid tumor microenvironment (TME) has been increasingly recognized to play a critical role in cancer progression and resistance to therapy, including in neuroblastoma. The Yes-Associated Protein (YAP) in the Hippo pathway can regulate cancer proliferation, tumor initiation, and therapy response in many cancer types and as such, its role in the TME has gained interest. In this review, we focus on YAP and its role in neuroblastoma and further describe its demonstrated and potential effects on the neuroblastoma TME. We also discuss the therapeutic strategies for inhibiting YAP in neuroblastoma.
Collapse
Affiliation(s)
- Jenny Shim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Kelly C. Goldsmith
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Correspondence: ; Tel.: +1-404-727-2655
| |
Collapse
|
42
|
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
|
43
|
Bowry A, Kelly RDW, Petermann E. Hypertranscription and replication stress in cancer. Trends Cancer 2021; 7:863-877. [PMID: 34052137 DOI: 10.1016/j.trecan.2021.04.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022]
Abstract
Replication stress results from obstacles to replication fork progression, including ongoing transcription, which can cause transcription-replication conflicts. Oncogenic signaling can promote global increases in transcription activity, also termed hypertranscription. Despite the widely accepted importance of oncogene-induced hypertranscription, its study remains neglected compared with other causes of replication stress and genomic instability in cancer. A growing number of recent studies are reporting that oncogenes, such as RAS, and targeted cancer treatments, such as bromodomain and extraterminal motif (BET) bromodomain inhibitors, increase global transcription, leading to R-loop accumulation, transcription-replication conflicts, and the activation of replication stress responses. Here we discuss our mechanistic understanding of hypertranscription-induced replication stress and the resulting cellular responses, in the context of oncogenes and targeted cancer therapies.
Collapse
Affiliation(s)
- Akhil Bowry
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Richard D W Kelly
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Eva Petermann
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
44
|
Lopez-Hernandez A, Sberna S, Campaner S. Emerging Principles in the Transcriptional Control by YAP and TAZ. Cancers (Basel) 2021; 13:cancers13164242. [PMID: 34439395 PMCID: PMC8391352 DOI: 10.3390/cancers13164242] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary YAP and TAZ are transcriptional cofactors that integrate several upstream signals to generate context-dependent transcriptional responses. This requires extensive integration with epigenetic regulators and other transcription factors. The molecular and genomic characterization of YAP and TAZ nuclear function has broad implications both in physiological and pathological settings. Abstract Yes-associated protein (YAP) and TAZ are transcriptional cofactors that sit at the crossroad of several signaling pathways involved in cell growth and differentiation. As such, they play essential functions during embryonic development, regeneration, and, once deregulated, in cancer progression. In this review, we will revise the current literature and provide an overview of how YAP/TAZ control transcription. We will focus on data concerning the modulation of the basal transcriptional machinery, their ability to epigenetically remodel the enhancer–promoter landscape, and the mechanisms used to integrate transcriptional cues from multiple pathways. This reveals how YAP/TAZ activation in cancer cells leads to extensive transcriptional control that spans several hallmarks of cancer. The definition of the molecular mechanism of transcriptional control and the identification of the pathways regulated by YAP/TAZ may provide therapeutic opportunities for the effective treatment of YAP/TAZ-driven tumors.
Collapse
|
45
|
Lavado A, Gangwar R, Paré J, Wan S, Fan Y, Cao X. YAP/TAZ maintain the proliferative capacity and structural organization of radial glial cells during brain development. Dev Biol 2021; 480:39-49. [PMID: 34419458 DOI: 10.1016/j.ydbio.2021.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/29/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
The Hippo pathway regulates the development and homeostasis of many tissues and in many species. It controls the activity of two paralogous transcriptional coactivators, YAP and TAZ (YAP/TAZ). Although previous studies have established that aberrant YAP/TAZ activation is detrimental to mammalian brain development, whether and how endogenous levels of YAP/TAZ activity regulate brain development remain unclear. Here, we show that during mammalian cortical development, YAP/TAZ are specifically expressed in apical neural progenitor cells known as radial glial cells (RGCs). The subcellular localization of YAP/TAZ undergoes dynamic changes as corticogenesis proceeds. YAP/TAZ are required for maintaining the proliferative potential and structural organization of RGCs, and their ablation during cortical development reduces the numbers of cortical projection neurons and causes the loss of ependymal cells, resulting in hydrocephaly. Transcriptomic analysis using sorted RGCs reveals gene expression changes in YAP/TAZ-depleted cells that correlate with mutant phenotypes. Thus, our study has uncovered essential functions of YAP/TAZ during mammalian brain development and revealed the transcriptional mechanism of their action.
Collapse
Affiliation(s)
- Alfonso Lavado
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ruchika Gangwar
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Joshua Paré
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shibiao Wan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xinwei Cao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
46
|
Bulut-Karslioglu A, Jin H, Kim YK, Cho B, Guzman-Ayala M, Williamson AJK, Hejna M, Stötzel M, Whetton AD, Song JS, Ramalho-Santos M. Chd1 protects genome integrity at promoters to sustain hypertranscription in embryonic stem cells. Nat Commun 2021; 12:4859. [PMID: 34381042 PMCID: PMC8357957 DOI: 10.1038/s41467-021-25088-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/20/2021] [Indexed: 11/09/2022] Open
Abstract
Stem and progenitor cells undergo a global elevation of nascent transcription, or hypertranscription, during key developmental transitions involving rapid cell proliferation. The chromatin remodeler Chd1 mediates hypertranscription in pluripotent cells but its mechanism of action remains poorly understood. Here we report a novel role for Chd1 in protecting genome integrity at promoter regions by preventing DNA double-stranded break (DSB) accumulation in ES cells. Chd1 interacts with several DNA repair factors including Atm, Parp1, Kap1 and Topoisomerase 2β and its absence leads to an accumulation of DSBs at Chd1-bound Pol II-transcribed genes and rDNA. Genes prone to DNA breaks in Chd1 KO ES cells are longer genes with GC-rich promoters, a more labile nucleosomal structure and roles in chromatin regulation, transcription and signaling. These results reveal a vulnerability of hypertranscribing stem cells to accumulation of endogenous DNA breaks, with important implications for developmental and cancer biology.
Collapse
Affiliation(s)
- Aydan Bulut-Karslioglu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences and Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
| | - Hu Jin
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
- Department of Physics, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yun-Kyo Kim
- Lunenfeld-Tanenbaum Research Institute and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Brandon Cho
- Lunenfeld-Tanenbaum Research Institute and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Marcela Guzman-Ayala
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences and Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
- Senti Biosciences, South San Francisco, CA, USA
| | - Andrew J K Williamson
- Stoller Biomarker Discovery Centre, The University of Manchester, Manchester, UK
- Thermo Fisher Scientific, Stafford House, UK
| | - Miroslav Hejna
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
- Department of Physics, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | | | - Anthony D Whetton
- Stoller Biomarker Discovery Centre, The University of Manchester, Manchester, UK
| | - Jun S Song
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
- Department of Physics, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Miguel Ramalho-Santos
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences and Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
- Lunenfeld-Tanenbaum Research Institute and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
47
|
Cellular feedback dynamics and multilevel regulation driven by the hippo pathway. Biochem Soc Trans 2021; 49:1515-1527. [PMID: 34374419 PMCID: PMC8421037 DOI: 10.1042/bst20200253] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
The Hippo pathway is a dynamic cellular signalling nexus that regulates differentiation and controls cell proliferation and death. If the Hippo pathway is not precisely regulated, the functionality of the upstream kinase module is impaired, which increases nuclear localisation and activity of the central effectors, the transcriptional co-regulators YAP and TAZ. Pathological YAP and TAZ hyperactivity consequently cause cancer, fibrosis and developmental defects. The Hippo pathway controls an array of fundamental cellular processes, including adhesion, migration, mitosis, polarity and secretion of a range of biologically active components. Recent studies highlight that spatio-temporal regulation of Hippo pathway components are central to precisely controlling its context-dependent dynamic activity. Several levels of feedback are integrated into the Hippo pathway, which is further synergized with interactors outside of the pathway that directly regulate specific Hippo pathway components. Likewise, Hippo core kinases also ‘moonlight’ by phosphorylating multiple substrates beyond the Hippo pathway and thereby integrates further flexibility and robustness in the cellular decision-making process. This topic is still in its infancy but promises to reveal new fundamental insights into the cellular regulation of this therapeutically important pathway. We here highlight recent advances emphasising feedback dynamics and multilevel regulation of the Hippo pathway with a focus on mitosis and cell migration, as well as discuss potential productive future research avenues that might reveal novel insights into the overall dynamics of the pathway.
Collapse
|
48
|
Wu Q, Miao X, Zhang J, Xiang L, Li X, Bao X, Du S, Wang M, Miao S, Fan Y, Wang W, Xu X, Shen X, Yang D, Wang X, Fang Y, Hu L, Pan X, Dong H, Wang H, Wang Y, Li J, Huang Z. Astrocytic YAP protects the optic nerve and retina in an experimental autoimmune encephalomyelitis model through TGF-β signaling. Theranostics 2021; 11:8480-8499. [PMID: 34373754 PMCID: PMC8344002 DOI: 10.7150/thno.60031] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Rationale: Optic neuritis is one of main symptoms in multiple sclerosis (MS) that causes visual disability. Astrocytes are pivotal regulators of neuroinflammation in MS, and astrocytic yes-associated protein (YAP) plays a critical role in neuroinflammation. Meanwhile, YAP signaling is involved in visual impairment, including glaucoma, retinal choroidal atrophy and retinal detachment. However, the roles and underlying mechanisms of astrocytic YAP in neuroinflammation and demyelination of MS-related optic neuritis (MS-ON) remains unclear. Methods: To assess the functions of YAP in MS-ON, experimental autoimmune encephalomyelitis (EAE, a common model of MS) was established, and mice that conditional knockout (CKO) of YAP in astrocytes, YAPGFAP-CKO mice, were successfully generated. Behavior tests, immunostaining, Nissl staining, Hematoxylin-Eosin (HE) staining, TUNEL staining, Luxol Fast Blue (LFB) staining, electron microscopy (EM), quantitative real-time PCR (qPCR), gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) by RNA sequencing were used to examine the function and mechanism of YAP signaling based on these YAPGFAP-CKO mice and EAE model mice. To further explore the potential treatment of YAP signaling in EAE, EAE mice were treated with various drugs, including SRI-011381 that is an agonist of transforming growth factor-β (TGF-β) pathway, and XMU-MP-1 which inhibits Hippo kinase MST1/2 to activate YAP. Results: We found that YAP was significantly upregulated and activated in the astrocytes of optic nerve in EAE mice. Conditional knockout of YAP in astrocytes caused more severe inflammatory infiltration and demyelination in optic nerve, and damage of retinal ganglion cells (RGCs) in EAE mice. Moreover, YAP deletion in astrocytes promoted the activation of astrocytes and microglia, but inhibited the proliferation of astrocytes of optic nerve in EAE mice. Mechanically, TGF-β signaling pathway was significantly down-regulated after YAP deletion in astrocytes. Additionally, both qPCR and immunofluorescence assays confirmed the reduction of TGF-β signaling pathway in YAPGFAP-CKO EAE mice. Interestingly, SRI-011381 partially rescued the deficits in optic nerve and retina of YAPGFAP-CKO EAE mice. Finally, activation of YAP signaling by XMU-MP-1 relieved the neuroinflammation and demyelination in optic nerve of EAE mice. Conclusions: These results suggest astrocytic YAP may prevent the neuroinflammatory infiltration and demyelination through upregulation of TGF-β signaling and provide targets for the development of therapeutic strategies tailored for MS-ON.
Collapse
Affiliation(s)
- Qian Wu
- School of pharmacy and Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xuemeng Miao
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jingjing Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ludan Xiang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiuchun Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaomei Bao
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Siyu Du
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Mianxian Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shuangda Miao
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yiren Fan
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xingxing Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiya Shen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Danlu Yang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiwu Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuanyuan Fang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lixin Hu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xuyi Pan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Haoru Dong
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Hui Wang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ying Wang
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310053, China
| | - Jia Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhihui Huang
- School of pharmacy and Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| |
Collapse
|
49
|
Li X, Li K, Chen Y, Fang F. The Role of Hippo Signaling Pathway in the Development of the Nervous System. Dev Neurosci 2021; 43:263-270. [PMID: 34350875 DOI: 10.1159/000515633] [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: 09/08/2020] [Accepted: 02/26/2021] [Indexed: 11/19/2022] Open
Abstract
Hippo signaling pathway is a highly conserved and crucial signaling pathway that controls the size of tissues and organs by regulating the proliferation, differentiation, and apoptosis of cells. The nervous system is a complicated system that participates in information collection, integration, and procession. The balance of various aspects of the nervous system is vital for the normal regulation of physiological conditions of the body, like the population and distribution of nerve cells, nerve connections, and so on. Defects in these aspects may lead to cognitive, behavioral, and neurological dysfunction, resulting in various nervous system diseases. Recently, accumulating evidence proposes that Hippo pathway maintains numerous biological functions in the nervous system development, including modulating the proliferation and differentiation of nerve cells and promoting the development of synapse, corpus callosum, and cortex. In this review, we will summarize recent findings of Hippo pathway in the nervous system to improve our understanding on its function and to provide potential therapeutic strategies of nervous system diseases in the future.
Collapse
Affiliation(s)
- Xifan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Kaixuan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Yu Chen
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Fang Fang
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| |
Collapse
|
50
|
Panizzutti B, Bortolasci CC, Spolding B, Kidnapillai S, Connor T, Richardson MF, Truong TTT, Liu ZSJ, Morris G, Gray L, Hyun Kim J, Dean OM, Berk M, Walder K. Transcriptional Modulation of the Hippo Signaling Pathway by Drugs Used to Treat Bipolar Disorder and Schizophrenia. Int J Mol Sci 2021; 22:7164. [PMID: 34281223 PMCID: PMC8268913 DOI: 10.3390/ijms22137164] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
Recent reports suggest a link between positive regulation of the Hippo pathway with bipolar disorder (BD), and the Hippo pathway is known to interact with multiple other signaling pathways previously associated with BD and other psychiatric disorders. In this study, neuronal-like NT2 cells were treated with amisulpride (10 µM), aripiprazole (0.1 µM), clozapine (10 µM), lamotrigine (50 µM), lithium (2.5 mM), quetiapine (50 µM), risperidone (0.1 µM), valproate (0.5 mM), or vehicle control for 24 h. Genome-wide mRNA expression was quantified and analyzed using gene set enrichment analysis (GSEA), with genes belonging to Hippo, Wnt, Notch, TGF- β, and Hedgehog retrieved from the KEGG database. Five of the eight drugs downregulated the genes of the Hippo pathway and modulated several genes involved in the interacting pathways. We speculate that the regulation of these genes, especially by aripiprazole, clozapine, and quetiapine, results in a reduction of MAPK and NFκB pro-inflammatory signaling through modulation of Hippo, Wnt, and TGF-β pathways. We also employed connectivity map analysis to identify compounds that act on these pathways in a similar manner to the known psychiatric drugs. Thirty-six compounds were identified. The presence of antidepressants and antipsychotics validates our approach and reveals possible new targets for drug repurposing.
Collapse
Affiliation(s)
- Bruna Panizzutti
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Chiara C. Bortolasci
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Briana Spolding
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Srisaiyini Kidnapillai
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Timothy Connor
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Mark F. Richardson
- Genomics Centre, School of Life and Environmental Sciences, Deakin University, Burwood 3125, Australia;
| | - Trang T. T. Truong
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Zoe S. J. Liu
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Gerwyn Morris
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Laura Gray
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
| | - Jee Hyun Kim
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| | - Olivia M. Dean
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
| | - Michael Berk
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville 3052, Australia
- Centre of Youth Mental Health, University of Melbourne, Parkville 3052, Australia
- Orygen Youth Health Research Centre, Parkville 3052, Australia
| | - Ken Walder
- Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, IMPACT, Geelong 3220, Australia; (B.P.); (C.C.B.); (B.S.); (S.K.); (T.C.); (T.T.T.T.); (Z.S.J.L.); (G.M.); (L.G.); (J.H.K.); (O.M.D.); (M.B.)
| |
Collapse
|