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] [MESH Headings] [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
|
Rosales-Muñoz GJ, Souza-Arroyo V, Bucio-Ortiz L, Miranda-Labra RU, Gomez-Quiroz LE, Gutiérrez-Ruiz MC. Acute pancreatitis experimental models, advantages and disadvantages. J Physiol Biochem 2025:10.1007/s13105-025-01091-w. [PMID: 40380027 DOI: 10.1007/s13105-025-01091-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
Acute pancreatitis represents a severe health problem, not only because of the number of people affected but also because of the severity of its clinical presentation that can eventually lead to the death of patients. The study of the disease is complex, and we lack optimized models that can approach the clinical presentation in patients, in addition to the significant vulnerability of the organ itself. In the present work, we undertook the task of reviewing and analyzing the experimental methods most currently used for the induction of acute pancreatitis, emphasizing the advantages and disadvantages of each model and their delimitation based on experimental objectives. We aimed to provide an actual and quick-access guide for researchers interested in experimental acute pancreatitis.
Collapse
Affiliation(s)
- Genaro J Rosales-Muñoz
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
- Departamento de Ciencias de La Salud, Área de Medicina Experimental y Traslacional, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Verónica Souza-Arroyo
- Departamento de Ciencias de La Salud, Área de Medicina Experimental y Traslacional, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Leticia Bucio-Ortiz
- Departamento de Ciencias de La Salud, Área de Medicina Experimental y Traslacional, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Roxana U Miranda-Labra
- Departamento de Ciencias de La Salud, Área de Medicina Experimental y Traslacional, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Luis E Gomez-Quiroz
- Departamento de Ciencias de La Salud, Área de Medicina Experimental y Traslacional, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - María Concepción Gutiérrez-Ruiz
- Departamento de Ciencias de La Salud, Área de Medicina Experimental y Traslacional, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|
3
|
Neahring L, Zallen JA. Three-dimensional rosettes in epithelial formation. Cells Dev 2025:204022. [PMID: 40120722 DOI: 10.1016/j.cdev.2025.204022] [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: 12/24/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Epithelia are ubiquitous tissues with essential structural, signaling, and barrier functions. How cells transition from individual to collective behaviors as they build and remodel epithelia throughout development is a fundamental question in developmental biology. Recent studies show that three-dimensional multicellular rosettes are key intermediates that provide a solution to the challenge of building tissue-scale epithelia by coordinating local interactions in small groups of cells. These radially polarized rosette structures facilitate epithelial formation by providing a protected environment for cells to acquire apical-basal polarity, establish cell adhesion, and coordinate intercellular signaling. Once formed, rosettes can dynamically expand, move, coalesce, and interact with surrounding tissues to generate a wide range of structures with specialized functions, including epithelial sheets, tubes, cavities, and branched networks. In this review, we describe the mechanisms that regulate rosette assembly and dynamics, and discuss how rosettes serve as versatile intermediates in epithelial morphogenesis. In addition, we present open questions about the molecular, cellular, and biophysical mechanisms that drive rosette behaviors, and discuss the implications of this widely used mode of epithelial formation for understanding embryonic development and human disease.
Collapse
Affiliation(s)
- Lila Neahring
- HHMI and Developmental Biology Program, Sloan Kettering Institute, New York, NY, United States of America
| | - Jennifer A Zallen
- HHMI and Developmental Biology Program, Sloan Kettering Institute, New York, NY, United States of America.
| |
Collapse
|
4
|
Mima A, Kimura A, Ito R, Hatano Y, Tsujimoto H, Mae SI, Yamane J, Fujibuchi W, Uza N, Toyoda T, Seno H, Osafune K. Mechanistic elucidation of human pancreatic acinar development using single-cell transcriptome analysis on a human iPSC differentiation model. Sci Rep 2025; 15:4668. [PMID: 39920294 PMCID: PMC11806057 DOI: 10.1038/s41598-025-88690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
Few effective treatments have been developed for intractable pancreatic exocrine disorders due to the lack of suitable disease models using human cells. Pancreatic acinar cells differentiated from human induced pluripotent stem cells (hiPSCs) have the potential to solve this issue. In this study, we aimed to elucidate the developmental mechanisms of pancreatic exocrine acinar lineages to establish a directed differentiation method for pancreatic acinar cells from hiPSCs. hiPSC-derived pancreatic endoderm cells were spontaneously differentiated into both pancreatic exocrine and endocrine tissues by implantation into the renal subcapsular space of NOD/SCID mice. Single-cell RNA-seq analysis of the retrieved grafts confirmed the differentiation of pancreatic acinar lineage cells and identified REG4 as a candidate marker for pancreatic acinar progenitor cells. Furthermore, differential gene expression analysis revealed upregulated pathways, including cAMP-related signals, involved in the differentiation of hiPSC-derived pancreatic acinar lineage cells in vivo, and we found that a cAMP activator, forskolin, facilitates the differentiation from hiPSC-derived pancreatic endoderm into pancreatic acinar progenitor cells in our in vitro differentiation culture. Therefore, this platform contributes to our understanding of the developmental mechanisms of pancreatic acinar lineage cells and the establishment of differentiation methods for acinar cells from hiPSCs.
Collapse
Affiliation(s)
- Atsushi Mima
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Azuma Kimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ryo Ito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yu Hatano
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Junko Yamane
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Wataru Fujibuchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
5
|
Romero A, Walker BL, Krneta-Stankic V, Gerner-Mauro K, Youmans L, Miller RK. The dynamics of tubulogenesis in development and disease. Development 2025; 152:DEV202820. [PMID: 39959988 PMCID: PMC11883272 DOI: 10.1242/dev.202820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025]
Abstract
Tubes are crucial for the function of many organs in animals given their fundamental roles in transporting and exchanging substances to maintain homeostasis within an organism. Therefore, the development and maintenance of these tube-like structures within organs is a vital process. Tubes can form in diverse ways, and advances in our understanding of the molecular and cellular mechanisms underpinning these different modes of tubulogenesis have significant impacts in many biological contexts, including development and disease. This Review discusses recent progress in understanding developmental mechanisms underlying tube formation.
Collapse
Affiliation(s)
- Adrian Romero
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | - Brandy L. Walker
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Program in Genetics and Epigenetics, Houston, TX 77030, USA
| | - Vanja Krneta-Stankic
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kamryn Gerner-Mauro
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Baylor College of Medicine, Program in Development, Disease Models & Therapeutics, Houston, TX 77030, USA
| | - Lydia Youmans
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | - Rachel K. Miller
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Program in Genetics and Epigenetics, Houston, TX 77030, USA
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Molecular and Translational Biology, Houston, TX 77030, USA
| |
Collapse
|
6
|
Montero-Herradón S, García-Ceca J, Zapata AG. Thymus Ontogeny and Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:21-49. [PMID: 40067583 DOI: 10.1007/978-3-031-77921-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The thymus is a primary lymphoid organ composed of a three-dimensional (3D) epithelial network that provides a specialized microenvironment for the phenotypical and functional maturation of lymphoid progenitors. The specification of the pharyngeal endoderm to thymus fate occurs during the early stages of thymic organogenesis, independent of the expression of the transcription factor Foxn1. However, Foxn1 governs the later organogenesis of thymus together with the colonizing lymphoid cells. In the present chapter, we will review recent evidence on the topic covered in our original chapter (Muñoz and Zapata 2019). It described the early development of thymus and its resemblance to the development of endoderm-derived epithelial organs based on tubulogenesis and branching morphogenesis as well as the molecules known to be involved in these processes.
Collapse
Affiliation(s)
- Sara Montero-Herradón
- Department of Cell Biology. Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain
- Health Research Institute, Madrid, Spain
| | - Javier García-Ceca
- Department of Cell Biology. Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain
- Health Research Institute, Madrid, Spain
| | - Agustín G Zapata
- Department of Cell Biology. Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain.
- Health Research Institute, Madrid, Spain.
| |
Collapse
|
7
|
Ingthorsson S, Traustadottir GA, Gudjonsson T. Breast Morphogenesis: From Normal Development to Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:29-44. [PMID: 39821019 DOI: 10.1007/978-3-031-70875-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The human breast gland is composed of branching epithelial ducts that culminate in milk-producing units known as terminal duct lobular units (TDLUs). The epithelial compartment comprises an inner layer of luminal epithelial cells (LEP) and an outer layer of contractile myoepithelial cells (MEP). Both LEP and MEP arise from a common stem cell population. The epithelial compartment undergoes dynamic branching morphogenesis and remodelling, which expands the surface area for milk production. The epithelial remodelling that starts at the onset of menarche is largely under hormonal control, first and foremost by estrogen and progesterone from ovaries, the production of which is stimulated by pituitary-derived hormones. Menopause leads to a significant decline in estrogen and progesterone levels, resulting in involution and senescence of the breast epithelium. The branching morphogenesis involves developmental events such as epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET). EMT and MET confer plasticity to the epithelial compartment enabling the migration of epithelial cells through the stroma and restoration of the epithelial phenotype. In the normal breast, the stroma, including the basement membrane (BM), collagen-rich extracellular matrix, and various stromal cells, supports the correct histoarchitecture of the glandular tree. However, in cancer, the stroma can acquire tumour-promoting properties and is referred to as the tumour microenvironment. This chapter will explore the developmental processes including branching morphogenesis in the normal breast gland and discuss the lineage relationship between LEPS and MEPs and their interactions with the surrounding stroma in the normal and neoplastic breast gland. Finally, we will review various in vitro and in vivo models employed in mammary gland research.
Collapse
Affiliation(s)
- Saevar Ingthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Faculty of Nursing and Midwifery, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Pathology, Landspitali University Hospital, Reykjavik, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
- Department of Laboratory Hematology, Landspitali University Hospital, Reykjavik, Iceland.
| |
Collapse
|
8
|
Jin LL, Yin YL, Li FW, Zhou YM, Chen W, Tian Y, Feng X, Xu Y, Chen PF, Zhang JS, Xu HJ. Effects of FGFR2b-ligand signaling on pancreatic branching morphogenesis and postnatal islet function. J Mol Histol 2024; 56:45. [PMID: 39692915 DOI: 10.1007/s10735-024-10328-9] [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: 09/25/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024]
Abstract
Pancreatic development is a complex process vital for maintaining metabolic balance, requiring intricate interactions among different cell types and signaling pathways. Fibroblast growth factor receptors 2b (FGFR2b)-ligands signaling from adjacent mesenchymal cells is crucial in initiating pancreatic development and differentiating exocrine and endocrine cells through a paracrine mechanism. However, the precise critical time window that affects pancreatic development remains unclear. To explore the roles of FGFR2b-ligands and identify the narrow window of time during which FGFR2b-ligand signaling affects pancreatic development, we used an inducible mouse model to control the expression of soluble dominant-negative FGFR2b (sFGFR2b) at various stages of pancreatic development. Our findings revealed a significant effect of FGFR2b-ligand signaling on epithelial morphology, lumen formation, and pancreatic branching during primary and secondary transition stages. Additionally, sFGFR2b expression reduced the number of Pdx1+ progenitor cells and altered the pancreatic islet structure. Furthermore, we examined the effect of mutation in FGF10, an FGFR2b ligand, on embryonic pancreatic β-cell function. FGF10 null mutant embryos exhibited reduced pancreatic size and a decrease number of islet-like structure. Although neonatal mice with haploinsufficiency for FGF10 exhibited abnormal glucose tolerance test results, indicating a potential diabetes predisposition, these abnormalities normalized with age, aligning with observations in wild type mice. Our study underscores the critical role of FGFR2b-ligand signaling in pancreatic development and postnatal islet function, offering insights into potential therapeutic targets for pancreatic disorders.
Collapse
Affiliation(s)
- Li-Li Jin
- National Clinical Research Center for Ocular Disease, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 West Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Yi-Ling Yin
- National Clinical Research Center for Ocular Disease, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 West Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Fei-Wei Li
- International Collaborative Center On Growth Factor Research, and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yu-Mei Zhou
- National Clinical Research Center for Ocular Disease, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 West Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Wen Chen
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Ye Tian
- International Collaborative Center On Growth Factor Research, and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiao Feng
- International Collaborative Center On Growth Factor Research, and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yi Xu
- National Clinical Research Center for Ocular Disease, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 West Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Peng-Fei Chen
- National Clinical Research Center for Ocular Disease, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 West Xueyuan Road, Wenzhou, 325027, Zhejiang, China
| | - Jin-San Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| | - Hui-Jing Xu
- National Clinical Research Center for Ocular Disease, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 West Xueyuan Road, Wenzhou, 325027, Zhejiang, China.
- International Collaborative Center On Growth Factor Research, and School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
9
|
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
|
10
|
Alvarez YD, van der Spuy M, Wang JX, Noordstra I, Tan SZ, Carroll M, Yap AS, Serralbo O, White MD. A Lifeact-EGFP quail for studying actin dynamics in vivo. J Cell Biol 2024; 223:e202404066. [PMID: 38913324 PMCID: PMC11194674 DOI: 10.1083/jcb.202404066] [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: 04/12/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Here, we report the generation of a transgenic Lifeact-EGFP quail line for the investigation of actin organization and dynamics during morphogenesis in vivo. This transgenic avian line allows for the high-resolution visualization of actin structures within the living embryo, from the subcellular filaments that guide cell shape to the supracellular assemblies that coordinate movements across tissues. The unique suitability of avian embryos to live imaging facilitates the investigation of previously intractable processes during embryogenesis. Using high-resolution live imaging approaches, we present the dynamic behaviors and morphologies of cellular protrusions in different tissue contexts. Furthermore, through the integration of live imaging with computational segmentation, we visualize cells undergoing apical constriction and large-scale actin structures such as multicellular rosettes within the neuroepithelium. These findings not only enhance our understanding of tissue morphogenesis but also demonstrate the utility of the Lifeact-EGFP transgenic quail as a new model system for live in vivo investigations of the actin cytoskeleton.
Collapse
Affiliation(s)
- Yanina D. Alvarez
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Marise van der Spuy
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jian Xiong Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ivar Noordstra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Siew Zhuan Tan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Murron Carroll
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Alpha S. Yap
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Olivier Serralbo
- Commonwealth Scientific and Industrial Research (CSIRO) Health and Biosecurity, Geelong, Australia
| | - Melanie D. White
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| |
Collapse
|
11
|
Randriamanantsoa SJ, Raich MK, Saur D, Reichert M, Bausch AR. Coexisting mechanisms of luminogenesis in pancreatic cancer-derived organoids. iScience 2024; 27:110299. [PMID: 39055943 PMCID: PMC11269295 DOI: 10.1016/j.isci.2024.110299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/02/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Lumens are crucial features of the tissue architecture in both the healthy exocrine pancreas, where ducts shuttle enzymes from the acini to the intestine, and in the precancerous lesions of the highly lethal pancreatic ductal adenocarcinoma (PDAC), similarly displaying lumens that can further develop into cyst-like structures. Branched pancreatic-cancer derived organoids capture key architectural features of both the healthy and diseased pancreas, including lumens. However, their transition from a solid mass of cells to a hollow tissue remains insufficiently explored. Here, we show that organoids display two orthogonal but complementary lumen formation mechanisms: one relying on fluid intake for multiple microlumen nucleation, swelling and fusion, and the other involving the death of a central cell population, thereby hollowing out cavities. These results shed further light on the processes of luminogenesis, deepening our understanding of the early formation of PDAC precancerous lesions, including cystic neoplasia.
Collapse
Affiliation(s)
- Samuel J. Randriamanantsoa
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| | - Marion K. Raich
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| | - Dieter Saur
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, 81675 Munich, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner site Munich, 69120 Heidelberg, Germany
| | - Maximilian Reichert
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, 81675 Munich, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner site Munich, 69120 Heidelberg, Germany
- Technical University of Munich, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, Translational Pancreatic Cancer Research Center, 81675 Munich, Germany
| | - Andreas R. Bausch
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| |
Collapse
|
12
|
Zhao J, Liang S, Cen HH, Li Y, Baker RK, Ruprai B, Gao G, Zhang C, Ren H, Tang C, Chen L, Liu Y, Lynn FC, Johnson JD, Kieffer TJ. PDX1+ cell budding morphogenesis in a stem cell-derived islet spheroid system. Nat Commun 2024; 15:5894. [PMID: 39003281 PMCID: PMC11246529 DOI: 10.1038/s41467-024-50109-2] [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: 01/06/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024] Open
Abstract
Remarkable advances in protocol development have been achieved to manufacture insulin-secreting islets from human pluripotent stem cells (hPSCs). Distinct from current approaches, we devised a tunable strategy to generate islet spheroids enriched for major islet cell types by incorporating PDX1+ cell budding morphogenesis into staged differentiation. In this process that appears to mimic normal islet morphogenesis, the differentiating islet spheroids organize with endocrine cells that are intermingled or arranged in a core-mantle architecture, accompanied with functional heterogeneity. Through in vitro modelling of human pancreas development, we illustrate the importance of PDX1 and the requirement for EphB3/4 signaling in eliciting cell budding morphogenesis. Using this new approach, we model Mitchell-Riley syndrome with RFX6 knockout hPSCs illustrating unexpected morphogenesis defects in the differentiation towards islet cells. The tunable differentiation system and stem cell-derived islet models described in this work may facilitate addressing fundamental questions in islet biology and probing human pancreas diseases.
Collapse
Affiliation(s)
- Jia Zhao
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada.
| | - Shenghui Liang
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Haoning Howard Cen
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Yanjun Li
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, China
| | - Robert K Baker
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Balwinder Ruprai
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Guang Gao
- Imaging Core Facility, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Chloe Zhang
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Huixia Ren
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, China
- Center for Quantitative Biology, Peking University, Beijing, China
| | - Chao Tang
- Center for Quantitative Biology, Peking University, Beijing, China
| | - Liangyi Chen
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yanmei Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, South China Normal University, 510631, Guangzhou, China
- Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, 510631, Guangzhou, China
| | - Francis C Lynn
- BC Children's Hospital Research Institute, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - James D Johnson
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Life Sciences Institute, Departments of Cellular & Physiological Sciences and Surgery, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
13
|
Edri S, Rosenthal V, Ginsburg O, Newman Frisch A, Pierreux CE, Sharon N, Levenberg S. 3D model of mouse embryonic pancreas and endocrine compartment using stem cell-derived mesoderm and pancreatic progenitors. iScience 2024; 27:109959. [PMID: 38832019 PMCID: PMC11144751 DOI: 10.1016/j.isci.2024.109959] [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: 11/27/2023] [Revised: 03/21/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024] Open
Abstract
The developing mouse pancreas is surrounded by mesoderm compartments providing signals that induce pancreas formation. Most pancreatic organoid protocols lack this mesoderm niche and only partially capture the pancreatic cell repertoire. This work aims to generate pancreatic aggregates by differentiating mouse embryonic stem cells (mESCs) into mesoderm progenitors (MPs) and pancreas progenitors (PPs), without using Matrigel. First, mESCs were differentiated into epiblast stem cells (EpiSCs) to enhance the PP differentiation rate. Next, PPs and MPs aggregated together giving rise to various pancreatic cell types, including endocrine, acinar, and ductal cells, and to endothelial cells. Single-cell RNA sequencing analysis revealed a larger endocrine population within the PP + MP aggregates, as compared to PPs alone or PPs in Matrigel aggregates. The PP + MP aggregate gene expression signatures and its endocrine population percentage closely resembled those of the endocrine population found in the mouse embryonic pancreas, which holds promise for studying pancreas development.
Collapse
Affiliation(s)
- Shlomit Edri
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Vardit Rosenthal
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Or Ginsburg
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Abigail Newman Frisch
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | | | - Nadav Sharon
- Faculty of Biology, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
14
|
Morales EA, Wang S. Salivary gland developmental mechanics. Curr Top Dev Biol 2024; 160:1-30. [PMID: 38937029 DOI: 10.1016/bs.ctdb.2024.05.002] [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] [Indexed: 06/29/2024]
Abstract
The salivary gland undergoes branching morphogenesis to elaborate into a tree-like structure with numerous saliva-secreting acinar units, all joined by a hierarchical ductal system. The expansive epithelial surface generated by branching morphogenesis serves as the structural basis for the efficient production and delivery of saliva. Here, we elucidate the process of salivary gland morphogenesis, emphasizing the role of mechanics. Structurally, the developing salivary gland is characterized by a stratified epithelium tightly encased by the basement membrane, which is in turn surrounded by a mesenchyme consisting of a dense network of interstitial matrix and mesenchymal cells. Diverse cell types and extracellular matrices bestow this developing organ with organized, yet spatially varied mechanical properties. For instance, the surface epithelial sheet of the bud is highly fluidic due to its high cell motility and weak cell-cell adhesion, rendering it highly pliable. In contrast, the inner core of the bud is more rigid, characterized by reduced cell motility and strong cell-cell adhesion, which likely provide structural support for the tissue. The interactions between the surface epithelial sheet and the inner core give rise to budding morphogenesis. Furthermore, the basement membrane and the mesenchyme offer mechanical constraints that could play a pivotal role in determining the higher-order architecture of a fully mature salivary gland.
Collapse
Affiliation(s)
- E Angelo Morales
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Shaohe Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States.
| |
Collapse
|
15
|
Villaca CBP, Mastracci TL. Pancreatic Crosstalk in the Disease Setting: Understanding the Impact of Exocrine Disease on Endocrine Function. Compr Physiol 2024; 14:5371-5387. [PMID: 39109973 PMCID: PMC11425433 DOI: 10.1002/cphy.c230008] [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] [Indexed: 08/15/2024]
Abstract
The exocrine and endocrine are functionally distinct compartments of the pancreas that have traditionally been studied as separate entities. However, studies of embryonic development, adult physiology, and disease pathogenesis suggest there may be critical communication between exocrine and endocrine cells. In fact, the incidence of the endocrine disease diabetes secondary to exocrine disease/dysfunction ranges from 25% to 80%, depending on the type and severity of the exocrine pathology. Therefore, it is necessary to investigate how exocrine-endocrine "crosstalk" may impact pancreatic function. In this article, we discuss common exocrine diseases, including cystic fibrosis, acute, hereditary, and chronic pancreatitis, and the impact of these exocrine diseases on endocrine function. Additionally, we review how obesity and fatty pancreas influence exocrine function and the impact on cellular communication between the exocrine and endocrine compartments. Interestingly, in all pathologies, there is evidence that signals from the exocrine disease contribute to endocrine dysfunction and the progression to diabetes. Continued research efforts to identify the mechanisms that underlie the crosstalk between various cell types in the pancreas are critical to understanding normal pancreatic physiology as well as disease states. © 2024 American Physiological Society. Compr Physiol 14:5371-5387, 2024.
Collapse
Affiliation(s)
| | - Teresa L Mastracci
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
16
|
Rojek KO, Wrzos A, Żukowski S, Bogdan M, Lisicki M, Szymczak P, Guzowski J. Long-term day-by-day tracking of microvascular networks sprouting in fibrin gels: From detailed morphological analyses to general growth rules. APL Bioeng 2024; 8:016106. [PMID: 38327714 PMCID: PMC10849774 DOI: 10.1063/5.0180703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/04/2024] [Indexed: 02/09/2024] Open
Abstract
Understanding and controlling of the evolution of sprouting vascular networks remains one of the basic challenges in tissue engineering. Previous studies on the vascularization dynamics have typically focused only on the phase of intense growth and often lacked spatial control over the initial cell arrangement. Here, we perform long-term day-by-day analysis of tens of isolated microvasculatures sprouting from endothelial cell-coated spherical beads embedded in an external fibrin gel. We systematically study the topological evolution of the sprouting networks over their whole lifespan, i.e., for at least 14 days. We develop a custom image analysis toolkit and quantify (i) the overall length and area of the sprouts, (ii) the distributions of segment lengths and branching angles, and (iii) the average number of branch generations-a measure of network complexity. We show that higher concentrations of vascular endothelial growth factor (VEGF) lead to earlier sprouting and more branched networks, yet without significantly affecting the speed of growth of individual sprouts. We find that the mean branching angle is weakly dependent on VEGF and typically in the range of 60°-75°, suggesting that, by comparison with the available diffusion-limited growth models, the bifurcating tips tend to follow local VEGF gradients. At high VEGF concentrations, we observe exponential distributions of segment lengths, which signify purely stochastic branching. Our results-due to their high statistical relevance-may serve as a benchmark for predictive models, while our new image analysis toolkit, offering unique features and high speed of operation, could be exploited in future angiogenic drug tests.
Collapse
Affiliation(s)
- Katarzyna O. Rojek
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Antoni Wrzos
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | | | - Michał Bogdan
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Lisicki
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Piotr Szymczak
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Jan Guzowski
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
17
|
Darrigrand JF, Salowka A, Torres-Cano A, Tapia-Rojo R, Zhu T, Garcia-Manyes S, Spagnoli FM. Acinar-ductal cell rearrangement drives branching morphogenesis of the murine pancreas in an IGF/PI3K-dependent manner. Dev Cell 2024; 59:326-338.e5. [PMID: 38237591 PMCID: PMC11805742 DOI: 10.1016/j.devcel.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 02/08/2024]
Abstract
During organ formation, progenitor cells need to acquire different cell identities and organize themselves into distinct structural units. How these processes are coordinated and how tissue architecture(s) is preserved despite the dramatic cell rearrangements occurring in developing organs remain unclear. Here, we identified cellular rearrangements between acinar and ductal progenitors as a mechanism to drive branching morphogenesis in the pancreas while preserving the integrity of the acinar-ductal functional unit. Using ex vivo and in vivo mouse models, we found that pancreatic ductal cells form clefts by protruding and pulling on the acinar basement membrane, which leads to acini splitting. Newly formed acini remain connected to the bifurcated branches generated by ductal cell rearrangement. Insulin growth factor (IGF)/phosphatidylinositol 3-kinase (PI3K) pathway finely regulates this process by controlling pancreatic ductal tissue fluidity, with a simultaneous impact on branching and cell fate acquisition. Together, our results explain how acinar structure multiplication and branch bifurcation are synchronized during pancreas organogenesis.
Collapse
Affiliation(s)
- Jean-Francois Darrigrand
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK
| | - Anna Salowka
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK
| | - Alejo Torres-Cano
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK
| | - Rafael Tapia-Rojo
- Department of Physics, London Centre for Nanotechnology, King's College London, London, UK
| | - Tong Zhu
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, London, UK; Single-Molecule Mechanobiology Laboratory, The Francis Crick Institute, London, UK
| | - Sergi Garcia-Manyes
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, London, UK; Single-Molecule Mechanobiology Laboratory, The Francis Crick Institute, London, UK
| | - Francesca M Spagnoli
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, Great Maze Pond, SE1 9RT London, UK.
| |
Collapse
|
18
|
Hrncir HR, Bombin S, Goodloe B, Hogan CB, Jadi O, Gracz AD. Sox9 links biliary maturation to branching morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.574730. [PMID: 38293117 PMCID: PMC10827067 DOI: 10.1101/2024.01.15.574730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Branching morphogenesis couples cellular differentiation with development of tissue architecture. Intrahepatic bile duct (IHBD) morphogenesis is initiated with biliary epithelial cell (BEC) specification and eventually forms a heterogeneous network of large ducts and small ductules. Here, we show that Sox9 is required for developmental establishment of small ductules. IHBDs emerge as a webbed structure by E15.5 and undergo morphological maturation through 2 weeks of age. Developmental knockout of Sox9 leads to decreased postnatal branching morphogenesis, manifesting as loss of ductules in adult livers. In the absence of Sox9, BECs fail to mature and exhibit elevated TGF-β signaling and Activin A. Activin A induces developmental gene expression and morphological defects in BEC organoids and represses ductule formation in postnatal livers. Our data demonstrate that adult IHBD morphology and BEC maturation is regulated by the Sox9-dependent formation of precursors to ductules during development, mediated in part by downregulation of Activin A.
Collapse
Affiliation(s)
- Hannah R Hrncir
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
| | - Sergei Bombin
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
| | - Brianna Goodloe
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
| | - Connor B Hogan
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
| | - Othmane Jadi
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Adam D Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University. Atlanta, GA USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University
- Lead contact:
| |
Collapse
|
19
|
Agerskov RH, Nyeng P. Innervation of the pancreas in development and disease. Development 2024; 151:dev202254. [PMID: 38265192 DOI: 10.1242/dev.202254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
The autonomic nervous system innervates the pancreas by sympathetic, parasympathetic and sensory branches during early organogenesis, starting with neural crest cell invasion and formation of an intrinsic neuronal network. Several studies have demonstrated that signals from pancreatic neural crest cells direct pancreatic endocrinogenesis. Likewise, autonomic neurons have been shown to regulate pancreatic islet formation, and have also been implicated in type I diabetes. Here, we provide an overview of recent progress in mapping pancreatic innervation and understanding the interactions between pancreatic neurons, epithelial morphogenesis and cell differentiation. Finally, we discuss pancreas innervation as a factor in the development of diabetes.
Collapse
Affiliation(s)
- Rikke Hoegsberg Agerskov
- Roskilde University, Department of Science and Environment, Universitetsvej 1, building 28, Roskilde 4000, Denmark
| | - Pia Nyeng
- Roskilde University, Department of Science and Environment, Universitetsvej 1, building 28, Roskilde 4000, Denmark
| |
Collapse
|
20
|
Dale DJ, Rutan CD, Mastracci TL. Development of the Pancreatic Ducts and Their Contribution to Organogenesis. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2024; 239:31-55. [PMID: 39283481 PMCID: PMC11934529 DOI: 10.1007/978-3-031-62232-8_2] [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] [Indexed: 09/25/2024]
Abstract
The pancreas is a dual-function organ, with exocrine cells that aid in digestion and endocrine cells that regulate glucose homeostasis. These cell types share common progenitors and arise from the embryonic ducts. Early signaling events in the embryonic ducts shape the neonatal, adolescent, and adult exocrine and endocrine pancreas. This chapter discusses recent advances in the tools used to study the ducts and our current understanding of how ductal development contributes to pancreatic organogenesis.
Collapse
Affiliation(s)
- Dorian J Dale
- Department of Biology, Indiana University-Indianapolis, Indianapolis, IN, USA
| | - Caleb D Rutan
- Department of Biology, Indiana University-Indianapolis, Indianapolis, IN, USA
| | - Teresa L Mastracci
- Department of Biology, Indiana University-Indianapolis, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
21
|
Miotto M, Rosito M, Paoluzzi M, de Turris V, Folli V, Leonetti M, Ruocco G, Rosa A, Gosti G. Collective behavior and self-organization in neural rosette morphogenesis. Front Cell Dev Biol 2023; 11:1134091. [PMID: 37635866 PMCID: PMC10448396 DOI: 10.3389/fcell.2023.1134091] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Neural rosettes develop from the self-organization of differentiating human pluripotent stem cells. This process mimics the emergence of the embryonic central nervous system primordium, i.e., the neural tube, whose formation is under close investigation as errors during such process result in severe diseases like spina bifida and anencephaly. While neural tube formation is recognized as an example of self-organization, we still do not understand the fundamental mechanisms guiding the process. Here, we discuss the different theoretical frameworks that have been proposed to explain self-organization in morphogenesis. We show that an explanation based exclusively on stem cell differentiation cannot describe the emergence of spatial organization, and an explanation based on patterning models cannot explain how different groups of cells can collectively migrate and produce the mechanical transformations required to generate the neural tube. We conclude that neural rosette development is a relevant experimental 2D in-vitro model of morphogenesis because it is a multi-scale self-organization process that involves both cell differentiation and tissue development. Ultimately, to understand rosette formation, we first need to fully understand the complex interplay between growth, migration, cytoarchitecture organization, and cell type evolution.
Collapse
Affiliation(s)
- Mattia Miotto
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Maria Rosito
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
- Department of Physiology and Pharmacology V. Erspamer, Sapienza University of Rome, Rome, Italy
| | - Matteo Paoluzzi
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Barcelona, Spain
| | - Valeria de Turris
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Viola Folli
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
- D-TAILS srl, Rome, Italy
| | - Marco Leonetti
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
- D-TAILS srl, Rome, Italy
- Soft and Living Matter Laboratory, Institute of Nanotechnology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Alessandro Rosa
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Giorgio Gosti
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
- Soft and Living Matter Laboratory, Institute of Nanotechnology, Consiglio Nazionale delle Ricerche, Rome, Italy
| |
Collapse
|
22
|
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
|
23
|
Gredler ML, Zallen JA. Multicellular rosettes link mesenchymal-epithelial transition to radial intercalation in the mouse axial mesoderm. Dev Cell 2023:S1534-5807(23)00134-X. [PMID: 37080203 DOI: 10.1016/j.devcel.2023.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
Mesenchymal-epithelial transitions are fundamental drivers of development and disease, but how these behaviors generate epithelial structure is not well understood. Here, we show that mesenchymal-epithelial transitions promote epithelial organization in the mouse node and notochordal plate through the assembly and radial intercalation of three-dimensional rosettes. Axial mesoderm rosettes acquire junctional and apical polarity, develop a central lumen, and dynamically expand, coalesce, and radially intercalate into the surface epithelium, converting mesenchymal-epithelial transitions into higher-order tissue structure. In mouse Par3 mutants, axial mesoderm rosettes establish central tight junction polarity but fail to form an expanded apical domain and lumen. These defects are associated with altered rosette dynamics, delayed radial intercalation, and formation of a small, fragmented surface epithelial structure. These results demonstrate that three-dimensional rosette behaviors translate mesenchymal-epithelial transitions into collective radial intercalation and epithelial formation, providing a strategy for building epithelial sheets from individual self-organizing units in the mammalian embryo.
Collapse
Affiliation(s)
- Marissa L Gredler
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.
| |
Collapse
|
24
|
Szlachcic WJ, Letai KC, Scavuzzo MA, Borowiak M. Deep into the niche: Deciphering local endoderm-microenvironment interactions in development, homeostasis, and disease of pancreas and intestine. Bioessays 2023; 45:e2200186. [PMID: 36871153 DOI: 10.1002/bies.202200186] [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: 09/16/2022] [Revised: 01/11/2023] [Accepted: 01/23/2023] [Indexed: 03/06/2023]
Abstract
Unraveling molecular and functional heterogeneity of niche cells within the developing endoderm could resolve mechanisms of tissue formation and maturation. Here, we discuss current unknowns in molecular mechanisms underlying key developmental events in pancreatic islet and intestinal epithelial formation. Recent breakthroughs in single-cell and spatial transcriptomics, paralleled with functional studies in vitro, reveal that specialized mesenchymal subtypes drive the formation and maturation of pancreatic endocrine cells and islets via local interactions with epithelium, neurons, and microvessels. Analogous to this, distinct intestinal niche cells regulate both epithelial development and homeostasis throughout life. We propose how this knowledge can be used to progress research in the human context using pluripotent stem cell-derived multilineage organoids. Overall, understanding the interactions between the multitude of microenvironmental cells and how they drive tissue development and function could help us make more therapeutically relevant in vitro models.
Collapse
Affiliation(s)
- Wojciech J Szlachcic
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Katherine C Letai
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Marissa A Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
25
|
Papadakos SP, Dedes N, Gkolemi N, Machairas N, Theocharis S. The EPH/Ephrin System in Pancreatic Ductal Adenocarcinoma (PDAC): From Pathogenesis to Treatment. Int J Mol Sci 2023; 24:3015. [PMID: 36769332 PMCID: PMC9917762 DOI: 10.3390/ijms24033015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/09/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major concern for health care systems worldwide, since its mortality remains unaltered despite the surge in cutting-edge science. The EPH/ephrin signaling system was first investigated in the 1980s. EPH/ephrins have been shown to exert bidirectional signaling and cell-to-cell communication, influencing cellular morphology, adhesion, migration and invasion. Recent studies have highlighted the critical role of the EPH/ephrin system in various physiologic processes, including cellular proliferation, survival, synaptic plasticity and angiogenesis. Thus, it has become evident that the EPH/ephrin signaling system may have compelling effects on cell homeostasis that contribute to carcinogenesis. In particular, the EPH/ephrins have an impact on pancreatic morphogenesis and development, whereas several EPHs and ephrins are altered in PDAC. Several clinical and preclinical studies have attempted to elucidate the effects of the EPH/ephrin pathway, with multilayered effects on PDAC development. These studies have highlighted its highly promising role in the diagnosis, prognosis and therapeutic management of PDAC. The aim of this review is to explore the obscure aspects of the EPH/ephrin system concerning the development, physiology and homeostasis of the pancreas.
Collapse
Affiliation(s)
- Stavros P. Papadakos
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Dedes
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolina Gkolemi
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Machairas
- Second Department of Propaedeutic Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 11527 Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
26
|
Bele S, Wokasch AS, Gannon M. Epigenetic modulation of cell fate during pancreas development. TRENDS IN DEVELOPMENTAL BIOLOGY 2023; 16:1-27. [PMID: 38873037 PMCID: PMC11173269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Epigenetic modifications to DNA and its associated proteins affect cell plasticity and cell fate restrictions throughout embryonic development. Development of the vertebrate pancreas is characterized by initial is an over-lapping expression of a set of transcriptional regulators in a defined region of the posterior foregut endoderm that collectively promote pancreas progenitor specification and proliferation. As development progresses, these transcription factors segregate into distinct pancreatic lineages, with some being maintained in specific subsets of terminally differentiated pancreas cell types throughout adulthood. Here we describe the progressive stages and cell fate restrictions that occur during pancreas development and the relevant known epigenetic regulatory events that drive the dynamic expression patterns of transcription factors that regulate pancreas development. In addition, we highlight how changes in epigenetic marks can affect susceptibility to pancreas diseases (such as diabetes), adult pancreas cell plasticity, and the ability to derive replacement insulin-producing β cells for the treatment of diabetes.
Collapse
Affiliation(s)
- Shilpak Bele
- Department of Medicine, Vanderbilt University Medical Center, 2213 Garland Avenue, Nashville, TN, 37232, USA
| | - Anthony S. Wokasch
- Department of Cell and Developmental Biology, Vanderbilt University, 2213 Garland Avenue, Nashville, TN, 37232, USA
| | - Maureen Gannon
- Department of Medicine, Vanderbilt University Medical Center, 2213 Garland Avenue, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, 2213 Garland Avenue, Nashville, TN, 37232, USA
- Department of Veterans Affairs Tennessee Valley Authority, Research Division, 1310 24 Avenue South, Nashville, TN, 37212, USA
- Department of Molecular Physiology and Biophysics, 2213 Garland Avenue, Nashville, TN, 37232, USA
| |
Collapse
|
27
|
Paramore SV, Goodwin K, Nelson CM. How to build an epithelial tree. Phys Biol 2022; 19. [DOI: 10.1088/1478-3975/ac9e38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022]
Abstract
Abstract
Nature has evolved a variety of mechanisms to build epithelial trees of diverse architectures within different organs and across species. Epithelial trees are elaborated through branch initiation and extension, and their morphogenesis ends with branch termination. Each of these steps of the branching process can be driven by the actions of epithelial cells themselves (epithelial-intrinsic mechanisms) or by the cells of their surrounding tissues (epithelial-extrinsic mechanisms). Here, we describe examples of how these mechanisms drive each stage of branching morphogenesis, drawing primarily from studies of the lung, kidney, salivary gland, mammary gland, and pancreas, all of which contain epithelial trees that form through collective cell behaviors. Much of our understanding of epithelial branching comes from experiments using mice, but we also include examples here from avian and reptilian models. Throughout, we highlight how distinct mechanisms are employed in different organs and species to build epithelial trees. We also highlight how similar morphogenetic motifs are used to carry out conserved developmental programs or repurposed to support novel ones. Understanding the unique strategies used by nature to build branched epithelia from across the tree of life can help to inspire creative solutions to problems in tissue engineering and regenerative medicine.
Collapse
|
28
|
Rodriguez UA, Dahiya S, Raymond ML, Gao C, Martins-Cargill CP, Piganelli JD, Gittes GK, Hu J, Esni F. Focal adhesion kinase-mediated signaling controls the onset of pancreatic cell differentiation. Development 2022; 149:dev200761. [PMID: 36017799 PMCID: PMC9482336 DOI: 10.1242/dev.200761] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022]
Abstract
Signals from the endothelium play a pivotal role in pancreatic lineage commitment. As such, the fate of the epithelial cells relies heavily on the spatiotemporal recruitment of the endothelial cells to the embryonic pancreas. Although it is known that VEGFA secreted by the epithelium recruits the endothelial cells to the specific domains within the developing pancreas, the mechanism that controls the timing of such recruitment is poorly understood. Here, we have assessed the role of focal adhesion kinase (FAK) in mouse pancreatic development based on our observation that the presence of the enzymatically active form of FAK (pFAK) in the epithelial cells is inversely correlated with vessel recruitment. To study the role of FAK in the pancreas, we conditionally deleted the gene encoding focal adhesion kinase in the developing mouse pancreas. We found that homozygous deletion of Fak (Ptk2) during embryogenesis resulted in ectopic epithelial expression of VEGFA, abnormal endothelial recruitment and a delay in endocrine and acinar cell differentiation. The heterozygous mutants were born with no pancreatic phenotype but displayed gradual acinar atrophy due to cell polarity defects in exocrine cells. Together, our findings imply a role for FAK in controlling the timing of pancreatic lineage commitment and/or differentiation in the embryonic pancreas by preventing endothelial recruitment to the embryonic pancreatic epithelium.
Collapse
Affiliation(s)
- Uylissa A. Rodriguez
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Shakti Dahiya
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Michelle L. Raymond
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Chenxi Gao
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA 15244, USA
| | - Christina P. Martins-Cargill
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Jon D. Piganelli
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - George K. Gittes
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Jing Hu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA 15244, USA
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15244, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15123, USA
| |
Collapse
|
29
|
Glorieux L, Sapala A, Willnow D, Moulis M, Salowka A, Darrigrand JF, Edri S, Schonblum A, Sakhneny L, Schaumann L, Gómez HF, Lang C, Conrad L, Guillemot F, Levenberg S, Landsman L, Iber D, Pierreux CE, Spagnoli FM. Development of a 3D atlas of the embryonic pancreas for topological and quantitative analysis of heterologous cell interactions. Development 2022; 149:274013. [PMID: 35037942 PMCID: PMC8918780 DOI: 10.1242/dev.199655] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023]
Abstract
Generating comprehensive image maps, while preserving spatial three-dimensional (3D) context, is essential in order to locate and assess quantitatively specific cellular features and cell-cell interactions during organ development. Despite recent advances in 3D imaging approaches, our current knowledge of the spatial organization of distinct cell types in the embryonic pancreatic tissue is still largely based on two-dimensional histological sections. Here, we present a light-sheet fluorescence microscopy approach to image the pancreas in three dimensions and map tissue interactions at key time points in the mouse embryo. We demonstrate the utility of the approach by providing volumetric data, 3D distribution of three main cellular components (epithelial, mesenchymal and endothelial cells) within the developing pancreas, and quantification of their relative cellular abundance within the tissue. Interestingly, our 3D images show that endocrine cells are constantly and increasingly in contact with endothelial cells forming small vessels, whereas the interactions with mesenchymal cells decrease over time. These findings suggest distinct cell-cell interaction requirements for early endocrine cell specification and late differentiation. Lastly, we combine our image data in an open-source online repository (referred to as the Pancreas Embryonic Cell Atlas). Summary: A light-sheet fluorescence microscopy approach is used for 3D imaging of the pancreas and to quantitatively map its interactions with surrounding tissues at key development time points in the mouse embryo.
Collapse
Affiliation(s)
- Laura Glorieux
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe 1200, Belgium
| | - Aleksandra Sapala
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - David Willnow
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Manon Moulis
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe 1200, Belgium
| | - Anna Salowka
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Jean-Francois Darrigrand
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Shlomit Edri
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Anat Schonblum
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lina Sakhneny
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Laura Schaumann
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Harold F Gómez
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Christine Lang
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Lisa Conrad
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | | | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dagmar Iber
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | | | - Francesca M Spagnoli
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
30
|
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
|
31
|
Waters BJ, Blum B. Axon Guidance Molecules in the Islets of Langerhans. Front Endocrinol (Lausanne) 2022; 13:869780. [PMID: 35498433 PMCID: PMC9048200 DOI: 10.3389/fendo.2022.869780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Abstract
The islets of Langerhans, responsible for regulating blood glucose in vertebrates, are clusters of endocrine cells distributed throughout the exocrine pancreas. The spatial architecture of the different cell types within the islets controls cell-cell communication and impacts their ability to collectively regulate glucose. Islets rely on a range of chemotactic and adhesive cues to establish and manage intercellular relationships. Growing evidence indicates that axon guidance molecules such as Slit-Robo, Semaphorin-Neuropilin, Ephrin-Eph, and Netrins, influence endocrine progenitors' cell migration to establish correct architecture during islet morphogenesis, as well as directly regulating physical cell-cell communication in the mature islet to coordinate hormone secretion. In this mini-review, we discuss what is known and not yet known about how axon guidance molecules contribute to islet morphogenesis and function.
Collapse
Affiliation(s)
| | - Barak Blum
- *Correspondence: Bayley J. Waters, ; Barak Blum,
| |
Collapse
|
32
|
Abstract
Fluid secretion by exocrine glandular organs is essential to the survival of mammals. Each glandular unit within the body is uniquely organized to carry out its own specific functions, with failure to establish these specialized structures resulting in impaired organ function. Here, we review glandular organs in terms of shared and divergent architecture. We first describe the structural organization of the diverse glandular secretory units (the end-pieces) and their fluid transporting systems (the ducts) within the mammalian system, focusing on how tissue architecture corresponds to functional output. We then highlight how defects in development of end-piece and ductal architecture impacts secretory function. Finally, we discuss how knowledge of exocrine gland structure-function relationships can be applied to the development of new diagnostics, regenerative approaches and tissue regeneration.
Collapse
Affiliation(s)
- Sameed Khan
- Department of Obstetrics Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Sarah Fitch
- Department of Obstetrics Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Sarah Knox
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA
| | - Ripla Arora
- Department of Obstetrics Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
33
|
Scoville DW, Jetten AM. GLIS3: A Critical Transcription Factor in Islet β-Cell Generation. Cells 2021; 10:cells10123471. [PMID: 34943978 PMCID: PMC8700524 DOI: 10.3390/cells10123471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/23/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022] Open
Abstract
Understanding of pancreatic islet biology has greatly increased over the past few decades based in part on an increased understanding of the transcription factors that guide this process. One such transcription factor that has been increasingly tied to both β-cell development and the development of diabetes in humans is GLIS3. Genetic deletion of GLIS3 in mice and humans induces neonatal diabetes, while single nucleotide polymorphisms (SNPs) in GLIS3 have been associated with both Type 1 and Type 2 diabetes. As a significant progress has been made in understanding some of GLIS3’s roles in pancreas development and diabetes, we sought to compare current knowledge on GLIS3 within the pancreas to that of other islet enriched transcription factors. While GLIS3 appears to regulate similar genes and pathways to other transcription factors, its unique roles in β-cell development and maturation make it a key target for future studies and therapy.
Collapse
|
34
|
Malinova A, Veghini L, Real FX, Corbo V. Cell Lineage Infidelity in PDAC Progression and Therapy Resistance. Front Cell Dev Biol 2021; 9:795251. [PMID: 34926472 PMCID: PMC8675127 DOI: 10.3389/fcell.2021.795251] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022] Open
Abstract
Infidelity to cell fate occurs when differentiated cells lose their original identity and either revert to a more multipotent state or transdifferentiate into a different cell type, either within the same embryonic lineage or in an entirely different one. Whilst in certain circumstances, such as in wound repair, this process is beneficial, it can be hijacked by cancer cells to drive disease initiation and progression. Cell phenotype switching has been shown to also serve as a mechanism of drug resistance in some epithelial cancers. In pancreatic ductal adenocarcinoma (PDAC), the role of lineage infidelity and phenotype switching is still unclear. Two consensus molecular subtypes of PDAC have been proposed that mainly reflect the existence of cell lineages with different degrees of fidelity to pancreatic endodermal precursors. Indeed, the classical subtype of PDAC is characterised by the expression of endodermal lineage specifying transcription factors, while the more aggressive basal-like/squamous subtype is defined by epigenetic downregulation of endodermal genes and alterations in chromatin modifiers. Here, we summarise the current knowledge of mechanisms (genetic and epigenetic) of cell fate switching in PDAC and discuss how pancreatic organoids might help increase our understanding of both cell-intrinsic and cell-extrinsic factors governing lineage infidelity during the distinct phases of PDAC evolution.
Collapse
Affiliation(s)
- Antonia Malinova
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Francisco X. Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain
- CIBERONC, Madrid, Spain
- Department de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
35
|
Shin JH, Jeong CW. Zipper Is Necessary for Branching Morphogenesis of the Terminal Cells in the Drosophila melanogaster's Tracheal System. BIOLOGY 2021; 10:biology10080729. [PMID: 34439961 PMCID: PMC8389600 DOI: 10.3390/biology10080729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary The Drosophila melanogaster, also commonly known as the fruit fly, has a relatively simple structure, allowing scientists to study its anatomy. This research was carried out to investigate how a protein called Zipper may be important for the development of the model organism during the early developmental stages. The study concentrated on the respiratory system, also known as the tracheal system, more specifically the leading cells in the tracheal system also known as terminal cells. Zipper was shown to be in the cytoplasm of terminal cells, indicating that it may function in the D. melanogaster’s tracheal system. Then, comparisons between normal fruit flies and those engineered so that the RNA for zipper does not function were made. Visual and quantitative comparisons demonstrated less branching of the terminal cells for the mutants, while no differences were found for lumenogenesis—tube formation within the branched structures. Therefore, this study demonstrates the role of Zipper in branching of the terminal cells in the D. melanogaster’s tracheal system. This study adds onto the existing scientific literature by demonstrating the role of a specific protein in an important biological process occurring in most living organisms. Abstract Branching morphogenesis and seamless tube formation in Drosophila melanogaster are essential for the development of vascular and tracheal systems, and instructive in studying complex branched structures such as human organs. Zipper is a myosin II’s actin-binding heavy chain; hence, it is important for contracting actin, cell proliferation, and cell sheet adhesion for branching of the tracheal system in post-larval development of the D. melanogaster. Nevertheless, the specific role of Zipper in the larva is still in question. This paper intended to investigate the specific role of Zipper in branching morphogenesis and lumenogenesis in early developmental stages. It did so by checking the localization of the protein in the cytoplasm of the terminal cells and also by analyzing the morphology of zipper RNAi loss-of-function mutants in regard to branching and lumen formation in the terminal cells. A rescue experiment of RNAi mutants was also performed to check the sufficiency of Zipper in branching morphogenesis. Confocal imaging showed the localization of Zipper in the cytoplasm of the terminal cells, and respective quantitative analyses demonstrated that zipper RNAi terminal cells develop significantly fewer branches. Such a result hinted that Zipper is required for the regulation of branching in the terminal cells of D. melanogaster. Nevertheless, Zipper is not significantly involved in the formation of seamless tubes. One hypothesis is that Zipper’s contractility at the lateral epidermis’ leading edge allows cell sheet movement and respective elongation; as a result of such an elongation, further branching may occur in the elongated region of the cell, hence defining branching morphogenesis in the terminal cells of the tracheal system.
Collapse
Affiliation(s)
- Jong-Hyeon Shin
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Correspondence:
| | - Chan-Woo Jeong
- Department of Medicine, Seoul National University, Gwanak-gu, Seoul 08826, Korea;
| |
Collapse
|
36
|
Heymans C, Delcorte O, Spourquet C, Villacorte-Tabelin M, Dupasquier S, Achouri Y, Mahibullah S, Lemoine P, Balda MS, Matter K, Pierreux CE. Spatio-temporal expression pattern and role of the tight junction protein MarvelD3 in pancreas development and function. Sci Rep 2021; 11:14519. [PMID: 34267243 PMCID: PMC8282860 DOI: 10.1038/s41598-021-93654-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/23/2021] [Indexed: 11/29/2022] Open
Abstract
Tight junction complexes are involved in the establishment and maintenance of cell polarity and the regulation of signalling pathways, controlling biological processes such as cell differentiation and cell proliferation. MarvelD3 is a tight junction protein expressed in adult epithelial and endothelial cells. In Xenopus laevis, MarvelD3 morphants present differentiation defects of several ectodermal derivatives. In vitro experiments further revealed that MarvelD3 couples tight junctions to the MEKK1-JNK pathway to regulate cell behaviour and survival. In this work, we found that MarvelD3 is expressed from early developmental stages in the exocrine and endocrine compartments of the pancreas, as well as in endothelial cells of this organ. We thoroughly characterized MarvelD3 expression pattern in developing pancreas and evaluated its function by genetic ablation. Surprisingly, inactivation of MarvelD3 in mice did not alter development and differentiation of the pancreatic tissue. Moreover, tight junction formation and organization, cell polarization, and activity of the JNK-pathway were not impacted by the deletion of MarvelD3.
Collapse
Affiliation(s)
| | - Ophélie Delcorte
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
| | | | - Mylah Villacorte-Tabelin
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
- PRISM, MSU-IIT, Iligan City, Philippines
| | | | | | - Siam Mahibullah
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
| | - Pascale Lemoine
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
| | | | | | | |
Collapse
|
37
|
Pierreux CE. Shaping the thyroid: From peninsula to de novo lumen formation. Mol Cell Endocrinol 2021; 531:111313. [PMID: 33961919 DOI: 10.1016/j.mce.2021.111313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 01/06/2023]
Abstract
A challenging and stimulating question in biology deals with the formation of organs from groups of undifferentiated progenitor cells. Most epithelial organs indeed derive from the endodermal monolayer and evolve into various shape and tridimensional organization adapted to their specialized adult function. Thyroid organogenesis is no exception. In most mammals, it follows a complex and sequential process initiated from the endoderm and leading to the development of a multitude of independent closed spheres equipped and optimized for the synthesis, storage and production of thyroid hormones. The first sign of thyroid organogenesis is visible as a thickening of the anterior foregut endoderm. This group of thyroid progenitors then buds and detaches from the foregut to migrate caudally and then laterally. Upon reaching their final destination in the upper neck region on both sides of the trachea, thyroid progenitors mix with C cell progenitors and finally organize into hormone-producing thyroid follicles. Intrinsic and extrinsic factors controlling thyroid organogenesis have been identified in several species, but the fundamental cellular processes are not sufficiently considered. This review focuses on the cellular aspects of the key morphogenetic steps during thyroid organogenesis and highlights similarities and common mechanisms with developmental steps elucidated in other endoderm-derived organs, despite different final architecture and functions.
Collapse
|
38
|
Hill W, Zaragkoulias A, Salvador-Barbero B, Parfitt GJ, Alatsatianos M, Padilha A, Porazinski S, Woolley TE, Morton JP, Sansom OJ, Hogan C. EPHA2-dependent outcompetition of KRASG12D mutant cells by wild-type neighbors in the adult pancreas. Curr Biol 2021; 31:2550-2560.e5. [PMID: 33891893 PMCID: PMC8231095 DOI: 10.1016/j.cub.2021.03.094] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/15/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022]
Abstract
As we age, our tissues are repeatedly challenged by mutational insult, yet cancer occurrence is a relatively rare event. Cells carrying cancer-causing genetic mutations compete with normal neighbors for space and survival in tissues. However, the mechanisms underlying mutant-normal competition in adult tissues and the relevance of this process to cancer remain incompletely understood. Here, we investigate how the adult pancreas maintains tissue health in vivo following sporadic expression of oncogenic Kras (KrasG12D), the key driver mutation in human pancreatic cancer. We find that when present in tissues in low numbers, KrasG12D mutant cells are outcompeted and cleared from exocrine and endocrine compartments in vivo. Using quantitative 3D tissue imaging, we show that before being cleared, KrasG12D cells lose cell volume, pack into round clusters, and E-cadherin-based cell-cell adhesions decrease at boundaries with normal neighbors. We identify EphA2 receptor as an essential signal in the clearance of KrasG12D cells from exocrine and endocrine tissues in vivo. In the absence of functional EphA2, KrasG12D cells do not alter cell volume or shape, E-cadherin-based cell-cell adhesions increase and KrasG12D cells are retained in tissues. The retention of KRasG12D cells leads to the early appearance of premalignant pancreatic intraepithelial neoplasia (PanINs) in tissues. Our data show that adult pancreas tissues remodel to clear KrasG12D cells and maintain tissue health. This study provides evidence to support a conserved functional role of EphA2 in Ras-driven cell competition in epithelial tissues and suggests that EphA2 is a novel tumor suppressor in pancreatic cancer.
Collapse
Affiliation(s)
- William Hill
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Andreas Zaragkoulias
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Beatriz Salvador-Barbero
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Geraint J Parfitt
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; School of Optometry & Vision Sciences, Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK
| | - Markella Alatsatianos
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Ana Padilha
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Sean Porazinski
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Thomas E Woolley
- School of Mathematics, Cardiff University, Senghennydd Road, Cardiff CF24 4AG, UK
| | - Jennifer P Morton
- CRUK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Owen J Sansom
- CRUK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Catherine Hogan
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK.
| |
Collapse
|
39
|
Leng S, Carlone DL, Guagliardo NA, Barrett PQ, Breault DT. Rosette morphology in zona glomerulosa formation and function. Mol Cell Endocrinol 2021; 530:111287. [PMID: 33891993 PMCID: PMC8159910 DOI: 10.1016/j.mce.2021.111287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/18/2022]
Abstract
How morphology informs function is a fundamental biological question. Here, we review the morphological features of the adrenal zona glomerulosa (zG), highlighting recent cellular and molecular discoveries that govern its formation. The zG consists of glomeruli enwrapped in a Laminin-β1-enriched basement membrane (BM). Within each glomerulus, zG cells are organized as rosettes, a multicellular structure widely used throughout development to mediate epithelial remodeling, but not often found in healthy adult tissues. Rosettes arise by constriction at a common cellular contact point mediated/facilitated by adherens junctions (AJs). In mice, small, dispersed AJs first appear postnatally and enrich along the entire cell-cell contact around 10 days after birth. Subsequently, these AJ-rich contacts contract, allowing rosettes to form. Concurrently, flat sheet-like domains in the nascent zG, undergo invagination and folding, gradually giving rise to the compact round glomeruli that comprise the adult zG. How these structures impact adrenal function is discussed.
Collapse
Affiliation(s)
- Sining Leng
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - Paula Q Barrett
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
40
|
Lang C, Conrad L, Iber D. Organ-Specific Branching Morphogenesis. Front Cell Dev Biol 2021; 9:671402. [PMID: 34150767 PMCID: PMC8212048 DOI: 10.3389/fcell.2021.671402] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/06/2021] [Indexed: 01/09/2023] Open
Abstract
A common developmental process, called branching morphogenesis, generates the epithelial trees in a variety of organs, including the lungs, kidneys, and glands. How branching morphogenesis can create epithelial architectures of very different shapes and functions remains elusive. In this review, we compare branching morphogenesis and its regulation in lungs and kidneys and discuss the role of signaling pathways, the mesenchyme, the extracellular matrix, and the cytoskeleton as potential organ-specific determinants of branch position, orientation, and shape. Identifying the determinants of branch and organ shape and their adaptation in different organs may reveal how a highly conserved developmental process can be adapted to different structural and functional frameworks and should provide important insights into epithelial morphogenesis and developmental disorders.
Collapse
Affiliation(s)
- Christine Lang
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Lisa Conrad
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Dagmar Iber
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
41
|
Flasse L, Yennek S, Cortijo C, Barandiaran IS, Kraus MRC, Grapin-Botton A. Apical Restriction of the Planar Cell Polarity Component VANGL in Pancreatic Ducts Is Required to Maintain Epithelial Integrity. Cell Rep 2021; 31:107677. [PMID: 32460029 DOI: 10.1016/j.celrep.2020.107677] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/31/2020] [Accepted: 04/30/2020] [Indexed: 12/17/2022] Open
Abstract
Cell polarity is essential for the architecture and function of numerous epithelial tissues. Here, we show that apical restriction of planar cell polarity (PCP) components is necessary for the maintenance of epithelial integrity. Using the mammalian pancreas as a model, we find that components of the core PCP pathway, such as the transmembrane protein Van Gogh-like (VANGL), become apically restricted over a period of several days. Expansion of VANGL localization to the basolateral membranes of progenitors leads to their death and disruption of the epithelial integrity. VANGL basolateral expansion does not affect apico-basal polarity but acts in the cells where Vangl is mislocalized by reducing Dishevelled and its downstream target ROCK. This reduction in ROCK activity culminates in progenitor cell egression, death, and eventually pancreatic hypoplasia. Thus, precise spatiotemporal modulation of VANGL-dependent PCP signaling is crucial for proper pancreatic morphogenesis.
Collapse
Affiliation(s)
- Lydie Flasse
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| | - Siham Yennek
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Cédric Cortijo
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausannne, Switzerland
| | | | - Marine R-C Kraus
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausannne, Switzerland
| | - Anne Grapin-Botton
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| |
Collapse
|
42
|
Mechanical plasticity of collagen directs branch elongation in human mammary gland organoids. Nat Commun 2021; 12:2759. [PMID: 33980857 PMCID: PMC8115695 DOI: 10.1038/s41467-021-22988-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/08/2021] [Indexed: 12/25/2022] Open
Abstract
Epithelial branch elongation is a central developmental process during branching morphogenesis in diverse organs. This fundamental growth process into large arborized epithelial networks is accompanied by structural reorganization of the surrounding extracellular matrix (ECM), well beyond its mechanical linear response regime. Here, we report that epithelial ductal elongation within human mammary organoid branches relies on the non-linear and plastic mechanical response of the surrounding collagen. Specifically, we demonstrate that collective back-and-forth motion of cells within the branches generates tension that is strong enough to induce a plastic reorganization of the surrounding collagen network which results in the formation of mechanically stable collagen cages. Such matrix encasing in turn directs further tension generation, branch outgrowth and plastic deformation of the matrix. The identified mechanical tension equilibrium sets a framework to understand how mechanical cues can direct ductal branch elongation. Mammary organoid growth from single primary human cells rely on distinct morphogenetic processes. Here, the authors observe by live cell imaging the importance of the plastic mechanical response of the extracellular matrix and cell migration for the underlying arborized structure formation process.
Collapse
|
43
|
Breunig M, Merkle J, Wagner M, Melzer MK, Barth TFE, Engleitner T, Krumm J, Wiedenmann S, Cohrs CM, Perkhofer L, Jain G, Krüger J, Hermann PC, Schmid M, Madácsy T, Varga Á, Griger J, Azoitei N, Müller M, Wessely O, Robey PG, Heller S, Dantes Z, Reichert M, Günes C, Bolenz C, Kuhn F, Maléth J, Speier S, Liebau S, Sipos B, Kuster B, Seufferlein T, Rad R, Meier M, Hohwieler M, Kleger A. Modeling plasticity and dysplasia of pancreatic ductal organoids derived from human pluripotent stem cells. Cell Stem Cell 2021; 28:1105-1124.e19. [PMID: 33915078 DOI: 10.1016/j.stem.2021.03.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 12/22/2020] [Accepted: 03/09/2021] [Indexed: 12/14/2022]
Abstract
Personalized in vitro models for dysplasia and carcinogenesis in the pancreas have been constrained by insufficient differentiation of human pluripotent stem cells (hPSCs) into the exocrine pancreatic lineage. Here, we differentiate hPSCs into pancreatic duct-like organoids (PDLOs) with morphological, transcriptional, proteomic, and functional characteristics of human pancreatic ducts, further maturing upon transplantation into mice. PDLOs are generated from hPSCs inducibly expressing oncogenic GNAS, KRAS, or KRAS with genetic covariance of lost CDKN2A and from induced hPSCs derived from a McCune-Albright patient. Each oncogene causes a specific growth, structural, and molecular phenotype in vitro. While transplanted PDLOs with oncogenic KRAS alone form heterogenous dysplastic lesions or cancer, KRAS with CDKN2A loss develop dedifferentiated pancreatic ductal adenocarcinomas. In contrast, transplanted PDLOs with mutant GNAS lead to intraductal papillary mucinous neoplasia-like structures. Conclusively, PDLOs enable in vitro and in vivo studies of pancreatic plasticity, dysplasia, and cancer formation from a genetically defined background.
Collapse
Affiliation(s)
- Markus Breunig
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Jessica Merkle
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Martin Wagner
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Michael K Melzer
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany; Department of Urology, Ulm University, Ulm, Germany
| | | | - Thomas Engleitner
- Institute of Molecular Oncology and Functional Genomics, Center for Translational Cancer Research and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Johannes Krumm
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Sandra Wiedenmann
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany; Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Christian M Cohrs
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Gaurav Jain
- Institute of Molecular Oncology and Functional Genomics, Center for Translational Cancer Research and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jana Krüger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Patrick C Hermann
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Maximilian Schmid
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Tamara Madácsy
- First Department of Internal Medicine, University of Szeged, Szeged, Hungary; MTA-SZTE Momentum Epithelial Cell Signalling and Secretion Research Group, University of Szeged, Szeged, Hungary
| | - Árpád Varga
- First Department of Internal Medicine, University of Szeged, Szeged, Hungary; MTA-SZTE Momentum Epithelial Cell Signalling and Secretion Research Group, University of Szeged, Szeged, Hungary
| | - Joscha Griger
- Institute of Molecular Oncology and Functional Genomics, Center for Translational Cancer Research and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Ninel Azoitei
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Martin Müller
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Oliver Wessely
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Sandra Heller
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Zahra Dantes
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | | | | | - Florian Kuhn
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - József Maléth
- First Department of Internal Medicine, University of Szeged, Szeged, Hungary; MTA-SZTE Momentum Epithelial Cell Signalling and Secretion Research Group, University of Szeged, Szeged, Hungary; HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bence Sipos
- Department of Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany; Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), Technical University of Munich, Freising, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, Center for Translational Cancer Research and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias Meier
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Meike Hohwieler
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany.
| |
Collapse
|
44
|
Generation of a Novel Nkx6-1 Venus Fusion Reporter Mouse Line. Int J Mol Sci 2021; 22:ijms22073434. [PMID: 33810480 PMCID: PMC8036392 DOI: 10.3390/ijms22073434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 11/17/2022] Open
Abstract
Nkx6-1 is a member of the Nkx family of homeodomain transcription factors (TFs) that regulates motor neuron development, neuron specification and pancreatic endocrine and β-cell differentiation. To facilitate the isolation and tracking of Nkx6-1-expressing cells, we have generated a novel Nkx6-1 Venus fusion (Nkx6-1-VF) reporter allele. The Nkx6-1-VF knock-in reporter is regulated by endogenous cis-regulatory elements of Nkx6-1 and the fluorescent protein fusion does not interfere with the TF function, as homozygous mice are viable and fertile. The nuclear localization of Nkx6-1-VF protein reflects the endogenous Nkx6-1 protein distribution. During embryonic pancreas development, the reporter protein marks the pancreatic ductal progenitors and the endocrine lineage, but is absent in the exocrine compartment. As expected, the levels of Nkx6-1-VF reporter are upregulated upon β-cell differentiation during the major wave of endocrinogenesis. In the adult islets of Langerhans, the reporter protein is exclusively found in insulin-secreting β-cells. Importantly, the Venus reporter activities allow successful tracking of β-cells in live-cell imaging and their specific isolation by flow sorting. In summary, the generation of the Nkx6-1-VF reporter line reflects the expression pattern and dynamics of the endogenous protein and thus provides a unique tool to study the spatio-temporal expression pattern of this TF during organ development and enables isolation and tracking of Nkx6-1-expressing cells such as pancreatic β-cells, but also neurons and motor neurons in health and disease.
Collapse
|
45
|
Abstract
The pancreas of adult mammals displays a branched structure which transports digestive enzymes produced in the distal acini through a tree-like network of ducts into the duodenum. In contrast to several other branched organs, its branching patterns are not stereotypic. Moreover, the branches do not grow from dichotomic splitting of an initial stem but rather from the formation of microlumen in a mass of cells. These lumen progressively assemble into a hyperconnected network that refines into a tree by the time of birth. We review the cell remodeling events and the molecular mechanisms governing pancreas branching, as well as the role of the surrounding tissues in this process. Furthermore, we draw parallels with other branched organs such as the salivary and mammary gland.
Collapse
Affiliation(s)
- Lydie Flasse
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Coline Schewin
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Anne Grapin-Botton
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany; The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark.
| |
Collapse
|
46
|
Scoville DW, Kang HS, Jetten AM. Transcription factor GLIS3: Critical roles in thyroid hormone biosynthesis, hypothyroidism, pancreatic beta cells and diabetes. Pharmacol Ther 2020; 215:107632. [PMID: 32693112 PMCID: PMC7606550 DOI: 10.1016/j.pharmthera.2020.107632] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022]
Abstract
GLI-Similar 3 (GLIS3) is a member of the GLIS subfamily of Krüppel-like zinc finger transcription factors that functions as an activator or repressor of gene expression. Study of GLIS3-deficiency in mice and humans revealed that GLIS3 plays a critical role in the regulation of several biological processes and is implicated in the development of various diseases, including hypothyroidism and diabetes. This was supported by genome-wide association studies that identified significant associations of common variants in GLIS3 with increased risk of these pathologies. To obtain insights into the causal mechanisms underlying these diseases, it is imperative to understand the mechanisms by which this protein regulates the development of these pathologies. Recent studies of genes regulated by GLIS3 led to the identification of a number of target genes and have provided important molecular insights by which GLIS3 controls cellular processes. These studies revealed that GLIS3 is essential for thyroid hormone biosynthesis and identified a critical function for GLIS3 in the generation of pancreatic β cells and insulin gene transcription. These observations raised the possibility that the GLIS3 signaling pathway might provide a potential therapeutic target in the management of diabetes, hypothyroidism, and other diseases. To develop such strategies, it will be critical to understand the upstream signaling pathways that regulate the activity, expression and function of GLIS3. Here, we review the recent progress on the molecular mechanisms by which GLIS3 controls key functions in thyroid follicular and pancreatic β cells and how this causally relates to the development of hypothyroidism and diabetes.
Collapse
Affiliation(s)
- David W Scoville
- Cell Biology Group, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Hong Soon Kang
- Cell Biology Group, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Anton M Jetten
- Cell Biology Group, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
47
|
Rens EG, Zeegers MT, Rabbers I, Szabó A, Merks RMH. Autocrine inhibition of cell motility can drive epithelial branching morphogenesis in the absence of growth. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190386. [PMID: 32713299 DOI: 10.1098/rstb.2019.0386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epithelial branching morphogenesis drives the development of organs such as the lung, salivary gland, kidney and the mammary gland. It involves cell proliferation, cell differentiation and cell migration. An elaborate network of chemical and mechanical signals between the epithelium and the surrounding mesenchymal tissues regulates the formation and growth of branching organs. Surprisingly, when cultured in isolation from mesenchymal tissues, many epithelial tissues retain the ability to exhibit branching morphogenesis even in the absence of proliferation. In this work, we propose a simple, experimentally plausible mechanism that can drive branching morphogenesis in the absence of proliferation and cross-talk with the surrounding mesenchymal tissue. The assumptions of our mathematical model derive from in vitro observations of the behaviour of mammary epithelial cells. These data show that autocrine secretion of the growth factor TGF[Formula: see text]1 inhibits the formation of cell protrusions, leading to curvature-dependent inhibition of sprouting. Our hybrid cellular Potts and partial-differential equation model correctly reproduces the experimentally observed tissue-geometry-dependent determination of the sites of branching, and it suffices for the formation of self-avoiding branching structures in the absence and also in the presence of cell proliferation. This article is part of the theme issue 'Multi-scale analysis and modelling of collective migration in biological systems'.
Collapse
Affiliation(s)
- Elisabeth G Rens
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Mathé T Zeegers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Iraes Rabbers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - András Szabó
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Roeland M H Merks
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|
48
|
Palmer MA, Nelson CM. Fusion of airways during avian lung development constitutes a novel mechanism for the formation of continuous lumena in multicellular epithelia. Dev Dyn 2020; 249:1318-1333. [DOI: 10.1002/dvdy.215] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/01/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Affiliation(s)
- Michael A. Palmer
- Department of Chemical & Biological Engineering Princeton University Princeton New Jersey USA
| | - Celeste M. Nelson
- Department of Chemical & Biological Engineering Princeton University Princeton New Jersey USA
- Department of Molecular Biology Princeton University Princeton New Jersey USA
| |
Collapse
|
49
|
Chela H, Romana BS, Madabattula M, Albarrak AA, Yousef MH, Samiullah S, Tahan V. Stem cell therapy: a potential for the perils of pancreatitis. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2020; 31:415-424. [PMID: 32721912 PMCID: PMC7433995 DOI: 10.5152/tjg.2020.19143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 09/01/2019] [Indexed: 12/17/2022]
Abstract
Acute and chronic pancreatitis carry a significant disease burden and there is no definite treatment that exists for either. They are associated with local and systemic inflammation and lead to numerous complications. Stem cell therapy has been explored for other disease processes and is a topic of research that has gained momentum with regards to implications for acute and chronic pancreatitis. They not only carry the potential to aid in regeneration but also prevent pancreatic injury as well as injury of other organs and hence the resultant complications. Stem cells appear to have immunomodulatory properties and clinical potential as evidenced by numerous studies in animal models. This review article discusses the types of stem cells commonly used and the properties that show promise in the field of pancreatitis.
Collapse
Affiliation(s)
- Harleen Chela
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Bhupinder S Romana
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Markandeya Madabattula
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Abdulmajeed A Albarrak
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Mohamad H Yousef
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Sami Samiullah
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Veysel Tahan
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
50
|
Leng S, Pignatti E, Khetani RS, Shah MS, Xu S, Miao J, Taketo MM, Beuschlein F, Barrett PQ, Carlone DL, Breault DT. β-Catenin and FGFR2 regulate postnatal rosette-based adrenocortical morphogenesis. Nat Commun 2020; 11:1680. [PMID: 32245949 PMCID: PMC7125176 DOI: 10.1038/s41467-020-15332-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/28/2020] [Indexed: 02/08/2023] Open
Abstract
Rosettes are widely used in epithelial morphogenesis during embryonic development and organogenesis. However, their role in postnatal development and adult tissue maintenance remains largely unknown. Here, we show zona glomerulosa cells in the adult adrenal cortex organize into rosettes through adherens junction-mediated constriction, and that rosette formation underlies the maturation of adrenal glomerular structure postnatally. Using genetic mouse models, we show loss of β-catenin results in disrupted adherens junctions, reduced rosette number, and dysmorphic glomeruli, whereas β-catenin stabilization leads to increased adherens junction abundance, more rosettes, and glomerular expansion. Furthermore, we uncover numerous known regulators of epithelial morphogenesis enriched in β-catenin-stabilized adrenals. Among these genes, we show Fgfr2 is required for adrenal rosette formation by regulating adherens junction abundance and aggregation. Together, our data provide an example of rosette-mediated postnatal tissue morphogenesis and a framework for studying the role of rosettes in adult zona glomerulosa tissue maintenance and function.
Collapse
Affiliation(s)
- Sining Leng
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, 02115, USA
| | - Emanuele Pignatti
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Radhika S Khetani
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Manasvi S Shah
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Simiao Xu
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Makoto M Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo, Kyoto, 606-8506, Japan
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, UniversitätsSpital Zürich, Zurich, Switzerland
- Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Paula Q Barrett
- Departments of Pharmacology, University of Virginia, Charlottesville, VA, 22947, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|