1
|
Dagher L, Descroix S, Maître JL. Intercellular fluid dynamics in tissue morphogenesis. Curr Biol 2024; 34:R1031-R1044. [PMID: 39437722 DOI: 10.1016/j.cub.2024.05.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
During embryonic development, cells shape our body, which is mostly made up of water. It is often forgotten that some of this water is found in intercellular fluid, which, for example, immerses the cells of developing embryos. Intercellular fluid contributes to the properties of tissues and influences cell behaviour, thereby participating in tissue morphogenesis. While our understanding of the role of cells in shaping tissues advances, the exploration of the contribution of intercellular fluid dynamics is just beginning. In this review, we delve into the intricate mechanisms employed by cells to control fluid movements both across and within sealed tissue compartments. These mechanisms encompass sealing by tight junctions and controlled leakage, osmotic pumping, hydraulic fracturing of cell adhesion, cell and tissue contractions, as well as beating cilia. We illustrate key concepts by drawing extensively from the early mouse embryo, which successively forms multiple lumens that play essential roles in its development. Finally, we detail experimental approaches and emerging techniques that allow for the quantitative characterization and the manipulation of intercellular fluids in vivo, as well as theoretical frameworks that are crucial for comprehending their dynamics.
Collapse
Affiliation(s)
- Louise Dagher
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University, 75005 Paris, France; Institut Curie, Laboratoire Physics of Cells and Cancer (CNRS UMR 168), Institut Pierre-Gilles de Gennes, Sorbonne Université, PSL Research University, 6 rue Jean Calvin, 75005 Paris, France
| | - Stéphanie Descroix
- Institut Curie, Laboratoire Physics of Cells and Cancer (CNRS UMR 168), Institut Pierre-Gilles de Gennes, Sorbonne Université, PSL Research University, 6 rue Jean Calvin, 75005 Paris, France
| | - Jean-Léon Maître
- Institut Curie, CNRS UMR3215, INSERM U934, PSL Research University, 75005 Paris, France.
| |
Collapse
|
2
|
Shim G, Breinyn IB, Martínez-Calvo A, Rao S, Cohen DJ. Bioelectric stimulation controls tissue shape and size. Nat Commun 2024; 15:2938. [PMID: 38580690 PMCID: PMC10997591 DOI: 10.1038/s41467-024-47079-w] [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: 01/30/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
Epithelial tissues sheath organs and electro-mechanically regulate ion and water transport to regulate development, homeostasis, and hydrostatic organ pressure. Here, we demonstrate how external electrical stimulation allows us to control these processes in living tissues. Specifically, we electrically stimulate hollow, 3D kidneyoids and gut organoids and find that physiological-strength electrical stimulation of ∼ 5 - 10 V/cm powerfully inflates hollow tissues; a process we call electro-inflation. Electro-inflation is mediated by increased ion flux through ion channels/transporters and triggers subsequent osmotic water flow into the lumen, generating hydrostatic pressure that competes against cytoskeletal tension. Our computational studies suggest that electro-inflation is strongly driven by field-induced ion crowding on the outer surface of the tissue. Electrically stimulated tissues also break symmetry in 3D resulting from electrotaxis and affecting tissue shape. The ability of electrical cues to regulate tissue size and shape emphasizes the role and importance of the electrical micro-environment for living tissues.
Collapse
Affiliation(s)
- Gawoon Shim
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, 08540, NJ, USA
| | - Isaac B Breinyn
- Department of Quantitative and Computational Biology, Princeton University, Princeton, 08540, NJ, USA
| | - Alejandro Martínez-Calvo
- Princeton Center for Theoretical Science, Princeton University, Princeton, 08540, NJ, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, 08540, NJ, USA
| | - Sameeksha Rao
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, 08540, NJ, USA
| | - Daniel J Cohen
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, 08540, NJ, USA.
| |
Collapse
|
3
|
Grandy S, Scur M, Dolan K, Nickerson R, Cheng Z. Using model systems to unravel host-Pseudomonas aeruginosa interactions. Environ Microbiol 2023; 25:1765-1784. [PMID: 37290773 DOI: 10.1111/1462-2920.16440] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/29/2023] [Indexed: 06/10/2023]
Abstract
Using model systems in infection biology has led to the discoveries of many pathogen-encoded virulence factors and critical host immune factors to fight pathogenic infections. Studies of the remarkable Pseudomonas aeruginosa bacterium that infects and causes disease in hosts as divergent as humans and plants afford unique opportunities to shed new light on virulence strategies and host defence mechanisms. One of the rationales for using model systems as a discovery tool to characterise bacterial factors driving human infection outcomes is that many P. aeruginosa virulence factors are required for pathogenesis in diverse different hosts. On the other side, many host signalling components, such as the evolutionarily conserved mitogen-activated protein kinases, are involved in immune signalling in a diverse range of hosts. Some model organisms that have less complex immune systems also allow dissection of the direct impacts of innate immunity on host defence without the interference of adaptive immunity. In this review, we start with discussing the occurrence of P. aeruginosa in the environment and the ability of this bacterium to cause disease in various hosts as a natural opportunistic pathogen. We then summarise the use of some model systems to study host defence and P. aeruginosa virulence.
Collapse
Affiliation(s)
- Shannen Grandy
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michal Scur
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kathleen Dolan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Rhea Nickerson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
4
|
Aljiboury AA, Ingram E, Krishnan N, Ononiwu F, Pal D, Manikas J, Taveras C, Hall NA, Da Silva J, Freshour J, Hehnly H. Rab8, Rab11, and Rab35 coordinate lumen and cilia formation during zebrafish left-right organizer development. PLoS Genet 2023; 19:e1010765. [PMID: 37186603 PMCID: PMC10212091 DOI: 10.1371/journal.pgen.1010765] [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: 01/10/2023] [Revised: 05/25/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
An essential process during Danio rerio's left-right organizer (Kupffer's Vesicle, KV) formation is the formation of a motile cilium by developing KV cells which extends into the KV lumen. Beating of motile cilia within the KV lumen directs fluid flow to establish the embryo's left-right axis. However, the timepoint at which KV cells start to form cilia and how cilia formation is coordinated with KV lumen formation have not been examined. We identified that nascent KV cells form cilia at their centrosomes at random intracellular positions that then move towards a forming apical membrane containing cystic fibrosis transmembrane conductance regulator (CFTR). Using optogenetic clustering approaches, we found that Rab35 positive membranes recruit Rab11 to modulate CFTR delivery to the apical membrane, which is required for lumen opening, and subsequent cilia extension into the lumen. Once the intracellular cilia reach the CFTR positive apical membrane, Arl13b-positive cilia extend and elongate in a Rab8 dependent manner into the forming lumen once the lumen reaches an area of 300 μm2. These studies demonstrate the need to acutely coordinate Rab8, Rab11, and Rab35-mediated membrane trafficking events to ensure appropriate timing in lumen and cilia formation during KV development.
Collapse
Affiliation(s)
- Abrar A. Aljiboury
- Biology Department, Syracuse University, Syracuse, New York, United States of America
- BioInspired Institute, Syracuse University, Syracuse, New York, United States of America
| | - Eric Ingram
- Biology Department, Syracuse University, Syracuse, New York, United States of America
- BioInspired Institute, Syracuse University, Syracuse, New York, United States of America
| | - Nikhila Krishnan
- Biology Department, Syracuse University, Syracuse, New York, United States of America
- BioInspired Institute, Syracuse University, Syracuse, New York, United States of America
| | - Favour Ononiwu
- Biology Department, Syracuse University, Syracuse, New York, United States of America
- BioInspired Institute, Syracuse University, Syracuse, New York, United States of America
| | - Debadrita Pal
- Biology Department, Syracuse University, Syracuse, New York, United States of America
- BioInspired Institute, Syracuse University, Syracuse, New York, United States of America
| | - Julie Manikas
- Biology Department, Syracuse University, Syracuse, New York, United States of America
| | - Christopher Taveras
- Biology Department, Syracuse University, Syracuse, New York, United States of America
| | - Nicole A. Hall
- Biology Department, Syracuse University, Syracuse, New York, United States of America
| | - Jonah Da Silva
- Biology Department, Syracuse University, Syracuse, New York, United States of America
| | - Judy Freshour
- Biology Department, Syracuse University, Syracuse, New York, United States of America
| | - Heidi Hehnly
- Biology Department, Syracuse University, Syracuse, New York, United States of America
- BioInspired Institute, Syracuse University, Syracuse, New York, United States of America
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 0] [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
|
6
|
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
|
7
|
Forrest K, Barricella AC, Pohar SA, Hinman AM, Amack JD. Understanding laterality disorders and the left-right organizer: Insights from zebrafish. Front Cell Dev Biol 2022; 10:1035513. [PMID: 36619867 PMCID: PMC9816872 DOI: 10.3389/fcell.2022.1035513] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Vital internal organs display a left-right (LR) asymmetric arrangement that is established during embryonic development. Disruption of this LR asymmetry-or laterality-can result in congenital organ malformations. Situs inversus totalis (SIT) is a complete concordant reversal of internal organs that results in a low occurrence of clinical consequences. Situs ambiguous, which gives rise to Heterotaxy syndrome (HTX), is characterized by discordant development and arrangement of organs that is associated with a wide range of birth defects. The leading cause of health problems in HTX patients is a congenital heart malformation. Mutations identified in patients with laterality disorders implicate motile cilia in establishing LR asymmetry. However, the cellular and molecular mechanisms underlying SIT and HTX are not fully understood. In several vertebrates, including mouse, frog and zebrafish, motile cilia located in a "left-right organizer" (LRO) trigger conserved signaling pathways that guide asymmetric organ development. Perturbation of LRO formation and/or function in animal models recapitulates organ malformations observed in SIT and HTX patients. This provides an opportunity to use these models to investigate the embryological origins of laterality disorders. The zebrafish embryo has emerged as an important model for investigating the earliest steps of LRO development. Here, we discuss clinical characteristics of human laterality disorders, and highlight experimental results from zebrafish that provide insights into LRO biology and advance our understanding of human laterality disorders.
Collapse
Affiliation(s)
- Kadeen Forrest
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Alexandria C. Barricella
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Sonny A. Pohar
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Anna Maria Hinman
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY, United States
| |
Collapse
|
8
|
Xia Y, Duca S, Perder B, Dündar F, Zumbo P, Qiu M, Yao J, Cao Y, Harrison MRM, Zangi L, Betel D, Cao J. Activation of a transient progenitor state in the epicardium is required for zebrafish heart regeneration. Nat Commun 2022; 13:7704. [PMID: 36513650 PMCID: PMC9747719 DOI: 10.1038/s41467-022-35433-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
The epicardium, a mesothelial cell tissue that encompasses vertebrate hearts, supports heart regeneration after injury through paracrine effects and as a source of multipotent progenitors. However, the progenitor state in the adult epicardium has yet to be defined. Through single-cell RNA-sequencing of isolated epicardial cells from uninjured and regenerating adult zebrafish hearts, we define the epithelial and mesenchymal subsets of the epicardium. We further identify a transiently activated epicardial progenitor cell (aEPC) subpopulation marked by ptx3a and col12a1b expression. Upon cardiac injury, aEPCs emerge from the epithelial epicardium, migrate to enclose the wound, undergo epithelial-mesenchymal transition (EMT), and differentiate into mural cells and pdgfra+hapln1a+ mesenchymal epicardial cells. These EMT and differentiation processes are regulated by the Tgfβ pathway. Conditional ablation of aEPCs blocks heart regeneration through reduced nrg1 expression and mesenchymal cell number. Our findings identify a transient progenitor population of the adult epicardium that is indispensable for heart regeneration and highlight it as a potential target for enhancing cardiac repair.
Collapse
Affiliation(s)
- Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Sierra Duca
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Björn Perder
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Friederike Dündar
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Applied Bioinformatics Core, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Applied Bioinformatics Core, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Jun Yao
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Yingxi Cao
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Michael R M Harrison
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Lior Zangi
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Institute for Computational Biomedicine, Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
9
|
Carleton AE, Duncan MC, Taniguchi K. Human epiblast lumenogenesis: From a cell aggregate to a lumenal cyst. Semin Cell Dev Biol 2022; 131:117-123. [PMID: 35637065 PMCID: PMC9529837 DOI: 10.1016/j.semcdb.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The formation of a central lumen in the human epiblast is a critical step for development. However, because the lumen forms in the epiblast coincident with implantation, the molecular and cellular events of this early lumenogenesis process cannot be studied in vivo. Recent developments using new model systems have revealed insight into the underpinnings of epiblast formation. To provide an up-to-date comprehensive review of human epiblast lumenogenesis, we highlight recent findings from human and mouse models with an emphasis on new molecular understanding of a newly described apicosome compartment, a novel 'formative' state of pluripotency that coordinates with epiblast polarization, and new evidence about the physical and polarized trafficking mechanisms contributing to lumenogenesis.
Collapse
Affiliation(s)
- Amber E. Carleton
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin USA
| | - Mara C. Duncan
- Departments of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan USA,Co-corresponding authors
| | - Kenichiro Taniguchi
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin USA,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin USA,Co-corresponding authors
| |
Collapse
|
10
|
Chugh M, Munjal A, Megason SG. Hydrostatic pressure as a driver of cell and tissue morphogenesis. Semin Cell Dev Biol 2022; 131:134-145. [PMID: 35534334 PMCID: PMC9529827 DOI: 10.1016/j.semcdb.2022.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 12/14/2022]
Abstract
Morphogenesis, the process by which tissues develop into functional shapes, requires coordinated mechanical forces. Most current literature ascribes contractile forces derived from actomyosin networks as the major driver of tissue morphogenesis. Recent works from diverse species have shown that pressure derived from fluids can generate deformations necessary for tissue morphogenesis. In this review, we discuss how hydrostatic pressure is generated at the cellular and tissue level and how the pressure can cause deformations. We highlight and review findings demonstrating the mechanical roles of pressures from fluid-filled lumens and viscous gel-like components of the extracellular matrix. We also emphasise the interactions and mechanochemical feedbacks between extracellular pressures and tissue behaviour in driving tissue remodelling. Lastly, we offer perspectives on the open questions in the field that will further our understanding to uncover new principles of tissue organisation during development.
Collapse
Affiliation(s)
- Mayank Chugh
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| | - Akankshi Munjal
- Department of Cell Biology, Duke University School of Medicine, Nanaline Duke Building, 307 Research Drive, Durham, NC 27710, USA.
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Lobert VH, Skardal ML, Malerød L, Simensen JE, Algra HA, Andersen AN, Fleischer T, Enserink HA, Liestøl K, Heath JK, Rusten TE, Stenmark HA. PHLPP1 regulates CFTR activity and lumen expansion through AMPK. Development 2022; 149:276412. [PMID: 35997536 PMCID: PMC9534488 DOI: 10.1242/dev.200955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/12/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Complex organ development depends on single lumen formation and its expansion during tubulogenesis. This can be achieved by correct mitotic spindle orientation during cell division, combined with luminal fluid filling that generates hydrostatic pressure. Using a human 3D cell culture model, we have identified two regulators of these processes. We find that pleckstrin homology leucine-rich repeat protein phosphatase (PHLPP) 2 regulates mitotic spindle orientation, and thereby midbody positioning and maintenance of a single lumen. Silencing the sole PHLPP family phosphatase in Drosophila melanogaster, phlpp, resulted in defective spindle orientation in Drosophila neuroblasts. Importantly, cystic fibrosis transmembrane conductance regulator (CFTR) is the main channel regulating fluid transport in this system, stimulated by phosphorylation by protein kinase A and inhibited by the AMP-activated protein kinase AMPK. During lumen expansion, CFTR remains open through the action of PHLPP1, which stops activated AMPK from inhibiting ion transport through CFTR. In the absence of PHLPP1, the restraint on AMPK activity is lost and this tips the balance in the favour of channel closing, resulting in the lack of lumen expansion and accumulation of mucus.
Collapse
Affiliation(s)
- Viola H. Lobert
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Maren L. Skardal
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Lene Malerød
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Julia E. Simensen
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Hermine A. Algra
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Aram N. Andersen
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Thomas Fleischer
- Institute for Cancer Research, Oslo University Hospital 3 Department of Cancer Genetics , , Montebello, Oslo 0379 , Norway
| | - Hilde A. Enserink
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Knut Liestøl
- University of Oslo 4 Department of Informatics , , Oslo 0316 , Norway
| | - Joan K. Heath
- Walter and Eliza Hall Institute of Medical Research 5 Epigenetics and Development Division , , Parkville, Victoria 3052 , Australia
| | - Tor Erik Rusten
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| | - Harald A. Stenmark
- Institute for Cancer Research, Oslo University Hospital 1 Department of Molecular Cell Biology , , Montebello, Oslo 0379 , Norway
- Centre for Cancer Cell Reprogramming 2 , Faculty of Medicine , , Oslo 0379 , Norway
- University of Oslo 2 , Faculty of Medicine , , Oslo 0379 , Norway
| |
Collapse
|
12
|
Bagnat M, Daga B, Di Talia S. Morphogenetic Roles of Hydrostatic Pressure in Animal Development. Annu Rev Cell Dev Biol 2022; 38:375-394. [PMID: 35804476 PMCID: PMC9675319 DOI: 10.1146/annurev-cellbio-120320-033250] [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: 11/09/2022]
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
|
13
|
Krishnan N, Swoger M, Rathbun LI, Fioramonti PJ, Freshour J, Bates M, Patteson AE, Hehnly H. Rab11 endosomes and Pericentrin coordinate centrosome movement during pre-abscission in vivo. Life Sci Alliance 2022; 5:e202201362. [PMID: 35304423 PMCID: PMC8933627 DOI: 10.26508/lsa.202201362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/24/2022] Open
Abstract
The last stage of cell division involves two daughter cells remaining interconnected by a cytokinetic bridge that is cleaved during abscission. Conserved between the zebrafish embryo and human cells, we found that the oldest centrosome moves in a Rab11-dependent manner towards the cytokinetic bridge sometimes followed by the youngest. Rab11-endosomes are organized in a Rab11-GTP dependent manner at the mother centriole during pre-abscission, with Rab11 endosomes at the oldest centrosome being more mobile compared with the youngest. The GTPase activity of Rab11 is necessary for the centrosome protein, Pericentrin, to be enriched at the centrosome. Reduction in Pericentrin expression or optogenetic disruption of Rab11-endosome function inhibited both centrosome movement towards the cytokinetic bridge and abscission, resulting in daughter cells prone to being binucleated and/or having supernumerary centrosomes. These studies suggest that Rab11-endosomes contribute to centrosome function during pre-abscission by regulating Pericentrin organization resulting in appropriate centrosome movement towards the cytokinetic bridge and subsequent abscission.
Collapse
Affiliation(s)
- Nikhila Krishnan
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Maxx Swoger
- Department of Physics, Syracuse University, Physics Building, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Lindsay I Rathbun
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Peter J Fioramonti
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Judy Freshour
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Michael Bates
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Alison E Patteson
- Department of Physics, Syracuse University, Physics Building, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Heidi Hehnly
- Department of Biology, Syracuse University, Syracuse, NY, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, USA
| |
Collapse
|
14
|
Shah VS, Chivukula RR, Lin B, Waghray A, Rajagopal J. Cystic Fibrosis and the Cells of the Airway Epithelium: What Are Ionocytes and What Do They Do? ANNUAL REVIEW OF PATHOLOGY 2022; 17:23-46. [PMID: 34437820 PMCID: PMC10837786 DOI: 10.1146/annurev-pathol-042420-094031] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cystic fibrosis (CF) is caused by defects in an anion channel, the cystic fibrosis transmembrane conductance regulator (CFTR). Recently, a new airway epithelial cell type has been discovered and dubbed the pulmonary ionocyte. Unexpectedly, these ionocytes express higher levels of CFTR than any other airway epithelial cell type. However, ionocytes are not the sole CFTR-expressing airway epithelial cells, and CF-associated disease genes are in fact expressed in multiple airway epithelial cell types. The experimental depletion of ionocytes perturbs epithelial physiology in the mouse trachea, but the role of these rare cells in the pathogenesis of human CF remains mysterious. Ionocytes have been described in diverse tissues(kidney and inner ear) and species (frog and fish). We draw on these prior studies to suggest potential roles of airway ionocytes in health and disease. A complete understanding of ionocytes in the mammalian airway will ultimately depend on cell type-specific genetic manipulation.
Collapse
Affiliation(s)
- Viral S Shah
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Raghu R Chivukula
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | - Brian Lin
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| | - Avinash Waghray
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| | - Jayaraj Rajagopal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Klarman Cell Observatory, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
15
|
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: 1.7] [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
|
16
|
Knockout of mafba Causes Inner-Ear Developmental Defects in Zebrafish via the Impairment of Proliferation and Differentiation of Ionocyte Progenitor Cells. Biomedicines 2021; 9:biomedicines9111699. [PMID: 34829928 PMCID: PMC8616026 DOI: 10.3390/biomedicines9111699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/18/2021] [Accepted: 10/28/2021] [Indexed: 12/03/2022] Open
Abstract
Zebrafish is an excellent model for exploring the development of the inner ear. Its inner ear has similar functions to that of humans, specifically in the maintenance of hearing and balance. Mafba is a component of the Maf transcription factor family. It participates in multiple biological processes, but its role in inner-ear development remains poorly understood. In this study, we constructed a mafba knockout (mafba−/−) zebrafish model using CRISPR/Cas9 technology. The mafba−/− mutant inner ear displayed severe impairments, such as enlarged otocysts, smaller or absent otoliths, and insensitivity to sound stimulation. The proliferation of p63+ epidermal stem cells and dlc+ ionocyte progenitors was inhibited in mafba−/− mutants. Moreover, the results showed that mafba deletion induces the apoptosis of differentiated K+-ATPase-rich (NR) cells and H+-ATPase-rich (HR) cells. The activation of p53 apoptosis and G0/G1 cell cycle arrest resulted from DNA damage in the inner-ear region, providing a mechanism to account for the inner ear deficiencies. The loss of homeostasis resulting from disorders of ionocyte progenitors resulted in structural defects in the inner ear and, consequently, loss of hearing. In conclusion, the present study elucidated the function of ionic channel homeostasis and inner-ear development using a zebrafish Mafba model and clarified the possible physiological roles.
Collapse
|
17
|
Pont S, Blanc-Potard AB. Zebrafish Embryo Infection Model to Investigate Pseudomonas aeruginosa Interaction With Innate Immunity and Validate New Therapeutics. Front Cell Infect Microbiol 2021; 11:745851. [PMID: 34660345 PMCID: PMC8515127 DOI: 10.3389/fcimb.2021.745851] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/08/2021] [Indexed: 12/26/2022] Open
Abstract
The opportunistic human pathogen Pseudomonas aeruginosa is responsible for a variety of acute infections and is a major cause of mortality in chronically infected patients with cystic fibrosis (CF). Considering the intrinsic and acquired resistance of P. aeruginosa to currently used antibiotics, new therapeutic strategies against this pathogen are urgently needed. Whereas virulence factors of P. aeruginosa are well characterized, the interplay between P. aeruginosa and the innate immune response during infection remains unclear. Zebrafish embryo is now firmly established as a potent vertebrate model for the study of infectious human diseases, due to strong similarities of its innate immune system with that of humans and the unprecedented possibilities of non-invasive real-time imaging. This model has been successfully developed to investigate the contribution of bacterial and host factors involved in P. aeruginosa pathogenesis, as well as rapidly assess the efficacy of anti-Pseudomonas molecules. Importantly, zebrafish embryo appears as the state-of-the-art model to address in vivo the contribution of innate immunity in the outcome of P. aeruginosa infection. Of interest, is the finding that the zebrafish encodes a CFTR channel closely related to human CFTR, which allowed to develop a model to address P. aeruginosa pathogenesis, innate immune response, and treatment evaluation in a CF context.
Collapse
Affiliation(s)
- Stéphane Pont
- Laboratory of Pathogen-Host Interactions (LPHI), Université Montpellier, Montpellier, France.,CNRS, UMR5235, Montpellier, France
| | - Anne-Béatrice Blanc-Potard
- Laboratory of Pathogen-Host Interactions (LPHI), Université Montpellier, Montpellier, France.,CNRS, UMR5235, Montpellier, France
| |
Collapse
|
18
|
Levic DS, Yamaguchi N, Wang S, Knaut H, Bagnat M. Knock-in tagging in zebrafish facilitated by insertion into non-coding regions. Development 2021; 148:dev199994. [PMID: 34495314 PMCID: PMC8513609 DOI: 10.1242/dev.199994] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022]
Abstract
Zebrafish provide an excellent model for in vivo cell biology studies because of their amenability to live imaging. Protein visualization in zebrafish has traditionally relied on overexpression of fluorescently tagged proteins from heterologous promoters, making it difficult to recapitulate endogenous expression patterns and protein function. One way to circumvent this problem is to tag the proteins by modifying their endogenous genomic loci. Such an approach is not widely available to zebrafish researchers because of inefficient homologous recombination and the error-prone nature of targeted integration in zebrafish. Here, we report a simple approach for tagging proteins in zebrafish on their N or C termini with fluorescent proteins by inserting PCR-generated donor amplicons into non-coding regions of the corresponding genes. Using this approach, we generated endogenously tagged alleles for several genes that are crucial for epithelial biology and organ development, including the tight junction components ZO-1 and Cldn15la, the trafficking effector Rab11a, the apical polarity protein aPKC and the ECM receptor Integrin β1b. Our approach facilitates the generation of knock-in lines in zebrafish, opening the way for accurate quantitative imaging studies.
Collapse
Affiliation(s)
- Daniel S. Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Naoya Yamaguchi
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, Department of Cell Biology, New York, NY 10016, USA
| | - Siyao Wang
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, Department of Cell Biology, New York, NY 10016, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
19
|
Oliveira I, Jacinto R, Pestana S, Nolasco F, Calado J, Lopes SS, Roxo-Rosa M. Zebrafish Model as a Screen to Prevent Cyst Inflation in Autosomal Dominant Polycystic Kidney Disease. Int J Mol Sci 2021; 22:ijms22169013. [PMID: 34445719 PMCID: PMC8396643 DOI: 10.3390/ijms22169013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022] Open
Abstract
In autosomal dominant polycystic kidney disease (ADPKD), kidney cyst growth requires the recruitment of CFTR (cystic fibrosis transmembrane conductance regulator), the chloride channel that is defective in cystic fibrosis. We have been studying cyst inflation using the zebrafish Kupffer’s vesicle (KV) as model system because we previously demonstrated that knocking down polycystin 2 (PC2) induced a CFTR-mediated enlargement of the organ. We have now quantified the PC2 knockdown by showing that it causes a 73% reduction in the number of KV cilia expressing PC2. According to the literature, this is an essential event in kidney cystogenesis in ADPKD mice. Additionally, we demonstrated that the PC2 knockdown leads to a significant accumulation of CFTR-GFP at the apical region of the KV cells. Furthermore, we determined that KV enlargement is rescued by the injection of Xenopus pkd2 mRNA and by 100 µM tolvaptan treatment, the unique and approved pharmacologic approach for ADPKD management. We expected vasopressin V2 receptor antagonist to lower the cAMP levels of KV-lining cells and, thus, to inactivate CFTR. These findings further support the use of the KV as an in vivo model for screening compounds that may prevent cyst enlargement in this ciliopathy, through CFTR inhibition.
Collapse
Affiliation(s)
- Inês Oliveira
- CEDOC, Chronic Diseases Research Center, NOVA Medical School|Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (I.O.); (R.J.); (S.P.)
| | - Raquel Jacinto
- CEDOC, Chronic Diseases Research Center, NOVA Medical School|Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (I.O.); (R.J.); (S.P.)
| | - Sara Pestana
- CEDOC, Chronic Diseases Research Center, NOVA Medical School|Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (I.O.); (R.J.); (S.P.)
| | - Fernando Nolasco
- Department of Nephrology, Centro Hospitalar e Universitário de Lisboa Central, Hospital de Curry Cabral, Rua da Beneficência, 8, 1069-166 Lisboa, Portugal; (F.N.); (J.C.)
| | - Joaquim Calado
- Department of Nephrology, Centro Hospitalar e Universitário de Lisboa Central, Hospital de Curry Cabral, Rua da Beneficência, 8, 1069-166 Lisboa, Portugal; (F.N.); (J.C.)
- ToxOmics, Center of ToxicoGenomics & Human Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
| | - Susana Santos Lopes
- CEDOC, Chronic Diseases Research Center, NOVA Medical School|Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (I.O.); (R.J.); (S.P.)
- Correspondence: (S.S.L.); (M.R.-R.)
| | - Mónica Roxo-Rosa
- CEDOC, Chronic Diseases Research Center, NOVA Medical School|Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (I.O.); (R.J.); (S.P.)
- Correspondence: (S.S.L.); (M.R.-R.)
| |
Collapse
|
20
|
Lee M, Betz C, Yin J, Paatero I, Schellinx N, Carte AN, Wilson CW, Ye W, Affolter M, Belting HG. Control of dynamic cell behaviors during angiogenesis and anastomosis by Rasip1. Development 2021; 148:271819. [PMID: 34383884 PMCID: PMC8380458 DOI: 10.1242/dev.197509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/08/2021] [Indexed: 11/23/2022]
Abstract
Organ morphogenesis is driven by a wealth of tightly orchestrated cellular behaviors, which ensure proper organ assembly and function. Many of these cell activities involve cell-cell interactions and remodeling of the F-actin cytoskeleton. Here, we analyze the requirement for Rasip1 (Ras-interacting protein 1), an endothelial-specific regulator of junctional dynamics, during blood vessel formation. Phenotype analysis of rasip1 mutants in zebrafish embryos reveals distinct functions of Rasip1 during sprouting angiogenesis, anastomosis and lumen formation. During angiogenic sprouting, loss of Rasip1 causes cell pairing defects due to a destabilization of tricellular junctions, indicating that stable tricellular junctions are essential to maintain multicellular organization within the sprout. During anastomosis, Rasip1 is required to establish a stable apical membrane compartment; rasip1 mutants display ectopic, reticulated junctions and the apical compartment is frequently collapsed. Loss of Ccm1 and Heg1 function mimics the junctional defects of rasip1 mutants. Furthermore, downregulation of ccm1 and heg1 leads to a delocalization of Rasip1 at cell junctions, indicating that junctional tethering of Rasip1 is required for its function in junction formation and stabilization during sprouting angiogenesis. Summary:In vivo analysis of rasip1 mutants reveals multiple roles for Rasip1 during angiogenic sprouting, anastomosis and lumen formation, including stabilization of tricellular junctions to permit coordinated cell rearrangements and multicellular tube formation.
Collapse
Affiliation(s)
- Minkyoung Lee
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Charles Betz
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Jianmin Yin
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Ilkka Paatero
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Niels Schellinx
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Adam N Carte
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Christopher W Wilson
- Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Weilan Ye
- Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Markus Affolter
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Heinz-Georg Belting
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| |
Collapse
|
21
|
Valenzuela L, Pacheco S, Rincón G, Pavez L, Lam N, Hernández AJ, Dantagnan P, González F, Jilberto F, Ravanal MC, Ramos C, Garcia H, Araneda C, Ulloa PE. Intestinal Transcriptome Analysis Reveals Enrichment of Genes Associated with Immune and Lipid Mechanisms, Favoring Soybean Meal Tolerance in High-Growth Zebrafish ( Danio Rerio). Genes (Basel) 2021; 12:genes12050700. [PMID: 34066767 PMCID: PMC8151431 DOI: 10.3390/genes12050700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 01/10/2023] Open
Abstract
The molecular mechanisms underlying fish tolerance to soybean meal (SBM) remain unclear. Identifying these mechanisms would be beneficial, as this trait favors growth. Two fish replicates from 19 experimental families were fed fishmeal-(100FM) or SBM-based diets supplemented with saponin (50SBM + 2SPN) from juvenile to adult stages. Individuals were selected from families with a genotype-by-environment interaction higher (HG-50SBM + 2SPN, 170 ± 18 mg) or lower (LG-50SBM + 2SPN, 76 ± 10 mg) weight gain on 50SBM + 2SPN for intestinal transcriptomic analysis. A histological evaluation confirmed middle intestinal inflammation in the LG- vs. HG-50SBM + 2SPN group. Enrichment analysis of 665 differentially expressed genes (DEGs) identified pathways associated with immunity and lipid metabolism. Genes linked to intestinal immunity were downregulated in HG fish (mpx, cxcr3.2, cftr, irg1l, itln2, sgk1, nup61l, il22), likely dampening inflammatory responses. Conversely, genes involved in retinol signaling were upregulated (rbp4, stra6, nr2f5), potentially favoring growth by suppressing insulin responses. Genes associated with lipid metabolism were upregulated, including key components of the SREBP (mbtps1, elov5l, elov6l) and cholesterol catabolism (cyp46a1), as well as the downregulation of cyp7a1. These results strongly suggest that transcriptomic changes in lipid metabolism mediate SBM tolerance. Genotypic variations in DEGs may become biomarkers for improving early selection of fish tolerant to SMB or others plant-based diets.
Collapse
Affiliation(s)
- Luis Valenzuela
- Omics Lab, Villavicencio 378, Oficina 32, Santiago 8320164, Chile;
| | - Sebastian Pacheco
- Programa de Doctorado en Inmunología y Microbiología, Universidad San Sebastian, Lota 2465, Santiago 7510157, Chile;
| | - Gonzalo Rincón
- Zoetis, VMRD Genetics R&D, 333 Portage Street, Kalamazoo, MI 49007, USA;
| | - Leonardo Pavez
- Núcleo de Investigaciones Aplicadas en Ciencias Veterinarias y Agronómicas, Universidad de Las Américas, Avenida Manuel Montt 948, Santiago 7500975, Chile; (L.P.); (F.G.); (C.R.)
| | - Natalia Lam
- Departamento de Producción Animal, Facultad de Ciencias Agronómicas, Universidad de Chile, Santa Rosa 11315, Santiago 8820808, Chile; (N.L.); (F.J.); (C.A.)
| | - Adrián J. Hernández
- Laboratorio de Nutrición y Fisiología de Peces, Núcleo de Investigación en Producción Alimentaria, Departamento de Ciencias Agropecuarias y Acuícolas, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco 4780000, Chile; (A.J.H.); (P.D.)
| | - Patricio Dantagnan
- Laboratorio de Nutrición y Fisiología de Peces, Núcleo de Investigación en Producción Alimentaria, Departamento de Ciencias Agropecuarias y Acuícolas, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco 4780000, Chile; (A.J.H.); (P.D.)
| | - Felipe González
- Núcleo de Investigaciones Aplicadas en Ciencias Veterinarias y Agronómicas, Universidad de Las Américas, Avenida Manuel Montt 948, Santiago 7500975, Chile; (L.P.); (F.G.); (C.R.)
| | - Felipe Jilberto
- Departamento de Producción Animal, Facultad de Ciencias Agronómicas, Universidad de Chile, Santa Rosa 11315, Santiago 8820808, Chile; (N.L.); (F.J.); (C.A.)
| | - M. Cristina Ravanal
- Instituto de Ciencia y Tecnología de los Alimentos (ICYTAL), Facultad de Ciencias Agrarias y Alimentarias, Universidad Austral de Chile, Avda. Julio Sarrazín s/n, Isla Teja, Valdivia 5090000, Chile;
| | - Cecilia Ramos
- Núcleo de Investigaciones Aplicadas en Ciencias Veterinarias y Agronómicas, Universidad de Las Américas, Avenida Manuel Montt 948, Santiago 7500975, Chile; (L.P.); (F.G.); (C.R.)
| | - Héctor Garcia
- Laboratorios Diagnofruit Ltd.a., Depto. Fitopatología Molecular, Santiago 7770273, Chile;
| | - Cristian Araneda
- Departamento de Producción Animal, Facultad de Ciencias Agronómicas, Universidad de Chile, Santa Rosa 11315, Santiago 8820808, Chile; (N.L.); (F.J.); (C.A.)
| | - Pilar E. Ulloa
- Núcleo de Investigaciones Aplicadas en Ciencias Veterinarias y Agronómicas, Universidad de Las Américas, Avenida Manuel Montt 948, Santiago 7500975, Chile; (L.P.); (F.G.); (C.R.)
- Departamento de Producción Animal, Facultad de Ciencias Agronómicas, Universidad de Chile, Santa Rosa 11315, Santiago 8820808, Chile; (N.L.); (F.J.); (C.A.)
- Correspondence: ; Tel.: +56-222-531-129
| |
Collapse
|
22
|
Gibson-Corley KN, Engelhardt JF. Animal Models and Their Role in Understanding the Pathophysiology of Cystic Fibrosis-Associated Gastrointestinal Lesions. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:51-67. [PMID: 33497264 DOI: 10.1146/annurev-pathol-022420-105133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The life expectancy of cystic fibrosis (CF) patients has greatly increased over the past decade, and researchers and clinicians must now navigate complex disease manifestations that were not a concern prior to the development of modern therapies. Explosive growth in the number of CF animal models has also occurred over this time span, clarifying CF disease pathophysiology and creating opportunities to understand more complex disease processes associated with an aging CF population. This review focuses on the CF-associated pathologies of the gastrointestinal system and how animal models have increased our understanding of this complex multisystemic disease. Although CF is primarily recognized as a pulmonary disease, gastrointestinal pathology occurs very commonly and can affect the quality of life for these patients. Furthermore, we discuss how next-generation genetic engineering of larger animal models will impact the field's understanding of CF disease pathophysiology and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Katherine N Gibson-Corley
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA.,Current affiliation: Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee 37232, USA;
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA;
| |
Collapse
|
23
|
Duong Phu M, Bross S, Burkhalter MD, Philipp M. Limitations and opportunities in the pharmacotherapy of ciliopathies. Pharmacol Ther 2021; 225:107841. [PMID: 33771583 DOI: 10.1016/j.pharmthera.2021.107841] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023]
Abstract
Ciliopathies are a family of rather diverse conditions, which have been grouped based on the finding of altered or dysfunctional cilia, potentially motile, small cellular antennae extending from the surface of postmitotic cells. Cilia-related disorders include embryonically arising conditions such as Joubert, Usher or Kartagener syndrome, but also afflictions with a postnatal or even adult onset phenotype, i.e. autosomal dominant polycystic kidney disease. The majority of ciliopathies are syndromic rather than affecting only a single organ due to cilia being found on almost any cell in the human body. Overall ciliopathies are considered rare diseases. Despite that, pharmacological research and the strive to help these patients has led to enormous therapeutic advances in the last decade. In this review we discuss new treatment options for certain ciliopathies, give an outlook on promising future therapeutic strategies, but also highlight the limitations in the development of therapeutic approaches of ciliopathies.
Collapse
Affiliation(s)
- Max Duong Phu
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Stefan Bross
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Martin D Burkhalter
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany
| | - Melanie Philipp
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University of Tübingen, 72074 Tübingen, Germany.
| |
Collapse
|
24
|
Collinet C, Lecuit T. Programmed and self-organized flow of information during morphogenesis. Nat Rev Mol Cell Biol 2021; 22:245-265. [PMID: 33483696 DOI: 10.1038/s41580-020-00318-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2020] [Indexed: 11/09/2022]
Abstract
How the shape of embryos and organs emerges during development is a fundamental question that has fascinated scientists for centuries. Tissue dynamics arise from a small set of cell behaviours, including shape changes, cell contact remodelling, cell migration, cell division and cell extrusion. These behaviours require control over cell mechanics, namely active stresses associated with protrusive, contractile and adhesive forces, and hydrostatic pressure, as well as material properties of cells that dictate how cells respond to active stresses. In this Review, we address how cell mechanics and the associated cell behaviours are robustly organized in space and time during tissue morphogenesis. We first outline how not only gene expression and the resulting biochemical cues, but also mechanics and geometry act as sources of morphogenetic information to ultimately define the time and length scales of the cell behaviours driving morphogenesis. Next, we present two idealized modes of how this information flows - how it is read out and translated into a biological effect - during morphogenesis. The first, akin to a programme, follows deterministic rules and is hierarchical. The second follows the principles of self-organization, which rests on statistical rules characterizing the system's composition and configuration, local interactions and feedback. We discuss the contribution of these two modes to the mechanisms of four very general classes of tissue deformation, namely tissue folding and invagination, tissue flow and extension, tissue hollowing and, finally, tissue branching. Overall, we suggest a conceptual framework for understanding morphogenetic information that encapsulates genetics and biochemistry as well as mechanics and geometry as information modules, and the interplay of deterministic and self-organized mechanisms of their deployment, thereby diverging considerably from the traditional notion that shape is fully encoded and determined by genes.
Collapse
Affiliation(s)
- Claudio Collinet
- Aix-Marseille Université & CNRS, IBDM - UMR7288 & Turing Centre for Living Systems, Campus de Luminy Case 907, Marseille, France
| | - Thomas Lecuit
- Aix-Marseille Université & CNRS, IBDM - UMR7288 & Turing Centre for Living Systems, Campus de Luminy Case 907, Marseille, France. .,Collège de France, Paris, France.
| |
Collapse
|
25
|
McCarron A, Parsons D, Donnelley M. Animal and Cell Culture Models for Cystic Fibrosis: Which Model Is Right for Your Application? THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:228-242. [PMID: 33232694 DOI: 10.1016/j.ajpath.2020.10.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/01/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Over the past 30 years, a range of cystic fibrosis (CF) animal models have been generated for research purposes. Different species, including mice, rats, ferrets, rabbits, pigs, sheep, zebrafish, and fruit flies, have all been used to model CF disease. While access to such a variety of animal models is a luxury for any research field, it also complicates the decision-making process when it comes to selecting the right model for an investigation. The purpose of this review is to provide a guide for selecting the most appropriate CF animal model for any given application. In this review, the characteristics and phenotypes of each animal model are described, along with a discussion of the key considerations that must be taken into account when choosing a suitable animal model. Available in vitro systems of CF are also described and can offer a useful alternative to using animal models. Finally, the future of CF animal model generation and its use in research are speculated upon.
Collapse
Affiliation(s)
- Alexandra McCarron
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia.
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| |
Collapse
|
26
|
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: 7.3] [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
|
27
|
Bernut A, Loynes CA, Floto RA, Renshaw SA. Deletion of cftr Leads to an Excessive Neutrophilic Response and Defective Tissue Repair in a Zebrafish Model of Sterile Inflammation. Front Immunol 2020; 11:1733. [PMID: 32849617 PMCID: PMC7412881 DOI: 10.3389/fimmu.2020.01733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/29/2020] [Indexed: 11/13/2022] Open
Abstract
Inflammation-related progressive lung destruction is the leading causes of premature death in cystic fibrosis (CF), a genetic disorder caused by a defective cystic fibrosis transmembrane conductance regulator (CFTR). However, therapeutic targeting of inflammation has been hampered by a lack of understanding of the links between a dysfunctional CFTR and the deleterious innate immune response in CF. Herein, we used a CFTR-depleted zebrafish larva, as an innovative in vivo vertebrate model, to understand how CFTR dysfunction leads to abnormal inflammatory status in CF. We show that impaired CFTR-mediated inflammation correlates with an exuberant neutrophilic response after injury: CF zebrafish exhibit enhanced and sustained accumulation of neutrophils at wounds. Excessive epithelial oxidative responses drive enhanced neutrophil recruitment towards wounds. Persistence of neutrophils at inflamed sites is associated with impaired reverse migration of neutrophils and reduction in neutrophil apoptosis. As a consequence, the increased number of neutrophils at wound sites causes tissue damage and abnormal tissue repair. Importantly, the molecule Tanshinone IIA successfully accelerates inflammation resolution and improves tissue repair in CF animal. Our findings bring important new understanding of the mechanisms underlying the inflammatory pathology in CF, which could be addressed therapeutically to prevent inflammatory lung damage in CF patients with potential improvements in disease outcomes.
Collapse
Affiliation(s)
- Audrey Bernut
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
| | - Catherine A. Loynes
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
| | - R. Andres Floto
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical, Cambridge, United Kingdom
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
| |
Collapse
|
28
|
Cell-Based Therapeutic Approaches for Cystic Fibrosis. Int J Mol Sci 2020; 21:ijms21155219. [PMID: 32718005 PMCID: PMC7432606 DOI: 10.3390/ijms21155219] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 01/01/2023] Open
Abstract
Cystic Fibrosis (CF) is a chronic autosomal recessive disease caused by defects in the cystic fibrosis transmembrane conductance regulator gene (CFTR). Cystic Fibrosis affects multiple organs but progressive remodeling of the airways, mucus accumulation, and chronic inflammation in the lung, result in lung disease as the major cause of morbidity and mortality. While advances in management of CF symptoms have increased the life expectancy of this devastating disease, and there is tremendous excitement about the potential of new agents targeting the CFTR molecule itself, there is still no curative treatment. With the recent advances in the identification of endogenous airway progenitor cells and in directed differentiation of pluripotent cell sources, cell-based therapeutic approaches for CF have become a plausible treatment method with the potential to ultimately cure the disease. In this review, we highlight the current state of cell therapy in the CF field focusing on the relevant autologous and allogeneic cell populations under investigation and the challenges associated with their use. In addition, we present advances in induced pluripotent stem (iPS) cell approaches and emerging new genetic engineering methods, which have the capacity to overcome the current limitations hindering cell therapy approaches.
Collapse
|
29
|
CFTR deficiency causes cardiac dysplasia during zebrafish embryogenesis and is associated with dilated cardiomyopathy. Mech Dev 2020; 163:103627. [PMID: 32574800 DOI: 10.1016/j.mod.2020.103627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
Mutations in the CFTR gene cause cystic fibrosis (CF) with myocardial dysfunction. However, it remains unknown whether CF-related heart disease is a secondary effect of pulmonary disease, or an intrinsic primary defect in the heart. Here, we used zebrafish, which lack lung tissue, to investigate the role of CFTR in cardiogenesis. Our findings demonstrated that the loss of CFTR impairs cardiac development from the cardiac progenitor stage, resulting in cardiac looping defects, a dilated atrium, pericardial edema, and a decrease in heart rate. Furthermore, we found that cardiac development was perturbed in wild-type embryos treated with a gating-specific CFTR channel inhibitor, CFTRinh-172, at the blastula stage of development, but not at later stages. Gene expression analysis of blastulas indicated that transcript levels, including mRNAs associated with cardiovascular diseases, were significantly altered in embryos derived from cftr mutants relative to controls. To evaluate the role of CFTR in human heart failure, we performed a genetic association study on individuals with dilated cardiomyopathy and found that the I556V mutation in CFTR, which causes a channel defect, was associated with the disease. Similar to other well-studied channel-defective CFTR mutants, CFTR I556V mRNA failed to restore cardiac dysplasia in mutant embryos. The present study revealed an important role for the CFTR ion channel in regulating cardiac development during early embryogenesis, supporting the hypothesis that CF-related heart disease results from an intrinsic primary defect in the heart.
Collapse
|
30
|
Tang Y, Yan Z, Engelhardt JF. Viral Vectors, Animal Models, and Cellular Targets for Gene Therapy of Cystic Fibrosis Lung Disease. Hum Gene Ther 2020; 31:524-537. [PMID: 32138545 PMCID: PMC7232698 DOI: 10.1089/hum.2020.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
After more than two decades since clinical trials tested the first use of recombinant adeno-associated virus (rAAV) to treat cystic fibrosis (CF) lung disease, gene therapy for this disorder has undergone a tremendous resurgence. Fueling this enthusiasm has been an enhanced understanding of rAAV transduction biology and cellular processes that limit transduction of airway epithelia, the development of new rAAV serotypes and other vector systems with high-level tropism for airway epithelial cells, an improved understanding of CF lung pathogenesis and the cellular targets for gene therapy, and the development of new animal models that reproduce the human CF disease phenotype. These advances have created a preclinical path for both assessing the efficacy of gene therapies in the CF lung and interrogating the target cell types in the lung required for complementation of the CF disease state. Lessons learned from early gene therapy attempts with rAAV in the CF lung have guided thinking for the testing of next-generation vector systems. Although unknown questions still remain regarding the cellular targets in the lung that are required or sufficient to complement CF lung disease, the field is now well positioned to tackle these challenges. This review will highlight the role that next-generation CF animal models are playing in the preclinical development of gene therapies for CF lung disease and the knowledge gaps in disease pathophysiology that these models are attempting to fill.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
31
|
What Role Does CFTR Play in Development, Differentiation, Regeneration and Cancer? Int J Mol Sci 2020; 21:ijms21093133. [PMID: 32365523 PMCID: PMC7246864 DOI: 10.3390/ijms21093133] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023] Open
Abstract
One of the key features associated with the substantial increase in life expectancy for individuals with CF is an elevated predisposition to cancer, firmly established by recent studies involving large cohorts. With the recent advances in cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapies and the increased long-term survival rate of individuals with cystic fibrosis (CF), this is a novel challenge emerging at the forefront of this disease. However, the mechanisms linking dysfunctional CFTR to carcinogenesis have yet to be unravelled. Clues to this challenging open question emerge from key findings in an increasing number of studies showing that CFTR plays a role in fundamental cellular processes such as foetal development, epithelial differentiation/polarization, and regeneration, as well as in epithelial–mesenchymal transition (EMT). Here, we provide state-of-the-art descriptions on the moonlight roles of CFTR in these processes, highlighting how they can contribute to novel therapeutic strategies. However, such roles are still largely unknown, so we need rapid progress in the elucidation of the underlying mechanisms to find the answers and thus tailor the most appropriate therapeutic approaches.
Collapse
|
32
|
Drosophila as a model for studying cystic fibrosis pathophysiology of the gastrointestinal system. Proc Natl Acad Sci U S A 2020; 117:10357-10367. [PMID: 32345720 DOI: 10.1073/pnas.1913127117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF) is a recessive disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The most common symptoms include progressive lung disease and chronic digestive conditions. CF is the first human genetic disease to benefit from having five different species of animal models. Despite the phenotypic differences among the animal models and human CF, these models have provided invaluable insight into understanding disease mechanisms at the organ-system level. Here, we identify a member of the ABCC4 family, CG5789, that has the structural and functional properties expected for encoding the Drosophila equivalent of human CFTR, and thus refer to it as Drosophila CFTR (Dmel\CFTR). We show that knockdown of Dmel\CFTR in the adult intestine disrupts osmotic homeostasis and displays CF-like phenotypes that lead to intestinal stem cell hyperplasia. We also show that expression of wild-type human CFTR, but not mutant variants of CFTR that prevent plasma membrane expression, rescues the mutant phenotypes of Dmel\CFTR Furthermore, we performed RNA sequencing (RNA-Seq)-based transcriptomic analysis using Dmel\CFTR fly intestine and identified a mucin gene, Muc68D, which is required for proper intestinal barrier protection. Altogether, our findings suggest that Drosophila can be a powerful model organism for studying CF pathophysiology.
Collapse
|
33
|
Bernut A, Dupont C, Ogryzko NV, Neyret A, Herrmann JL, Floto RA, Renshaw SA, Kremer L. CFTR Protects against Mycobacterium abscessus Infection by Fine-Tuning Host Oxidative Defenses. Cell Rep 2020; 26:1828-1840.e4. [PMID: 30759393 DOI: 10.1016/j.celrep.2019.01.071] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/17/2018] [Accepted: 01/17/2019] [Indexed: 01/03/2023] Open
Abstract
Infection by rapidly growing Mycobacterium abscessus is increasingly prevalent in cystic fibrosis (CF), a genetic disease caused by a defective CF transmembrane conductance regulator (CFTR). However, the potential link between a dysfunctional CFTR and vulnerability to M. abscessus infection remains unknown. Herein, we exploit a CFTR-depleted zebrafish model, recapitulating CF immuno-pathogenesis, to study the contribution of CFTR in innate immunity against M. abscessus infection. Loss of CFTR increases susceptibility to infection through impaired NADPH oxidase-dependent restriction of intracellular growth and reduced neutrophil chemotaxis, which together compromise granuloma formation and integrity. As a consequence, extracellular multiplication of M. abscessus expands rapidly, inducing abscess formation and causing lethal infections. Because these phenotypes are not observed with other mycobacteria, our findings highlight the crucial and specific role of CFTR in the immune control of M. abscessus by mounting effective oxidative responses.
Collapse
Affiliation(s)
- Audrey Bernut
- CNRS, UMR9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France; Bateson Centre, University of Sheffield, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK.
| | - Christian Dupont
- CNRS, UMR9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Nikolay V Ogryzko
- Bateson Centre, University of Sheffield, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
| | - Aymeric Neyret
- CNRS, UMR9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | | | - R Andres Floto
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Stephen A Renshaw
- Bateson Centre, University of Sheffield, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
| | - Laurent Kremer
- CNRS, UMR9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France; INSERM, IRIM, Montpellier, France.
| |
Collapse
|
34
|
Rathbun LI, Colicino EG, Manikas J, O'Connell J, Krishnan N, Reilly NS, Coyne S, Erdemci-Tandogan G, Garrastegui A, Freshour J, Santra P, Manning ML, Amack JD, Hehnly H. Cytokinetic bridge triggers de novo lumen formation in vivo. Nat Commun 2020; 11:1269. [PMID: 32152267 PMCID: PMC7062744 DOI: 10.1038/s41467-020-15002-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 02/14/2020] [Indexed: 02/03/2023] Open
Abstract
Multicellular rosettes are transient epithelial structures that serve as intermediates during diverse organ formation. We have identified a unique contributor to rosette formation in zebrafish Kupffer's vesicle (KV) that requires cell division, specifically the final stage of mitosis termed abscission. KV utilizes a rosette as a prerequisite before forming a lumen surrounded by ciliated epithelial cells. Our studies identify that KV-destined cells remain interconnected by cytokinetic bridges that position at the rosette's center. These bridges act as a landmark for directed Rab11 vesicle motility to deliver an essential cargo for lumen formation, CFTR (cystic fibrosis transmembrane conductance regulator). Here we report that premature bridge cleavage through laser ablation or inhibiting abscission using optogenetic clustering of Rab11 result in disrupted lumen formation. We present a model in which KV mitotic cells strategically place their cytokinetic bridges at the rosette center, where Rab11-associated vesicles transport CFTR to aid in lumen establishment.
Collapse
Affiliation(s)
- L I Rathbun
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - E G Colicino
- Biology Department, Syracuse University, Syracuse, New York, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - J Manikas
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - J O'Connell
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - N Krishnan
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - N S Reilly
- Department of Physics and Astronomy, University of Rochester, Rochester, New York, USA
| | - S Coyne
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
- Department of Biology, SUNY Geneseo, Geneseo, New York, USA
| | | | - A Garrastegui
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - J Freshour
- Biology Department, Syracuse University, Syracuse, New York, USA
| | - P Santra
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
| | - M L Manning
- Department of Physics, Syracuse University, Syracuse, New York, USA
| | - J D Amack
- Department of Cell and Developmental Biology, SUNY Upstate Medical School, Syracuse, New York, USA
| | - H Hehnly
- Biology Department, Syracuse University, Syracuse, New York, USA.
| |
Collapse
|
35
|
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: 5.5] [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
|
36
|
Ye L, Mueller O, Bagwell J, Bagnat M, Liddle RA, Rawls JF. High fat diet induces microbiota-dependent silencing of enteroendocrine cells. eLife 2019; 8:48479. [PMID: 31793875 PMCID: PMC6937151 DOI: 10.7554/elife.48479] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Enteroendocrine cells (EECs) are specialized sensory cells in the intestinal epithelium that sense and transduce nutrient information. Consumption of dietary fat contributes to metabolic disorders, but EEC adaptations to high fat feeding were unknown. Here, we established a new experimental system to directly investigate EEC activity in vivo using a zebrafish reporter of EEC calcium signaling. Our results reveal that high fat feeding alters EEC morphology and converts them into a nutrient insensitive state that is coupled to endoplasmic reticulum (ER) stress. We called this novel adaptation 'EEC silencing'. Gnotobiotic studies revealed that germ-free zebrafish are resistant to high fat diet induced EEC silencing. High fat feeding altered gut microbiota composition including enrichment of Acinetobacter bacteria, and we identified an Acinetobacter strain sufficient to induce EEC silencing. These results establish a new mechanism by which dietary fat and gut microbiota modulate EEC nutrient sensing and signaling.
Collapse
Affiliation(s)
- Lihua Ye
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States.,Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Olaf Mueller
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States
| | - Jennifer Bagwell
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Michel Bagnat
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Rodger A Liddle
- Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States.,Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, United States
| |
Collapse
|
37
|
Laselva O, Erwood S, Du K, Ivakine Z, Bear CE. Activity of lumacaftor is not conserved in zebrafish Cftr bearing the major cystic fibrosis-causing mutation. FASEB Bioadv 2019; 1:661-670. [PMID: 32123813 PMCID: PMC6996396 DOI: 10.1096/fba.2019-00039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/02/2019] [Accepted: 08/30/2019] [Indexed: 12/11/2022] Open
Abstract
F508del-cystic fibrosis transmembrane conductance regulator (CFTR) is the major mutant responsible for cystic fibrosis (CF). ORKAMBI®, approved for patients bearing this mutant, contains lumacaftor (VX-809) that partially corrects F508del-CFTR's processing defect and ivacaftor (VX-770) that potentiates its defective channel activity. Unfortunately, the clinical efficacy of ORKAMBI® is modest, highlighting the need to understand how the small molecules work so that superior compounds can be developed. Because, human CFTR (hCFTR) and zebrafish Cftr (zCftr) are structurally conserved as determined in recent cryo-EM structural models, we hypothesized that the consequences of the major mutation and small molecule modulators would be similar for the two species of protein. As expected, like the F508del mutation in hCFTR, the homologous mutation in zCftr (F507del) is misprocessed, yet not as severely as the human mutant and this defect was restored by low-temperature (27°C) culture conditions. After rescue to the cell surface, F507del-zCftr exhibited regulated channel activity that was potentiated by ivacaftor. Surprisingly, lumacaftor failed to rescue misprocessing of the F507del-zCftr at either 37 or 27°C suggesting that future comparative studies with F508del-hCFTR would provide insight into its structure: function relationships. Interestingly, the robust rescue of F508del-zCftr at 27°C and availability of methods for in vivo screening in zebrafish present the opportunity to define the cellular pathways underlying rescue.
Collapse
Affiliation(s)
- Onofrio Laselva
- Programme in Molecular MedicineHospital for Sick ChildrenTorontoCanada
- Department of PhysiologyUniversity of TorontoTorontoCanada
| | - Steven Erwood
- Programme in Genetics and Genome BiologyHospital for Sick ChildrenTorontoCanada
| | - Kai Du
- Programme in Molecular MedicineHospital for Sick ChildrenTorontoCanada
| | - Zhenya Ivakine
- Programme in Genetics and Genome BiologyHospital for Sick ChildrenTorontoCanada
| | - Christine E. Bear
- Programme in Molecular MedicineHospital for Sick ChildrenTorontoCanada
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Department of BiochemistryUniversity of TorontoTorontoCanada
| |
Collapse
|
38
|
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: 8.0] [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
|
39
|
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: 2.0] [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]
|
40
|
Fillatre J, Fauny JD, Fels JA, Li C, Goll M, Thisse C, Thisse B. TEADs, Yap, Taz, Vgll4s transcription factors control the establishment of Left-Right asymmetry in zebrafish. eLife 2019; 8:45241. [PMID: 31513014 PMCID: PMC6759317 DOI: 10.7554/elife.45241] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 09/11/2019] [Indexed: 12/15/2022] Open
Abstract
In many vertebrates, establishment of Left-Right (LR) asymmetry results from the activity of a ciliated organ functioning as the LR Organizer (LRO). While regulation of the formation of this structure by major signaling pathways has been described, the transcriptional control of LRO formation is poorly understood. Using the zebrafish model, we show that the transcription factors and cofactors mediating or regulating the transcriptional outcome of the Hippo signaling pathway play a pivotal role in controlling the expression of genes essential to the formation of the LRO including ligands and receptors of signaling pathways involved in this process and most genes required for motile ciliogenesis. Moreover, the transcription cofactor, Vgll4l regulates epigenetic programming in LRO progenitors by controlling the expression of writers and readers of DNA methylation marks. Altogether, our study uncovers a novel and essential role for the transcriptional effectors and regulators of the Hippo pathway in establishing LR asymmetry.
Collapse
Affiliation(s)
- Jonathan Fillatre
- Department of Cell Biology, University of Virginia, Charlottesville, United States
| | - Jean-Daniel Fauny
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, Illkirch-Graffenstaden, France.,Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | | | - Cheng Li
- Department of Genetics, University of Georgia, Athens, United States
| | - Mary Goll
- Department of Genetics, University of Georgia, Athens, United States
| | - Christine Thisse
- Department of Cell Biology, University of Virginia, Charlottesville, United States.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Bernard Thisse
- Department of Cell Biology, University of Virginia, Charlottesville, United States.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, Illkirch-Graffenstaden, France
| |
Collapse
|
41
|
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: 60] [Impact Index Per Article: 12.0] [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
|
42
|
Cafora M, Deflorian G, Forti F, Ferrari L, Binelli G, Briani F, Ghisotti D, Pistocchi A. Phage therapy against Pseudomonas aeruginosa infections in a cystic fibrosis zebrafish model. Sci Rep 2019; 9:1527. [PMID: 30728389 PMCID: PMC6365511 DOI: 10.1038/s41598-018-37636-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Cystic fibrosis (CF) is a hereditary disease due to mutations in the CFTR gene and causes mortality in humans mainly due to respiratory infections caused by Pseudomonas aeruginosa. In a previous work we used phage therapy, which is a treatment with a mix of phages, to actively counteract acute P. aeruginosa infections in mice and Galleria mellonella larvae. In this work we apply phage therapy to the treatment of P. aeruginosa PAO1 infections in a CF zebrafish model. The structure of the CFTR channel is evolutionary conserved between fish and mammals and cftr-loss-of-function zebrafish embryos show a phenotype that recapitulates the human disease, in particular with destruction of the pancreas. We show that phage therapy is able to decrease lethality, bacterial burden, and the pro-inflammatory response caused by PAO1 infection. In addition, phage administration relieves the constitutive inflammatory state of CF embryos. To our knowledge, this is the first time that phage therapy is used to cure P. aeruginosa infections in a CF animal model. We also find that the curative effect against PAO1 infections is improved by combining phages and antibiotic treatments, opening a useful therapeutic approach that could reduce antibiotic doses and time of administration.
Collapse
Affiliation(s)
- Marco Cafora
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano - LITA, via Fratelli Cervi 93, 20090, Segrate, MI, Italy
| | - Gianluca Deflorian
- Istituto FIRC di Oncologia Molecolare - IFOM, Via Adamello 16, 20139, Milano, Italy
| | - Francesca Forti
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Laura Ferrari
- Istituto FIRC di Oncologia Molecolare - IFOM, Via Adamello 16, 20139, Milano, Italy
| | - Giorgio Binelli
- Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, Via J.H. Dunant 3, Varese, Italy
| | - Federica Briani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Daniela Ghisotti
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Anna Pistocchi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano - LITA, via Fratelli Cervi 93, 20090, Segrate, MI, Italy.
| |
Collapse
|
43
|
Functional characterization reveals that zebrafish CFTR prefers to occupy closed channel conformations. PLoS One 2018; 13:e0209862. [PMID: 30596737 PMCID: PMC6312236 DOI: 10.1371/journal.pone.0209862] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR), the culprit behind the genetic disease cystic fibrosis (CF), is a phosphorylation-activated, but ATP-gated anion channel. Studies of human CFTR over the past two decades have provided an in-depth understanding of how CFTR works as an ion channel despite its structural resemblance to ABC transporters. Recently-solved cryo-EM structures of unphosphorylated human and zebrafish CFTR (hCFTR and zCFTR), as well as phosphorylated ATP-bound zebrafish and human CFTR offer an unprecedented opportunity to understand CFTR's function at a molecular level. Interestingly, despite millions of years of phylogenetic distance between human and zebrafish, the structures of zCFTR and hCFTR exhibit remarkable similarities. In the current study, we characterized biophysical and pharmacological properties of zCFTR with the patch-clamp technique, and showed surprisingly very different functional properties between these two orthologs. First, while hCFTR has a single-channel conductance of 8.4 pS with a linear I-V curve, zCFTR shows an inwardly-rectified I-V relationship with a single-channel conductance of ~3.5 pS. Second, single-channel gating behaviors of phosphorylated zCFTR are very different from those of hCFTR, featuring a very low open probability Po (0.03 ± 0.02, vs. ~0.50 for hCFTR) with exceedingly long closed events and brief openings. In addition, unlike hCFTR where each open burst is clearly defined with rare short-lived flickery closures, the open bursts of zCFTR are not easily resolved. Third, although abolishing ATP hydrolysis by replacing the catalytic glutamate with glutamine (i.e., E1372Q) drastically prolongs the open bursts defined by the macroscopic relaxation analysis in zCFTR, the Po within a "locked-open" burst of E1372Q-zCFTR is only ~ 0.35 (vs. Po > 0.94 in E1371Q-hCFTR). Collectively, our data not only provide a reasonable explanation for the unexpected closed-state structure of phosphorylated E1372Q-zCFTR with a canonical ATP-bound dimer of the nucleotide binding domains (NBDs), but also implicate significant structural and functional differences between these two evolutionarily distant orthologs.
Collapse
|
44
|
Liao H, Chen Y, Li Y, Xue S, Liu M, Lin Z, Liu Y, Chan HC, Zhang X, Sun H. CFTR is required for the migration of primordial germ cells during zebrafish early embryogenesis. Reproduction 2018; 156:261-268. [PMID: 29930176 PMCID: PMC6106808 DOI: 10.1530/rep-17-0681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 06/21/2018] [Indexed: 12/12/2022]
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene affect fertility in both sexes. However, the involvement of CFTR in regulating germ cell development remains largely unknown. Here, we used zebrafish model to investigate the role of CFTR in primordial germ cells (PGCs) development. We generated a cftr frameshift mutant zebrafish line using CRISPR/Cas9 technique and investigated the migration of PGCs during early embryo development. Our results showed that loss of Cftr impairs the migration of PGCs from dome stages onward. The migration of PGCs was also perturbed by treatment of CFTRinh-172, a gating-specific CFTR channel inhibitor. Moreover, defected PGCs migration in cftr mutant embryos can be partially rescued by injection of WT but not other channel-defective mutant cftr mRNAs. Finally, we observed the elevation of cxcr4b, cxcl12a, rgs14a and ca15b, key factors involved in zebrafish PGCs migration, in cftr-mutant zebrafish embryos. Taken together, the present study revealed an important role of CFTR acting as an ion channel in regulating PGCs migration during early embryogenesis. Defect of which may impair germ cell development through elevation of key factors involved in cell motility and response to chemotactic gradient in PGCs.
Collapse
Affiliation(s)
- Huijuan Liao
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yan Chen
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yulong Li
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- The School of Life ScienceShandong University, Jinan, Shandong, People’s Republic of China
| | - Shaolong Xue
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Epithelial Cell Biology Research CenterSchool of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Mingfeng Liu
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of EducationCollege of Life Science, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Ziyuan Lin
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yanyan Liu
- Prenatal Diagnosis CenterDepartment of Obstetrics & Gynecologic, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Hsiao Chang Chan
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Epithelial Cell Biology Research CenterSchool of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Xiaohu Zhang
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Huaqin Sun
- SCU-CUHK Joint Laboratory for Reproductive MedicineKey Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
45
|
Zhou X, Xiao C, Li Y, Shang Y, Yin D, Li S, Xiang B, Lu R, Ji Y, Wu Y, Meng W, Zhu H, Liu J, Hu H, Mo X, Xu H. Mid1ip1b modulates apical reorientation of non-centrosomal microtubule organizing center in epithelial cells. J Genet Genomics 2018; 45:433-442. [PMID: 30174135 DOI: 10.1016/j.jgg.2018.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 08/03/2018] [Accepted: 08/05/2018] [Indexed: 02/05/2023]
Abstract
In most kinds of animal cells, the centrosome serves as the main microtubule organizing center (MTOC) that nucleates microtubule arrays throughout the cytoplasm to maintain cell structure, cell division and intracellular transport. Whereas in epithelial cells, non-centrosomal MTOCs are established in the apical domain for generating asymmetric microtubule fibers and cilia in epithelial cells for the organ morphogenesis during embryonic development. However, the mechanism by which MTOCs localize to the apical domain in epithelial cells remains largely unknown. Here, we show that Mid1ip1b has a close interaction with γ-tubulin protein, the central component of MTOC, and modulates lumen opening of the neural tube, gut, intestine, and kidney of zebrafish. Knockdown or dominant negative effect of Mid1ip1b resulted in failure of lumen formation of the organs as aforementioned. Moreover, the non-centrosomal MTOCs were unable to orientate to the apical domain in Mid1ip1b knockdown epithelial cells, and the centrosomal MTOCs were inaccurately placed in the apical domain, resulting in defective formation of asymmetric microtubules and misplacement of cilia in the apical domain. These data uncover a molecule that controls the proper localization of MTOCs in the apical domain in epithelial cells for organ morphogenesis during embryonic development.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Chun Xiao
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Li
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yanna Shang
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Dongqin Yin
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Siying Li
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bo Xiang
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ran Lu
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yi Ji
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yang Wu
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Wentong Meng
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Hongyan Zhu
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jin Liu
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Huozhen Hu
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xianming Mo
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Hong Xu
- Department of Pediatric Surgery and Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
46
|
Saydmohammed M, Yagi H, Calderon M, Clark MJ, Feinstein T, Sun M, Stolz DB, Watkins SC, Amack JD, Lo CW, Tsang M. Vertebrate myosin 1d regulates left-right organizer morphogenesis and laterality. Nat Commun 2018; 9:3381. [PMID: 30139971 PMCID: PMC6107537 DOI: 10.1038/s41467-018-05866-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 07/28/2018] [Indexed: 11/25/2022] Open
Abstract
Establishing left-right asymmetry is a fundamental process essential for arrangement of visceral organs during development. In vertebrates, motile cilia-driven fluid flow in the left-right organizer (LRO) is essential for initiating symmetry breaking event. Here, we report that myosin 1d (myo1d) is essential for establishing left-right asymmetry in zebrafish. Using super-resolution microscopy, we show that the zebrafish LRO, Kupffer's vesicle (KV), fails to form a spherical lumen and establish proper unidirectional flow in the absence of myo1d. This process requires directed vacuolar trafficking in KV epithelial cells. Interestingly, the vacuole transporting function of zebrafish Myo1d can be substituted by myosin1C derived from an ancient eukaryote, Acanthamoeba castellanii, where it regulates the transport of contractile vacuoles. Our findings reveal an evolutionary conserved role for an unconventional myosin in vacuole trafficking, lumen formation, and determining laterality.
Collapse
Affiliation(s)
- Manush Saydmohammed
- Department of Developmental Biology, University of Pittsburgh, 3501 5th Avenue, Pittsburgh, PA, 5213, USA.
| | - Hisato Yagi
- Department of Developmental Biology, University of Pittsburgh, 3501 5th Avenue, Pittsburgh, PA, 5213, USA
| | - Michael Calderon
- Department of Cell Biology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Madeline J Clark
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Timothy Feinstein
- Department of Developmental Biology, University of Pittsburgh, 3501 5th Avenue, Pittsburgh, PA, 5213, USA
| | - Ming Sun
- Department of Cell Biology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Jeffrey D Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh, 3501 5th Avenue, Pittsburgh, PA, 5213, USA
| | - Michael Tsang
- Department of Developmental Biology, University of Pittsburgh, 3501 5th Avenue, Pittsburgh, PA, 5213, USA.
| |
Collapse
|
47
|
Juan T, Géminard C, Coutelis JB, Cerezo D, Polès S, Noselli S, Fürthauer M. Myosin1D is an evolutionarily conserved regulator of animal left-right asymmetry. Nat Commun 2018; 9:1942. [PMID: 29769531 PMCID: PMC5955935 DOI: 10.1038/s41467-018-04284-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/13/2018] [Indexed: 12/30/2022] Open
Abstract
The establishment of left-right (LR) asymmetry is fundamental to animal development, but the identification of a unifying mechanism establishing laterality across different phyla has remained elusive. A cilia-driven, directional fluid flow is important for symmetry breaking in numerous vertebrates, including zebrafish. Alternatively, LR asymmetry can be established independently of cilia, notably through the intrinsic chirality of the acto-myosin cytoskeleton. Here, we show that Myosin1D (Myo1D), a previously identified regulator of Drosophila LR asymmetry, is essential for the formation and function of the zebrafish LR organizer (LRO), Kupffer's vesicle (KV). Myo1D controls the orientation of LRO cilia and interacts functionally with the planar cell polarity (PCP) pathway component VanGogh-like2 (Vangl2), to shape a productive LRO flow. Our findings identify Myo1D as an evolutionarily conserved regulator of animal LR asymmetry, and show that functional interactions between Myo1D and PCP are central to the establishment of animal LR asymmetry.
Collapse
Affiliation(s)
- Thomas Juan
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, F-06108, France
| | - Charles Géminard
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, F-06108, France
| | - Jean-Baptiste Coutelis
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, F-06108, France
| | - Delphine Cerezo
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, F-06108, France
| | - Sophie Polès
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, F-06108, France
| | - Stéphane Noselli
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, F-06108, France.
| | - Maximilian Fürthauer
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, F-06108, France.
| |
Collapse
|
48
|
CFTR mutation enhances Dishevelled degradation and results in impairment of Wnt-dependent hematopoiesis. Cell Death Dis 2018; 9:275. [PMID: 29449653 PMCID: PMC5833403 DOI: 10.1038/s41419-018-0311-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 01/16/2023]
Abstract
Mutations of cystic fibrosis transmembrane conductance regulator (CFTR) cause cystic fibrosis (CF) with a multitude of clinical manifestations. Some CF patients develop clinically significant anemia, suggesting that CFTR may regulate hematopoiesis. Here, we report that cftr mutant zebrafish model exhibits primitive and definitive hematopoietic defects with impaired Wnt signaling. Cftr is found to interact, via its PDZ-binding domain (PDZBD), with Dishevelled (Dvl), a key component of Wnt signaling required for hematopoietic progenitor specification, thus protecting Dvl from Dapper1 (Dpr1)-induced lysosomal degradation. Defective hematopoiesis and impaired Wnt signaling in cftr mutant can be rescued by overexpression of wild-type or channel function-defective G551D mutant CFTR with an intact PDZBD, but not Cftr with mutations in the PDZBD. Analysis of human database (http://r2.amc.nl) shows that CFTR is positively correlated with DVL2 and Wnt-related hematopoietic factors in human blood system. The results reveal a previously unrecognized role of CFTR, which is independent of its channel function, in regulating DVL degradation and thus Wnt signaling required for hematopoiesis in both zebrafish and humans, providing an explanation for the anemic phenotype of CF patients.
Collapse
|
49
|
Dasgupta A, Merkel M, Clark MJ, Jacob AE, Dawson JE, Manning ML, Amack JD. Cell volume changes contribute to epithelial morphogenesis in zebrafish Kupffer's vesicle. eLife 2018; 7:30963. [PMID: 29376824 PMCID: PMC5800858 DOI: 10.7554/elife.30963] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 01/26/2018] [Indexed: 02/07/2023] Open
Abstract
How epithelial cell behaviors are coordinately regulated to sculpt tissue architecture is a fundamental question in biology. Kupffer’s vesicle (KV), a transient organ with a fluid-filled lumen, provides a simple system to investigate the interplay between intrinsic cellular mechanisms and external forces during epithelial morphogenesis. Using 3-dimensional (3D) analyses of single cells we identify asymmetric cell volume changes along the anteroposterior axis of KV that coincide with asymmetric cell shape changes. Blocking ion flux prevents these cell volume changes and cell shape changes. Vertex simulations suggest cell shape changes do not depend on lumen expansion. Consistent with this prediction, asymmetric changes in KV cell volume and shape occur normally when KV lumen growth fails due to leaky cell adhesions. These results indicate ion flux mediates cell volume changes that contribute to asymmetric cell shape changes in KV, and that these changes in epithelial morphology are separable from lumen-generated forces.
Collapse
Affiliation(s)
- Agnik Dasgupta
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, United States
| | - Matthias Merkel
- Department of Physics, Syracuse University, Syracuse, United States
| | - Madeline J Clark
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, United States
| | - Andrew E Jacob
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, United States
| | | | - M Lisa Manning
- Department of Physics, Syracuse University, Syracuse, United States
| | - Jeffrey D Amack
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, United States
| |
Collapse
|
50
|
Rosa JB, Metzstein MM, Ghabrial AS. An Ichor-dependent apical extracellular matrix regulates seamless tube shape and integrity. PLoS Genet 2018; 14:e1007146. [PMID: 29309404 PMCID: PMC5774827 DOI: 10.1371/journal.pgen.1007146] [Citation(s) in RCA: 20] [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: 10/13/2017] [Revised: 01/19/2018] [Accepted: 12/09/2017] [Indexed: 01/25/2023] Open
Abstract
During sprouting angiogenesis in the vertebrate vascular system, and primary branching in the Drosophila tracheal system, specialized tip cells direct branch outgrowth and network formation. When tip cells lumenize, they form subcellular (seamless) tubes. How these seamless tubes are made, shaped and maintained remains poorly understood. Here we characterize a Drosophila mutant called ichor (ich), and show that ich is essential for the integrity and shape of seamless tubes in tracheal terminal cells. We find that Ich regulates seamless tubulogenesis via its role in promoting the formation of a mature apical extracellular matrix (aECM) lining the lumen of the seamless tubes. We determined that ich encodes a zinc finger protein (CG11966) that acts, as a transcriptional activator required for the expression of multiple aECM factors, including a novel membrane-anchored trypsin protease (CG8213). Thus, the integrity and shape of seamless tubes are regulated by the aECM that lines their lumens.
Collapse
Affiliation(s)
- Jeffrey B. Rosa
- Department of Cell & Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Mark M. Metzstein
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America
| | - Amin S. Ghabrial
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|