1
|
Ruan ZR, Yu Z, Xing C, Chen EH. Inter-organ steroid hormone signaling promotes myoblast fusion via direct transcriptional regulation of a single key effector gene. Curr Biol 2024; 34:1438-1452.e6. [PMID: 38513654 PMCID: PMC11003854 DOI: 10.1016/j.cub.2024.02.056] [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: 07/09/2023] [Revised: 12/24/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Steroid hormones regulate tissue development and physiology by modulating the transcription of a broad spectrum of genes. In insects, the principal steroid hormones, ecdysteroids, trigger the expression of thousands of genes through a cascade of transcription factors (TFs) to coordinate developmental transitions such as larval molting and metamorphosis. However, whether ecdysteroid signaling can bypass transcriptional hierarchies to exert its function in individual developmental processes is unclear. Here, we report that a single non-TF effector gene mediates the transcriptional output of ecdysteroid signaling in Drosophila myoblast fusion, a critical step in muscle development and differentiation. Specifically, we show that the 20-hydroxyecdysone (commonly referred to as "ecdysone") secreted from an extraembryonic tissue, amnioserosa, acts on embryonic muscle cells to directly activate the expression of antisocial (ants), which encodes an essential scaffold protein enriched at the fusogenic synapse. Not only is ants transcription directly regulated by the heterodimeric ecdysone receptor complex composed of ecdysone receptor (EcR) and ultraspiracle (USP) via ecdysone-response elements but also more strikingly, expression of ants alone is sufficient to rescue the myoblast fusion defect in ecdysone signaling-deficient mutants. We further show that EcR/USP and a muscle-specific TF Twist synergistically activate ants expression in vitro and in vivo. Taken together, our study provides the first example of a steroid hormone directly activating the expression of a single key non-TF effector gene to regulate a developmental process via inter-organ signaling and provides a new paradigm for understanding steroid hormone signaling in other developmental and physiological processes.
Collapse
Affiliation(s)
- Zhi-Rong Ruan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ze Yu
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabeth H Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
2
|
Milosavljevic J, Lempicki C, Lang K, Heinkele H, Kampf LL, Leroy C, Chen M, Gerstner L, Spitz D, Wang M, Knob AU, Kayser S, Helmstädter M, Walz G, Pollak MR, Hermle T. Nephrotic Syndrome Gene TBC1D8B Is Required for Endosomal Maturation and Nephrin Endocytosis in Drosophila. J Am Soc Nephrol 2022; 33:2174-2193. [PMID: 36137753 PMCID: PMC9731638 DOI: 10.1681/asn.2022030275] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 09/01/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Variants in TBC1D8B cause nephrotic syndrome. TBC1D8B is a GTPase-activating protein for Rab11 (RAB11-GAP) that interacts with nephrin, but how it controls nephrin trafficking or other podocyte functions remains unclear. METHODS We generated a stable deletion in Tbc1d8b and used microhomology-mediated end-joining for genome editing. Ex vivo functional assays utilized slit diaphragms in podocyte-like Drosophila nephrocytes. Manipulation of endocytic regulators and transgenesis of murine Tbc1d8b provided a comprehensive functional analysis of Tbc1d8b. RESULTS A null allele of Drosophila TBC1D8B exhibited a nephrocyte-restricted phenotype of nephrin mislocalization, similar to patients with isolated nephrotic syndrome who have variants in the gene. The protein was required for rapid nephrin turnover in nephrocytes and for endocytosis of nephrin induced by excessive Rab5 activity. The protein expressed from the Tbc1d8b locus bearing the edited tag predominantly localized to mature early and late endosomes. Tbc1d8b was required for endocytic cargo processing and degradation. Silencing Hrs, a regulator of endosomal maturation, phenocopied loss of Tbc1d8b. Low-level expression of murine TBC1D8B rescued loss of the Drosophila gene, indicating evolutionary conservation. Excessive murine TBC1D8B selectively disturbed nephrin dynamics. Finally, we discovered four novel TBC1D8B variants within a cohort of 363 patients with FSGS and validated a functional effect of two variants in Drosophila, suggesting a personalized platform for TBC1D8B-associated FSGS. CONCLUSIONS Variants in TBC1D8B are not infrequent among patients with FSGS. TBC1D8B, functioning in endosomal maturation and degradation, is essential for nephrin trafficking.
Collapse
Affiliation(s)
- Julian Milosavljevic
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Camille Lempicki
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Konrad Lang
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Helena Heinkele
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Lina L. Kampf
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Claire Leroy
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Mengmeng Chen
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Lea Gerstner
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Dominik Spitz
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Minxian Wang
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Andrea U. Knob
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Séverine Kayser
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Helmstädter
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- CIBSS–Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Martin R. Pollak
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Tobias Hermle
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
3
|
Rujano MA, Briand D, Ðelić B, Marc J, Spéder P. An interplay between cellular growth and atypical fusion defines morphogenesis of a modular glial niche in Drosophila. Nat Commun 2022; 13:4999. [PMID: 36008397 PMCID: PMC9411534 DOI: 10.1038/s41467-022-32685-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Neural stem cells (NSCs) live in an intricate cellular microenvironment supporting their activity, the niche. Whilst shape and function are inseparable, the morphogenetic aspects of niche development are poorly understood. Here, we use the formation of a glial niche to investigate acquisition of architectural complexity. Cortex glia (CG) in Drosophila regulate neurogenesis and build a reticular structure around NSCs. We first show that individual CG cells grow tremendously to ensheath several NSC lineages, employing elaborate proliferative mechanisms which convert these cells into syncytia rich in cytoplasmic bridges. CG syncytia further undergo homotypic cell-cell fusion, using defined cell surface receptors and actin regulators. Cellular exchange is however dynamic in space and time. This atypical cell fusion remodels cellular borders, restructuring the CG syncytia. Ultimately, combined growth and fusion builds the multi-level architecture of the niche, and creates a modular, spatial partition of the NSC population. Our findings provide insights into how a niche forms and organises while developing intimate contacts with a stem cell population.
Collapse
Affiliation(s)
| | | | - Bojana Ðelić
- Institut Pasteur, CNRS UMR3738, Paris, France
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Cell Division and Neurogenesis, Ecole Normale Supérieure, CNRS, Inserm, PSL Université Paris, Paris, France
| | - Julie Marc
- Institut Pasteur, CNRS UMR3738, Paris, France
| | | |
Collapse
|
4
|
Lang K, Milosavljevic J, Heinkele H, Chen M, Gerstner L, Spitz D, Kayser S, Helmstädter M, Walz G, Köttgen M, Spracklen A, Poulton J, Hermle T. Selective endocytosis controls slit diaphragm maintenance and dynamics in Drosophila nephrocytes. eLife 2022; 11:79037. [PMID: 35876643 PMCID: PMC9355562 DOI: 10.7554/elife.79037] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/24/2022] [Indexed: 11/28/2022] Open
Abstract
The kidneys generate about 180 l of primary urine per day by filtration of plasma. An essential part of the filtration barrier is the slit diaphragm, a multiprotein complex containing nephrin as major component. Filter dysfunction typically manifests with proteinuria and mutations in endocytosis regulating genes were discovered as causes of proteinuria. However, it is unclear how endocytosis regulates the slit diaphragm and how the filtration barrier is maintained without either protein leakage or filter clogging. Here, we study nephrin dynamics in podocyte-like nephrocytes of Drosophila and show that selective endocytosis either by dynamin- or flotillin-mediated pathways regulates a stable yet highly dynamic architecture. Short-term manipulation of endocytic functions indicates that dynamin-mediated endocytosis of ectopic nephrin restricts slit diaphragm formation spatially while flotillin-mediated turnover of nephrin within the slit diaphragm is needed to maintain filter permeability by shedding of molecules bound to nephrin in endosomes. Since slit diaphragms cannot be studied in vitro and are poorly accessible in mouse models, this is the first analysis of their dynamics within the slit diaphragm multiprotein complex. Identification of the mechanisms of slit diaphragm maintenance will help to develop novel therapies for proteinuric renal diseases that are frequently limited to symptomatic treatment.
Collapse
Affiliation(s)
- Konrad Lang
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Helena Heinkele
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Mengmeng Chen
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Lea Gerstner
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Dominik Spitz
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Severine Kayser
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Gerd Walz
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Köttgen
- Department of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrew Spracklen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - John Poulton
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Tobias Hermle
- Department of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Bali N, Lee HK(P, Zinn K. Sticks and Stones, a conserved cell surface ligand for the Type IIa RPTP Lar, regulates neural circuit wiring in Drosophila. eLife 2022; 11:e71469. [PMID: 35356892 PMCID: PMC9000958 DOI: 10.7554/elife.71469] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Type IIa receptor-like protein tyrosine phosphatases (RPTPs) are essential for neural development. They have cell adhesion molecule (CAM)-like extracellular domains that interact with cell-surface ligands and coreceptors. We identified the immunoglobulin superfamily CAM Sticks and Stones (Sns) as a new partner for the Drosophila Type IIa RPTP Lar. Lar and Sns bind to each other in embryos and in vitro, and the human Sns ortholog, Nephrin, binds to human Type IIa RPTPs. Genetic analysis shows that Lar and Sns function together to regulate larval neuromuscular junction development, axon guidance in the mushroom body (MB), and innervation of the optic lobe (OL) medulla by R7 photoreceptors. In the neuromuscular system, Lar and Sns are both required in motor neurons, and may function as coreceptors. In the MB and OL, however, the relevant Lar-Sns interactions are in trans (between neurons), so Sns functions as a Lar ligand in these systems.
Collapse
Affiliation(s)
- Namrata Bali
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Hyung-Kook (Peter) Lee
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| |
Collapse
|
6
|
Paul A, Annunziato S, Lu B, Sun T, Evrova O, Planas-Paz L, Orsini V, Terracciano LM, Charlat O, Loureiro ZY, Ji L, Zamponi R, Sigoillot F, Lei H, Lindeman A, Russ C, Reece-Hoyes JS, Nicholson TB, Tchorz JS, Cong F. Cell adhesion molecule KIRREL1 is a feedback regulator of Hippo signaling recruiting SAV1 to cell-cell contact sites. Nat Commun 2022; 13:930. [PMID: 35177623 PMCID: PMC8854406 DOI: 10.1038/s41467-022-28567-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
The Hippo/YAP pathway controls cell proliferation through sensing physical and spatial organization of cells. How cell-cell contact is sensed by Hippo signaling is poorly understood. Here, we identified the cell adhesion molecule KIRREL1 as an upstream positive regulator of the mammalian Hippo pathway. KIRREL1 physically interacts with SAV1 and recruits SAV1 to cell-cell contact sites. Consistent with the hypothesis that KIRREL1-mediated cell adhesion suppresses YAP activity, knockout of KIRREL1 increases YAP activity in neighboring cells. Analyzing pan-cancer CRISPR proliferation screen data reveals KIRREL1 as the top plasma membrane protein showing strong correlation with known Hippo regulators, highlighting a critical role of KIRREL1 in regulating Hippo signaling and cell proliferation. During liver regeneration in mice, KIRREL1 is upregulated, and its genetic ablation enhances hepatic YAP activity, hepatocyte reprogramming and biliary epithelial cell proliferation. Our data suggest that KIRREL1 functions as a feedback regulator of the mammalian Hippo pathway through sensing cell-cell interaction and recruiting SAV1 to cell-cell contact sites.
Collapse
Affiliation(s)
- Atanu Paul
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Stefano Annunziato
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Bo Lu
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Tianliang Sun
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Olivera Evrova
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Lara Planas-Paz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Vanessa Orsini
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Luigi M Terracciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy.,IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Olga Charlat
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Zinger Yang Loureiro
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Lei Ji
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Raffaella Zamponi
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Frederic Sigoillot
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Hong Lei
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Alicia Lindeman
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Carsten Russ
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - John S Reece-Hoyes
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Thomas B Nicholson
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Jan S Tchorz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Feng Cong
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA.
| |
Collapse
|
7
|
Junion G, Jagla K. Diversification of muscle types in Drosophila embryos. Exp Cell Res 2022; 410:112950. [PMID: 34838813 DOI: 10.1016/j.yexcr.2021.112950] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/31/2022]
Abstract
Drosophila embryonic somatic muscles represent a simple and tractable model system to study the gene regulatory networks that control diversification of cell types. Somatic myogenesis in Drosophila is initiated by intrinsic action of the mesodermal master gene twist, which activates a cascade of transcriptional outputs including myogenic differentiation factor Mef2, which triggers all aspects of the myogenic differentiation program. In parallel, the expression of a combinatorial code of identity transcription factors (iTFs) defines discrete particular features of each muscle fiber, such as number of fusion events, and specific attachment to tendon cells or innervation, thus ensuring diversification of muscle types. Here, we take the example of a subset of lateral transverse (LT) muscles and discuss how the iTF code and downstream effector genes progressively define individual LT properties such as fusion program, attachment and innervation. We discuss new challenges in the field including the contribution of posttranscriptional and epitranscriptomic regulation of gene expression in the diversification of cell types.
Collapse
Affiliation(s)
- Guillaume Junion
- Genetics Reproduction and Development Institute (iGReD), CNRS UMR6293, INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Krzysztof Jagla
- Genetics Reproduction and Development Institute (iGReD), CNRS UMR6293, INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand, France.
| |
Collapse
|
8
|
Sengupta T, Koonce NL, Vázquez-Martínez N, Moyle MW, Duncan LH, Emerson SE, Han X, Shao L, Wu Y, Santella A, Fan L, Bao Z, Mohler W, Shroff H, Colón-Ramos DA. Differential adhesion regulates neurite placement via a retrograde zippering mechanism. eLife 2021; 10:71171. [PMID: 34783657 PMCID: PMC8843091 DOI: 10.7554/elife.71171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
During development, neurites and synapses segregate into specific neighborhoods or layers within nerve bundles. The developmental programs guiding placement of neurites in specific layers, and hence their incorporation into specific circuits, are not well understood. We implement novel imaging methods and quantitative models to document the embryonic development of the C. elegans brain neuropil, and discover that differential adhesion mechanisms control precise placement of single neurites onto specific layers. Differential adhesion is orchestrated via developmentally-regulated expression of the IgCAM SYG-1, and its partner ligand SYG-2. Changes in SYG-1 expression across neuropil layers result in changes in adhesive forces, which sort SYG-2-expressing neurons. Sorting to layers occurs, not via outgrowth from the neurite tip, but via an alternate mechanism of retrograde zippering, involving interactions between neurite shafts. Our study indicates that biophysical principles from differential adhesion govern neurite placement and synaptic specificity in vivo in developing neuropil bundles.
Collapse
Affiliation(s)
- Titas Sengupta
- Yale University School of Medicine, New Haven, United States
| | - Noelle L Koonce
- Yale University School of Medicine, New Haven, United States
| | | | - Mark W Moyle
- Yale University School of Medicine, New Haven, United States
| | | | - Sarah E Emerson
- Yale University School of Medicine, New Haven, United States
| | - Xiaofei Han
- National Institutes of Health, Bethesda, United States
| | - Lin Shao
- Yale University School of Medicine, New Haven, United States
| | - Yicong Wu
- National Institutes of Health, Bethesda, United States
| | - Anthony Santella
- Developmental Biology Program, Molecular Cytology Core, Sloan-Kettering Institute, New York, United States
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Institute, Weill Cornell Medicine, New York, United States
| | - Zhirong Bao
- Developmental Biology Program, Sloan-Kettering Institute, New York, United States
| | - William Mohler
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, United States
| | - Hari Shroff
- National Institutes of Health, Bethesda, United States
| | | |
Collapse
|
9
|
Chan EHY, Zhou Y, Aerne BL, Holder MV, Weston A, Barry DJ, Collinson L, Tapon N. RASSF8-mediated transport of Echinoid via the exocyst promotes Drosophila wing elongation and epithelial ordering. Development 2021; 148:dev199731. [PMID: 34532737 PMCID: PMC8572004 DOI: 10.1242/dev.199731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/13/2021] [Indexed: 01/14/2023]
Abstract
Cell-cell junctions are dynamic structures that maintain cell cohesion and shape in epithelial tissues. During development, junctions undergo extensive rearrangements to drive the epithelial remodelling required for morphogenesis. This is particularly evident during axis elongation, where neighbour exchanges, cell-cell rearrangements and oriented cell divisions lead to large-scale alterations in tissue shape. Polarised vesicle trafficking of junctional components by the exocyst complex has been proposed to promote junctional rearrangements during epithelial remodelling, but the receptors that allow exocyst docking to the target membranes remain poorly understood. Here, we show that the adherens junction component Ras Association domain family 8 (RASSF8) is required for the epithelial re-ordering that occurs during Drosophila pupal wing proximo-distal elongation. We identify the exocyst component Sec15 as a RASSF8 interactor. Loss of RASSF8 elicits cytoplasmic accumulation of Sec15 and Rab11-containing vesicles. These vesicles also contain the nectin-like homophilic adhesion molecule Echinoid, the depletion of which phenocopies the wing elongation and epithelial packing defects observed in RASSF8 mutants. Thus, our results suggest that RASSF8 promotes exocyst-dependent docking of Echinoid-containing vesicles during morphogenesis.
Collapse
Affiliation(s)
- Eunice H. Y. Chan
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Yanxiang Zhou
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Birgit L. Aerne
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Maxine V. Holder
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Anne Weston
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - David J. Barry
- Advanced Light Microscopy Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Lucy Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nicolas Tapon
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
10
|
Rodríguez-Pérez F, Manford AG, Pogson A, Ingersoll AJ, Martínez-González B, Rape M. Ubiquitin-dependent remodeling of the actin cytoskeleton drives cell fusion. Dev Cell 2021; 56:588-601.e9. [PMID: 33609460 DOI: 10.1016/j.devcel.2021.01.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/14/2020] [Accepted: 01/24/2021] [Indexed: 12/11/2022]
Abstract
Cell-cell fusion is a frequent and essential event during development, and its dysregulation causes diseases ranging from infertility to muscle weakness. Fusing cells need to repeatedly remodel their plasma membrane through orchestrated formation and disassembly of actin filaments, but how the dynamic reorganization of the cortical actin cytoskeleton is controlled is still poorly understood. Here, we identified a ubiquitin-dependent toggle switch that establishes reversible actin bundling during mammalian cell fusion. We found that EPS8-IRSp53 complexes stabilize cortical actin bundles at sites of cell contact to promote close membrane alignment. EPS8 monoubiquitylation by CUL3KCTD10 displaces EPS8-IRSp53 from membranes and counteracts actin bundling, a dual activity that restricts actin bundling to allow paired cells to progress with fusion. We conclude that cytoskeletal rearrangements during development are precisely controlled by ubiquitylation, raising the possibility of modulating the efficiency of cell-cell fusion for therapeutic benefit.
Collapse
Affiliation(s)
- Fernando Rodríguez-Pérez
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Andrew G Manford
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Angela Pogson
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Andrew J Ingersoll
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Brenda Martínez-González
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Michael Rape
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
11
|
Finegan TM, Bergstralh DT. Neuronal immunoglobulin superfamily cell adhesion molecules in epithelial morphogenesis: insights from Drosophila. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190553. [PMID: 32829687 PMCID: PMC7482216 DOI: 10.1098/rstb.2019.0553] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2020] [Indexed: 12/25/2022] Open
Abstract
In this review, we address the function of immunoglobulin superfamily cell adhesion molecules (IgCAMs) in epithelia. Work in the Drosophila model system in particular has revealed novel roles for calcium-independent adhesion molecules in the morphogenesis of epithelial tissues. We review the molecular composition of lateral junctions with a focus on their IgCAM components and reconsider the functional roles of epithelial lateral junctions. The epithelial IgCAMs discussed in this review have well-defined roles in the nervous system, particularly in the process of axon guidance, suggesting functional overlap and conservation in mechanism between that process and epithelial remodelling. We expand on the hypothesis that epithelial occluding junctions and synaptic junctions are compositionally equivalent and present a novel hypothesis that the mechanism of epithelial cell (re)integration and synaptic junction formation are shared. We highlight the importance of considering non-cadherin-based adhesion in our understanding of the mechanics of epithelial tissues and raise questions to direct future work. This article is part of the discussion meeting issue 'Contemporary morphogenesis'.
Collapse
|
12
|
Abstract
Cell-cell fusion is indispensable for creating life and building syncytial tissues and organs. Ever since the discovery of cell-cell fusion, how cells join together to form zygotes and multinucleated syncytia has remained a fundamental question in cell and developmental biology. In the past two decades, Drosophila myoblast fusion has been used as a powerful genetic model to unravel mechanisms underlying cell-cell fusion in vivo. Many evolutionarily conserved fusion-promoting factors have been identified and so has a surprising and conserved cellular mechanism. In this review, we revisit key findings in Drosophila myoblast fusion and highlight the critical roles of cellular invasion and resistance in driving cell membrane fusion.
Collapse
Affiliation(s)
- Donghoon M Lee
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| | - Elizabeth H Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
13
|
Geisbrecht ER, Baylies MK. In memoriam: Susan Abmayr (1956–2019) – “What do we do? Whatever it takes!”. Skelet Muscle 2019. [PMCID: PMC6882191 DOI: 10.1186/s13395-019-0215-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
14
|
Kampf LL, Schneider R, Gerstner L, Thünauer R, Chen M, Helmstädter M, Amar A, Onuchic-Whitford AC, Loza Munarriz R, Berdeli A, Müller D, Schrezenmeier E, Budde K, Mane S, Laricchia KM, Rehm HL, MacArthur DG, Lifton RP, Walz G, Römer W, Bergmann C, Hildebrandt F, Hermle T. TBC1D8B Mutations Implicate RAB11-Dependent Vesicular Trafficking in the Pathogenesis of Nephrotic Syndrome. J Am Soc Nephrol 2019; 30:2338-2353. [PMID: 31732614 DOI: 10.1681/asn.2019040414] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mutations in about 50 genes have been identified as monogenic causes of nephrotic syndrome, a frequent cause of CKD. These genes delineated the pathogenetic pathways and rendered significant insight into podocyte biology. METHODS We used whole-exome sequencing to identify novel monogenic causes of steroid-resistant nephrotic syndrome (SRNS). We analyzed the functional significance of an SRNS-associated gene in vitro and in podocyte-like Drosophila nephrocytes. RESULTS We identified hemizygous missense mutations in the gene TBC1D8B in five families with nephrotic syndrome. Coimmunoprecipitation assays indicated interactions between TBC1D8B and active forms of RAB11. Silencing TBC1D8B in HEK293T cells increased basal autophagy and exocytosis, two cellular functions that are independently regulated by RAB11. This suggests that TBC1D8B plays a regulatory role by inhibiting endogenous RAB11. Coimmunoprecipitation assays showed TBC1D8B also interacts with the slit diaphragm protein nephrin, and colocalizes with it in immortalized cell lines. Overexpressed murine Tbc1d8b with patient-derived mutations had lower affinity for endogenous RAB11 and nephrin compared with wild-type Tbc1d8b protein. Knockdown of Tbc1d8b in Drosophila impaired function of the podocyte-like nephrocytes, and caused mistrafficking of Sns, the Drosophila ortholog of nephrin. Expression of Rab11 RNAi in nephrocytes entailed defective delivery of slit diaphragm protein to the membrane, whereas RAB11 overexpression revealed a partial phenotypic overlap to Tbc1d8b loss of function. CONCLUSIONS Novel mutations in TBC1D8B are monogenic causes of SRNS. This gene inhibits RAB11. Our findings suggest that RAB11-dependent vesicular nephrin trafficking plays a role in the pathogenesis of nephrotic syndrome.
Collapse
Affiliation(s)
- Lina L Kampf
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Ronen Schneider
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lea Gerstner
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Roland Thünauer
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Freiburg, Germany.,Advanced Light and Fluorescence Microscopy Facility, Centre for Structural Systems Biology (CSSB) and University of Hamburg, Hamburg, Germany
| | - Mengmeng Chen
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Helmstädter
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Ali Amar
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ana C Onuchic-Whitford
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Afig Berdeli
- Department of Pediatrics, Molecular Medicine Laboratory, Ege University, Izmir, Turkey
| | - Dominik Müller
- Department of Pediatric Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - Kristen M Laricchia
- Broad Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge
| | - Heidi L Rehm
- Broad Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge
| | - Daniel G MacArthur
- Broad Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.,Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Gerd Walz
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Winfried Römer
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Carsten Bergmann
- Center for Human Genetics, Mainz, Germany.,Center for Human Genetics, Bioscientia, Ingelheim, Germany; and.,Department of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts;
| | - Tobias Hermle
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany;
| |
Collapse
|
15
|
Abstract
Cell-cell fusion is a fundamental process underlying fertilization, development, regeneration and physiology of metazoans. It is a multi-step process involving cell recognition and adhesion, actin cytoskeletal rearrangements, fusogen engagement, lipid mixing and fusion pore formation, ultimately resulting in the integration of two fusion partners. Here, we focus on the asymmetric actin cytoskeletal rearrangements at the site of fusion, known as the fusogenic synapse, which was first discovered during myoblast fusion in Drosophila embryos and later also found in mammalian muscle and non-muscle cells. At the asymmetric fusogenic synapse, actin-propelled invasive membrane protrusions from an attacking fusion partner trigger actomyosin-based mechanosensory responses in the receiving cell. The interplay between the invasive and resisting forces generated by the two fusion partners puts the fusogenic synapse under high mechanical tension and brings the two cell membranes into close proximity, promoting the engagement of fusogens to initiate fusion pore formation. In this Cell Science at a Glance article and the accompanying poster, we highlight the molecular, cellular and biophysical events at the asymmetric fusogenic synapse using Drosophila myoblast fusion as a model.
Collapse
Affiliation(s)
- Ji Hoon Kim
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth H Chen
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA .,Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
16
|
Carrasco-Rando M, Prieto-Sánchez S, Culi J, Tutor AS, Ruiz-Gómez M. A specific isoform of Pyd/ZO-1 mediates junctional remodeling and formation of slit diaphragms. J Cell Biol 2019; 218:2294-2308. [PMID: 31171632 PMCID: PMC6605796 DOI: 10.1083/jcb.201810171] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/21/2019] [Accepted: 05/14/2019] [Indexed: 12/18/2022] Open
Abstract
Podocyte slit diaphragms are key components of the glomerular filtration barrier. Using Drosophila nephrocytes, Carrasco-Rando et al. propose a conserved role for Pyd/ZO-1 in triggering junctional remodeling leading to the formation of slit diaphragms. The podocyte slit diaphragm (SD), responsible for blood filtration in vertebrates, is a major target of injury in chronic kidney disease. The damage includes severe morphological changes with destabilization of SDs and their replacement by junctional complexes between abnormally broadened foot processes. In Drosophila melanogaster, SDs are present in nephrocytes, which filter the fly's hemolymph. Here, we show that a specific isoform of Polychaetoid/ZO-1, Pyd-P, is essential for Drosophila SDs, since, in pyd mutants devoid of Pyd-P, SDs do not form and the SD component Dumbfounded accumulates at ectopic septate-like junctions between abnormally aggregated nephrocytes. Reintroduction of Pyd-P leads to junctional remodeling and their progressive normalization toward SDs. This transition requires the coiled-coil domain of Pyd-P and implies formation of nonclathrin vesicles containing SD components and their trafficking to the nephrocyte external membrane, where SDs assemble. Analyses in zebrafish suggest a conserved role for Tjp1a/ZO-1 in promoting junctional remodeling in podocytes.
Collapse
Affiliation(s)
- Marta Carrasco-Rando
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Silvia Prieto-Sánchez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Joaquim Culi
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio S Tutor
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Mar Ruiz-Gómez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
17
|
Ben-Zvi DS, Volk T. Escort cell encapsulation of Drosophila germline cells is maintained by irre cell recognition module proteins. Biol Open 2019; 8:bio039842. [PMID: 30837217 PMCID: PMC6451344 DOI: 10.1242/bio.039842] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/06/2019] [Indexed: 12/17/2022] Open
Abstract
Differentiation of germline stem cells (GSCs) in the Drosophila ovary is induced by somatic escort cells (ECs), which extend membrane protrusions encapsulating the germline cells (GCs). Germline encapsulation requires activated epidermal growth factor receptor (Egfr) signaling within the ECs, following secretion of its ligands from the GCs. We show that the conserved family of irre cell recognition module (IRM) proteins is essential for GC encapsulation by ECs, with a requirement for roughest (rst) and kin of irre (kirre) in the germline and for sticks and stones (sns) and hibris (hbs) in ECs. In the absence of IRM components in their respective cell types, EC extensions are reduced concomitantly with a decrease in Egfr signaling in these cells. Reintroducing either activated Egfr in the ECs, or overexpressing its ligand Spitz (Spi) from the germline, rescued the requirement for IRM proteins in both cell types. These experiments introduce novel essential components, the IRM proteins, into the process of inductive interactions between GCs and ECs, and imply that IRM-mediated activity is required upstream of the Egfr signaling.
Collapse
Affiliation(s)
- Doreen S Ben-Zvi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
18
|
Hermle T, Schneider R, Schapiro D, Braun DA, van der Ven AT, Warejko JK, Daga A, Widmeier E, Nakayama M, Jobst-Schwan T, Majmundar AJ, Ashraf S, Rao J, Finn LS, Tasic V, Hernandez JD, Bagga A, Jalalah SM, El Desoky S, Kari JA, Laricchia KM, Lek M, Rehm HL, MacArthur DG, Mane S, Lifton RP, Shril S, Hildebrandt F. GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome. J Am Soc Nephrol 2018; 29:2123-2138. [PMID: 29959197 PMCID: PMC6065084 DOI: 10.1681/asn.2017121312] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/24/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of CKD. The discovery of monogenic causes of SRNS has revealed specific pathogenetic pathways, but these monogenic causes do not explain all cases of SRNS. METHODS To identify novel monogenic causes of SRNS, we screened 665 patients by whole-exome sequencing. We then evaluated the in vitro functional significance of two genes and the mutations therein that we discovered through this sequencing and conducted complementary studies in podocyte-like Drosophila nephrocytes. RESULTS We identified conserved, homozygous missense mutations of GAPVD1 in two families with early-onset NS and a homozygous missense mutation of ANKFY1 in two siblings with SRNS. GAPVD1 and ANKFY1 interact with the endosomal regulator RAB5. Coimmunoprecipitation assays indicated interaction between GAPVD1 and ANKFY1 proteins, which also colocalized when expressed in HEK293T cells. Silencing either protein diminished the podocyte migration rate. Compared with wild-type GAPVD1 and ANKFY1, the mutated proteins produced upon ectopic expression of GAPVD1 or ANKFY1 bearing the patient-derived mutations exhibited altered binding affinity for active RAB5 and reduced ability to rescue the knockout-induced defect in podocyte migration. Coimmunoprecipitation assays further demonstrated a physical interaction between nephrin and GAPVD1, and immunofluorescence revealed partial colocalization of these proteins in rat glomeruli. The patient-derived GAPVD1 mutations reduced nephrin-GAPVD1 binding affinity. In Drosophila, silencing Gapvd1 impaired endocytosis and caused mistrafficking of the nephrin ortholog. CONCLUSIONS Mutations in GAPVD1 and probably in ANKFY1 are novel monogenic causes of NS. The discovery of these genes implicates RAB5 regulation in the pathogenesis of human NS.
Collapse
Affiliation(s)
- Tobias Hermle
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Renal Division, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ronen Schneider
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David Schapiro
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amelie T van der Ven
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jillian K Warejko
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ankana Daga
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eugen Widmeier
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Makiko Nakayama
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tilman Jobst-Schwan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amar J Majmundar
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shazia Ashraf
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jia Rao
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Laura S Finn
- Department of Pathology, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - Velibor Tasic
- Department of Pediatric Nephrology, Medical Faculty Skopje, University Children's Hospital, Skopje, Macedonia
| | - Joel D Hernandez
- Department of Pediatric Nephrology, Providence Sacred Heart Medical Center and Children's Hospital, Spokane, Washington
| | - Arvind Bagga
- Division of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Sherif El Desoky
- Pediatric Nephrology Center of Excellence and Pediatric Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Jameela A Kari
- Pediatric Nephrology Center of Excellence and Pediatric Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Kristen M Laricchia
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Monkol Lek
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Heidi L Rehm
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Daniel G MacArthur
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut; and
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut; and
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Shirlee Shril
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
19
|
Duan R, Kim JH, Shilagardi K, Schiffhauer ES, Lee DM, Son S, Li S, Thomas C, Luo T, Fletcher DA, Robinson DN, Chen EH. Spectrin is a mechanoresponsive protein shaping fusogenic synapse architecture during myoblast fusion. Nat Cell Biol 2018; 20:688-698. [PMID: 29802406 PMCID: PMC6397639 DOI: 10.1038/s41556-018-0106-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 04/18/2018] [Indexed: 12/24/2022]
Abstract
Spectrin is a membrane skeletal protein best known for its structural role in maintaining cell shape and protecting cells from mechanical damage. Here, we report that α/βH-spectrin (βH is also called karst) dynamically accumulates and dissolves at the fusogenic synapse between fusing Drosophila muscle cells, where an attacking fusion partner invades its receiving partner with actin-propelled protrusions to promote cell fusion. Using genetics, cell biology, biophysics and mathematical modelling, we demonstrate that spectrin exhibits a mechanosensitive accumulation in response to shear deformation, which is highly elevated at the fusogenic synapse. The transiently accumulated spectrin network functions as a cellular fence to restrict the diffusion of cell-adhesion molecules and a cellular sieve to constrict the invasive protrusions, thereby increasing the mechanical tension of the fusogenic synapse to promote cell membrane fusion. Our study reveals a function of spectrin as a mechanoresponsive protein and has general implications for understanding spectrin function in dynamic cellular processes.
Collapse
Affiliation(s)
- Rui Duan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Laboratory of Regenerative Medicine in Sports Science, School of Sports Science, South China Normal University, Guangzhou, China
| | - Ji Hoon Kim
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Khurts Shilagardi
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric S Schiffhauer
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donghoon M Lee
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sungmin Son
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Shuo Li
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Claire Thomas
- Departments of Biology and of Biochemistry and Molecular Biology, Penn State University, University Park, PA, USA
| | - Tianzhi Luo
- Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Daniel A Fletcher
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth H Chen
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
20
|
Abstract
Cell-cell fusion is essential for fertilization and organ development. Dedicated proteins known as fusogens are responsible for mediating membrane fusion. However, until recently, these proteins either remained unidentified or were poorly understood at the mechanistic level. Here, we review how fusogens surmount multiple energy barriers to mediate cell-cell fusion. We describe how early preparatory steps bring membranes to a distance of ∼10 nm, while fusogens act in the final approach between membranes. The mechanical force exerted by cell fusogens and the accompanying lipidic rearrangements constitute the hallmarks of cell-cell fusion. Finally, we discuss the relationship between viral and eukaryotic fusogens, highlight a classification scheme regrouping a superfamily of fusogens called Fusexins, and propose new questions and avenues of enquiry.
Collapse
Affiliation(s)
- Javier M Hernández
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, D-44227 Dortmund, Germany
| | - Benjamin Podbilewicz
- Department of Biology, Technion - Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
21
|
Hermle T, Braun DA, Helmstädter M, Huber TB, Hildebrandt F. Modeling Monogenic Human Nephrotic Syndrome in the Drosophila Garland Cell Nephrocyte. J Am Soc Nephrol 2017; 28:1521-1533. [PMID: 27932481 PMCID: PMC5407722 DOI: 10.1681/asn.2016050517] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 11/05/2016] [Indexed: 11/03/2022] Open
Abstract
Steroid-resistant nephrotic syndrome is characterized by podocyte dysfunction. Drosophila garland cell nephrocytes are podocyte-like cells and thus provide a potential in vivo model in which to study the pathogenesis of nephrotic syndrome. However, relevant pathomechanisms of nephrotic syndrome have not been studied in nephrocytes. Here, we discovered that two Drosophila slit diaphragm proteins, orthologs of the human genes encoding nephrin and nephrin-like protein 1, colocalize within a fingerprint-like staining pattern that correlates with ultrastructural morphology. Using RNAi and conditional CRISPR/Cas9 in nephrocytes, we found this pattern depends on the expression of both orthologs. Tracer endocytosis by nephrocytes required Cubilin and reflected size selectivity analogous to that of glomerular function. Using RNAi and tracer endocytosis as a functional read-out, we screened Drosophila orthologs of human monogenic causes of nephrotic syndrome and observed conservation of the central pathogenetic alterations. We focused on the coenzyme Q10 (CoQ10) biosynthesis gene Coq2, the silencing of which disrupted slit diaphragm morphology. Restoration of CoQ10 synthesis by vanillic acid partially rescued the phenotypic and functional alterations induced by Coq2-RNAi. Notably, Coq2 colocalized with mitochondria, and Coq2 silencing increased the formation of reactive oxygen species (ROS). Silencing of ND75, a subunit of the mitochondrial respiratory chain that controls ROS formation independently of CoQ10, phenocopied the effect of Coq2-RNAi. Moreover, the ROS scavenger glutathione partially rescued the effects of Coq2-RNAi. In conclusion, Drosophila garland cell nephrocytes provide a model with which to study the pathogenesis of nephrotic syndrome, and ROS formation may be a pathomechanism of COQ2-nephropathy.
Collapse
Affiliation(s)
- Tobias Hermle
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts;
| | - Daniela A Braun
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Martin Helmstädter
- Renal Division, University Medical Center Freiburg, Freiburg, Germany, Faculty of Medicine, University of Freiburg, Freiburg, Germany; and
| | - Tobias B Huber
- Renal Division, University Medical Center Freiburg, Freiburg, Germany, Faculty of Medicine, University of Freiburg, Freiburg, Germany; and
- BIOSS Center for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
22
|
Krauss RS, Joseph GA, Goel AJ. Keep Your Friends Close: Cell-Cell Contact and Skeletal Myogenesis. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029298. [PMID: 28062562 DOI: 10.1101/cshperspect.a029298] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Development of skeletal muscle is a multistage process that includes lineage commitment of multipotent progenitor cells, differentiation and fusion of myoblasts into multinucleated myofibers, and maturation of myofibers into distinct types. Lineage-specific transcriptional regulation lies at the core of this process, but myogenesis is also regulated by extracellular cues. Some of these cues are initiated by direct cell-cell contact between muscle precursor cells themselves or between muscle precursors and cells of other lineages. Examples of the latter include interaction of migrating neural crest cells with multipotent muscle progenitor cells, muscle interstitial cells with myoblasts, and neurons with myofibers. Among the signaling factors involved are Notch ligands and receptors, cadherins, Ig superfamily members, and Ephrins and Eph receptors. In this article we describe recent progress in this area and highlight open questions raised by the findings.
Collapse
Affiliation(s)
- Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Giselle A Joseph
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Aviva J Goel
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
23
|
Pérez-Moreno JJ, Espina-Zambrano AG, García-Calderón CB, Estrada B. Kon-tiki/Perdido enhances PS2 integrin adhesion and localizes its ligand, Thrombospondin, in the myotendinous junction. J Cell Sci 2017; 130:950-962. [DOI: 10.1242/jcs.197459] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/13/2017] [Indexed: 12/13/2022] Open
Abstract
Cell-extracellular matrix adhesion is mediated by cell receptors, mainly integrins and transmembrane proteoglycans, which can functionally interact. How these receptors are regulated and coordinated is largely unknown and key to understand cell adhesion in development. We show that the conserved transmembrane proteoglycan Kon-tiki/Perdido (Kon) interacts with αPS2βPS integrin to mediate muscle-tendon adhesion. Double mutant embryos for kon and inflated show a synergistic increase in muscle detachment. Furthermore, Kon modulates αPS2βPS signaling at the muscle attachment, since P-Fak is reduced in kon mutants. This reduction in integrin signaling can be rescued by the expression of a truncated Kon protein containing the transmembrane and extracellular domains, suggesting that these domains are sufficient to mediate this signaling. We show that these domains are sufficient to properly localize the αPS2βPS ligand, Thrombospondin, to the muscle attachment, and to partially rescue Kon dependent muscle-tendon adhesion. We propose that Kon can engage in a protein complex with αPS2βPS and enhance integrin-mediated signaling and adhesion by recruiting its ligand, which would increase integrin-binding affinity to the extracellular matrix, resulting in the consolidation of the myotendinous junction.
Collapse
Affiliation(s)
- J. J. Pérez-Moreno
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Seville, Spain
- Present address: Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - A. G. Espina-Zambrano
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Seville, Spain
| | - C. B. García-Calderón
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Seville, Spain
- Present address: Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - B. Estrada
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Seville, Spain
| |
Collapse
|
24
|
Dhanyasi N, Segal D, Shimoni E, Shinder V, Shilo BZ, VijayRaghavan K, Schejter ED. Surface apposition and multiple cell contacts promote myoblast fusion in Drosophila flight muscles. J Cell Biol 2016; 211:191-203. [PMID: 26459604 PMCID: PMC4602036 DOI: 10.1083/jcb.201503005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transmission EM methods reveal that cell–cell fusion of individual myoblasts with growing Drosophila flight muscles is a stepwise process in which the cell adhesion and branched actin machineries mediate tight apposition and formation of multiple contacts and pores between the surfaces of the fusing cells. Fusion of individual myoblasts to form multinucleated myofibers constitutes a widely conserved program for growth of the somatic musculature. We have used electron microscopy methods to study this key form of cell–cell fusion during development of the indirect flight muscles (IFMs) of Drosophila melanogaster. We find that IFM myoblast–myotube fusion proceeds in a stepwise fashion and is governed by apparent cross talk between transmembrane and cytoskeletal elements. Our analysis suggests that cell adhesion is necessary for bringing myoblasts to within a minimal distance from the myotubes. The branched actin polymerization machinery acts subsequently to promote tight apposition between the surfaces of the two cell types and formation of multiple sites of cell–cell contact, giving rise to nascent fusion pores whose expansion establishes full cytoplasmic continuity. Given the conserved features of IFM myogenesis, this sequence of cell interactions and membrane events and the mechanistic significance of cell adhesion elements and the actin-based cytoskeleton are likely to represent general principles of the myoblast fusion process.
Collapse
Affiliation(s)
- Nagaraju Dhanyasi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India Manipal University, Manipal, Karnataka 576104, India
| | - Dagan Segal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eyal Shimoni
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Vera Shinder
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - K VijayRaghavan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
25
|
Linneweber GA, Winking M, Fischbach KF. The Cell Adhesion Molecules Roughest, Hibris, Kin of Irre and Sticks and Stones Are Required for Long Range Spacing of the Drosophila Wing Disc Sensory Sensilla. PLoS One 2015; 10:e0128490. [PMID: 26053791 PMCID: PMC4459997 DOI: 10.1371/journal.pone.0128490] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 04/27/2015] [Indexed: 12/18/2022] Open
Abstract
Most animal tissues and organ systems are comprised of highly ordered arrays of varying cell types. The development of external sensory organs requires complex cell-cell communication in order to give each cell a specific identity and to ensure a regular distributed pattern of the sensory bristles. This involves both long and short range signaling mediated by either diffusible or cell anchored factors. In a variety of processes the heterophilic Irre Cell Recognition Module, consisting of the Neph-like proteins: Roughest, Kin of irre and of the Nephrin-like proteins: Sticks and Stones, Hibris, plays key roles in the recognition events of different cell types throughout development. In the present study these proteins are apically expressed in the adhesive belt of epithelial cells participating in sense organ development in a partially exclusive and asymmetric manner. Using mutant analysis the GAL4/UAS system, RNAi and gain of function we found an involvement of all four Irre Cell Recognition Module-proteins in the development of a highly structured array of sensory organs in the wing disc. The proteins secure the regular spacing of sensory organs showing partial redundancy and may function in early lateral inhibition events as well as in cell sorting processes. Comparisons with other systems suggest that the Irre Cell Recognition module is a key organizer of highly repetitive structures.
Collapse
Affiliation(s)
- Gerit Arne Linneweber
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| | - Mathis Winking
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| | - Karl-Friedrich Fischbach
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| |
Collapse
|
26
|
Haralalka S, Abmayr SM. Tracing myoblast fusion in Drosophila embryos by fluorescent actin probes. Methods Mol Biol 2015; 1313:149-64. [PMID: 25947663 DOI: 10.1007/978-1-4939-2703-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Myoblast fusion in the Drosophila embryo is a highly elaborate process that is initiated by Founder Cells and Fusion-Competent Myoblasts (FCMs). It occurs through an asymmetric event in which actin foci assemble in the FCMs at points of cell-cell contact and direct the formation of membrane protrusions that drive fusion. Herein, we describe the approach that we have used to image in living embryos the highly dynamic actin foci and actin-rich projections that precede myoblast fusion. We discuss resources currently available for imaging actin and myogenesis, and our experience with these resources if available. This technical report is not intended to be comprehensive on providing instruction on standard microscopy practices or software utilization. However, we discuss microscope parameters that we have used in data collection, and our experience with image processing tools in data analysis.
Collapse
Affiliation(s)
- Shruti Haralalka
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO, 64110, USA
| | | |
Collapse
|
27
|
Haralalka S, Shelton C, Cartwright HN, Guo F, Trimble R, Kumar RP, Abmayr SM. Live imaging provides new insights on dynamic F-actin filopodia and differential endocytosis during myoblast fusion in Drosophila. PLoS One 2014; 9:e114126. [PMID: 25474591 PMCID: PMC4256407 DOI: 10.1371/journal.pone.0114126] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/28/2014] [Indexed: 11/29/2022] Open
Abstract
The process of myogenesis includes the recognition, adhesion, and fusion of committed myoblasts into multinucleate syncytia. In the larval body wall muscles of Drosophila, this elaborate process is initiated by Founder Cells and Fusion-Competent Myoblasts (FCMs), and cell adhesion molecules Kin-of-IrreC (Kirre) and Sticks-and-stones (Sns) on their respective surfaces. The FCMs appear to provide the driving force for fusion, via the assembly of protrusions associated with branched F-actin and the WASp, SCAR and Arp2/3 pathways. In the present study, we utilize the dorsal pharyngeal musculature that forms in the Drosophila embryo as a model to explore myoblast fusion and visualize the fusion process in live embryos. These muscles rely on the same cell types and genes as the body wall muscles, but are amenable to live imaging since they do not undergo extensive morphogenetic movement during formation. Time-lapse imaging with F-actin and membrane markers revealed dynamic FCM-associated actin-enriched protrusions that rapidly extend and retract into the myotube from different sites within the actin focus. Ultrastructural analysis of this actin-enriched area showed that they have two morphologically distinct structures: wider invasions and/or narrow filopodia that contain long linear filaments. Consistent with this, formin Diaphanous (Dia) and branched actin nucleator, Arp3, are found decorating the filopodia or enriched at the actin focus, respectively, indicating that linear actin is present along with branched actin at sites of fusion in the FCM. Gain-of-function Dia and loss-of-function Arp3 both lead to fusion defects, a decrease of F-actin foci and prominent filopodia from the FCMs. We also observed differential endocytosis of cell surface components at sites of fusion, with actin reorganizing factors, WASp and SCAR, and Kirre remaining on the myotube surface and Sns preferentially taken up with other membrane proteins into early endosomes and lysosomes in the myotube.
Collapse
Affiliation(s)
- Shruti Haralalka
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, United States of America
| | - Claude Shelton
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, United States of America
| | - Heather N. Cartwright
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, United States of America
| | - Fengli Guo
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, United States of America
| | - Rhonda Trimble
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, United States of America
| | - Ram P. Kumar
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, United States of America
| | - Susan M. Abmayr
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, United States of America
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas, 66160, United States of America
- * E-mail:
| |
Collapse
|
28
|
Önel SF, Rust MB, Jacob R, Renkawitz-Pohl R. Tethering membrane fusion: common and different players in myoblasts and at the synapse. J Neurogenet 2014; 28:302-15. [PMID: 24957080 PMCID: PMC4245166 DOI: 10.3109/01677063.2014.936014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Drosophila Membrane fusion is essential for the communication of membrane-defined compartments, development of multicellular organisms and tissue homeostasis. Although membrane fusion has been studied extensively, still little is known about the molecular mechanisms. Especially the intercellular fusion of cells during development and tissue homeostasis is poorly understood. Somatic muscle formation in Drosophila depends on the intercellular fusion of myoblasts. In this process, myoblasts recognize each other and adhere, thereby triggering a protein machinery that leads to electron-dense plaques, vesicles and F-actin formation at apposing membranes. Two models of how local membrane stress is achieved to induce the merging of the myoblast membranes have been proposed: the electron-dense vesicles transport and release a fusogen and F-actin bends the plasma membrane. In this review, we highlight cell-adhesion molecules and intracellular proteins known to be involved in myoblast fusion. The cell-adhesion proteins also mediate the recognition and adhesion of other cell types, such as neurons that communicate with each other via special intercellular junctions, termed chemical synapses. At these synapses, neurotransmitters are released through the intracellular fusion of synaptic vesicles with the plasma membrane. As the targeting of electron-dense vesicles in myoblasts shares some similarities with the targeting of synaptic vesicle fusion, we compare molecules required for synaptic vesicle fusion to recently identified molecules involved in myoblast fusion.
Collapse
Affiliation(s)
- Susanne Filiz Önel
- Developmental Biology, Philipps University of Marburg , 35043 Marburg , Germany
| | | | | | | |
Collapse
|
29
|
Abstract
The description of the Rst protein by Karl-Friedrich Fischbach and colleagues was a milestone in the discovery of the irre cell recognition module (IRM). IRM proteins represent a family of immunoglobulin superfamily cell adhesion proteins that orchestrate intercellular adhesion and signaling events necessary for the development of various tissues. This review briefly summarizes the fundamental role of IRM proteins for neuronal wiring and filtration in organisms spanning the evolutionary distance from Drosophila (nephrocyte diaphragm) to humans (slit diaphragm).
Collapse
|
30
|
Costa MSA, Machado MCR, Vieceli FM, Amistá L, Baroneza JE, Yan CYI, Ramos RGP. The Rst-Neph family of cell adhesion molecules in Gallus gallus. J Neurogenet 2014; 28:270-81. [PMID: 24914768 DOI: 10.3109/01677063.2014.933220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Rst-Neph family comprises an evolutionarily conserved group of single-pass transmembrane glycoproteins that belong to the immunoglobulin superfamily and participate in a wide range of cell adhesion and recognition events in both vertebrates and invertebrates. In mammals and fish, three Rst-Neph members, named Neph1-3, are present. Besides being widely expressed in the embryo, particularly in the developing nervous system, they also contribute to the formation and integrity of the urine filtration apparatus in the slit diaphragm of kidney glomerular podocytes, where they form homodimers, as well as heterodimers with Nephrin, another immunoglobulin-like cell adhesion molecule. In mice, absence of Neph1 causes severe proteinuria, podocyte effacement and perinatal death, while in humans, a mutated form of Nephrin leads to congenital nephrotic syndrome of the Finnish type. Intriguingly, neither Nephrin nor Neph3 are present in birds, which nevertheless have typical vertebrate kidneys with mammalian-like slit diaphragms. These characteristics make, in principle, avian systems very helpful for understanding the evolution and functional significance of the complex interactions displayed by Rst-Neph proteins. To this end we have started a systematic study of chicken Neph embryonic and post-embryonic expression, both at mRNA and protein level. RT-qPCR mRNA quantification of the two Neph paralogues in adult tissues showed that both are expressed in heart, brain, and retina. Neph1 is additionally present in kidney, liver, pancreas, lungs, and testicles, while Neph2 mRNA is barely detected in kidney, testicles, pancreas and absent in liver and lungs. In embryos, mRNA from both genes can already be detected at as early as stage HH14, and remain expressed until at least HH28. Finally, we used a specific antibody to examine the spatial dynamics and subcellular distribution of ggNeph2 between stages HH20-28, particularly in the mesonephros, dermomyotomes, developing heart, and retina.
Collapse
Affiliation(s)
- Mara Silvia A Costa
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | | | | | | | | | | | | |
Collapse
|
31
|
Bothe I, Deng S, Baylies M. PI(4,5)P2 regulates myoblast fusion through Arp2/3 regulator localization at the fusion site. Development 2014; 141:2289-301. [PMID: 24821989 DOI: 10.1242/dev.100743] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cell-cell fusion is a regulated process that requires merging of the opposing membranes and underlying cytoskeletons. However, the integration between membrane and cytoskeleton signaling during fusion is not known. Using Drosophila, we demonstrate that the membrane phosphoinositide PI(4,5)P2 is a crucial regulator of F-actin dynamics during myoblast fusion. PI(4,5)P2 is locally enriched and colocalizes spatially and temporally with the F-actin focus that defines the fusion site. PI(4,5)P2 enrichment depends on receptor engagement but is upstream or parallel to actin remodeling. Regulators of actin branching via Arp2/3 colocalize with PI(4,5)P2 in vivo and bind PI(4,5)P2 in vitro. Manipulation of PI(4,5)P2 availability leads to impaired fusion, with a reduction in the F-actin focus size and altered focus morphology. Mechanistically, the changes in the actin focus are due to a failure in the enrichment of actin regulators at the fusion site. Moreover, improper localization of these regulators hinders expansion of the fusion interface. Thus, PI(4,5)P2 enrichment at the fusion site encodes spatial and temporal information that regulates fusion progression through the localization of activators of actin polymerization.
Collapse
Affiliation(s)
- Ingo Bothe
- Program in Developmental Biology, Sloan Kettering Institute, New York, NY 10065, USA
| | - Su Deng
- Graduate Program in Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Mary Baylies
- Program in Developmental Biology, Sloan Kettering Institute, New York, NY 10065, USA Graduate Program in Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| |
Collapse
|
32
|
Özkan E, Chia PH, Wang RR, Goriatcheva N, Borek D, Otwinowski Z, Walz T, Shen K, Garcia KC. Extracellular architecture of the SYG-1/SYG-2 adhesion complex instructs synaptogenesis. Cell 2014; 156:482-94. [PMID: 24485456 PMCID: PMC3962013 DOI: 10.1016/j.cell.2014.01.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 09/04/2013] [Accepted: 01/06/2014] [Indexed: 01/29/2023]
Abstract
SYG-1 and SYG-2 are multipurpose cell adhesion molecules (CAMs) that have evolved across all major animal taxa to participate in diverse physiological functions, ranging from synapse formation to formation of the kidney filtration barrier. In the crystal structures of several SYG-1 and SYG-2 orthologs and their complexes, we find that SYG-1 orthologs homodimerize through a common, bispecific interface that similarly mediates an unusual orthogonal docking geometry in the heterophilic SYG-1/SYG-2 complex. C. elegans SYG-1's specification of proper synapse formation in vivo closely correlates with the heterophilic complex affinity, which appears to be tuned for optimal function. Furthermore, replacement of the interacting domains of SYG-1 and SYG-2 with those from CAM complexes that assume alternative docking geometries or the introduction of segmental flexibility compromised synaptic function. These results suggest that SYG extracellular complexes do not simply act as "molecular velcro" and that their distinct structural features are important in instructing synaptogenesis. PAPERFLICK:
Collapse
Affiliation(s)
- Engin Özkan
- Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Poh Hui Chia
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Ruiqi Rachel Wang
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Natalia Goriatcheva
- Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dominika Borek
- Departments of Biochemistry and Biophysics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Zbyszek Otwinowski
- Departments of Biochemistry and Biophysics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Thomas Walz
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kang Shen
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
33
|
Lüthy K, Ahrens B, Rawal S, Lu Z, Tarnogorska D, Meinertzhagen IA, Fischbach KF. The irre cell recognition module (IRM) protein Kirre is required to form the reciprocal synaptic network of L4 neurons in the Drosophila lamina. J Neurogenet 2014; 28:291-301. [PMID: 24697410 DOI: 10.3109/01677063.2014.883390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Each neuropil module, or cartridge, in the fly's lamina has a fixed complement of cells. Of five types of monopolar cell interneurons, only L4 has collaterals that invade neighboring cartridges. In the proximal lamina, these collaterals form reciprocal synapses with both the L2 of their own cartridge and the L4 collateral branches from two other neighboring cartridges. During synaptogenesis, L4 collaterals strongly express the cell adhesion protein Kirre, a member of the irre cell recognition module (IRM) group of proteins ( Fischbach et al., 2009 , J Neurogenet, 23, 48-67). The authors show by mutant analysis and gene knockdown techniques that L4 neurons develop their lamina collaterals in the absence of this cell adhesion protein. Using electron microscopy (EM), the authors demonstrate, however, that without Kirre protein these L4 collaterals selectively form fewer synapses. The collaterals of L4 neurons of various genotypes reconstructed from serial-section EM revealed that the number of postsynaptic sites was dramatically reduced in the absence of Kirre, almost eliminating any synaptic input to L4 neurons. A significant reduction of presynaptic sites was also detected in kirre(0) mutants and gene knockdown flies using RNA interference. L4 neuron reciprocal synapses are thus almost eliminated. A presynaptic marker, Brp-short(GFP) confirmed these data using confocal microscopy. This study reveals that removing Kirre protein specifically disrupts the functional L4 synaptic network in the Drosophila lamina.
Collapse
Affiliation(s)
- Kevin Lüthy
- Faculty of Biology, Albert-Ludwigs University Freiburg , Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Differential adhesion provides a mechanical force to drive cells into stable configurations during the assembly of tissues and organs. This is well illustrated in the Drosophila eye where differential adhesion plays a role in sequential recruitment of all support cells. Cell adhesion, on the other hand, is linked to the cytoskeleton and subject to regulation by cell signaling. The integration of cell adhesion with the cytoskeleton and cell signaling may provide a more thorough explanation for the diversity of forms and shapes seen in tissues and organs.
Collapse
Affiliation(s)
- Sujin Bao
- Saint James School of Medicine , Bonaire , Caribbean Netherlands
| |
Collapse
|
35
|
Durcan PJ, Al-Shanti N, Stewart CE. Identification and characterization of novel Kirrel isoform during myogenesis. Physiol Rep 2013; 1:e00044. [PMID: 24303129 PMCID: PMC3835000 DOI: 10.1002/phy2.44] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 07/03/2013] [Indexed: 12/31/2022] Open
Abstract
Somatic cell fusion is an essential component of skeletal muscle development and growth and repair from injury. Additional cell types such as trophoblasts and osteoclasts also require somatic cell fusion events to perform their physiological functions. Currently we have rudimentary knowledge on molecular mechanisms regulating somatic cell fusion events in mammals. We therefore investigated during in vitro murine myogenesis a mammalian homolog, Kirrel, of the Drosophila Melanogaster genes Roughest (Rst) and Kin of Irre (Kirre) which regulate somatic muscle cell fusion during embryonic development. Our results demonstrate the presence of a novel murine Kirrel isoform containing a truncated cytoplasmic domain which we term Kirrel B. Protein expression levels of Kirrel B are inverse to the occurrence of cell fusion events during in vitro myogenesis which is in stark contrast to the expression profile of Rst and Kirre during myogenesis in Drosophila. Furthermore, chemical inhibition of cell fusion confirmed the inverse expression pattern of Kirrel B protein levels in relation to cell fusion events. The discovery of a novel Kirrel B protein isoform during myogenesis highlights the need for more thorough investigation of the similarities and potential differences between fly and mammals with regards to the muscle cell fusion process.
Collapse
Affiliation(s)
- Peter J Durcan
- Department of Physiological Sciences, Stellenbosch University Merriman avenue, Stellenbosch, 7600, Western Cape, South Africa ; Institute for Biomedical Research into Human movement, School of Healthcare Science, Manchester Metropolitan University Oxford road, M1 5GD, Manchester, U.K
| | | | | |
Collapse
|
36
|
Shilagardi K, Li S, Luo F, Marikar F, Duan R, Jin P, Kim JH, Murnen K, Chen EH. Actin-propelled invasive membrane protrusions promote fusogenic protein engagement during cell-cell fusion. Science 2013; 340:359-63. [PMID: 23470732 DOI: 10.1126/science.1234781] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cell-cell fusion is critical for the conception, development, and physiology of multicellular organisms. Although cellular fusogenic proteins and the actin cytoskeleton are implicated in cell-cell fusion, it remains unclear whether and how they coordinate to promote plasma membrane fusion. We reconstituted a high-efficiency, inducible cell fusion culture system in the normally nonfusing Drosophila S2R+ cells. Both fusogenic proteins and actin cytoskeletal rearrangements were necessary for cell fusion, and in combination they were sufficient to impart fusion competence. Localized actin polymerization triggered by specific cell-cell or cell-matrix adhesion molecules propelled invasive cell membrane protrusions, which in turn promoted fusogenic protein engagement and plasma membrane fusion. This de novo cell fusion culture system reveals a general role for actin-propelled invasive membrane protrusions in driving fusogenic protein engagement during cell-cell fusion.
Collapse
Affiliation(s)
- Khurts Shilagardi
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gildor B, Schejter ED, Shilo BZ. Bidirectional Notch activation represses fusion competence in swarming adult Drosophila myoblasts. Development 2012; 139:4040-50. [PMID: 23048185 DOI: 10.1242/dev.077495] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A major aspect of indirect flight muscle formation during adult Drosophila myogenesis involves transition of a semi-differentiated and proliferating pool of myoblasts to a mature myoblast population, capable of fusing with nascent myotubes and generating mature muscle fibers. Here we examine the molecular genetic programs underlying these two phases of myoblast differentiation. We show that the cell adhesion proteins Dumbfounded (Duf) and Sticks and stones (Sns), together with their paralogs Roughest (Rst) and Hibris (Hbs), respectively, are required for adhesion of migrating myoblasts to myotubes and initiation of myoblast-myotube fusion. As myoblasts approach their myotube targets, they are maintained in a semi-differentiated state by continuous Notch activation, where each myoblast provides the ligand Delta to its neighbors. This unique form of bidirectional Notch activation is achieved by finely tuning the levels of the ligand and receptor. Activation of Notch signaling in myoblasts represses expression of key fusion elements such as Sns. Only upon reaching the vicinity of the myotubes does Notch signaling decay, leading to terminal differentiation of the myoblasts. The ensuing induction of proteins required for fusion enables myoblasts to fuse with the myotubes and give rise to subsequent muscle fiber growth.
Collapse
Affiliation(s)
- Boaz Gildor
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | |
Collapse
|
38
|
Bonn BR, Rudolf A, Hornbruch-Freitag C, Daum G, Kuckwa J, Kastl L, Buttgereit D, Renkawitz-Pohl R. Myosin heavy chain-like localizes at cell contact sites during Drosophila myoblast fusion and interacts in vitro with Rolling pebbles 7. Exp Cell Res 2012; 319:402-16. [PMID: 23246571 DOI: 10.1016/j.yexcr.2012.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 12/04/2012] [Accepted: 12/05/2012] [Indexed: 11/29/2022]
Abstract
Besides representing the sarcomeric thick filaments, myosins are involved in many cellular transport and motility processes. Myosin heavy chains are grouped into 18 classes. Here we show that in Drosophila, the unconventional group XVIII myosin heavy chain-like (Mhcl) is transcribed in the mesoderm of embryos, most prominently in founder cells (FCs). An ectopically expressed GFP-tagged Mhcl localizes in the growing muscle at cell-cell contacts towards the attached fusion competent myoblast (FCM). We further show that Mhcl interacts in vitro with the essential fusion protein Rolling pebbles 7 (Rols7), which is part of a protein complex established at cell contact sites (Fusion-restricted Myogenic-Adhesive Structure or FuRMAS). Here, branched F-actin is likely needed to widen the fusion pore and to integrate the myoblast into the growing muscle. We show that the localization of Mhcl is dependent on the presence of Rols7, and we postulate that Mhcl acts at the FuRMAS as an actin motor protein. We further show that Mhcl deficient embryos develop a wild-type musculature. We thus propose that Mhcl functions redundantly to other myosin heavy chains in myoblasts. Lastly, we found that the protein is detectable adjacent to the sarcomeric Z-discs, suggesting an additional function in mature muscles.
Collapse
Affiliation(s)
- Bettina R Bonn
- Developmental Biology, Department of Biology, Philipps-Universität Marburg, Karl-von-Frisch-Strasse 8, 35037 Marburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Duan R, Jin P, Luo F, Zhang G, Anderson N, Chen EH. Group I PAKs function downstream of Rac to promote podosome invasion during myoblast fusion in vivo. ACTA ACUST UNITED AC 2012; 199:169-85. [PMID: 23007650 PMCID: PMC3461515 DOI: 10.1083/jcb.201204065] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Group I p21-activated kinases organize actin filaments in myoblasts into dense foci, which promote podosome invasion and subsequent myoblast fusion. The p21-activated kinases (PAKs) play essential roles in diverse cellular processes and are required for cell proliferation, apoptosis, polarity establishment, migration, and cell shape changes. Here, we have identified a novel function for the group I PAKs in cell–cell fusion. We show that the two Drosophila group I PAKs, DPak3 and DPak1, have partially redundant functions in myoblast fusion in vivo, with DPak3 playing a major role. DPak3 is enriched at the site of fusion colocalizing with the F-actin focus within a podosome-like structure (PLS), and promotes actin filament assembly during PLS invasion. Although the small GTPase Rac is involved in DPak3 activation and recruitment to the PLS, the kinase activity of DPak3 is required for effective PLS invasion. We propose a model whereby group I PAKs act downstream of Rac to organize the actin filaments within the PLS into a dense focus, which in turn promotes PLS invasion and fusion pore initiation during myoblast fusion.
Collapse
Affiliation(s)
- Rui Duan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
40
|
Kaipa BR, Shao H, Schäfer G, Trinkewitz T, Groth V, Liu J, Beck L, Bogdan S, Abmayr SM, Önel SF. Dock mediates Scar- and WASp-dependent actin polymerization through interaction with cell adhesion molecules in founder cells and fusion-competent myoblasts. J Cell Sci 2012; 126:360-72. [PMID: 22992459 DOI: 10.1242/jcs.113860] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The formation of the larval body wall musculature of Drosophila depends on the asymmetric fusion of two myoblast types, founder cells (FCs) and fusion-competent myoblasts (FCMs). Recent studies have established an essential function of Arp2/3-based actin polymerization during myoblast fusion, formation of a dense actin focus at the site of fusion in FCMs, and a thin sheath of actin in FCs and/or growing muscles. The formation of these actin structures depends on recognition and adhesion of myoblasts that is mediated by cell surface receptors of the immunoglobulin superfamily. However, the connection of the cell surface receptors with Arp2/3-based actin polymerization is poorly understood. To date only the SH2-SH3 adaptor protein Crk has been suggested to link cell adhesion with Arp2/3-based actin polymerization in FCMs. Here, we propose that the SH2-SH3 adaptor protein Dock, like Crk, links cell adhesion with actin polymerization. We show that Dock is expressed in FCs and FCMs and colocalizes with the cell adhesion proteins Sns and Duf at cell-cell contact points. Biochemical data in this study indicate that different domains of Dock are involved in binding the cell adhesion molecules Duf, Rst, Sns and Hbs. We emphasize the importance of these interactions by quantifying the enhanced myoblast fusion defects in duf dock, sns dock and hbs dock double mutants. Additionally, we show that Dock interacts biochemically and genetically with Drosophila Scar, Vrp1 and WASp. Based on these data, we propose that Dock links cell adhesion in FCs and FCMs with either Scar- or Vrp1-WASp-dependent Arp2/3 activation.
Collapse
Affiliation(s)
- Balasankara Reddy Kaipa
- Fachbereich Biologie, Entwicklungsbiologie, Philipps-Universität Marburg, Karl-von-Frisch Str. 8, D-35043 Marburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Powell GT, Wright GJ. Genomic organisation, embryonic expression and biochemical interactions of the zebrafish junctional adhesion molecule family of receptors. PLoS One 2012; 7:e40810. [PMID: 22815827 PMCID: PMC3399880 DOI: 10.1371/journal.pone.0040810] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 06/13/2012] [Indexed: 12/17/2022] Open
Abstract
The mammalian JAM family is composed of three cell surface receptors. Interactions between the proteins have well-characterised roles in inflammation and tight junction formation, but little is known about their function in early development. Recently, we identified a role for jamb and jamc in zebrafish myocyte fusion. Genome duplication in the teleost lineage raised the possibility that additional JAM family paralogues may also function in muscle development. To address this, we searched the zebrafish genome to identify potential paralogues and confirmed their homology, bringing the total number of zebrafish jam family members to six. We then compared the physical binding properties of each paralogue by surface plasmon resonance and determined the gene expression patterns of all zebrafish jam genes at different stages of development. Our results suggest a significant sub-functionalisation of JAM-B and JAM-C orthologues with respect to binding strength (but not specificity) and gene expression. The paralogous genes, jamb2 and jamc2, were not detected in the somites or myotome of wild-type embryos. We conclude that it is unlikely that the paralogues have a function in primary myogenesis.
Collapse
Affiliation(s)
- Gareth T. Powell
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- * E-mail: (GTP); (GJW)
| | - Gavin J. Wright
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- * E-mail: (GTP); (GJW)
| |
Collapse
|
42
|
Do muscle founder cells exist in vertebrates? Trends Cell Biol 2012; 22:391-6. [PMID: 22710008 DOI: 10.1016/j.tcb.2012.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 05/17/2012] [Accepted: 05/21/2012] [Indexed: 11/21/2022]
Abstract
Skeletal muscle is formed by the iterative fusion of precursor cells (myocytes) into long multinuclear fibres. Extensive studies of fusion in Drosophila embryos have lead to a paradigm in which myoblasts are divided into two distinct subtypes - founder and fusion-competent myoblasts (FCMs) - that can fuse to each other, but not among themselves. Only founder cells can direct the formation of muscle fibres, while FCMs act as a cellular substrate. Recent studies in zebrafish and mice have demonstrated conservation of the molecules originally identified in Drosophila, but an important question remains: is vertebrate fusion regulated by specifying myocyte subtypes? Stated simply: do vertebrate founder cells exist? In light of recent findings, we argue that a different regulatory mechanism has evolved in vertebrates.
Collapse
|
43
|
Abstract
The fusion of myoblasts into multinucleate syncytia plays a fundamental role in muscle function, as it supports the formation of extended sarcomeric arrays, or myofibrils, within a large volume of cytoplasm. Principles learned from the study of myoblast fusion not only enhance our understanding of myogenesis, but also contribute to our perspectives on membrane fusion and cell-cell fusion in a wide array of model organisms and experimental systems. Recent studies have advanced our views of the cell biological processes and crucial proteins that drive myoblast fusion. Here, we provide an overview of myoblast fusion in three model systems that have contributed much to our understanding of these events: the Drosophila embryo; developing and regenerating mouse muscle; and cultured rodent muscle cells.
Collapse
Affiliation(s)
- Susan M Abmayr
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.
| | | |
Collapse
|
44
|
Powell GT, Wright GJ. Jamb and jamc are essential for vertebrate myocyte fusion. PLoS Biol 2011; 9:e1001216. [PMID: 22180726 PMCID: PMC3236736 DOI: 10.1371/journal.pbio.1001216] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 10/27/2011] [Indexed: 12/02/2022] Open
Abstract
Jamb and Jamc are an essential cell surface receptor pair that interact to drive fusion between muscle precursor cells during zebrafish development. Cellular fusion is required in the development of several tissues, including skeletal muscle. In vertebrates, this process is poorly understood and lacks an in vivo-validated cell surface heterophilic receptor pair that is necessary for fusion. Identification of essential cell surface interactions between fusing cells is an important step in elucidating the molecular mechanism of cellular fusion. We show here that the zebrafish orthologues of JAM-B and JAM-C receptors are essential for fusion of myocyte precursors to form syncytial muscle fibres. Both jamb and jamc are dynamically co-expressed in developing muscles and encode receptors that physically interact. Heritable mutations in either gene prevent myocyte fusion in vivo, resulting in an overabundance of mononuclear, but otherwise overtly normal, functional fast-twitch muscle fibres. Transplantation experiments show that the Jamb and Jamc receptors must interact between neighbouring cells (in trans) for fusion to occur. We also show that jamc is ectopically expressed in prdm1a mutant slow muscle precursors, which inappropriately fuse with other myocytes, suggesting that control of myocyte fusion through regulation of jamc expression has important implications for the growth and patterning of muscles. Our discovery of a receptor-ligand pair critical for fusion in vivo has important implications for understanding the molecular mechanisms responsible for myocyte fusion and its regulation in vertebrate myogenesis. The fusion of precursor cells is a crucial step in many biological processes, one of which is the development of skeletal muscle. The molecular and cell biology of fusion of muscle precursors has been well described in Drosophila melanogaster larvae, leading to insights into the process in vertebrates. However, the identity and mechanism of action of essential cell surface proteins for fusion between vertebrate muscle precursors has previously been lacking. Here, we describe a vertebrate-specific cell surface receptor pair that is essential for fusion in zebrafish: Jamb and Jamc. Loss of function of either receptor causes a near-complete block in fusion, resulting in an overabundance of mononucleate muscle fibres that are otherwise overtly normal. We demonstrate that Jamb and Jamc physically interact and are co-expressed by muscle precursors. Moreover, we show that the interaction between them is essential for fusion between neighbouring precursors in an embryo. We hypothesise that binding of Jamb to Jamc is a necessary recognition and adhesion step permissive for, but not sufficient to cause, myocyte fusion. Knowledge of these molecular components in vertebrates will lead to better understanding of how fusion is controlled to pattern skeletal muscle tissue.
Collapse
Affiliation(s)
- Gareth T. Powell
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Gavin J. Wright
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW The functioning kidney requires proper organization in multiple cell types that mediate filtration and removal of wastes. Interest has increasingly focused on the podocyte as an important mediator of kidney function; defects in podocyte function likely mediate a broad palate of kidney dysfunctions. Here I explore recent work that establishes the Drosophila nephrocyte as a useful model for podocyte function and dysfunction. RECENT FINDINGS Although described many decades in the past, recent evidence has emphasized important similarities in the molecules that construct the 'nephrocyte diaphragm' and its vertebrate cousin the 'podocyte diaphragm'. For example, loss of Nephrin and its associated proteins lead to collapse of these structures and loss of proper filtration. SUMMARY These data emphasize both differences between the podocyte and nephrocyte and also useful similarities. These similarities provide the promise of bringing Drosophila genetics--strongly successful in other disciplines--to the complex problem of how podocyte dysfunction leads to disease. To further explore this point I discuss work on Nephrin in a better understood tissue, the Drosophila eye, in which the role of Nephrin and its connection to actin dynamics is under intense study.
Collapse
|
46
|
Hornbruch-Freitag C, Griemert B, Buttgereit D, Renkawitz-Pohl R. Drosophila Swiprosin-1/EFHD2 accumulates at the prefusion complex stage during Drosophila myoblast fusion. J Cell Sci 2011; 124:3266-78. [PMID: 21896648 DOI: 10.1242/jcs.083907] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In the Drosophila embryo, transient cell adhesion during myoblast fusion is known to lead to the formation of fusion-restricted myogenic-adhesive structures (FuRMASs). Here, we report that within these FuRMASs, a Drosophila homologue of human and mouse swiprosins (EF-hand-domain-containing proteins) is expressed, which we named Drosophila Swiprosin-1 (Drosophila Swip-1). Drosophila Swip-1 is highly conserved and is closely related to the calcium-binding proteins swiprosin-1 and swiprosin-2 that have a role in the immune system in humans and mice. Our study shows that Drosophila Swip-1 is also expressed in corresponding cells of the Drosophila immune system. During myoblast fusion, Drosophila Swip-1 accumulates transiently in the foci of fusion-competent myoblasts (FCMs). Both the EF-hand and the coiled-coil domain of Drosophila Swip-1 are required to localise the protein to these foci. The formation of Drosophila Swip-1 foci requires successful cell adhesion between FCMs and founder cells (FCs) or growing myotubes. Moreover, Drosophila Swip-1 foci were found to increase in number in sing(22) mutants, which arrest myoblast fusion after prefusion complex formation. By contrast, Drosophila Swip-1 foci are not significantly enriched in blow(2) and kette(J4-48) mutants, which stop myogenesis beyond the prefusion complex stage but before plasma membrane merging. Therefore, we hypothesise that Drosophila Swip-1 participates in the breakdown of the prefusion complex during the progression of myoblast fusion.
Collapse
|
47
|
Functional and spatial analysis of C. elegans SYG-1 and SYG-2, orthologs of the Neph/nephrin cell adhesion module directing selective synaptogenesis. PLoS One 2011; 6:e23598. [PMID: 21858180 PMCID: PMC3156230 DOI: 10.1371/journal.pone.0023598] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 07/20/2011] [Indexed: 01/25/2023] Open
Abstract
The assembly of specific synaptic connections represents a prime example of cellular recognition. Members of the Ig superfamily are among the most ancient proteins represented in the genomes of both mammalian and invertebrate organisms, where they constitute a trans-synaptic adhesion system. The correct connectivity patterns of the highly conserved immunoglobulin superfamily proteins nephrin and Neph1 are crucial for the assembly of functional neuronal circuits and the formation of the kidney slit diaphragm, a synapse-like structure forming the filtration barrier. Here, we utilize the nematode C. elegans model for studying the requirements of synaptic specificity mediated by nephrin-Neph proteins. In C. elegans, the nephrin/Neph1 orthologs SYG-2 and SYG-1 form intercellular contacts strictly in trans between epithelial guidepost cells and neurons specifying the localization of synapses. We demonstrate a functional conservation between mammalian nephrin and SYG-2. Expression of nephrin effectively compensated loss of syg-2 function in C. elegans and restored defective synaptic connectivity further establishing the C. elegans system as a valuable model for slit diaphragm proteins. Next, we investigated the effect of SYG-1 and SYG-2 trans homodimerization respectively. Strikingly, synapse assembly could be induced by homophilic SYG-1 but not SYG-2 binding indicating a critical role of SYG-1 intracellular signalling for morphogenetic events and pointing toward the dynamic and stochastic nature of extra- and intracellular nephrin-Neph interactions to generate reproducible patterns of synaptic connectivity.
Collapse
|
48
|
Mukherjee P, Gildor B, Shilo BZ, VijayRaghavan K, Schejter ED. The actin nucleator WASp is required for myoblast fusion during adult Drosophila myogenesis. Development 2011; 138:2347-57. [PMID: 21558381 DOI: 10.1242/dev.055012] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Myoblast fusion provides a fundamental, conserved mechanism for muscle fiber growth. We demonstrate here that the functional contribution of Wsp, the Drosophila homolog of the conserved actin nucleation-promoting factor (NPF) WASp, is essential for myoblast fusion during the formation of muscles of the adult fly. Disruption of Wsp function results in complete arrest of myoblast fusion in all muscles examined. Wsp activity during adult Drosophila myogenesis is specifically required for muscle cell fusion and is crucial both for the formation of new muscle fibers and for the growth of muscles derived from persistent larval templates. Although Wsp is expressed both in fibers and individual myoblasts, its activity in either one of these cell types is sufficient. SCAR, a second major Arp2/3 NPF, is also required during adult myoblast fusion. Formation of fusion-associated actin 'foci' is dependent on Arp2/3 complex function, but appears to rely on a distinct, unknown nucleator. The comprehensive nature of these requirements identifies Arp2/3-based branched actin polymerization as a universal mechanism underlying myoblast fusion.
Collapse
Affiliation(s)
- Priyankana Mukherjee
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | | | | | | | | |
Collapse
|
49
|
Takemura M, Adachi-Yamada T. Cell death and selective adhesion reorganize the dorsoventral boundary for zigzag patterning of Drosophila wing margin hairs. Dev Biol 2011; 357:336-46. [PMID: 21781959 DOI: 10.1016/j.ydbio.2011.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Revised: 06/30/2011] [Accepted: 07/05/2011] [Indexed: 11/17/2022]
Abstract
Animal tissues and organs are comprised of several types of cells, which are often arranged in a well-ordered pattern. The posterior part of the Drosophila wing margin is covered with a double row of long hairs, which are equally and alternately derived from the dorsal and ventral sides of the wing, exhibiting a zigzag pattern in the lateral view. How this geometrically regular pattern is formed has not been fully understood. In this study, we show that this zigzag pattern is created by rearrangement of wing margin cells along the dorsoventral boundary flanked by the double row of hair cells during metamorphosis. This cell rearrangement is induced by selective apoptosis of wing margin cells that are spatially separated from hair cells. As a result of apoptosis, the remaining wing margin cells are rearranged in a well-ordered manner, which shapes corrugated lateral sides of both dorsal and ventral edges to interlock them for zigzag patterning. We further show that the corrugated topology of the wing edges is achieved by cell-type specific expression and localization of four kinds of NEPH1/nephrin family proteins through heterophilic adhesion between wing margin cells and hair cells. Homophilic E-cadherin adhesion is also required for attachment of the corrugated dorsoventral edges. Taken together, our results demonstrate that sequential coordination of apoptosis and epithelial architecture with selective adhesion creates the zigzag hair alignment. This may be a common mechanism for geometrically ordered repetitive packing of several types of cells in similarly patterned developmental fields such as the mammalian organ of Corti.
Collapse
Affiliation(s)
- Masahiko Takemura
- Department of Biology, Graduate School of Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | | |
Collapse
|
50
|
Haralalka S, Shelton C, Cartwright HN, Katzfey E, Janzen E, Abmayr SM. Asymmetric Mbc, active Rac1 and F-actin foci in the fusion-competent myoblasts during myoblast fusion in Drosophila. Development 2011; 138:1551-62. [PMID: 21389053 DOI: 10.1242/dev.057653] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Myoblast fusion is an intricate process that is initiated by cell recognition and adhesion, and culminates in cell membrane breakdown and formation of multinucleate syncytia. In the Drosophila embryo, this process occurs asymmetrically between founder cells that pattern the musculature and fusion-competent myoblasts (FCMs) that account for the bulk of the myoblasts. The present studies clarify and amplify current models of myoblast fusion in several important ways. We demonstrate that the non-conventional guanine nucleotide exchange factor (GEF) Mbc plays a fundamental role in the FCMs, where it functions to activate Rac1, but is not required in the founder cells for fusion. Mbc, active Rac1 and F-actin foci are highly enriched in the FCMs, where they localize to the Sns:Kirre junction. Furthermore, Mbc is crucial for the integrity of the F-actin foci and the FCM cytoskeleton, presumably via its activation of Rac1 in these cells. Finally, the local asymmetric distribution of these proteins at adhesion sites is reminiscent of invasive podosomes and, consistent with this model, they are enriched at sites of membrane deformation, where the FCM protrudes into the founder cell/myotube. These data are consistent with models promoting actin polymerization as the driving force for myoblast fusion.
Collapse
Affiliation(s)
- Shruti Haralalka
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | | | | | | | | |
Collapse
|