1
|
Shi W, Duclut C, Xu Y, Ma Y, Qiao J, Lin B, Yang D, Prost J, Dong B. Physics of notochord tube expansion in ascidians. Proc Natl Acad Sci U S A 2025; 122:e2419960122. [PMID: 40472029 DOI: 10.1073/pnas.2419960122] [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: 09/30/2024] [Accepted: 04/09/2025] [Indexed: 06/18/2025] Open
Abstract
Interaction of cells and the surrounding lumen drives the formation of tubular system that plays the transport and exchange functions within an organism. The physical and biological mechanisms of lumen expansion have been explored. However, how cells communicate and coordinate with the surrounding lumen, leading to continuous tube expansion to a defined geometry, is crucial but remains elusive. In this study, we utilized the ascidian notochord tube as a model to address the underlying mechanisms. We first quantitatively measured and calculated the geometric parameters and found that tube expansion experienced three distinct phases. During the growth processes, we identified and experimentally demonstrated that both Rho GTPase Cdc42 signaling-mediated cell cortex distribution and the stability of tight junctions (TJs) were essential for lumen opening and tube expansion. Based on these experimental data, a conservation-laws-based tube expansion theory was developed, considering critical cell communication pathways, including secretory activity through vesicles, asymmetric cortex tension driven anisotropic lumen geometry, as well as the TJs gate barrier function. Moreover, by estimating the critical tube expansion parameters from experimental observation, we successfully predicted tube growth kinetics under different conditions through the combination of computational and experimental approaches, highlighting the coupling between actomyosin-based active mechanics and hydraulic processes. Taken together, our findings identify the critical cellular regulatory factors that drive the biological tube expansion and maintain its stability.
Collapse
Affiliation(s)
- Wenjie Shi
- Fang Zongxi Center for Marine EvoDevo, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Charlie Duclut
- Laboratoire Physique des Cellules et Cancer (PCC), CNRS UMR 168, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, Paris 75005, France
| | - Yan Xu
- Fang Zongxi Center for Marine EvoDevo, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yuanting Ma
- Fang Zongxi Center for Marine EvoDevo, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jinghan Qiao
- Fang Zongxi Center for Marine EvoDevo, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Boyan Lin
- Fang Zongxi Center for Marine EvoDevo, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Dongyu Yang
- Fang Zongxi Center for Marine EvoDevo, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jacques Prost
- Laboratoire Physique des Cellules et Cancer (PCC), CNRS UMR 168, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, Paris 75005, France
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Bo Dong
- Fang Zongxi Center for Marine EvoDevo, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Meirelles MG, Fénero CIM, Nornberg BF, Camara NOS, Marins LF. In Vivo Measurement of Intestinal Permeability to Macromolecules in Adult Zebrafish ( Danio rerio). Zebrafish 2025; 22:1-10. [PMID: 39895322 DOI: 10.1089/zeb.2024.0167] [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: 02/04/2025] Open
Abstract
Intestinal permeability plays a crucial role in intestinal barrier function. Altered intestinal permeability is well documented in numerous chronic diseases and may serve as a risk factor for disease onset as well as a target for innovative therapeutic strategies. While reliable and sensitive approaches for studying intestinal permeability have been established in animal models, such as mice and zebrafish larvae, methods for investigating this in adult zebrafish remain a considerable challenge. The zebrafish has emerged as a valuable model for studying intestinal development, physiology, and disease. Moreover, zebrafish offer certain advantages over rodent models, such as the ability to evaluate the dynamic interactions of labeled markers in vivo and in real time. In this study, we present a comprehensive pipeline for assessing in vivo intestinal permeability in adult zebrafish using fluorescent-labeled dextran. Detailed protocols for fish handling, reagent preparation, optimization of reagent dosage and delivery routes, and quantification of fluorescent markers in extraintestinal sites are provided. Our findings suggest that zebrafish hold promise as an alternative model for in vivo investigations of intestinal permeability induced by genetic, pathophysiological, and/or pharmacological events.
Collapse
Affiliation(s)
- Marcela G Meirelles
- LEGENE-Research Group in Genetic Engineering and Biotechnology, Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Rio Grande-FURG, Rio Grande, Brazil
| | - Camila I M Fénero
- Department of Immunology, Institute of Biomedical Science, University of São Paulo-USP, São Paulo, Brazil
| | - Bruna F Nornberg
- LEGENE-Research Group in Genetic Engineering and Biotechnology, Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Rio Grande-FURG, Rio Grande, Brazil
| | - Niels Olsen S Camara
- Department of Immunology, Institute of Biomedical Science, University of São Paulo-USP, São Paulo, Brazil
| | - Luis Fernando Marins
- LEGENE-Research Group in Genetic Engineering and Biotechnology, Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Rio Grande-FURG, Rio Grande, Brazil
| |
Collapse
|
5
|
Uribe-Montes LC, Sanabria-Camargo CA, Piñeros-Romero CC, Otálora-Tarazona S, Ávila-Jiménez E, Acosta-Virgüez E, Garavito-Aguilar ZV. Fibronectin and Hand2 influence tubulogenesis during pronephros development and mesonephros regeneration in zebrafish (Danio rerio). PLoS One 2024; 19:e0307390. [PMID: 39240899 PMCID: PMC11379296 DOI: 10.1371/journal.pone.0307390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 09/08/2024] Open
Abstract
Worldwide incidence of kidney diseases has been rising. Thus, recent research has focused on zebrafish, whose fast development and innate regeneration capacity allow identifying factors influencing renal processes. Among these poorly studied factors are extracellular matrix (ECM) proteins like Fibronectin (Fn) essential in various tissues but not yet evaluated in a renal context. We utilized early nat and han zebrafish mutant embryos and carrier adults to investigate Fn's role during kidney development and regeneration. The locus natter (nat) encodes Fn and the locus han encodes Hand2, which results in increased Fn deposition. Our results show that Fn impacts identity maintenance and morphogenesis during development and influences conditions for neonephrogenic cluster formation during regeneration. Histological analysis revealed disrupted pronephric structures and increased blood cell accumulation in Fn mutants. Despite normal expression of specification markers (pax2, ATPα1a.1), structural abnormalities were evident. Differences between wild-type and mutation-carriers suggest a haploinsufficiency scenario. These findings reveal a novel function for ECM in renal development and regeneration, with potential implications for understanding and treating kidney diseases.
Collapse
Affiliation(s)
- Lucia Carolina Uribe-Montes
- Laboratorio de Biología del Desarrollo-BIOLDES, Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Camilo Alfonso Sanabria-Camargo
- Laboratorio de Biología del Desarrollo-BIOLDES, Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Cristian Camilo Piñeros-Romero
- Laboratorio de Biología del Desarrollo-BIOLDES, Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Sebastián Otálora-Tarazona
- Laboratorio de Biología del Desarrollo-BIOLDES, Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Estefanía Ávila-Jiménez
- Laboratorio de Biología del Desarrollo-BIOLDES, Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Edwin Acosta-Virgüez
- Departamento de Biología, Universidad Nacional de Colombia-Sede Bogotá, Bogotá, Colombia
| | - Zayra Viviana Garavito-Aguilar
- Laboratorio de Biología del Desarrollo-BIOLDES, Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
6
|
Liang Z, Dondorp DC, Chatzigeorgiou M. The ion channel Anoctamin 10/TMEM16K coordinates organ morphogenesis across scales in the urochordate notochord. PLoS Biol 2024; 22:e3002762. [PMID: 39173068 PMCID: PMC11341064 DOI: 10.1371/journal.pbio.3002762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 07/20/2024] [Indexed: 08/24/2024] Open
Abstract
During embryonic development, tissues and organs are gradually shaped into their functional morphologies through a series of spatiotemporally tightly orchestrated cell behaviors. A highly conserved organ shape across metazoans is the epithelial tube. Tube morphogenesis is a complex multistep process of carefully choreographed cell behaviors such as convergent extension, cell elongation, and lumen formation. The identity of the signaling molecules that coordinate these intricate morphogenetic steps remains elusive. The notochord is an essential tubular organ present in the embryonic midline region of all members of the chordate phylum. Here, using genome editing, pharmacology and quantitative imaging in the early chordate Ciona intestinalis we show that Ano10/Tmem16k, a member of the evolutionarily ancient family of transmembrane proteins called Anoctamin/TMEM16 is essential for convergent extension, lumen expansion, and connection during notochord morphogenesis. We find that Ano10/Tmem16k works in concert with the plasma membrane (PM) localized Na+/Ca2+ exchanger (NCX) and the endoplasmic reticulum (ER) residing SERCA, RyR, and IP3R proteins to establish developmental stage specific Ca2+ signaling molecular modules that regulate notochord morphogenesis and Ca2+ dynamics. In addition, we find that the highly conserved Ca2+ sensors calmodulin (CaM) and Ca2+/calmodulin-dependent protein kinase (CaMK) show an Ano10/Tmem16k-dependent subcellular localization. Their pharmacological inhibition leads to convergent extension, tubulogenesis defects, and deranged Ca2+ dynamics, suggesting that Ano10/Tmem16k is involved in both the "encoding" and "decoding" of developmental Ca2+ signals. Furthermore, Ano10/Tmem16k mediates cytoskeletal reorganization during notochord morphogenesis, likely by altering the localization of 2 important cytoskeletal regulators, the small GTPase Ras homolog family member A (RhoA) and the actin binding protein Cofilin. Finally, we use electrophysiological recordings and a scramblase assay in tissue culture to demonstrate that Ano10/Tmem16k likely acts as an ion channel but not as a phospholipid scramblase. Our results establish Ano10/Tmem16k as a novel player in the prevertebrate molecular toolkit that controls organ morphogenesis across scales.
Collapse
Affiliation(s)
- Zonglai Liang
- Michael Sars Centre, University of Bergen, Bergen, Norway
| | | | | |
Collapse
|
7
|
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
|
8
|
Thottacherry JJ, Chen J, Johnston DS. Apical-basal polarity in the gut. Semin Cell Dev Biol 2023; 150-151:15-22. [PMID: 36670034 DOI: 10.1016/j.semcdb.2022.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/24/2022] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
Apical-Basal polarity is a fundamental property of all epithelial cells that underlies both their form and function. The gut is made up of a single layer of intestinal epithelial cells, with distinct apical, lateral and basal domains. Occluding junctions at the apical side of the lateral domains create a barrier between the gut lumen and the body, which is crucial for tissue homeostasis, protection against gastrointestinal pathogens and for the maintenance of the immune response. Apical-basal polarity in most epithelia is established by conserved polarity factors, but recent evidence suggests that the gut epithelium in at least some organisms polarises by novel mechanisms. In this review, we discuss the recent advances in understanding polarity factors by focussing on work in C. elegans, Drosophila, Zebrafish and Mouse.
Collapse
Affiliation(s)
- Joseph Jose Thottacherry
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge CB2 1QN, United Kingdom
| | - Jia Chen
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge CB2 1QN, United Kingdom
| | - Daniel St Johnston
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge CB2 1QN, United Kingdom.
| |
Collapse
|
9
|
Zhang L, Wei X. Stepwise modulation of apical orientational cell adhesions for vertebrate neurulation. Biol Rev Camb Philos Soc 2023; 98:2271-2283. [PMID: 37534608 DOI: 10.1111/brv.13006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Neurulation transforms the neuroectoderm into the neural tube. This transformation relies on reorganising the configurational relationships between the orientations of intrinsic polarities of neighbouring cells. These orientational intercellular relationships are established, maintained, and modulated by orientational cell adhesions (OCAs). Here, using zebrafish (Danio rerio) neurulation as a major model, we propose a new perspective on how OCAs contribute to the parallel, antiparallel, and opposing intercellular relationships that underlie the neural plate-keel-rod-tube transformation, a stepwise process of cell aggregation followed by cord hollowing. We also discuss how OCAs in neurulation may be regulated by various adhesion molecules, including cadherins, Eph/Ephrins, Claudins, Occludins, Crumbs, Na+ /K+ -ATPase, and integrins. By comparing neurulation among species, we reveal that antiparallel OCAs represent a conserved mechanism for the fusion of the neural tube. Throughout, we highlight some outstanding questions regarding OCAs in neurulation. Answers to these questions will help us understand better the mechanisms of tubulogenesis of many tissues.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Psychology, Dalian Medical University, 9 South LvShun Road, Dalian, 116044, China
| | - Xiangyun Wei
- Departments of Ophthalmology, Developmental Biology, and Microbiology & Molecular Genetics, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
| |
Collapse
|
10
|
Dinet C, Torres-Sánchez A, Lanfranco R, Di Michele L, Arroyo M, Staykova M. Patterning and dynamics of membrane adhesion under hydraulic stress. Nat Commun 2023; 14:7445. [PMID: 37978292 PMCID: PMC10656516 DOI: 10.1038/s41467-023-43246-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Hydraulic fracturing plays a major role in cavity formation during embryonic development, when pressurized fluid opens microlumens at cell-cell contacts, which evolve to form a single large lumen. However, the fundamental physical mechanisms behind these processes remain masked by the complexity and specificity of biological systems. Here, we show that adhered lipid vesicles subjected to osmotic stress form hydraulic microlumens similar to those in cells. Combining vesicle experiments with theoretical modelling and numerical simulations, we provide a physical framework for the hydraulic reconfiguration of cell-cell adhesions. We map the conditions for microlumen formation from a pristine adhesion, the emerging dynamical patterns and their subsequent maturation. We demonstrate control of the fracturing process depending on the applied pressure gradients and the type and density of membrane bonds. Our experiments further reveal an unexpected, passive transition of microlumens to closed buds that suggests a physical route to adhesion remodeling by endocytosis.
Collapse
Affiliation(s)
- Céline Dinet
- Department of Physics, Durham University, Durham, UK
- Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, CNRS-Aix-Marseille University, 31 Chemin Joseph Aiguier, 13009, Marseille, France
| | - Alejandro Torres-Sánchez
- Universitat Politècnica de Catalunya-BarcelonaTech, 08034, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
- European Molecular Biology Laboratory (EMBL-Barcelona), 08003, Barcelona, Spain
| | - Roberta Lanfranco
- Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
| | - Lorenzo Di Michele
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
- Department of Chemistry, Imperial College of London, London, UK
| | - Marino Arroyo
- Universitat Politècnica de Catalunya-BarcelonaTech, 08034, Barcelona, Spain.
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.
- Centre Internacional de Mètodes Numèrics en Enginyeria (CIMNE), 08034, Barcelona, Spain.
| | | |
Collapse
|
11
|
Chen Z, Zhao J, Wang C, Liu X, Chen Z, Zhou J, Zhang L, Zhang C, Li H. Epithelial polarity-driven membrane separation but not cavitation regulates lumen formation of rat eccrine sweat glands. Acta Histochem 2023; 125:152093. [PMID: 37757514 DOI: 10.1016/j.acthis.2023.152093] [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: 05/31/2023] [Revised: 08/31/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Each eccrine sweat gland (ESG) is a single-tubular structure with a central lumen, and the formation of hollow lumen in the initial solid cell mass is a key developmental process. To date, there are no reports on the mechanism of native ESG lumen formation. METHODS To investigate the lumen morphogenesis and the lumen formation mechanisms of Sprague-Dawley (SD) rat ESGs, SD rat hind-footpads at E20.5, P1-P5, P7, P9, P12, P21, P28 and P56 were obtained. The lumen morphogenesis of ESGs was examined by HE staining and immunofluorescence staining for polarity markers. The possible mechanisms of lumen formation were detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) apoptosis assay and autophagy marker LC3B immunofluorescence staining, and further explored by ouabain intervention experiment. RESULTS In SD rat ESGs, the microlumen was formed at P1, and the small intact lumen with apical-basal polarity appeared at P3. The expression of apical marker F-actin, basal marker Laminin, basolateral marker E-cadherin was consistent with the timing of lumen formation of SD rat ESGs. During rat ESG development, apoptosis and autophagy were not detected. However, inhibition of Na+-K+-ATPase (NKA) with ouabain resulted in decreased lumen size, although neither the timing of lumen formation nor the expression of polarity proteins was altered. CONCLUSIONS Epithelial polarity-driven membrane separation but not cavitation regulates lumen formation of SD rat ESGs. NKA-regulated fluid accumulation drives lumen expansion.
Collapse
Affiliation(s)
- Zixiu Chen
- Department of Wound Repair and Dermatologic Surgery, Taihe Hospital, Hubei University of Medicine, Jinzhou Medical University Graduate Training Base, Shiyan, Hubei Province, China
| | - Junhong Zhao
- Department of Wound Repair and Dermatologic Surgery, Taihe Hospital, Hubei University of Medicine, Jinzhou Medical University Graduate Training Base, Shiyan, Hubei Province, China
| | - Cangyu Wang
- Department of Wound Repair and Dermatologic Surgery, Taihe Hospital, Hubei University of Medicine, Jinzhou Medical University Graduate Training Base, Shiyan, Hubei Province, China
| | - Xiang Liu
- Department of Wound Repair and Dermatologic Surgery, Taihe Hospital, Hubei University of Medicine, Jinzhou Medical University Graduate Training Base, Shiyan, Hubei Province, China
| | - Zihua Chen
- Department of Wound Repair and Dermatologic Surgery, Taihe Hospital, Hubei University of Medicine, Jinzhou Medical University Graduate Training Base, Shiyan, Hubei Province, China
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, The Third Hospital of Central South University, Changsha, Hunan, China
| | - Lei Zhang
- Mental Health Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| | - Cuiping Zhang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and Fourth Medical Center of PLA General Hospital, Beijing, China.
| | - Haihong Li
- Department of Wound Repair and Dermatologic Surgery, Taihe Hospital, Hubei University of Medicine, Jinzhou Medical University Graduate Training Base, Shiyan, Hubei Province, China; Department of Burns and Plastic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| |
Collapse
|
12
|
Qi X, Zhang Y, Zhang Y, Luo F, Song K, Wang G, Ling F. Vitamin B 12 produced by Cetobacterium somerae improves host resistance against pathogen infection through strengthening the interactions within gut microbiota. MICROBIOME 2023; 11:135. [PMID: 37322528 PMCID: PMC10268390 DOI: 10.1186/s40168-023-01574-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/15/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Pathogen infections seriously affect host health, and the use of antibiotics increases the risk of the emergence of drug-resistant bacteria and also increases environmental and health safety risks. Probiotics have received much attention for their excellent ability to prevent pathogen infections. Particularly, explaining mechanism of action of probiotics against pathogen infections is important for more efficient and rational use of probiotics and the maintenance of host health. RESULTS Here, we describe the impacts of probiotic on host resistance to pathogen infections. Our findings revealed that (I) the protective effect of oral supplementation with B. velezensis against Aeromonas hydrophila infection was dependent on gut microbiota, specially the anaerobic indigenous gut microbe Cetobacterium; (II) Cetobacterium was a sensor of health, especially for fish infected with pathogenic bacteria; (III) the genome resolved the ability of Cetobacterium somerae CS2105-BJ to synthesize vitamin B12 de novo, while in vivo and in vitro metabolism assays also showed the ability of Cetobacterium somerae CS2105-BJ to produce vitamin B12; (IV) the addition of vitamin B12 significantly altered the gut redox status and the gut microbiome structure and function, and then improved the stability of the gut microbial ecological network, and enhanced the gut barrier tight junctions to prevent the pathogen infection. CONCLUSION Collectively, this study found that the effect of probiotics in enhancing host resistance to pathogen infections depended on function of B12 produced by an anaerobic indigenous gut microbe, Cetobacterium. Furthermore, as a gut microbial regulator, B12 exhibited the ability to strengthen the interactions within gut microbiota and gut barrier tight junctions, thereby improving host resistance against pathogen infection. Video Abstract.
Collapse
Affiliation(s)
- Xiaozhou Qi
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yong Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Fei Luo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Kaige Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Gaoxue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| | - Fei Ling
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
13
|
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
|
14
|
Zhang S, Chen A, Jiang L, Liu X, Chai L. Copper-mediated shifts in transcriptomic responses of intestines in Bufo gargarizans tadpoles to lead stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:50144-50161. [PMID: 36790706 DOI: 10.1007/s11356-023-25801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/04/2023] [Indexed: 04/16/2023]
Abstract
The differential transcriptomic responses of intestines in Bufo gargarizans tadpoles to Pb alone or in the presence of Cu were evaluated. Tadpoles were exposed to 30 μg/L Pb individually and in combination with Cu at 16 or 64 μg/L from Gosner stage (Gs) 26 to Gs 38. After de novo assembly, 105,107 unigenes were generated. Compared to the control group, 7387, 6937, and 11139 differentially expressed genes (DEGs) were identified in the treatment of Pb + Cu0, Pb + Cu16, and Pb + Cu64, respectively. In addition, functional annotation and enrichment analysis of DEGs revealed substantial transcriptional reprogramming of diverse molecular and biological pathways were induced in all heavy metal treatments. The relative expression levels of genes associated with intestinal epithelial barrier and bile acids (BAs) metabolism, such as mucin2, claudin5, ZO-1, Asbt, and Ost-β, were validated by qPCR. This study demonstrated that Pb exposure induced transcriptional responses in tadpoles, and the responses could be modulated by Cu.
Collapse
Affiliation(s)
- Siliang Zhang
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Aixia Chen
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Ling Jiang
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Xiaoli Liu
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China
| | - Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an, 710054, People's Republic of China.
- Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an, 710054, People's Republic of China.
| |
Collapse
|
15
|
Levic DS, Bagnat M. Polarized transport of membrane and secreted proteins during lumen morphogenesis. Semin Cell Dev Biol 2023; 133:65-73. [PMID: 35307284 PMCID: PMC9481742 DOI: 10.1016/j.semcdb.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
Abstract
A ubiquitous feature of animal development is the formation of fluid-filled cavities or lumina, which transport gases and fluids across tissues and organs. Among different species, lumina vary drastically in size, scale, and complexity. However, all lumen formation processes share key morphogenetic principles that underly their development. Fundamentally, a lumen simply consists of epithelial cells that encapsulate a continuous internal space, and a common way of building a lumen is via opening and enlarging by filling it with fluid and/or macromolecules. Here, we discuss how polarized targeting of membrane and secreted proteins regulates lumen formation, mainly focusing on ion transporters in vertebrate model systems. We also discuss mechanistic differences observed among invertebrates and vertebrates and describe how the unique properties of the Na+/K+-ATPase and junctional proteins can promote polarization of immature epithelia to build lumina de novo in developing organs.
Collapse
Affiliation(s)
- Daniel S Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
16
|
Nuclear Factor of Activated T Cells-5 Regulates Notochord Lumenogenesis in Chordate Larval Development. Int J Mol Sci 2022; 23:ijms232214407. [PMID: 36430885 PMCID: PMC9698811 DOI: 10.3390/ijms232214407] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Osmoregulation is essential for organisms to adapt to the exterior environment and plays an important role in embryonic organogenesis. Tubular organ formation usually involves a hyperosmotic lumen environment. The mechanisms of how the cells respond and regulate lumen formation remain largely unknown. Here, we reported that the nuclear factor of activated T cells-5 (NFAT5), the only transcription factor in the NFAT family involved in the cellular responses to hypertonic stress, regulated notochord lumen formation in chordate Ciona. Ciona NFAT5 (Ci-NFAT5) was expressed in notochord, and its expression level increased during notochord lumen formation and expansion. Knockout and expression of the dominant negative of NFAT5 in Ciona embryos resulted in the failure of notochord lumen expansion. We further demonstrated that the Ci-NFAT5 transferred from the cytoplasm into nuclei in HeLa cells under the hyperosmotic medium, indicating Ci-NFAT5 can respond the hypertonicity. To reveal the underly mechanisms, we predicted potential downstream genes of Ci-NFAT5 and further validated Ci-NFAT5-interacted genes by the luciferase assay. The results showed that Ci-NFAT5 promoted SLC26A6 expression. Furthermore, expression of a transport inactivity mutant of SLC26A6 (L421P) in notochord led to the failure of lumen expansion, phenocopying that of Ci-NFAT5 knockout. These results suggest that Ci-NFAT5 regulates notochord lumen expansion via the SLC26A6 axis. Taken together, our results reveal that the chordate NFAT5 responds to hypertonic stress and regulates lumen osmotic pressure via an ion channel pathway on luminal organ formation.
Collapse
|
17
|
Fuji K, Tanida S, Sano M, Nonomura M, Riveline D, Honda H, Hiraiwa T. Computational approaches for simulating luminogenesis. Semin Cell Dev Biol 2022; 131:173-185. [PMID: 35773151 DOI: 10.1016/j.semcdb.2022.05.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022]
Abstract
Lumens, liquid-filled cavities surrounded by polarized tissue cells, are elementary units involved in the morphogenesis of organs. Theoretical modeling and computations, which can integrate various factors involved in biophysics of morphogenesis of cell assembly and lumens, may play significant roles to elucidate the mechanisms in formation of such complex tissue with lumens. However, up to present, it has not been documented well what computational approaches or frameworks can be applied for this purpose and how we can choose the appropriate approach for each problem. In this review, we report some typical lumen morphologies and basic mechanisms for the development of lumens, focusing on three keywords - mechanics, hydraulics and geometry - while outlining pros and cons of the current main computational strategies. We also describe brief guidance of readouts, i.e., what we should measure in experiments to make the comparison with the model's assumptions and predictions.
Collapse
Affiliation(s)
- Kana Fuji
- Universal Biology Institute, Graduate School of Science, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sakurako Tanida
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, Japan
| | - Masaki Sano
- Institute of Natural Sciences, School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Makiko Nonomura
- Department of Mathematical Information Engineering, College of Industrial Technology, Nihon University, 1-2-1 Izumicho, Narashino-shi, Chiba 275-8575, Japan
| | - Daniel Riveline
- Laboratory of Cell Physics IGBMC, CNRS, INSERM and Université de Strasbourg, Strasbourg, France
| | - Hisao Honda
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine Kobe University, Kobe, Hyogo, Japan
| | - Tetsuya Hiraiwa
- Mechanobiology Institute, Singapore, National University of Singapore, 117411, Singapore.
| |
Collapse
|
18
|
Choudhury MI, Benson MA, Sun SX. Trans-epithelial fluid flow and mechanics of epithelial morphogenesis. Semin Cell Dev Biol 2022; 131:146-159. [PMID: 35659163 DOI: 10.1016/j.semcdb.2022.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/14/2022]
Abstract
Active fluid transport across epithelial monolayers is emerging as a major driving force of tissue morphogenesis in a variety of healthy and diseased systems, as well as during embryonic development. Cells use directional transport of ions and osmotic gradients to drive fluid flow across the cell surface, in the process also building up fluid pressure. The basic physics of this process is described by the osmotic engine model, which also underlies actin-independent cell migration. Recently, the trans-epithelial fluid flux and the hydraulic pressure gradient have been explicitly measured for a variety of cellular and tissue model systems across various species. For the kidney, it was shown that tubular epithelial cells behave as active mechanical fluid pumps: the trans-epithelial fluid flux depends on the hydraulic pressure difference across the epithelial layer. When a stall pressure is reached, the fluid flux vanishes. Hydraulic forces generated from active fluid pumping are important in tissue morphogenesis and homeostasis, and could also underlie multiple morphogenic events seen in other developmental contexts. In this review, we highlight findings that examined the role of trans-epithelial fluid flux and hydraulic pressure gradient in driving tissue-scale morphogenesis. We also review organ pathophysiology due to impaired fluid pumping and the loss of hydraulic pressure sensing at the cellular scale. Finally, we draw an analogy between cellular fluidic pumps and a connected network of water pumps in a city. The dynamics of fluid transport in an active and adaptive network is determined globally at the systemic level, and transport in such a network is best when each pump is operating at its optimal efficiency.
Collapse
Affiliation(s)
- Mohammad Ikbal Choudhury
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States; Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| | - Morgan A Benson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States; Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD 21218, United States.
| |
Collapse
|
19
|
Abstract
During organismal development, organs and systems are built following a genetic blueprint that produces structures capable of performing specific physiological functions. Interestingly, we have learned that the physiological activities of developing tissues also contribute to their own morphogenesis. Specifically, physiological activities such as fluid secretion and cell contractility generate hydrostatic pressure that can act as a morphogenetic force. Here, we first review the role of hydrostatic pressure in tube formation during animal development and discuss mathematical models of lumen formation. We then illustrate specific roles of the notochord as a hydrostatic scaffold in anterior-posterior axis development in chordates. Finally, we cover some examples of how fluid flows influence morphogenetic processes in other developmental contexts. Understanding how fluid forces act during development will be key for uncovering the self-organizing principles that control morphogenesis.
Collapse
Affiliation(s)
- Michel Bagnat
- Department of Cell Biology, Duke University, Durham, North Carolina, USA;
| | - Bijoy Daga
- Department of Cell Biology, Duke University, Durham, North Carolina, USA;
| | - Stefano Di Talia
- Department of Cell Biology, Duke University, Durham, North Carolina, USA;
- Department of Orthopaedic Surgery, Duke University, Durham, North Carolina, USA
| |
Collapse
|
20
|
Shashikanth N, France MM, Xiao R, Haest X, Rizzo HE, Yeste J, Reiner J, Turner JR. Tight junction channel regulation by interclaudin interference. Nat Commun 2022; 13:3780. [PMID: 35773259 PMCID: PMC9246906 DOI: 10.1038/s41467-022-31587-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 06/23/2022] [Indexed: 02/06/2023] Open
Abstract
Tight junctions form selectively permeable seals across the paracellular space. Both barrier function and selective permeability have been attributed to members of the claudin protein family, which can be categorized as pore-forming or barrier-forming. Here, we show that claudin-4, a prototypic barrier-forming claudin, reduces paracellular permeability by a previously unrecognized mechanism. Claudin-4 knockout or overexpression has minimal effects on tight junction permeability in the absence of pore-forming claudins. However, claudin-4 selectively inhibits flux across cation channels formed by claudins 2 or 15. Claudin-4-induced loss of claudin channel function is accompanied by reduced anchoring and subsequent endocytosis of pore-forming claudins. Analyses in nonepithelial cells show that claudin-4, which is incapable of independent polymerization, disrupts polymeric strands and higher order meshworks formed by claudins 2, 7, 15, and 19. This process of interclaudin interference, in which one claudin disrupts higher order structures and channels formed by a different claudin, represents a previously unrecognized mechanism of barrier regulation.
Collapse
Affiliation(s)
- Nitesh Shashikanth
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Marion M France
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruyue Xiao
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Heather E Rizzo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jose Yeste
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Instituto de Microelectrónica de Barcelona, IMB-CNM (CSIC), Bellaterra, Spain
| | - Johannes Reiner
- Division of Gastroenterology and Endocrinology, Department of Medicine II, Rostock University Medical Center, Ernst-Heydemann-Str. 6, Rostock, Germany
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Hamilton MK, Wall ES, Robinson CD, Guillemin K, Eisen JS. Enteric nervous system modulation of luminal pH modifies the microbial environment to promote intestinal health. PLoS Pathog 2022; 18:e1009989. [PMID: 35143593 PMCID: PMC8830661 DOI: 10.1371/journal.ppat.1009989] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/07/2022] [Indexed: 01/02/2023] Open
Abstract
The enteric nervous system (ENS) controls many aspects of intestinal homeostasis, including parameters that shape the habitat of microbial residents. Previously we showed that zebrafish lacking an ENS, due to deficiency of the sox10 gene, develop intestinal inflammation and bacterial dysbiosis, with an expansion of proinflammatory Vibrio strains. To understand the primary defects resulting in dysbiosis in sox10 mutants, we investigated how the ENS shapes the intestinal environment in the absence of microbiota and associated inflammatory responses. We found that intestinal transit, intestinal permeability, and luminal pH regulation are all aberrant in sox10 mutants, independent of microbially induced inflammation. Treatment with the proton pump inhibitor, omeprazole, corrected the more acidic luminal pH of sox10 mutants to wild type levels. Omeprazole treatment also prevented overabundance of Vibrio and ameliorated inflammation in sox10 mutant intestines. Treatment with the carbonic anhydrase inhibitor, acetazolamide, caused wild type luminal pH to become more acidic, and increased both Vibrio abundance and intestinal inflammation. We conclude that a primary function of the ENS is to regulate luminal pH, which plays a critical role in shaping the resident microbial community and regulating intestinal inflammation.
Collapse
Affiliation(s)
- M. Kristina Hamilton
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Elena S. Wall
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Catherine D. Robinson
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
- Humans and the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- * E-mail: (KG); (JSE)
| | - Judith S. Eisen
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
- * E-mail: (KG); (JSE)
| |
Collapse
|
22
|
La Charité-Harbec S, Lachance JFB, Ryan AK, Gupta IR. Claudin-3 regulates luminal fluid accumulation in the developing chick lung. Differentiation 2022; 124:52-59. [DOI: 10.1016/j.diff.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 12/31/2021] [Accepted: 01/28/2022] [Indexed: 11/03/2022]
|
23
|
Bugda Gwilt K, Thiagarajah JR. Membrane Lipids in Epithelial Polarity: Sorting out the PIPs. Front Cell Dev Biol 2022; 10:893960. [PMID: 35712665 PMCID: PMC9197455 DOI: 10.3389/fcell.2022.893960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
The development of cell polarity in epithelia, is critical for tissue morphogenesis and vectorial transport between the environment and the underlying tissue. Epithelial polarity is defined by the development of distinct plasma membrane domains: the apical membrane interfacing with the exterior lumen compartment, and the basolateral membrane directly contacting the underlying tissue. The de novo generation of polarity is a tightly regulated process, both spatially and temporally, involving changes in the distribution of plasma membrane lipids, localization of apical and basolateral membrane proteins, and vesicular trafficking. Historically, the process of epithelial polarity has been primarily described in relation to the localization and function of protein 'polarity complexes.' However, a critical and foundational role is emerging for plasma membrane lipids, and in particular phosphoinositide species. Here, we broadly review the evidence for a primary role for membrane lipids in the generation of epithelial polarity and highlight key areas requiring further research. We discuss the complex interchange that exists between lipid species and briefly examine how major membrane lipid constituents are generated and intersect with vesicular trafficking to be preferentially localized to different membrane domains with a focus on some of the key protein-enzyme complexes involved in these processes.
Collapse
Affiliation(s)
- Katlynn Bugda Gwilt
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
24
|
Palmer MA, Nerger BA, Goodwin K, Sudhakar A, Lemke SB, Ravindran PT, Toettcher JE, Košmrlj A, Nelson CM. Stress ball morphogenesis: How the lizard builds its lung. SCIENCE ADVANCES 2021; 7:eabk0161. [PMID: 34936466 PMCID: PMC8694616 DOI: 10.1126/sciadv.abk0161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/08/2021] [Indexed: 06/14/2023]
Abstract
The function of the lung is closely coupled to its structural anatomy, which varies greatly across vertebrates. Although architecturally simple, a complex pattern of airflow is thought to be achieved in the lizard lung due to its cavernous central lumen and honeycomb-shaped wall. We find that the wall of the lizard lung is generated from an initially smooth epithelial sheet, which is pushed through holes in a hexagonal smooth muscle meshwork by forces from fluid pressure, similar to a stress ball. Combining transcriptomics with time-lapse imaging reveals that the hexagonal meshwork self-assembles in response to circumferential and axial stresses downstream of pressure. A computational model predicts the pressure-driven changes in epithelial topology, which we probe using optogenetically driven contraction of 3D-printed engineered muscle. These results reveal the physical principles used to sculpt the unusual architecture of the lizard lung, which could be exploited as a novel strategy to engineer tissues.
Collapse
Affiliation(s)
- Michael A. Palmer
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Bryan A. Nerger
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Anvitha Sudhakar
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Sandra B. Lemke
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | | | - Jared E. Toettcher
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Andrej Košmrlj
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
- Princeton Institute for the Science and Technology of Materials, Princeton University, Princeton, NJ 08544,USA
| | - Celeste M. Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
25
|
Blastocoel morphogenesis: A biophysics perspective. Semin Cell Dev Biol 2021; 130:12-23. [PMID: 34756494 DOI: 10.1016/j.semcdb.2021.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/21/2021] [Accepted: 10/08/2021] [Indexed: 11/22/2022]
Abstract
The blastocoel is a fluid-filled cavity characteristic of animal embryos at the blastula stage. Its emergence is commonly described as the result of cleavage patterning, but this historical view conceals a large diversity of mechanisms and overlooks many unsolved questions from a biophysics perspective. In this review, we describe generic mechanisms for blastocoel morphogenesis, rooted in biological literature and simple physical principles. We propose novel directions of study and emphasize the importance to study blastocoel morphogenesis as an evolutionary and physical continuum.
Collapse
|
26
|
Vasquez CG, Vachharajani VT, Garzon-Coral C, Dunn AR. Physical basis for the determination of lumen shape in a simple epithelium. Nat Commun 2021; 12:5608. [PMID: 34556639 PMCID: PMC8460836 DOI: 10.1038/s41467-021-25050-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
The formation of a hollow lumen in a formerly solid mass of cells is a key developmental process whose dysregulation leads to diseases of the kidney and other organs. Hydrostatic pressure has been proposed to drive lumen expansion, a view that is supported by experiments in the mouse blastocyst. However, lumens formed in other tissues adopt irregular shapes with cell apical faces that are bowed inward, suggesting that pressure may not be the dominant contributor to lumen shape in all cases. Here we use live-cell imaging to study the physical mechanism of lumen formation in Madin-Darby Canine Kidney cell spheroids, a canonical cell-culture model for lumenogenesis. We find that in this system, lumen shape reflects basic geometrical considerations tied to the establishment of apico-basal polarity. A physical model incorporating both cell geometry and intraluminal pressure can account for our observations as well as cases in which pressure plays a dominant role.
Collapse
Affiliation(s)
| | | | | | - Alexander R Dunn
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
27
|
Ivanovics B, Gazsi G, Reining M, Berta I, Poliska S, Toth M, Domokos A, Nagy B, Staszny A, Cserhati M, Csosz E, Bacsi A, Csenki-Bakos Z, Acs A, Urbanyi B, Czimmerer Z. Embryonic exposure to low concentrations of aflatoxin B1 triggers global transcriptomic changes, defective yolk lipid mobilization, abnormal gastrointestinal tract development and inflammation in zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2021; 416:125788. [PMID: 33838512 DOI: 10.1016/j.jhazmat.2021.125788] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/19/2021] [Accepted: 03/29/2021] [Indexed: 06/12/2023]
Abstract
Aflatoxin B1-contaminated feeds and foods induce various health problems in domesticated animals and humans, including tumor development and hepatotoxicity. Aflatoxin B1 also has embryotoxic effects in different livestock species and humans. However, it is difficult to distinguish between the indirect, maternally-mediated toxic effects and the direct embryotoxicity of aflatoxin B1 in mammals. In the present study, we investigated the aflatoxin B1-induced direct embryotoxic effects in a zebrafish embryo model system combining toxicological, transcriptomic, immunological, and biochemical approaches. Embryonic exposure to aflatoxin B1 induced significant changes at the transcriptome level resulting in elevated expression of inflammatory gene network and repression of lipid metabolism and gastrointestinal tract development-related gene sets. According to the gene expression changes, massive neutrophil granulocyte influx, elevated nitric oxide production, and yolk lipid accumulation were observed in the abdominal region of aflatoxin B1-exposed larvae. In parallel, aflatoxin B1-induced defective gastrointestinal tract development and reduced L-arginine level were found in our model system. Our results revealed the complex direct embryotoxic effects of aflatoxin B1, including inhibited lipid utilization, defective intestinal development, and inflammation.
Collapse
Affiliation(s)
- Bence Ivanovics
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Gyongyi Gazsi
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Marta Reining
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Izabella Berta
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Szilard Poliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Marta Toth
- Department of Immunology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Apolka Domokos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; Molecular Cell and Immunobiology Doctoral School, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary
| | - Bela Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Adam Staszny
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Matyas Cserhati
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Eva Csosz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Attila Bacsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Zsolt Csenki-Bakos
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Andras Acs
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary
| | - Bela Urbanyi
- Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, H-2100 Godollo, Hungary.
| | - Zsolt Czimmerer
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| |
Collapse
|
28
|
Torres-Sánchez A, Winter MK, Salbreux G. Tissue hydraulics: Physics of lumen formation and interaction. Cells Dev 2021; 168:203724. [PMID: 34339904 DOI: 10.1016/j.cdev.2021.203724] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/08/2021] [Accepted: 07/20/2021] [Indexed: 11/29/2022]
Abstract
Lumen formation plays an essential role in the morphogenesis of tissues during development. Here we review the physical principles that play a role in the growth and coarsening of lumens. Solute pumping by the cell, hydraulic flows driven by differences of osmotic and hydrostatic pressures, balance of forces between extracellular fluids and cell-generated cytoskeletal forces, and electro-osmotic effects have been implicated in determining the dynamics and steady-state of lumens. We use the framework of linear irreversible thermodynamics to discuss the relevant force, time and length scales involved in these processes. We focus on order of magnitude estimates of physical parameters controlling lumen formation and coarsening.
Collapse
Affiliation(s)
| | - Max Kerr Winter
- The Francis Crick Institute, 1 Midland Road, NW1 1AT, United Kingdom
| | - Guillaume Salbreux
- The Francis Crick Institute, 1 Midland Road, NW1 1AT, United Kingdom; University of Geneva, Quai Ernest Ansermet 30, 1205 Genève, Switzerland.
| |
Collapse
|
29
|
Smoothelin-like 2 Inhibits Coronin-1B to Stabilize the Apical Actin Cortex during Epithelial Morphogenesis. Curr Biol 2021; 31:696-706.e9. [PMID: 33275893 DOI: 10.1016/j.cub.2020.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 09/24/2020] [Accepted: 11/03/2020] [Indexed: 11/22/2022]
Abstract
The actin cortex is involved in many biological processes and needs to be significantly remodeled during cell differentiation. Developing epithelial cells construct a dense apical actin cortex to carry out their barrier and exchange functions. The apical cortex assembles in response to three-dimensional (3D) extracellular cues, but the regulation of this process during epithelial morphogenesis remains unknown. Here, we describe the function of Smoothelin-like 2 (SMTNL2), a member of the smooth-muscle-related Smoothelin protein family, in apical cortex maturation. SMTNL2 is induced during development in multiple epithelial tissues and localizes to the apical and junctional actin cortex in intestinal and kidney epithelial cells. SMTNL2 deficiency leads to membrane herniations in the apical domain of epithelial cells, indicative of cortex abnormalities. We find that SMTNL2 binds to actin filaments and is required to slow down the turnover of apical actin. We also characterize the SMTNL2 proximal interactome and find that SMTNL2 executes its functions partly through inhibition of coronin-1B. Although coronin-1B-mediated actin dynamics are required for early morphogenesis, its sustained activity is detrimental for the mature apical shape. SMTNL2 binds to coronin-1B through its N-terminal coiled-coil region and negates its function to stabilize the apical cortex. In sum, our results unveil a mechanism for regulating actin dynamics during epithelial morphogenesis, providing critical insights on the developmental control of the cellular cortex.
Collapse
|
30
|
Fiorentino A, Christophorou A, Massa F, Garbay S, Chiral M, Ramsing M, Rasmussen M, Gubler MC, Bessieres B, Heidet L, Fischer E, Pontoglio M. Developmental Renal Glomerular Defects at the Origin of Glomerulocystic Disease. Cell Rep 2020; 33:108304. [PMID: 33113370 DOI: 10.1016/j.celrep.2020.108304] [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] [Received: 05/11/2020] [Revised: 07/28/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022] Open
Abstract
The architecture of renal glomeruli is acquired through intricate and still poorly understood developmental steps. In our study we identify a crucial glomerular morphogenetic event in nephrogenesis that drives the remodeling/separation of the prospective vascular pole (the future entrance of the glomerular arterioles) and the urinary pole (the tubular outflow). We demonstrate that this remodeling is genetically programmed. In fact, in mouse and human, the absence of HNF1B impairs the remodeling/separation of the two poles, leading to trapping and constriction of the tubular outflow inside the glomerulus. This aberration gives rise to obstructive glomerular dilations upon the initiation of primary urine production. In this context, we show that pharmacological decrease of glomerular filtration significantly contains cystic expansion. From a developmental point of view, our study discloses a crucial event on glomerular patterning affecting the "inside-outside" fate of the epithelia in the renal glomerulus.
Collapse
Affiliation(s)
- Arianna Fiorentino
- Université de Paris, Institut Necker-Enfants Malades (INEM), Epigenetics and Development Team, INSERM U1151, CNRS UMR 8253, 75015 Paris, France
| | - Armelle Christophorou
- Université de Paris, Institut Necker-Enfants Malades (INEM), Epigenetics and Development Team, INSERM U1151, CNRS UMR 8253, 75015 Paris, France
| | - Filippo Massa
- Université de Paris, Institut Necker-Enfants Malades (INEM), Epigenetics and Development Team, INSERM U1151, CNRS UMR 8253, 75015 Paris, France
| | - Serge Garbay
- Université de Paris, Institut Necker-Enfants Malades (INEM), Epigenetics and Development Team, INSERM U1151, CNRS UMR 8253, 75015 Paris, France
| | - Magali Chiral
- Université de Paris, Institut Necker-Enfants Malades (INEM), Epigenetics and Development Team, INSERM U1151, CNRS UMR 8253, 75015 Paris, France
| | - Mette Ramsing
- Department of Pathology, Randers Regional Hospital, 8930 Randers, Denmark
| | - Maria Rasmussen
- Department of Genetics, Vejle Hospital, Lillebælt Hospital, University of Southern Denmark, 7100 Vejle, Denmark
| | - Marie-Claire Gubler
- INSERM U1163/Centre de Référence MARHEA, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Bettina Bessieres
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Laurence Heidet
- INSERM U1163/Centre de Référence MARHEA, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Evelyne Fischer
- Université de Paris, Institut Necker-Enfants Malades (INEM), Epigenetics and Development Team, INSERM U1151, CNRS UMR 8253, 75015 Paris, France.
| | - Marco Pontoglio
- Université de Paris, Institut Necker-Enfants Malades (INEM), Epigenetics and Development Team, INSERM U1151, CNRS UMR 8253, 75015 Paris, France.
| |
Collapse
|
31
|
Cohen JD, Sparacio AP, Belfi AC, Forman-Rubinsky R, Hall DH, Maul-Newby H, Frand AR, Sundaram MV. A multi-layered and dynamic apical extracellular matrix shapes the vulva lumen in Caenorhabditis elegans. eLife 2020; 9:e57874. [PMID: 32975517 PMCID: PMC7544507 DOI: 10.7554/elife.57874] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Biological tubes must develop and maintain their proper diameter to transport materials efficiently. These tubes are molded and protected in part by apical extracellular matrices (aECMs) that line their lumens. Despite their importance, aECMs are difficult to image in vivo and therefore poorly understood. The Caenorhabditis elegans vulva has been a paradigm for understanding many aspects of organogenesis. Here we describe the vulva luminal matrix, which contains chondroitin proteoglycans, Zona Pellucida (ZP) domain proteins, and other glycoproteins and lipid transporters related to those in mammals. Confocal and transmission electron microscopy revealed, with unprecedented detail, a complex and dynamic aECM. Different matrix factors assemble on the apical surfaces of each vulva cell type, with clear distinctions seen between Ras-dependent (1°) and Notch-dependent (2°) cell types. Genetic perturbations suggest that chondroitin and other aECM factors together generate a structured scaffold that both expands and constricts lumen shape.
Collapse
Affiliation(s)
- Jennifer D Cohen
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Alessandro P Sparacio
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Alexandra C Belfi
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Rachel Forman-Rubinsky
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Hannah Maul-Newby
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Alison R Frand
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| |
Collapse
|
32
|
Characterization and molecular evolution of claudin genes in the Pungitius sinensis. J Comp Physiol B 2020; 190:749-759. [PMID: 32778926 DOI: 10.1007/s00360-020-01301-5] [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: 02/20/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 10/23/2022]
Abstract
Claudins are a family of integrated membrane-bound proteins involving in paracellular tightness, barrier forming, ion permeability, and substrate selection at tight junctions of chordate epithelial and endothelial cells. Here, 39 putative claudin genes were identified in the Pungitius sinensis based on the high throughput RNA-seq. Conservative motif distribution in each group suggested functional relevance. Divergence of duplicated genes implied the species' adaptation to the environment. In addition, selective pressure analyses identified one site, which may accelerate functional divergence in this protein family. Pesticides cause environmental pollution and have a serious impact on aquatic organisms when entering the water. The expression pattern of most claudin genes was affected by organophosphorus pesticide, indicating that they may be involved in the immune regulation of organisms and the detoxification of xenobiotics. Protein-protein network analyses also exhibited 439 interactions, which implied the functional diversity. It will provide some references for the functional study on claudin genes.
Collapse
|
33
|
Brzezinski K, MacMillan HA. Chilling induces unidirectional solute leak through the locust gut epithelia. J Exp Biol 2020; 223:jeb215475. [PMID: 32532867 DOI: 10.1242/jeb.215475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 06/03/2020] [Indexed: 08/26/2023]
Abstract
Chill-susceptible insects, like the migratory locust, often die when exposed to low temperatures from an accumulation of tissue damage that is unrelated to freezing (chilling injury). Chilling injury is often associated with a loss of ion balance across the gut epithelia. It has recently been suggested that this imbalance is at least partly caused by a cold-induced disruption of epithelial barrier function. Here, we aimed to test this hypothesis in the migratory locust (Locustamigratoria). First, chill tolerance was quantified by exposing locusts to -2°C and recording chill coma recovery time and survival 24 h post-cold exposure. Longer exposure times significantly increased recovery time and caused injury and death. Ion-selective microelectrodes were also used to test for a loss of ion balance in the cold. We found a significant increase of haemolymph K+ and decrease of haemolymph Na+ concentration over time. Next, barrier failure along the gut was tested by monitoring the movement of an epithelial barrier marker (FITC-dextran) across the gut epithelia during exposure to -2°C. We found a significant increase in haemolymph FITC-dextran concentration over time in the cold when assayed in the mucosal to serosal direction. However, when tested in the serosal to mucosal direction, we saw minimal marker movement across the gut epithelia. This suggests that while cold-induced barrier disruption is present, it is apparently unidirectional. It is important to note that these data reveal only the phenomenon itself. The location of this leak as well as the underlying mechanisms remain unclear and require further investigation.
Collapse
Affiliation(s)
- Kaylen Brzezinski
- Department of Biology, Carleton University, Ottawa, ON, Canada K1S 5B6
| | - Heath A MacMillan
- Department of Biology, Carleton University, Ottawa, ON, Canada K1S 5B6
| |
Collapse
|
34
|
Madsen SS, Bollinger RJ, Brauckhoff M, Engelund MB. Gene expression profiling of proximal and distal renal tubules in Atlantic salmon ( Salmo salar) acclimated to fresh water and seawater. Am J Physiol Renal Physiol 2020; 319:F380-F393. [PMID: 32628538 DOI: 10.1152/ajprenal.00557.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Euryhaline teleost kidneys undergo a major functional switch from being filtratory in freshwater (FW) to being predominantly secretory in seawater (SW) conditions. The transition involves both vascular and tubular effects. There is consensus that the glomerular filtration rate is greatly reduced upon exposure to hyperosmotic conditions. Yet, regulation at the tubular level has only been examined sporadically in a few different species. This study aimed to obtain a broader understanding of transcriptional regulation in proximal versus distal tubular segments during osmotic transitions. Proximal and distal tubule cells were dissected separately by laser capture microdissection, RNA was extracted, and relative mRNA expression levels of >30 targets involved in solute and water transport were quantified by quantitative PCR in relation to segment type in fish acclimated to FW or SW. The gene categories were aquaporins, solute transporters, fxyd proteins, and tight junction proteins. aqp8bb1, aqp10b1, nhe3, sglt1, slc41a1, cnnm3, fxyd12a, cldn3b, cldn10b, cldn15a, and cldn12 were expressed at a higher level in proximal compared with distal tubules. aqp1aa, aqp1ab, nka-a1a, nka-a1b, nkcc1a, nkcc2, ncc, clc-k, slc26a6C, sglt2, fxyd2, cldn3a, and occln were expressed at a higher level in distal compared with proximal tubules. Expression of aqp1aa, aqp3a1, aqp10b1, ncc, nhe3, cftr, sglt1, slc41a1, fxyd12a, cldn3a, cldn3b, cldn3c, cldn10b, cldn10e, cldn28a, and cldn30c was higher in SW- than in FW-acclimated salmon, whereas the opposite was the case for aqp1ab, slc26a6C, and fxyd2. The data show distinct segmental distribution of transport genes and a significant regulation of tubular transcripts when kidney function is modulated during salinity transitions.
Collapse
Affiliation(s)
- Steffen S Madsen
- Department of Biology, University of Southern Denmark, Odense M, Denmark
| | | | - Melanie Brauckhoff
- Department of Biology, University of Southern Denmark, Odense M, Denmark
| | | |
Collapse
|
35
|
Zhang D, Sun B, Zhao X, Sun H, An J, Lin X, Zhu D, Zhao X, Wang X, Liu F, Zhang Y, Liu J, Gu Q, Dong X, Qiu Z, Liu Z, Qi H, Che N, Li J, Cheng R, Zheng X. Twist1 accelerates tumour vasculogenic mimicry by inhibiting Claudin15 expression in triple-negative breast cancer. J Cell Mol Med 2020; 24:7163-7174. [PMID: 32469152 PMCID: PMC7339217 DOI: 10.1111/jcmm.15167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 07/16/2019] [Accepted: 08/21/2019] [Indexed: 12/18/2022] Open
Abstract
The up-regulation of EMT regulator Twist1 has been implicated in vasculogenic mimicry (VM) formation in human triple-negative breast cancer (TNBC). Twist1 targets the Claudin15 promoter in hepatocellular carcinoma cells. Claudin family members are related with TNBC. However, the relationship between Claudin15 and VM formation is not clear. In this study, we first found that Claudin15 expression was frequently down-regulated in human TNBC, and Claudin15 down-regulation was significantly associated with VM and Twist1 nuclear expression. Claudin15 down-regulation correlated with shorter survival compared with high levels. Claudin15 silence significantly enhanced cell motility, invasiveness and VM formation in the non-TNBC MCF-7 cells. Conversely, an up-regulation of Claudin15 remarkably reduced TNBC MDA-MB-231 cell migration, invasion and VM formation. We also showed that down-regulation of Claudin15 was Twist1-dependent, and Twist1 repressed Claudin15 promoter activity. Furthermore, GeneChip analyses of mammary glands of Claudin15-deficient mice indicated that Claudin18 and Jun might be downstream factors of Twist1-Claudin15. Our results suggest that Twist1 induced VM through Claudin15 suppression in TNBC, and Twist1 inhibition of Claudin15 might involve Claudin18 and Jun expression.
Collapse
Affiliation(s)
- Danfang Zhang
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Baocun Sun
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
- Department of PathologyCancer Hospital of Tianjin Medical UniversityTianjinChina
| | - Xiulan Zhao
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Huizhi Sun
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Jindan An
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Xian Lin
- Department of PathologyTianjin Medical UniversityTianjinChina
| | - Dongwang Zhu
- Department of PathologyTianjin Medical UniversityTianjinChina
| | - Xueming Zhao
- Department of PathologyTianjin Medical UniversityTianjinChina
| | - Xudong Wang
- Department of PathologyCancer Hospital of Tianjin Medical UniversityTianjinChina
| | - Fang Liu
- Department of PathologyTianjin Medical UniversityTianjinChina
| | - Yanhui Zhang
- Department of PathologyCancer Hospital of Tianjin Medical UniversityTianjinChina
| | - Jiameng Liu
- Department of PathologyTianjin Medical UniversityTianjinChina
| | - Qiang Gu
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Xueyi Dong
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Zhiqiang Qiu
- Department of PathologyCancer Hospital of Tianjin Medical UniversityTianjinChina
| | - Zhiyong Liu
- Department of PathologyCancer Hospital of Tianjin Medical UniversityTianjinChina
| | - Hong Qi
- Department of PathologyTianjin Medical UniversityTianjinChina
| | - Na Che
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Jing Li
- Department of PathologyTianjin Medical UniversityTianjinChina
- Department of PathologyGeneral Hospital of Tianjin Medical UniversityTianjinChina
| | - Runfen Cheng
- Department of PathologyCancer Hospital of Tianjin Medical UniversityTianjinChina
| | - Xu Zheng
- Department of PathologyTianjin Medical UniversityTianjinChina
| |
Collapse
|
36
|
Meoli L, Günzel D. Channel functions of claudins in the organization of biological systems. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183344. [PMID: 32442419 DOI: 10.1016/j.bbamem.2020.183344] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Claudins are tight junction proteins mostly appreciated in their function of paracellular barrier-formation. Compared to a virtual absence of any tight junctions, their paracellular sealing role certainly stands out. Yet, it was recognized immediately after the discovery of the first claudins, that some members of the claudin protein family were able to convey size and charge selectivity to the paracellular pathway. Thus, paracellular permeability can be fine-tuned according to the physiological needs of a tissue by inserting these channel-forming claudins into tight junction strands. Precise permeability adjustment is further suggested by the presence of numerous isoforms of channel-forming claudins (claudin-10b-, -15-, -16-like isoforms) in various vertebrate taxa. Moreover, their expression and localization are controlled by multiple transcriptional and posttranslational mechanisms. Consequently, mutation or dysregulation of channel-forming claudins can cause severe diseases. The present review therefore aims at providing an up-to-date report of the current research on these aspects of channel-forming claudins and their possible implications on future developments.
Collapse
Affiliation(s)
- Luca Meoli
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Dorothee Günzel
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, 12203 Berlin, Germany.
| |
Collapse
|
37
|
Durel JF, Nerurkar NL. Mechanobiology of vertebrate gut morphogenesis. Curr Opin Genet Dev 2020; 63:45-52. [PMID: 32413823 DOI: 10.1016/j.gde.2020.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/09/2020] [Indexed: 01/15/2023]
Abstract
Approximately a century after D'Arcy Thompson's On Growth and Form, there continues to be widespread interest in the biophysical and mathematical basis of morphogenesis. Particularly over the past 20 years, this interest has led to great advances in our understanding of a broad range of processes in embryonic development through a quantitative, mechanically driven framework. Nowhere in vertebrate development is this more apparent than the development of endodermally derived organs. Here, we discuss recent advances in the study of gut development that have emerged primarily from mechanobiology-motivated approaches that span from gut tube morphogenesis and later organogenesis of the respiratory and gastrointestinal systems.
Collapse
Affiliation(s)
- John F Durel
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States; Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, United States.
| |
Collapse
|
38
|
Tipsmark CK, Nielsen AM, Bossus MC, Ellis LV, Baun C, Andersen TL, Dreier J, Brewer JR, Madsen SS. Drinking and Water Handling in the Medaka Intestine: A Possible Role of Claudin-15 in Paracellular Absorption? Int J Mol Sci 2020; 21:ijms21051853. [PMID: 32182691 PMCID: PMC7085193 DOI: 10.3390/ijms21051853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/27/2022] Open
Abstract
When euryhaline fish move between fresh water (FW) and seawater (SW), the intestine undergoes functional changes to handle imbibed SW. In Japanese medaka, the potential transcellular aquaporin-mediated conduits for water are paradoxically downregulated during SW acclimation, suggesting paracellular transport to be of principal importance in hyperosmotic conditions. In mammals, intestinal claudin-15 (CLDN15) forms paracellular channels for small cations and water, which may participate in water transport. Since two cldn15 paralogs, cldn15a and cldn15b, have previously been identified in medaka, we examined the salinity effects on their mRNA expression and immunolocalization in the intestine. In addition, we analyzed the drinking rate and intestinal water handling by adding non-absorbable radiotracers, 51-Cr-EDTA or 99-Tc-DTPA, to the water. The drinking rate was >2-fold higher in SW than FW-acclimated fish, and radiotracer experiments showed anterior accumulation in FW and posterior buildup in SW intestines. Salinity had no effect on expression of cldn15a, while cldn15b was approximately 100-fold higher in FW than SW. Despite differences in transcript dynamics, Cldn15a and Cldn15b proteins were both similarly localized in the apical tight junctions of enterocytes, co-localizing with occludin and with no apparent difference in localization and abundance between FW and SW. The stability of the Cldn15 protein suggests a physiological role in water transport in the medaka intestine.
Collapse
Affiliation(s)
- Christian K. Tipsmark
- Department of Biological Sciences, University of Arkansas, SCEN 601, Fayetteville, AR 72701, USA; (M.C.B.); (L.V.E.); (S.S.M.)
- Correspondence: ; Tel.: +1-479-575-8436
| | - Andreas M. Nielsen
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark;
| | - Maryline C. Bossus
- Department of Biological Sciences, University of Arkansas, SCEN 601, Fayetteville, AR 72701, USA; (M.C.B.); (L.V.E.); (S.S.M.)
- Department of Math and Sciences, Lyon College, 2300 Highland Rd, Batesville, AR 72501, USA
| | - Laura V. Ellis
- Department of Biological Sciences, University of Arkansas, SCEN 601, Fayetteville, AR 72701, USA; (M.C.B.); (L.V.E.); (S.S.M.)
| | - Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000 Odense C, Denmark; (C.B.); (T.L.A.)
| | - Thomas L. Andersen
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000 Odense C, Denmark; (C.B.); (T.L.A.)
| | - Jes Dreier
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; (J.D.); (J.R.B.)
| | - Jonathan R. Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; (J.D.); (J.R.B.)
| | - Steffen S. Madsen
- Department of Biological Sciences, University of Arkansas, SCEN 601, Fayetteville, AR 72701, USA; (M.C.B.); (L.V.E.); (S.S.M.)
- Department of Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark;
| |
Collapse
|
39
|
Chan CJ, Hiiragi T. Integration of luminal pressure and signalling in tissue self-organization. Development 2020; 147:147/5/dev181297. [DOI: 10.1242/dev.181297] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
ABSTRACT
Many developmental processes involve the emergence of intercellular fluid-filled lumina. This process of luminogenesis results in a build up of hydrostatic pressure and signalling molecules in the lumen. However, the potential roles of lumina in cellular functions, tissue morphogenesis and patterning have yet to be fully explored. In this Review, we discuss recent findings that describe how pressurized fluid expansion can provide both mechanical and biochemical cues to influence cell proliferation, migration and differentiation. We also review emerging techniques that allow for precise quantification of fluid pressure in vivo and in situ. Finally, we discuss the intricate interplay between luminogenesis, tissue mechanics and signalling, which provide a new dimension for understanding the principles governing tissue self-organization in embryonic development.
Collapse
Affiliation(s)
- Chii J. Chan
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Takashi Hiiragi
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
40
|
A morphogenetic EphB/EphrinB code controls hepatopancreatic duct formation. Nat Commun 2019; 10:5220. [PMID: 31745086 PMCID: PMC6864101 DOI: 10.1038/s41467-019-13149-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022] Open
Abstract
The hepatopancreatic ductal (HPD) system connects the intrahepatic and intrapancreatic ducts to the intestine and ensures the afferent transport of the bile and pancreatic enzymes. Yet the molecular and cellular mechanisms controlling their differentiation and morphogenesis into a functional ductal system are poorly understood. Here, we characterize HPD system morphogenesis by high-resolution microscopy in zebrafish. The HPD system differentiates from a rod of unpolarized cells into mature ducts by de novo lumen formation in a dynamic multi-step process. The remodeling step from multiple nascent lumina into a single lumen requires active cell intercalation and myosin contractility. We identify key functions for EphB/EphrinB signaling in this dynamic remodeling step. Two EphrinB ligands, EphrinB1 and EphrinB2a, and two EphB receptors, EphB3b and EphB4a, control HPD morphogenesis by remodeling individual ductal compartments, and thereby coordinate the morphogenesis of this multi-compartment ductal system.
Collapse
|
41
|
Ryan AQ, Chan CJ, Graner F, Hiiragi T. Lumen Expansion Facilitates Epiblast-Primitive Endoderm Fate Specification during Mouse Blastocyst Formation. Dev Cell 2019; 51:684-697.e4. [PMID: 31735667 PMCID: PMC6912163 DOI: 10.1016/j.devcel.2019.10.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/29/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
Abstract
Epithelial tissues typically form lumina. In mammalian blastocysts, in which the first embryonic lumen forms, many studies have investigated how the cell lineages are specified through genetics and signaling, whereas potential roles of the fluid lumen have yet to be investigated. We discover that in mouse pre-implantation embryos at the onset of lumen formation, cytoplasmic vesicles are secreted into intercellular space. The segregation of epiblast and primitive endoderm directly follows lumen coalescence. Notably, pharmacological and biophysical perturbation of lumen expansion impairs the specification and spatial segregation of primitive endoderm cells within the blastocyst. Luminal deposition of FGF4 expedites fate specification and partially rescues the reduced specification in blastocysts with smaller cavities. Combined, our results suggest that blastocyst lumen expansion plays a critical role in guiding cell fate specification and positioning, possibly mediated by luminally deposited FGF4. Lumen expansion may provide a general mechanism for tissue pattern formation. Lumenogenesis coincides with cytoplasmic vesicle release into intercellular space Mouse blastocyst epiblast-primitive endoderm segregation follows lumen expansion Reduced lumen expansion impairs cell fate specification and segregation Luminally deposited FGF4 expedites epiblast-primitive endoderm specification
Collapse
Affiliation(s)
- Allyson Quinn Ryan
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Laboratoire Matière et Systèmes Complexes, Université Denis Diderot, Paris 7, CNRS UMR 7057, Condorcet Building 10 rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Chii Jou Chan
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - François Graner
- Laboratoire Matière et Systèmes Complexes, Université Denis Diderot, Paris 7, CNRS UMR 7057, Condorcet Building 10 rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Takashi Hiiragi
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
42
|
Mosaliganti KR, Swinburne IA, Chan CU, Obholzer ND, Green AA, Tanksale S, Mahadevan L, Megason SG. Size control of the inner ear via hydraulic feedback. eLife 2019; 8:39596. [PMID: 31571582 PMCID: PMC6773445 DOI: 10.7554/elife.39596] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/26/2019] [Indexed: 01/05/2023] Open
Abstract
Animals make organs of precise size, shape, and symmetry but how developing embryos do this is largely unknown. Here, we combine quantitative imaging, physical theory, and physiological measurement of hydrostatic pressure and fluid transport in zebrafish to study size control of the developing inner ear. We find that fluid accumulation creates hydrostatic pressure in the lumen leading to stress in the epithelium and expansion of the otic vesicle. Pressure, in turn, inhibits fluid transport into the lumen. This negative feedback loop between pressure and transport allows the otic vesicle to change growth rate to control natural or experimentally-induced size variation. Spatiotemporal patterning of contractility modulates pressure-driven strain for regional tissue thinning. Our work connects molecular-driven mechanisms, such as osmotic pressure driven strain and actomyosin tension, to the regulation of tissue morphogenesis via hydraulic feedback to ensure robust control of organ size. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
| | - Ian A Swinburne
- Department of Systems Biology, Harvard Medical School, Boston, United States
| | - Chon U Chan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, United States
| | - Nikolaus D Obholzer
- Department of Systems Biology, Harvard Medical School, Boston, United States
| | - Amelia A Green
- Department of Systems Biology, Harvard Medical School, Boston, United States
| | - Shreyas Tanksale
- Department of Systems Biology, Harvard Medical School, Boston, United States
| | - L Mahadevan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, United States.,Department of Organismal and Evolutionary Biology, Harvard University, Cambridge, United States.,Department of Physics, Harvard University, Cambridge, United States.,Kavli Institute for NanoBio Science and Technology, Harvard University, Cambridge, United States
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, United States
| |
Collapse
|
43
|
Micropattern-based platform as a physiologically relevant model to study epithelial morphogenesis and nephrotoxicity. Biomaterials 2019; 218:119339. [DOI: 10.1016/j.biomaterials.2019.119339] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 01/09/2023]
|
44
|
Nasr Esfahani S, Shao Y, Resto Irizarry AM, Li Z, Xue X, Gumucio DL, Fu J. Microengineered human amniotic ectoderm tissue array for high-content developmental phenotyping. Biomaterials 2019; 216:119244. [PMID: 31207406 PMCID: PMC6658735 DOI: 10.1016/j.biomaterials.2019.119244] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/01/2019] [Indexed: 02/09/2023]
Abstract
During early post-implantation human embryogenesis, the epiblast (EPI) within the blastocyst polarizes to generate a cyst with a central lumen. Cells at the uterine pole of the EPI cyst then undergo differentiation to form the amniotic ectoderm (AM), a tissue essential for further embryonic development. While the causes of early pregnancy failure are complex, improper lumenogenesis or amniogenesis of the EPI represent possible contributing factors. Here we report a novel AM microtissue array platform that allows quantitative phenotyping of lumenogenesis and amniogenesis of the EPI and demonstrate its potential application for embryonic toxicity profiling. Specifically, a human pluripotent stem cell (hPSC)-based amniogenic differentiation protocol was developed using a two-step micropatterning technique to generate a regular AM microtissue array with defined tissue sizes. A computer-assisted analysis pipeline was developed to automatically process imaging data and quantify morphological and biological features of AM microtissues. Analysis of the effects of cell density, cyst size and culture conditions revealed a clear connection between cyst size and amniogenesis of hPSC. Using this platform, we demonstrated that pharmacological inhibition of ROCK signaling, an essential mechanotransductive pathway, suppressed lumenogenesis but did not perturb amniogenic differentiation of hPSC, suggesting uncoupled regulatory mechanisms for AM morphogenesis vs. cytodifferentiation. The AM microtissue array was further applied to screen a panel of clinically relevant drugs, which successfully detected their differential teratogenecity. This work provides a technological platform for toxicological screening of clinically relevant drugs for their effects on lumenogenesis and amniogenesis during early human peri-implantation development, processes that have been previously inaccessible to study.
Collapse
Affiliation(s)
- Sajedeh Nasr Esfahani
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | | | - Zida Li
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
45
|
Lysosome-Rich Enterocytes Mediate Protein Absorption in the Vertebrate Gut. Dev Cell 2019; 51:7-20.e6. [PMID: 31474562 PMCID: PMC6783362 DOI: 10.1016/j.devcel.2019.08.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/06/2019] [Accepted: 07/29/2019] [Indexed: 12/25/2022]
Abstract
The guts of neonatal mammals and stomachless fish have a limited capacity for luminal protein digestion, which allows oral acquisition of antibodies and antigens. However, how dietary protein is absorbed during critical developmental stages when the gut is still immature is unknown. Here, we show that specialized intestinal cells, which we call lysosome-rich enterocytes (LREs), internalize dietary protein via receptor-mediated and fluid-phase endocytosis for intracellular digestion and trans-cellular transport. In LREs, we identify a conserved endocytic machinery, composed of the scavenger receptor complex Cubilin/Amnionless and Dab2, that is required for protein uptake by LREs and for growth and survival of larval zebrafish. Moreover, impairing LRE function in suckling mice, via conditional deletion of Dab2, leads to stunted growth and severe protein malnutrition reminiscent of kwashiorkor, a devastating human malnutrition syndrome. These findings identify digestive functions and conserved molecular mechanisms in LREs that are crucial for vertebrate growth and survival.
Collapse
|
46
|
Nowotschin S, Hadjantonakis AK, Campbell K. The endoderm: a divergent cell lineage with many commonalities. Development 2019; 146:146/11/dev150920. [PMID: 31160415 PMCID: PMC6589075 DOI: 10.1242/dev.150920] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The endoderm is a progenitor tissue that, in humans, gives rise to the majority of internal organs. Over the past few decades, genetic studies have identified many of the upstream signals specifying endoderm identity in different model systems, revealing them to be divergent from invertebrates to vertebrates. However, more recent studies of the cell behaviours driving endodermal morphogenesis have revealed a surprising number of shared features, including cells undergoing epithelial-to-mesenchymal transitions (EMTs), collective cell migration, and mesenchymal-to-epithelial transitions (METs). In this Review, we highlight how cross-organismal studies of endoderm morphogenesis provide a useful perspective that can move our understanding of this fascinating tissue forward.
Collapse
Affiliation(s)
- Sonja Nowotschin
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kyra Campbell
- Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK .,Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
47
|
Coronado M, Solis CJ, Hernandez PP, Feijóo CG. Soybean Meal-Induced Intestinal Inflammation in Zebrafish Is T Cell-Dependent and Has a Th17 Cytokine Profile. Front Immunol 2019; 10:610. [PMID: 31001250 PMCID: PMC6454071 DOI: 10.3389/fimmu.2019.00610] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 03/07/2019] [Indexed: 01/02/2023] Open
Abstract
Currently, inflammatory bowel disease (IBD) is a serious public health problem on the rise worldwide. In this work, we utilized the zebrafish to introduce a new model of intestinal inflammation triggered by food intake. Taking advantage of the translucency of the larvae and the availability of transgenic zebrafish lines with fluorescently labeled macrophages, neutrophils, or lymphocytes, we studied the behavior of these cell types in vivo during the course of inflammation. We established two feeding strategies, the first using fish that were not previously exposed to food (naïve strategy) and the second in which fish were initially exposed to normal food (developed strategy). In both strategies, we analyzed the effect of subsequent intake of a control or a soybean meal diet. Our results showed increased numbers of innate immune cells in the gut in both the naïve or developed protocols. Likewise, macrophages underwent drastic morphological changes after feeding, switching from a small and rounded contour to a larger and dendritic shape. Lymphocytes colonized the intestine as early as 5 days post fertilization and increased in numbers during the inflammatory process. Gene expression analysis indicated that lymphocytes present in the intestine correspond to T helper cells. Interestingly, control diet only induced a regulatory T cell profile in the developed model. On the contrary, soybean meal diet induced a Th17 response both in naïve and developed model. In addition, when feeding was performed in rag1-deficient fish, intestinal inflammation was not induced indicating that inflammation induced by soybean meal is T cell-dependent.
Collapse
Affiliation(s)
- Maximo Coronado
- Departamento de Ciencias Biologicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Camila J. Solis
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- Escuela de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad San Sebastian, Santiago, Chile
| | - Pedro P. Hernandez
- Macrophages and Development of Immunity, Institute Pasteur, Paris, France
| | - Carmen G. Feijóo
- Departamento de Ciencias Biologicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
| |
Collapse
|
48
|
Zha JM, Li HS, Lin Q, Kuo WT, Jiang ZH, Tsai PY, Ding N, Wu J, Xu SF, Wang YT, Pan J, Zhou XM, Chen K, Tao M, Odenwald MA, Tamura A, Tsukita S, Turner JR, He WQ. Interleukin 22 Expands Transit-Amplifying Cells While Depleting Lgr5 + Stem Cells via Inhibition of Wnt and Notch Signaling. Cell Mol Gastroenterol Hepatol 2018; 7:255-274. [PMID: 30686779 PMCID: PMC6352747 DOI: 10.1016/j.jcmgh.2018.09.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/24/2018] [Accepted: 09/06/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Epithelial regeneration is essential for homeostasis and repair of the mucosal barrier. In the context of infectious and immune-mediated intestinal disease, interleukin (IL) 22 is thought to augment these processes. We sought to define the mechanisms by which IL22 promotes mucosal healing. METHODS Intestinal stem cell cultures and mice were treated with recombinant IL22. Cell proliferation, death, and differentiation were assessed in vitro and in vivo by morphometric analysis, quantitative reverse transcriptase polymerase chain reaction, and immunohistochemistry. RESULTS IL22 increased the size and number of proliferating cells within enteroids but decreased the total number of enteroids. Enteroid size increases required IL22-dependent up-regulation of the tight junction cation and water channel claudin-2, indicating that enteroid enlargement reflected paracellular flux-induced swelling. However, claudin-2 did not contribute to IL22-dependent enteroid loss, depletion of Lgr5+ stem cells, or increased epithelial proliferation. IL22 induced stem cell apoptosis but, conversely, enhanced proliferation within and expanded numbers of transit-amplifying cells. These changes were associated with reduced wnt and notch signaling, both in vitro and in vivo, as well as skewing of epithelial differentiation, with increases in Paneth cells and reduced numbers of enteroendocrine cells. CONCLUSIONS IL22 promotes transit-amplifying cell proliferation but reduces Lgr5+ stem cell survival by inhibiting notch and wnt signaling. IL22 can therefore promote or inhibit mucosal repair, depending on whether effects on transit-amplifying or stem cells predominate. These data may explain why mucosal healing is difficult to achieve in some inflammatory bowel disease patients despite markedly elevated IL22 production.
Collapse
Affiliation(s)
- Juan-Min Zha
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China; Department of Pathology, University of Chicago, Chicago, Illinois
| | - Hua-Shan Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei-Ting Kuo
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zhi-Hui Jiang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Pei-Yun Tsai
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Ning Ding
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shao-Fang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi-Tang Wang
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Jian Pan
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, China
| | - Xiu-Min Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Atsushi Tamura
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Sachiko Tsukita
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Jerrold R Turner
- Department of Pathology, University of Chicago, Chicago, Illinois; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Wei-Qi He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genome Resource Center, Soochow University, and Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China; Department of Pathology, University of Chicago, Chicago, Illinois.
| |
Collapse
|
49
|
Yap AS, Duszyc K, Viasnoff V. Mechanosensing and Mechanotransduction at Cell-Cell Junctions. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028761. [PMID: 28778874 DOI: 10.1101/cshperspect.a028761] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cell adhesion systems are defined by their ability to resist detachment force. Our understanding of the biology of cell-cell adhesions has recently been transformed by the realization that many of the forces that act on those adhesions are generated by the cells that they couple together; and that force at adhesive junctions can be sensed to regulate cell behavior. Here, we consider the mechanisms responsible for applying force to cell-cell junctions and the mechanosensory pathways that detect those forces. We focus on cadherins, as these are the best-studied examples to date, but it is likely that similar principles will apply to other molecular systems that can engage with force-generators within cells and physically couple those cells together.
Collapse
Affiliation(s)
- Alpha S Yap
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, The University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Kinga Duszyc
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, The University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore 117411.,CNRS, Singapore 117411
| |
Collapse
|
50
|
Riddle MR, Boesmans W, Caballero O, Kazwiny Y, Tabin CJ. Morphogenesis and motility of the Astyanax mexicanus gastrointestinal tract. Dev Biol 2018; 441:285-296. [PMID: 29883660 DOI: 10.1016/j.ydbio.2018.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/03/2018] [Accepted: 06/04/2018] [Indexed: 01/01/2023]
Abstract
Through the course of evolution, the gastrointestinal (GI) tract has been modified to maximize nutrient absorption, forming specialized segments that are morphologically and functionally distinct. Here we show that the GI tract of the Mexican tetra, Astyanax mexicanus, has distinct regions, exhibiting differences in morphology, motility, and absorption. We found that A. mexicanus populations adapted for life in subterranean caves exhibit differences in the GI segments compared to those adapted to surface rivers. Cave-adapted fish exhibit bi-directional churning motility in the stomach region that is largely absent in river-adapted fish. We investigated how this motility pattern influences intestinal transit of powdered food and live prey. We found that powdered food is more readily emptied from the cavefish GI tract. In contrast, the transit of live rotifers from the stomach region to the midgut occurs more slowly in cavefish compared to surface fish, consistent with the presence of churning motility. Differences in intestinal motility and transit likely reflect adaptation to unique food sources available to post-larval A. mexicanus in the cave and river environments. We found that cavefish grow more quickly than surface fish when fed ad libitum, suggesting that altered GI function may aid in nutrient consumption or absorption. We did not observe differences in enteric neuron density or smooth muscle organization between cavefish and surface fish. Altered intestinal motility in cavefish could instead be due to changes in the activity or patterning of the enteric nervous system. Exploring this avenue will lead to a better understanding of how the GI tract evolves to maximize energy assimilation from novel food sources.
Collapse
Affiliation(s)
- Misty R Riddle
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Werend Boesmans
- Laboratory for Enteric Neuroscience, Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Olivya Caballero
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Ophthalmology, SUNY Downstate, Brooklyn, NY 11203, USA
| | - Youcef Kazwiny
- Laboratory for Enteric Neuroscience, Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Clifford J Tabin
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|