1
|
Ahuja N, Maynard C, Bierschenck T, Cleaver O. Characterization of Hippo signaling components in the early dorsal pancreatic bud. Gene Expr Patterns 2025; 55:119392. [PMID: 40081783 DOI: 10.1016/j.gep.2025.119392] [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: 11/08/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
All pancreatic lineages originate from a transitory structure known as the multipotent progenitor epithelium (MPE), which is an endodermal placode formed via epithelial stratification. Cells within the MPE undergo de novo lumenogenesis to give rise to an epithelial plexus, which serves as a progenitor niche for subsequent development of endocrine, ductal and acinar cell types. Recent evidence suggests that Hippo signaling is required for pancreatic cell differentiation, but little is known about the function of Hippo signaling in the development of the MPE. Here, we characterize the expression of YAP1, TAZ, and the Hippo regulators LATS1/2 kinases and MERLIN in early murine pancreatic epithelium, during epithelial stratification, plexus development and emergence of endocrine cells. We find that YAP1 expression is relatively low in the pancreas bud during stratification but increases by E11.5. Intriguingly, we find differing patterns of TAZ and YAP1 immunoreactivty throughout pancreatic development. We further find that MERLIN and LATS1/2 kinases are expressed during the period of rapid stratification and become markedly apical at nascent lumens. To gain a better understanding of how Hippo signaling and lumen formation are connected, we analyzed the subcellular localization of Hippo signaling components during varying stages of lumen formation and found that they are dynamically localized during lumenogenesis. Together, our results point to a previously unsuspected relationship between Hippo signaling and lumen formation during pancreatic development.
Collapse
Affiliation(s)
- Neha Ahuja
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Caitlin Maynard
- Department of Biology, The University of Texas at Arlington, 501 S. Nedderman Drive, Arlington, TX, 76019, USA
| | - Tyler Bierschenck
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
2
|
Ahuja N, Maynard C, Bierschenck T, Cleaver O. Characterization of Hippo Signaling Components in the Early Dorsal Pancreatic Bud. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.26.619721. [PMID: 39484500 PMCID: PMC11527122 DOI: 10.1101/2024.10.26.619721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
All pancreatic lineages originate from a transitory structure known as the multipotent progenitor epithelium (MPE), which is a placode formed via epithelial stratification. Cells within the MPE undergo de novo lumenogenesis to give rise to an epithelial plexus, which serves as a progenitor niche for subsequent development of endocrine, ductal and acinar cell types. Recent evidence suggests that Hippo signaling is required for pancreatic cell differentiation, but little is known about the function of Hippo signaling in the development of the MPE. Here, we characterize the expression of YAP1, TAZ, and the Hippo regulators LATS1/2 kinases and MERLIN in early murine pancreatic epithelium, during epithelial stratification, plexus development and emergence of endocrine cells. We find that YAP1 expression is relatively low in the pancreas bud during stratification, but increases by E11.5. Intriguingly, we find that TAZ, but not YAP1, is expressed in early endocrine cells. We further find that MERLIN and LATS1/2 kinases are robustly expressed during the period of rapid stratification and become markedly apical at nascent lumens. To gain a better understanding of how Hippo signaling and lumen formation are connected, we analyzed the expression of Hippo signaling components in an in vitro model of lumen formation and found that they are dynamically regulated during lumenogenesis. Together, our results point to a relationship between Hippo signaling and lumen formation during pancreatic development. HIGHLIGHTS YAP1 expression in the early mouse pancreatic anlagen is low until approximately E11.5, when it becomes localized to cell nuclei in multipotent progenitor cells. At E14.5, we find nuclear YAP1 in ductal cells.YAP1 is not expressed in early and midgestation endocrine cells. By contrast, TAZ is expressed in first transition endocrine cells.Hippo regulators MERLIN and LATS1/2 kinases are robustly expressed in the early pancreatic bud by E10.5. Both MERLIN and LATS1/2 exhibit strong apical localization in epithelial cells at nascent microlumens. Using in vitro models of de novo pancreas lumen formation, we show that YAP1 nuclear localization is high in early phases of lumen formation and gradually decreases as lumens matures.
Collapse
|
3
|
Wu Y, Qin K, Xu Y, Rajhans S, Vo T, Lopez KM, Liu J, Nipper MH, Deng J, Yin X, Ramjit LR, Ye Z, Luan Y, Arda HE, Wang P. Hippo pathway-mediated YAP1/TAZ inhibition is essential for proper pancreatic endocrine specification and differentiation. eLife 2024; 13:e84532. [PMID: 39051998 PMCID: PMC11272159 DOI: 10.7554/elife.84532] [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: 10/28/2022] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
The Hippo pathway plays a central role in tissue development and homeostasis. However, the function of Hippo in pancreatic endocrine development remains obscure. Here, we generated novel conditional genetically engineered mouse models to examine the roles of Hippo pathway-mediated YAP1/TAZ inhibition in the development stages of endocrine specification and differentiation. While YAP1 protein was localized to the nuclei in bipotent progenitor cells, Neurogenin 3 expressing endocrine progenitors completely lost YAP1 expression. Using genetically engineered mouse models, we found that inactivation of YAP1 requires both an intact Hippo pathway and Neurogenin 3 protein. Gene deletion of Lats1 and 2 kinases (Lats1&2) in endocrine progenitor cells of developing mouse pancreas using Neurog3Cre blocked endocrine progenitor cell differentiation and specification, resulting in reduced islets size and a disorganized pancreas at birth. Loss of Lats1&2 in Neurogenin 3 expressing cells activated YAP1/TAZ transcriptional activity and recruited macrophages to the developing pancreas. These defects were rescued by deletion of Yap1/Wwtr1 genes, suggesting that tight regulation of YAP1/TAZ by Hippo signaling is crucial for pancreatic endocrine specification. In contrast, deletion of Lats1&2 using β-cell-specific Ins1CreER resulted in a phenotypically normal pancreas, indicating that Lats1&2 are indispensable for differentiation of endocrine progenitors but not for that of β-cells. Our results demonstrate that loss of YAP1/TAZ expression in the pancreatic endocrine compartment is not a passive consequence of endocrine specification. Rather, Hippo pathway-mediated inhibition of YAP1/TAZ in endocrine progenitors is a prerequisite for endocrine specification and differentiation.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
- Department of Obstetrics, The Second Xiangya Hospital, Central South UniversityChangshaChina
| | - Kunhua Qin
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
- Department of Molecular Medicine, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Yi Xu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Shreya Rajhans
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Truong Vo
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Kevin M Lopez
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Jun Liu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Michael H Nipper
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Janice Deng
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Xue Yin
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Logan R Ramjit
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Zhenqing Ye
- Department of Population Health Sciences, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Yu Luan
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - H Efsun Arda
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Pei Wang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San AntonioSan AntonioUnited States
| |
Collapse
|
4
|
Abou Nader N, Charrier L, Meisnsohn MC, Banville L, Deffrennes B, St-Jean G, Boerboom D, Zamberlam G, Brind'Amour J, Pépin D, Boyer A. Lats1 and Lats2 regulate YAP and TAZ activity to control the development of mouse Sertoli cells. FASEB J 2024; 38:e23633. [PMID: 38690712 DOI: 10.1096/fj.202400346r] [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: 02/19/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
Recent reports suggest that the Hippo signaling pathway regulates testis development, though its exact roles in Sertoli cell differentiation remain unknown. Here, we examined the functions of the main Hippo pathway kinases, large tumor suppressor homolog kinases 1 and 2 (Lats1 and Lats2) in developing mouse Sertoli cells. Conditional inactivation of Lats1/2 in Sertoli cells resulted in the disorganization and overgrowth of the testis cords, the induction of a testicular inflammatory response and germ cell apoptosis. Stimulated by retinoic acid 8 (STRA8) expression in germ cells additionally suggested that germ cells may have been preparing to enter meiosis prior to their loss. Gene expression analyses of the developing testes of conditional knockout animals further suggested impaired Sertoli cell differentiation, epithelial-to-mesenchymal transition, and the induction of a specific set of genes associated with Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)-mediated integrin signaling. Finally, the involvement of YAP/TAZ in Sertoli cell differentiation was confirmed by concomitantly inactivating Yap/Taz in Lats1/2 conditional knockout model, which resulted in a partial rescue of the testicular phenotypic changes. Taken together, these results identify Hippo signaling as a crucial pathway for Sertoli cell development and provide novel insight into Sertoli cell fate maintenance.
Collapse
Affiliation(s)
- Nour Abou Nader
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Laureline Charrier
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Marie-Charlotte Meisnsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Laurence Banville
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Bérengère Deffrennes
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
- École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Guillaume St-Jean
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Gustavo Zamberlam
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Julie Brind'Amour
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexandre Boyer
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
5
|
Jonischkies K, del Angel M, Demiray YE, Loaiza Zambrano A, Stork O. The NDR family of kinases: essential regulators of aging. Front Mol Neurosci 2024; 17:1371086. [PMID: 38803357 PMCID: PMC11129689 DOI: 10.3389/fnmol.2024.1371086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Aging is defined as a progressive decline of cognitive and physiological functions over lifetime. Since the definition of the nine hallmarks of aging in 2013 by López-Otin, numerous studies have attempted to identify the main regulators and contributors in the aging process. One interesting group of proteins whose participation has been implicated in several aging hallmarks are the nuclear DBF2-related (NDR) family of serine-threonine AGC kinases. They are one of the core components of the Hippo signaling pathway and include NDR1, NDR2, LATS1 and LATS2 in mammals, along with its highly conserved metazoan orthologs; Trc in Drosophila melanogaster, SAX-1 in Caenorhabditis elegans, CBK1, DBF20 in Saccharomyces cerevisiae and orb6 in Saccharomyces pombe. These kinases have been independently linked to the regulation of widely diverse cellular processes disrupted during aging such as the cell cycle progression, transcription, intercellular communication, nutrient homeostasis, autophagy, apoptosis, and stem cell differentiation. However, a comprehensive overview of the state-of-the-art knowledge regarding the post-translational modifications of and by NDR kinases in aging has not been conducted. In this review, we summarize the current understanding of the NDR family of kinases, focusing on their relevance to various aging hallmarks, and emphasize the growing body of evidence that suggests NDR kinases are essential regulators of aging across species.
Collapse
Affiliation(s)
- Kevin Jonischkies
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Miguel del Angel
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Yunus Emre Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Allison Loaiza Zambrano
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Science, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
6
|
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
|
7
|
Barlow HR, Ahuja N, Bierschenk T, Htike Y, Fassetta L, Azizoglu DB, Flores J, Gao N, de la O S, Sneddon JB, Marciano DK, Cleaver O. Rab11 is essential to pancreas morphogenesis, lumen formation and endocrine mass. Dev Biol 2023; 499:59-74. [PMID: 37172642 DOI: 10.1016/j.ydbio.2023.05.002] [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: 11/17/2022] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
The molecular links between tissue-level morphogenesis and the differentiation of cell lineages in the pancreas remain elusive despite a decade of studies. We previously showed that in pancreas both processes depend on proper lumenogenesis. The Rab GTPase Rab11 is essential for epithelial lumen formation in vitro, however few studies have addressed its functions in vivo and none have tested its requirement in pancreas. Here, we show that Rab11 is critical for proper pancreas development. Co-deletion of the Rab11 isoforms Rab11A and Rab11B in the developing pancreatic epithelium (Rab11pancDKO) results in ∼50% neonatal lethality and surviving adult Rab11pancDKO mice exhibit defective endocrine function. Loss of both Rab11A and Rab11B in the embryonic pancreas results in morphogenetic defects of the epithelium, including defective lumen formation and lumen interconnection. In contrast to wildtype cells, Rab11pancDKO cells initiate the formation of multiple ectopic lumens, resulting in a failure to coordinate a single apical membrane initiation site (AMIS) between groups of cells. This results in a failure to form ducts with continuous lumens. Here, we show that these defects are due to failures in vesicle trafficking, as apical and junctional components remain trapped within Rab11pancDKO cells. Together, these observations suggest that Rab11 directly regulates epithelial lumen formation and morphogenesis. Our report links intracellular trafficking to organ morphogenesis in vivo and presents a novel framework for decoding pancreatic development.
Collapse
Affiliation(s)
- Haley R Barlow
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA.
| | - Neha Ahuja
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - Tyler Bierschenk
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - Yadanar Htike
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - Luke Fassetta
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - D Berfin Azizoglu
- Department of Developmental Biology, Beckman Center, 279 W. Campus Drive, B300, Stanford, CA, 94305, USA
| | - Juan Flores
- Rutgers University Microbiome Program, 679 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Nan Gao
- Rutgers University Microbiome Program, 679 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Sean de la O
- Department of Cell and Tissue Biology, Department of Anatomy, Diabetes Center, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Julie B Sneddon
- Department of Cell and Tissue Biology, Department of Anatomy, Diabetes Center, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Denise K Marciano
- Internal Medicine and Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA.
| |
Collapse
|
8
|
Martínez Traverso IM, Steimle JD, Zhao X, Wang J, Martin JF. LATS1/2 control TGFB-directed epithelial-to-mesenchymal transition in the murine dorsal cranial neuroepithelium through YAP regulation. Development 2022; 149:dev200860. [PMID: 36125128 PMCID: PMC9587805 DOI: 10.1242/dev.200860] [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: 04/22/2022] [Accepted: 08/12/2022] [Indexed: 11/20/2022]
Abstract
Hippo signaling, an evolutionarily conserved kinase cascade involved in organ size control, plays key roles in various tissue developmental processes, but its role in craniofacial development remains poorly understood. Using the transgenic Wnt1-Cre2 driver, we inactivated the Hippo signaling components Lats1 and Lats2 in the cranial neuroepithelium of mouse embryos and found that the double conditional knockout (DCKO) of Lats1/2 resulted in neural tube and craniofacial defects. Lats1/2 DCKO mutant embryos had microcephaly with delayed and defective neural tube closure. Furthermore, neuroepithelial cell shape and architecture were disrupted within the cranial neural tube in Lats1/2 DCKO mutants. RNA sequencing of embryonic neural tubes revealed increased TGFB signaling in Lats1/2 DCKO mutants. Moreover, markers of epithelial-to-mesenchymal transition (EMT) were upregulated in the cranial neural tube. Inactivation of Hippo signaling downstream effectors, Yap and Taz, suppressed neuroepithelial defects, aberrant EMT and TGFB upregulation in Lats1/2 DCKO embryos, indicating that LATS1/2 function via YAP and TAZ. Our findings reveal important roles for Hippo signaling in modulating TGFB signaling during neural crest EMT.
Collapse
Affiliation(s)
- Idaliz M. Martínez Traverso
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey D. Steimle
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - James F. Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX 77030, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, TX 77030 , USA
| |
Collapse
|
9
|
Liu P, Li Y, Wang W, Bai Y, Jia H, Yuan Z, Yang Z. Role and mechanisms of the NF-ĸB signaling pathway in various developmental processes. Biomed Pharmacother 2022; 153:113513. [DOI: 10.1016/j.biopha.2022.113513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022] Open
|
10
|
Ahuja N, Hiltabidle MS, Rajasekhar H, Voss S, Lu SZ, Barlow HR, Cowdin MA, Daniel E, Vaddaraju V, Anandakumar T, Black E, Cleaver O, Maynard C. Endothelial Cyp26b1 restrains murine heart valve growth during development. Dev Biol 2022; 486:81-95. [DOI: 10.1016/j.ydbio.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 11/28/2022]
|
11
|
Ahuja N, Cleaver O. The cell cortex as mediator of pancreatic epithelial development and endocrine differentiation. Curr Opin Genet Dev 2022; 72:118-127. [PMID: 34929610 PMCID: PMC8915777 DOI: 10.1016/j.gde.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/03/2023]
Abstract
Organogenesis is the complex process of cells coordinating their own proliferation with changes to their shape, cell migration and cell-cell signaling, so that they transform into a three dimensional functional tissue, with its own custom range of differentiated cell types. Understanding when and where critical signals emanate from, and how those signals are transduced and interpreted, is the fundamental challenge of developmental biology. Here, we review recent findings regarding how progenitor cells interpret cues during pancreatic morphogenesis and how they coordinate cell fate determination. Recent evidence suggests that molecules located in the cell cortex play a crticial role in determining cellular behavior during pancreatic morphogenesis. Specifically, we find that control of cell adhesion, polarity, and constriction are all integral to both initiation of epithelial development and to later cell differentiation. Here, we review key molecules that coordinate these processes and suggest that the cell cortex acts as a signaling center that relays cues during pancreas development.
Collapse
Affiliation(s)
- Neha Ahuja
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
12
|
Deng H, Li M, Zheng R, Qiu H, Yuan T, Wang W, Yang Q, Long Z, Huang X. YAP Promotes Cell Proliferation and Epithelium-Derived Cytokine Expression via NF-κB Pathway in Nasal Polyps. J Asthma Allergy 2021; 14:839-850. [PMID: 34276219 PMCID: PMC8277454 DOI: 10.2147/jaa.s315707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/05/2021] [Indexed: 11/23/2022] Open
Abstract
Background Hippo-Yes-associated protein (YAP) pathway plays an important role in epithelial cell proliferation and inflammation development in chronic rhinosinusitis with nasal polyps (CRSwNP). However, the underlying mechanisms remain unclear. Objective This study intends to investigate the role of YAP and the nuclear factor kappa-B (NF-κB) pathway in cell proliferation and the expression of epithelium-derived cytokines in nasal polyps (NP). Methods The expression levels of YAP, TEA domain family member 1 (TEAD1), Ki-67, and NF-κB as well as interleukin (IL-) 33, IL-25 and thymic stromal lymphopoietin (TSLP) in sinonasal mucosa, primary nasal epithelial cells (NPECs), and human nasal epithelial RPMI 2650 cells were detected. NPECs were cultured and treated with verteporfin (VP), YAP shRNA or BAY 11–7082. Results The hippo pathway effector YAP, Ki-67, p65 NF-κB, and cyclin D1 were significantly increased in NP compared with control mucosa, which was accompanied by overexpression of IL-33, IL-25, and TSLP. Pharmaceutical inhibition of YAP by VP suppressed cell proliferation of RPMI 2650 cells by blocking cell cycle progression at G0/G1 without inducing obvious cell apoptosis. Furthermore, lentiviral transfection-mediated knockdown of hippo pathway activity reduced the expression of IL-33, IL-25, TSLP as well as p65 NF-κB in RPMI 2650 cells. Downregulation of NF-κB pathway with BAY 11–7082 in NPECs could decrease the mRNA level of TSLP, IL-33 and IL-25 accordingly. Conclusion Inhibition of hippo pathway suppressed nasal epithelial cell proliferation and declined the expression of epithelium-derived cytokines via the NF-κB pathway in NPECs.
Collapse
Affiliation(s)
- Huiyi Deng
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meijiao Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Tung wah Hospital of Sun Yat-sen University, Dongguan, People's Republic of China
| | - Rui Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huijun Qiu
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Tian Yuan
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weihao Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qintai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zijie Long
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xuekun Huang
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
13
|
Wu Y, Aegerter P, Nipper M, Ramjit L, Liu J, Wang P. Hippo Signaling Pathway in Pancreas Development. Front Cell Dev Biol 2021; 9:663906. [PMID: 34079799 PMCID: PMC8165189 DOI: 10.3389/fcell.2021.663906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo signaling pathway is a vital regulator of pancreatic development and homeostasis, directing cell fate decisions, morphogenesis, and adult pancreatic cellular plasticity. Through loss-of-function research, Hippo signaling has been found to play key roles in maintaining the proper balance between progenitor cell renewal, proliferation, and differentiation in pancreatic organogenesis. Other studies suggest that overactivation of YAP, a downstream effector of the pathway, promotes ductal cell development and suppresses endocrine cell fate specification via repression of Ngn3. After birth, disruptions in Hippo signaling have been found to lead to de-differentiation of acinar cells and pancreatitis-like phenotype. Further, Hippo signaling directs pancreatic morphogenesis by ensuring proper cell polarization and branching. Despite these findings, the mechanisms through which Hippo governs cell differentiation and pancreatic architecture are yet to be fully understood. Here, we review recent studies of Hippo functions in pancreatic development, including its crosstalk with NOTCH, WNT/β-catenin, and PI3K/Akt/mTOR signaling pathways.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States.,Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pauline Aegerter
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Michael Nipper
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Logan Ramjit
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jun Liu
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| | - Pei Wang
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
14
|
Abstract
Over 50 years after its discovery in early chick embryos, the concept of epithelial-mesenchymal transition (EMT) is now widely applied to morphogenetic studies in both physiological and pathological contexts. Indeed, the EMT field has witnessed exponential growth in recent years, driven primarily by a rapid expansion of cancer-oriented EMT research. This has led to EMT-based therapeutic interventions that bear the prospect of fighting cancer, and has given developmental biologists new impetus to investigate EMT phenomena more closely and to find suitable models to address emerging EMT-related questions. Here, and in the accompanying poster, I provide a brief summary of the current status of EMT research and give an overview of EMT models that have been used in developmental studies. I also highlight dynamic epithelialization and de-epithelialization events that are involved in many developmental processes and that should be considered to provide a broader perspective of EMT. Finally, I put forward a set of criteria to separate morphogenetic phenomena that are EMT-related from those that are not.
Collapse
Affiliation(s)
- Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| |
Collapse
|
15
|
Wang G, Hiramoto K, Ma N, Yoshikawa N, Ohnishi S, Murata M, Kawanishi S. Glycyrrhizin Attenuates Carcinogenesis by Inhibiting the Inflammatory Response in a Murine Model of Colorectal Cancer. Int J Mol Sci 2021; 22:ijms22052609. [PMID: 33807620 PMCID: PMC7961658 DOI: 10.3390/ijms22052609] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Glycyrrhizin (GL), an important active ingredient of licorice root, which weakens the proinflammatory effects of high-mobility group box 1 (HMGB1) by blocking HMGB1 signaling. In this study, we investigated whether GL could suppress inflammation and carcinogenesis in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced murine model of colorectal cancer. ICR mice were divided into four groups (n = 5, each)—control group, GL group, colon cancer (CC) group, and GL-treated CC (CC + GL) group, and sacrificed after 20 weeks. Plasma levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α were measured using an enzyme-linked immunosorbent assay. The colonic tissue samples were immunohistochemically stained with DNA damage markers (8-nitroguanine and 8-oxo-7,8-dihydro-2′-deoxy-guanosine), inflammatory markers (COX-2 and HMGB1), and stem cell markers (YAP1 and SOX9). The average number of colonic tumors and the levels of IL-6 and TNF-α in the CC + GL group were significantly lower than those in the CC group. The levels of all inflammatory and cancer markers were significantly reduced in the CC + GL group. These results suggest that GL inhibits the inflammatory response by binding HMGB1, thereby inhibiting DNA damage and cancer stem cell proliferation and dedifferentiation. In conclusion, GL significantly attenuates the pathogenesis of AOM/DSS-induced colorectal cancer by inhibiting HMGB1-TLR4-NF-κB signaling.
Collapse
Affiliation(s)
- Guifeng Wang
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan;
- Sakuranomori Shiroko Home, Social Service Elderly Facilities, Suzuka University of Medical Science, Suzuka, Mie 513-0816, Japan
| | - Keiichi Hiramoto
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan; (K.H.); (S.O.)
| | - Ning Ma
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan;
- Institute of Traditional Chinese Medicine, Suzuka University of Medical Science, Suzuka, Mie 510-0226, Japan
| | - Nobuji Yoshikawa
- Matsusaka R&D Center, Cokey Co., Ltd., Matsusaka, Mie 515-0041, Japan;
| | - Shiho Ohnishi
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan; (K.H.); (S.O.)
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan;
- Correspondence: (M.M.); (S.K.); Tel.: +81-59-231-5011 (M.M.); +81-59-340-0550 (S.K.)
| | - Shosuke Kawanishi
- Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan
- Correspondence: (M.M.); (S.K.); Tel.: +81-59-231-5011 (M.M.); +81-59-340-0550 (S.K.)
| |
Collapse
|
16
|
Wang F, Fan M, Zhou X, Yu Y, Cai Y, Wu H, Zhang Y, Liu J, Huang S, He N, Hu Z, Ding G, Jin X. A positive feedback loop between TAZ and miR-942-3p modulates proliferation, angiogenesis, epithelial-mesenchymal transition process, glycometabolism and ROS homeostasis in human bladder cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:44. [PMID: 33499877 PMCID: PMC7836562 DOI: 10.1186/s13046-021-01846-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/14/2021] [Indexed: 12/26/2022]
Abstract
Background Transcriptional coactivator with PDZ-binding motif (TAZ) has been reported to be involved in tumor progression, angiogenesis, epithelial-mesenchymal transition (EMT), glycometabolic modulation and reactive oxygen species (ROS) buildup. Herein, the underlying molecular mechanisms of the TAZ-induced biological effects in bladder cancer were discovered. Methods qRT-PCR, western blotting and immunohistochemistry were performed to determine the levels of TAZ in bladder cancer cells and tissues. CCK-8, colony formation, tube formation, wound healing and Transwell assays and flow cytometry were used to evaluate the biological functions of TAZ, miR-942-3p and growth arrest-specific 1 (GAS1). QRT-PCR and western blotting were used to determine the expression levels of related genes. Chromatin immunoprecipitation and a dual-luciferase reporter assay were performed to confirm the interaction between TAZ and miR-942. In vivo tumorigenesis and colorimetric glycolytic assays were also conducted. Results We confirmed the upregulation and vital roles of TAZ in bladder cancer. TAZ-induced upregulation of miR-942-3p expression amplified upstream signaling by inhibiting the expression of large tumor suppressor 2 (LATS2, a TAZ inhibitor). MiR-942-3p attenuated the impacts on cell proliferation, angiogenesis, EMT, glycolysis and ROS levels induced by TAZ knockdown. Furthermore, miR-942-3p restrained the expression of GAS1 to modulate biological behaviors. Conclusion Our study identified a novel positive feedback loop between TAZ and miR-942-3p that regulates biological functions in bladder cancer cells via GAS1 expression and illustrated that TAZ, miR-942-3p and GAS1 might be potential therapeutic targets for bladder cancer treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01846-5.
Collapse
Affiliation(s)
- Feifan Wang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Mengjing Fan
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, P.R. China
| | - Xuejian Zhou
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Yanlan Yu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, P. R. China
| | - Yueshu Cai
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Hongshen Wu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Yan Zhang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Jiaxin Liu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Shihan Huang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Ning He
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Zhenghui Hu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Guoqing Ding
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, P. R. China.
| | - Xiaodong Jin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China.
| |
Collapse
|
17
|
Guo P, Wang Z, Zhou Z, Tai Y, Zhang A, Wei W, Wang Q. Immuno-hippo: Research progress of the hippo pathway in autoimmune disease. Immunol Lett 2020; 230:11-20. [PMID: 33345861 DOI: 10.1016/j.imlet.2020.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 10/22/2022]
Abstract
Extensive research in Drosophila and mammals has identified the core components of Hippo signaling, which controls gene expression. Studies of Drosophila have demonstrated the highly conserved Hippo pathway controls tissue homeostasis and organ size by regulating the balance between cell proliferation and apoptosis. Recent work has indicated a potential role of the Hippo pathway in regulating the immune system, which is the key player in autoimmune disease (AID). Therefore, the Hippo pathway may become a novel target for curing AID. Although the pivotal role of both the Hippo pathway in tumorigenesis has been thoroughly investigated, the role of it in AID is still poorly understood. Elucidating the role of Hippo signaling pathways in the activation and expression of specific molecules involved in immune regulation is important for understanding the pathogenesis of AID and exploring novel therapeutic targets. To aid in further research, this review describes the relationship between the Hippo pathway and inflammatory signals such as NF-κB and JAK-STAT, the function of the Hippo pathway in the formation and differentiation of immune cells, and the regulatory role of the Hippo pathway in AID.
Collapse
Affiliation(s)
- Paipai Guo
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, Anhui 230032, China
| | - Zhen Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, Anhui 230032, China
| | - Zhengwei Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, Anhui 230032, China
| | - Yu Tai
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, Anhui 230032, China
| | - Aijun Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, Anhui 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, Anhui 230032, China.
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, Anhui 230032, China.
| |
Collapse
|
18
|
Liu J, Zhang Y, Li Q, Wang Y. Transgelins: Cytoskeletal Associated Proteins Implicated in the Metastasis of Colorectal Cancer. Front Cell Dev Biol 2020; 8:573859. [PMID: 33117801 PMCID: PMC7575706 DOI: 10.3389/fcell.2020.573859] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022] Open
Abstract
Transgelins, including transgelin-1 (T-1), transgelin-2 (T-2), and transgelin-3 (T-3), are a family of actin-binding proteins (ABPs) that can alter the structure and morphology of the cytoskeleton. These proteins function by regulating migration, proliferation and apoptosis in many different cancers. Several studies have shown that in various types of tumor cells, including colorectal cancer (CRC) cells, and in the tumor microenvironment, the expression and biological effects of transgelins are diverse and may transform during tumor progression. Previous researches have demonstrated that transgelin levels are positively correlated with metastasis in CRC, and down-regulating their expression can inhibit this process. In advanced disease, T-1 is a tumor activator with increasing expression, and T-2 expression increases with the progression of CRC. Finally, T-3 is only expressed in neurons and is not associated with CRC. This evidence suggests that T-1 and T-2 are potential biomarkers and therapeutic targets for CRC metastasis.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
19
|
Cao X, Wang C, Liu J, Zhao B. Regulation and functions of the Hippo pathway in stemness and differentiation. Acta Biochim Biophys Sin (Shanghai) 2020; 52:736-748. [DOI: 10.1093/abbs/gmaa048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract
The Hippo pathway plays important roles in organ development, tissue regeneration, and human diseases, such as cancer. In the canonical Hippo pathway, the MST1/2-LATS1/2 kinase cascade phosphorylates and inhibits transcription coactivators Yes-associated protein and transcription coactivator with PDZ-binding motif and thus regulates transcription of genes important for cell proliferation and apoptosis. However, recent studies have depicted a much more complicate picture of the Hippo pathway with many new components and regulatory stimuli involving both chemical and mechanical signals. Furthermore, accumulating evidence indicates that the Hippo pathway also plays important roles in the determination of cell fates, such as self-renewal and differentiation. Here, we review regulations of the Hippo pathway and its functions in stemness and differentiation emphasizing recent discoveries.
Collapse
Affiliation(s)
- Xiaolei Cao
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Chenliang Wang
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Jiyang Liu
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Bin Zhao
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
20
|
Wang W, Chen H, Gao W, Wang S, Wu K, Lu C, Luo X, Li L, Yu C. Girdin interaction with vimentin induces EMT and promotes the growth and metastasis of pancreatic ductal adenocarcinoma. Oncol Rep 2020; 44:637-649. [PMID: 32467989 PMCID: PMC7336503 DOI: 10.3892/or.2020.7615] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant cancer of the digestive tract that has a high potential for metastasis and a poor prognosis. Girdin was first reported in 2005 as an actin-binding protein and was designated as Akt-phosphorylation enhancer (APE); thus, Girdin has been revealed to have an important role in regulating cancer development. There is additional evidence indicating that Girdin is associated with cell proliferation, migration, invasion and survival in certain cancers. However, the potential mechanisms involving Girdin and mobility in pancreatic cancer have not been elucidated. In the present study, it was revealed that Girdin was highly expressed in pancreatic cancer tissue and was associated with tumor grade. The present study, to the best of our knowledge, is the first aimed at investigating the unknown role of Girdin in PDAC metastasis. A short hairpin RNA for Girdin (sh-Girdin) was successfully constructed with recombinant adenoviral vectors to suppress the expression of Girdin in pancreatic cancer cell lines (PANC-1 and BXPC-3). The silencing efficiency of the Girdin shRNA was determined by RT-qPCR and western blot analysis, and decreased Girdin expression in the cytoplasm was revealed by immunofluorescence detection. Then, sulforhodamine B (SRB) and colony formation assays were used to confirm that the knockdown of Girdin inhibited proliferation in vitro, and Transwell assays were used to examine the influence of Girdin knockdown on cellular mobility. Animal experiments also confirmed that silencing the expression of Girdin in pancreatic cancer cells inhibited the growth and metastasis of pancreatic cancer in vivo. Transforming growth factor-β (TGF-β) is a common inducer of epithelial-mesenchymal transition (EMT) and can effectively induce EMT in PDAC. Notably, TGF-β-treated cells exhibited changes in the classic biological markers of EMT. The expression of E-cadherin, a marker of the epithelial phenotype, increased, and the expression of N-cadherin and vimentin, markers of the interstitial phenotype, decreased in response to sh-Girdin. According to these experiments, Girdin may affect pancreatic cancer progression and development by interacting with vimentin. Therefore, there is evidence indicating that Girdin could be designated as a prognostic biological indicator and a candidate therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Wulin Wang
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Hao Chen
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Wenjie Gao
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Sheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Kai Wu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Chen Lu
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Xiagang Luo
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Lianhong Li
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Chunzhao Yu
- Department of General Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
21
|
Liao L, Liu C, Xie X, Zhou J. Betulinic acid induces apoptosis and impairs migration and invasion in a mouse model of ovarian cancer. J Food Biochem 2020; 44:e13278. [PMID: 32412117 DOI: 10.1111/jfbc.13278] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 01/01/2023]
Abstract
Betulinic acid (BA) was verified to possess plenty of biological activities including anti-tumor, anti-inflammatory and so on. In our research, we studied the growth inhibition, apoptosis promotion and metastasis resistance of ovarian cancer cells by BA. The result showed that BA showed a time- and dose-dependent inhibitory effect on ovarian cancer cell lines. SKOV3 cell line proliferation has a concentration- and time-dependently, which may be inhibited by BA. Furthermore, BA inhibited the metastasis of tumor cells remarkably by inhibiting epithelial-mesenchymal transition process. Beyond that, the weight and volume of subcutaneous tumor was distinctly suppressed by administration of BA in tumor-bearing mice of SKOV3 cells. Pathological and immunohistochemical tests showed that Ki-67+ and MMP-2+ cells were dramatically decreased after BA administration, indicating that BA can not only suppress proliferation, but also inhibit migration of tumor cells. Taken together, BA can be a valuable candidate drug for the treatment of ovarian cancer. PRACTICAL APPLICATIONS: Betulinic acid (BA) isolated from natural plants such as fenugreek, eucalyptus bulb and mulberry has been reported with many biological activities. Results from this study revealed that in vitro and in vivo BA-induced apoptosis and inhibited migration and invasion of human ovarian cancer cells. Therefore, BA from natural plants may be developed as a potential drug for inhibition the development of ovarian cancer cells.
Collapse
Affiliation(s)
- Lingyun Liao
- Department of obstetrics and gynecology, First Affiliated Hospital of Gannan Medical College, Ganzhou, P.R. China
| | - Chi Liu
- College of Medical & Life Sciences, Chengdu University of TCM, Chengdu, P.R. China
| | - Xiaoying Xie
- Department of obstetrics and gynecology, First Affiliated Hospital of Gannan Medical College, Ganzhou, P.R. China
| | - Jieli Zhou
- Department of obstetrics and gynecology, First Affiliated Hospital of Gannan Medical College, Ganzhou, P.R. China
| |
Collapse
|
22
|
|