1
|
Avilés EC, Wang SK, Patel S, Cordero S, Shi S, Lin L, Kefalov VJ, Goodrich LV, Cepko CL, Xue Y. ERG responses to high-frequency flickers require FAT3 signaling in mouse retinal bipolar cells. J Gen Physiol 2025; 157:e202413642. [PMID: 39903280 PMCID: PMC11793021 DOI: 10.1085/jgp.202413642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/17/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Vision is initiated by the reception of light by photoreceptors and subsequent processing via downstream retinal neurons. Proper circuit organization depends on the multifunctional tissue polarity protein FAT3, which is required for amacrine cell connectivity and retinal lamination. Here, we investigated the retinal function of Fat3 mutant mice and found decreases in both electroretinography and perceptual responses to high-frequency flashes. These defects did not correlate with abnormal amacrine cell wiring, pointing instead to a role in bipolar cell subtypes that also express FAT3. The role of FAT3 in the response to high temporal frequency flashes depends upon its ability to transduce an intracellular signal. Mechanistically, FAT3 binds to the synaptic protein PTPσ intracellularly and is required to localize GRIK1 to OFF-cone bipolar cell synapses with cone photoreceptors. These findings expand the repertoire of FAT3's functions and reveal its importance in bipolar cells for high-frequency light response.
Collapse
Affiliation(s)
- Evelyn C. Avilés
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sean K. Wang
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Sarina Patel
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Sebastian Cordero
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Shuxiang Shi
- Lingang Laboratory, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lucas Lin
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Vladimir J. Kefalov
- Gavin Herbert Eye Institute and Center for Translational Vision Research, University of California, Irvine, Irvine, CA, USA
| | - Lisa V. Goodrich
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Constance L. Cepko
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Yunlu Xue
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Lingang Laboratory, Shanghai, China
| |
Collapse
|
2
|
Majeed M, Liao CP, Hobert O. Nervous system-wide analysis of all C. elegans cadherins reveals neuron-specific functions across multiple anatomical scales. SCIENCE ADVANCES 2025; 11:eads2852. [PMID: 39983000 PMCID: PMC11844738 DOI: 10.1126/sciadv.ads2852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/22/2025] [Indexed: 02/23/2025]
Abstract
Differential expression of cell adhesion proteins is a hallmark of cell-type diversity across the animal kingdom. Gene family-wide characterization of their organismal expression and function is, however, lacking. Using genome-engineered reporter alleles, we established an atlas of expression of the entire set of 12 cadherin gene family members in the nematode Caenorhabditis elegans, revealing differential expression across neuronal classes, a dichotomy between broadly and narrowly expressed cadherins, and several context-dependent temporal transitions in expression across development. Engineered mutant null alleles of cadherins were analyzed for defects in morphology, behavior, neuronal soma positions, neurite neighborhood topology and fasciculation, and localization of synapses in many parts of the nervous system. This analysis revealed a restricted pattern of neuronal differentiation defects at discrete subsets of anatomical scales, including a novel role of cadherins in experience-dependent electrical synapse formation. In total, our analysis results in previously little explored perspectives on cadherin deployment and function.
Collapse
Affiliation(s)
| | - Chien-Po Liao
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Bhatt A, Ruffine V, Töpfer U, Ryu J, Fischer-Friedrich E, Dahmann C. The WIRS motifs in Fat2 are required for Drosophila egg chamber rotation but not for elongation. Development 2025; 152:DEV204201. [PMID: 39823598 PMCID: PMC11829772 DOI: 10.1242/dev.204201] [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: 06/27/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
The elongation of tissues and organs is important for proper morphogenesis in animal development. In Drosophila ovaries, the elongation of egg chambers involves aligned Collagen IV fiber-like structures, a gradient of extracellular matrix stiffness and actin-based protrusion-driven collective cell migration, leading to the rotation of the egg chamber. Egg chamber elongation and rotation depend on the atypical cadherin Fat2. Fat2 contains in its intracellular region three WRC interacting receptor sequence (WIRS) motifs, which previously had been shown to bind to the WAVE regulatory complex (WRC), a conserved actin regulator. Here, we show that in fat2 mutant flies lacking the WIRS motifs, egg chambers fail to rotate and Collagen IV fiber-like structures are impaired, yet a gradient of extracellular matrix stiffness is established and egg chambers properly elongate. We conclude that the WIRS motifs are required for egg chamber rotation and that egg chamber rotation might be a prerequisite for proper formation of Collagen IV fiber-like structures. Egg chamber rotation, however, is dispensable for extracellular matrix stiffness gradient formation and for egg chamber elongation.
Collapse
Affiliation(s)
- Akanksha Bhatt
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
| | - Valentin Ruffine
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany
| | - Uwe Töpfer
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
| | - Jinhee Ryu
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
| | | | - Christian Dahmann
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany
| |
Collapse
|
4
|
Töpfer U, Ryu J, Guerra Santillán KY, Schulze J, Fischer-Friedrich E, Tanentzapf G, Dahmann C. AdamTS proteases control basement membrane heterogeneity and organ shape in Drosophila. Cell Rep 2024; 43:114399. [PMID: 38944833 DOI: 10.1016/j.celrep.2024.114399] [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: 11/15/2023] [Revised: 05/07/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024] Open
Abstract
The basement membrane (BM) is an extracellular matrix that plays important roles in animal development. A spatial heterogeneity in composition and structural properties of the BM provide cells with vital cues for morphogenetic processes such as cell migration or cell polarization. Here, using the Drosophila egg chamber as a model system, we show that the BM becomes heterogeneous during development, with a reduction in Collagen IV density at the posterior pole and differences in the micropattern of aligned fiber-like structures. We identified two AdamTS matrix proteases required for the proper elongated shape of the egg chamber, yet the molecular mechanisms by which they act are different. Stall is required to establish BM heterogeneity by locally limiting Collagen IV protein density, whereas AdamTS-A alters the micropattern of fiber-like structures within the BM at the posterior pole. Our results suggest that AdamTS proteases control BM heterogeneity required for organ shape.
Collapse
Affiliation(s)
- Uwe Töpfer
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; School of Science, Technische Universität Dresden, 01062 Dresden, Germany.
| | - Jinhee Ryu
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
| | - Karla Yanín Guerra Santillán
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany
| | - Jana Schulze
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
| | - Elisabeth Fischer-Friedrich
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany; Biotechnology Center, Technische Universität Dresden, 01062 Dresden, Germany
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Christian Dahmann
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
5
|
Vien KM, Duan Q, Yeung C, Barish S, Volkan PC. Atypical cadherin, Fat2, regulates axon terminal organization in the developing Drosophila olfactory receptor neurons. iScience 2024; 27:110340. [PMID: 39055932 PMCID: PMC11269957 DOI: 10.1016/j.isci.2024.110340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/08/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
The process of how neuronal identity confers circuit organization is intricately related to the mechanisms underlying neurodegeneration and neuropathologies. Modeling this process, the olfactory circuit builds a functionally organized topographic map, which requires widely dispersed neurons with the same identity to converge their axons into one a class-specific neuropil, a glomerulus. In this article, we identified Fat2 (also known as Kugelei) as a regulator of class-specific axon organization. In fat2 mutants, axons belonging to the highest fat2-expressing classes present with a more severe phenotype compared to axons belonging to low fat2-expressing classes. In extreme cases, mutations lead to neural degeneration. Lastly, we found that Fat2 intracellular domain interactors, APC1/2 (Adenomatous polyposis coli) and dop (Drop out), likely orchestrate the cytoskeletal remodeling required for axon condensation. Altogether, we provide a potential mechanism for how cell surface proteins' regulation of cytoskeletal remodeling necessitates identity specific circuit organization.
Collapse
Affiliation(s)
- Khanh M. Vien
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Qichen Duan
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Chun Yeung
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Scott Barish
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Pelin Cayirlioglu Volkan
- Department of Biology, Duke University, Durham, NC 27708, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
6
|
Avilés EC, Wang SK, Patel S, Shi S, Lin L, Kefalov VJ, Goodrich LV, Cepko CL, Xue Y. High temporal frequency light response in mouse retina requires FAT3 signaling in bipolar cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.02.565326. [PMID: 37961274 PMCID: PMC10635074 DOI: 10.1101/2023.11.02.565326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Vision is initiated by the reception of light by photoreceptors and subsequent processing via downstream retinal neurons. Proper cellular organization depends on the multi-functional tissue polarity protein FAT3, which is required for amacrine cell connectivity and retinal lamination. Here we investigated the retinal function of Fat3 mutant mice and found decreases in physiological and perceptual responses to high frequency flashes. These defects did not correlate with abnormal amacrine cell wiring, pointing instead to a role in bipolar cell subtypes that also express FAT3. The role of FAT3 in the response to high temporal frequency flashes depends upon its ability to transduce an intracellular signal. Mechanistically, FAT3 binds to the synaptic protein PTPσ, intracellularly, and is required to localize GRIK1 to OFF-cone bipolar cell synapses with cone photoreceptors. These findings expand the repertoire of FAT3's functions and reveal its importance in bipolar cells for high frequency light response.
Collapse
Affiliation(s)
- Evelyn C. Avilés
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
- Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Sean K. Wang
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Boston, MA 02115
| | - Sarina Patel
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Shuxiang Shi
- Lingang Laboratory, Shanghai, China, 200031
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China, 201210
| | - Lucas Lin
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Vladimir J. Kefalov
- Gavin Herbert Eye Institute & Center for Translational Vision Research, University of California, Irvine, CA 92697
| | - Lisa V. Goodrich
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Constance L. Cepko
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Boston, MA 02115
| | - Yunlu Xue
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
- Lingang Laboratory, Shanghai, China, 200031
- Lead contact
| |
Collapse
|
7
|
Yue T, Guo Y, Qi X, Zheng W, Zhang H, Wang B, Liu K, Zhou B, Zeng X, Ouzhuluobu, He Y, Su B. Sex-biased regulatory changes in the placenta of native highlanders contribute to adaptive fetal development. eLife 2024; 12:RP89004. [PMID: 38869160 PMCID: PMC11175615 DOI: 10.7554/elife.89004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Compared with lowlander migrants, native Tibetans have a higher reproductive success at high altitude though the underlying mechanism remains unclear. Here, we compared the transcriptome and histology of full-term placentas between native Tibetans and Han migrants. We found that the placental trophoblast shows the largest expression divergence between Tibetans and Han, and Tibetans show decreased immune response and endoplasmic reticulum stress. Remarkably, we detected a sex-biased expression divergence, where the male-infant placentas show a greater between-population difference than the female-infant placentas. The umbilical cord plays a key role in the sex-biased expression divergence, which is associated with the higher birth weight of the male newborns of Tibetans. We also identified adaptive histological changes in the male-infant placentas of Tibetans, including larger umbilical artery wall and umbilical artery intima and media, and fewer syncytial knots. These findings provide valuable insights into the sex-biased adaptation of human populations, with significant implications for medical and genetic studies of human reproduction.
Collapse
Affiliation(s)
- Tian Yue
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Yongbo Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang HospitalKunmingChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
| | - Wangshan Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
| | - Hui Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
| | - Bin Wang
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang HospitalKunmingChina
| | - Kai Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
| | - Bin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Xuerui Zeng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Ouzhuluobu
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang HospitalKunmingChina
| | - Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of SciencesKunmingChina
| |
Collapse
|
8
|
Williams AM, Horne-Badovinac S. Fat2 polarizes Lar and Sema5c to coordinate the motility of collectively migrating epithelial cells. J Cell Sci 2024; 137:jcs261173. [PMID: 37593878 PMCID: PMC10508692 DOI: 10.1242/jcs.261173] [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: 03/16/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023] Open
Abstract
Migrating epithelial cells globally align their migration machinery to achieve tissue-level movement. Biochemical signaling across leading-trailing cell-cell interfaces can promote this alignment by partitioning migratory behaviors like protrusion and retraction to opposite sides of the interface. However, how signaling proteins become organized at interfaces to accomplish this is poorly understood. The follicular epithelial cells of Drosophila melanogaster have two signaling modules at their leading-trailing interfaces - one composed of the atypical cadherin Fat2 (also known as Kugelei) and the receptor tyrosine phosphatase Lar, and one composed of Semaphorin5c and its receptor Plexin A. Here, we show that these modules form one interface signaling system with Fat2 at its core. Trailing edge-enriched Fat2 concentrates both Lar and Semaphorin5c at leading edges of cells, but Lar and Semaphorin5c play little role in the localization of Fat2. Fat2 is also more stable at interfaces than Lar or Semaphorin5c. Once localized, Lar and Semaphorin5c act in parallel to promote collective migration. We propose that Fat2 serves as the organizer of this interface signaling system by coupling and polarizing the distributions of multiple effectors that work together to align the migration machinery of neighboring cells.
Collapse
Affiliation(s)
- Audrey Miller Williams
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
9
|
Happel L, Voigt A. Coordinated Motion of Epithelial Layers on Curved Surfaces. PHYSICAL REVIEW LETTERS 2024; 132:078401. [PMID: 38427891 DOI: 10.1103/physrevlett.132.078401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/21/2023] [Indexed: 03/03/2024]
Abstract
Coordinated cellular movements are key processes in tissue morphogenesis. Using a cell-based modeling approach we study the dynamics of epithelial layers lining surfaces with constant and varying curvature. We demonstrate that extrinsic curvature effects can explain the alignment of cell elongation with the principal directions of curvature. Together with specific self-propulsion mechanisms and cell-cell interactions this effect gets enhanced and can explain observed large-scale, persistent, and circumferential rotation on cylindrical surfaces. On toroidal surfaces the resulting curvature coupling is an interplay of intrinsic and extrinsic curvature effects. These findings unveil the role of curvature and postulate its importance for tissue morphogenesis.
Collapse
Affiliation(s)
- L Happel
- Institute of Scientific Computing, TU Dresden, 01062 Dresden, Germany
| | - A Voigt
- Institute of Scientific Computing, TU Dresden, 01062 Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstr. 108, 01307 Dresden, Germany
- Cluster of Excellence, Physics of Life, TU Dresden, Arnoldstr. 18, 01307 Dresden, Germany
| |
Collapse
|
10
|
Berg C, Sieber M, Sun J. Finishing the egg. Genetics 2024; 226:iyad183. [PMID: 38000906 PMCID: PMC10763546 DOI: 10.1093/genetics/iyad183] [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: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/26/2023] Open
Abstract
Gamete development is a fundamental process that is highly conserved from early eukaryotes to mammals. As germ cells develop, they must coordinate a dynamic series of cellular processes that support growth, cell specification, patterning, the loading of maternal factors (RNAs, proteins, and nutrients), differentiation of structures to enable fertilization and ensure embryonic survival, and other processes that make a functional oocyte. To achieve these goals, germ cells integrate a complex milieu of environmental and developmental signals to produce fertilizable eggs. Over the past 50 years, Drosophila oogenesis has risen to the forefront as a system to interrogate the sophisticated mechanisms that drive oocyte development. Studies in Drosophila have defined mechanisms in germ cells that control meiosis, protect genome integrity, facilitate mRNA trafficking, and support the maternal loading of nutrients. Work in this system has provided key insights into the mechanisms that establish egg chamber polarity and patterning as well as the mechanisms that drive ovulation and egg activation. Using the power of Drosophila genetics, the field has begun to define the molecular mechanisms that coordinate environmental stresses and nutrient availability with oocyte development. Importantly, the majority of these reproductive mechanisms are highly conserved throughout evolution, and many play critical roles in the development of somatic tissues as well. In this chapter, we summarize the recent progress in several key areas that impact egg chamber development and ovulation. First, we discuss the mechanisms that drive nutrient storage and trafficking during oocyte maturation and vitellogenesis. Second, we examine the processes that regulate follicle cell patterning and how that patterning impacts the construction of the egg shell and the establishment of embryonic polarity. Finally, we examine regulatory factors that control ovulation, egg activation, and successful fertilization.
Collapse
Affiliation(s)
- Celeste Berg
- Department of Genome Sciences, University of Washington, Seattle, WA 98195-5065 USA
| | - Matthew Sieber
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jianjun Sun
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269 USA
| |
Collapse
|
11
|
Hernández-López C, Puliafito A, Xu Y, Lu Z, Di Talia S, Vergassola M. Two-fluid dynamics and micron-thin boundary layers shape cytoplasmic flows in early Drosophila embryos. Proc Natl Acad Sci U S A 2023; 120:e2302879120. [PMID: 37878715 PMCID: PMC10622894 DOI: 10.1073/pnas.2302879120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/22/2023] [Indexed: 10/27/2023] Open
Abstract
Cytoplasmic flows are widely emerging as key functional players in development. In early Drosophila embryos, flows drive the spreading of nuclei across the embryo. Here, we combine hydrodynamic modeling with quantitative imaging to develop a two-fluid model that features an active actomyosin gel and a passive viscous cytosol. Gel contractility is controlled by the cell cycle oscillator, the two fluids being coupled by friction. In addition to recapitulating experimental flow patterns, our model explains observations that remained elusive and makes a series of predictions. First, the model captures the vorticity of cytosolic flows, which highlights deviations from Stokes' flow that were observed experimentally but remained unexplained. Second, the model reveals strong differences in the gel and cytosol motion. In particular, a micron-sized boundary layer is predicted close to the cortex, where the gel slides tangentially while the cytosolic flow cannot slip. Third, the model unveils a mechanism that stabilizes the spreading of nuclei with respect to perturbations of their initial positions. This self-correcting mechanism is argued to be functionally important for proper nuclear spreading. Fourth, we use our model to analyze the effects of flows on the transport of the morphogen Bicoid and the establishment of its gradients. Finally, the model predicts that the flow strength should be reduced if the shape of the domain is more round, which is experimentally confirmed in Drosophila mutants. Thus, our two-fluid model explains flows and nuclear positioning in early Drosophila, while making predictions that suggest novel future experiments.
Collapse
Affiliation(s)
| | | | - Yitong Xu
- Department of Cell Biology, Duke University Medical Center, Durham, NC27710
| | - Ziqi Lu
- Department of Cell Biology, Duke University Medical Center, Durham, NC27710
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC27710
| | - Massimo Vergassola
- Department of Physics, École Normale Supérieure, Paris75005, France
- Department of Physics, University of California, San Diego, CA92075
| |
Collapse
|
12
|
Oyagi S, Nakamura R, Katsuno T, Sogami T, Kawai Y, Kishimoto Y, Omori K. Local coordination of epithelial planar polarity in the maintenance and regeneration of the adult rat airway. Cell Tissue Res 2023; 394:163-175. [PMID: 37460682 DOI: 10.1007/s00441-023-03809-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/05/2023] [Indexed: 10/07/2023]
Abstract
The maintenance of planar polarity in airway multiciliated cells (MCCs) has been poorly characterized. We recently reported that the direction of ciliary beating in a surgically inverted tracheal segment remained inverted beyond the time required for the turnover of cells, without adjustment to global distal-to-proximal polarity. We hypothesized that the local maintenance of tissue-level polarity occurs via locally reproduced cells. To provide further insight regarding this hypothetical property, we performed allotransplantation of an inverted tracheal segment between wild-type (donor) and tdTomato-expressing (host) rats, with and without scratching the mucosa of the transplants. The origin of cells in the transplants was assessed using tdTomato-specific immunostaining. Ciliary movement and structures were observed by high-speed video and electron microscopy to analyze MCC orientations. Variabilities in the orientations of closely and distantly located MCCs were analyzed to evaluate the local- and broad-scale coordination of polarity, respectively. The epithelium was maintained by donor-derived cells in the non-scratched inverted transplant over 6 months, beyond one cycle of turnover. The inverted orientation of MCCs was also maintained throughout the non-scratched transplant. MCCs regenerated in the scratched transplant were derived from the host and exhibited diverse orientations across the transplant. However, the orientations of adjacent regenerated MCCs were often coordinated, indicating that airway MCCs can locally coordinate their orientations. A steady-state airway may maintain MCC orientation by locally reproducing MCCs via the local coordination of polarity. This local coordination enables the formation and maintenance of tissue-level polarity in small regions after mucosal injury.
Collapse
Affiliation(s)
- Seiji Oyagi
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Min-iren Chuo Hospital, Kyoto, Japan
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryosuke Nakamura
- Department of Rehabilitation Medicine, New York University Grossman School of Medicine, New York, USA
| | - Tatsuya Katsuno
- Center of Anatomical, Pathological and Forensic Medical Researches, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tohru Sogami
- Department of Otolaryngology-Head and Neck Surgery, SOSEIKAI hospital, Kyoto, Japan
| | - Yoshitaka Kawai
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
13
|
Bai Y, Zhao F, Wu T, Chen F, Pang X. Actin polymerization and depolymerization in developing vertebrates. Front Physiol 2023; 14:1213668. [PMID: 37745245 PMCID: PMC10515290 DOI: 10.3389/fphys.2023.1213668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Development is a complex process that occurs throughout the life cycle. F-actin, a major component of the cytoskeleton, is essential for the morphogenesis of tissues and organs during development. F-actin is formed by the polymerization of G-actin, and the dynamic balance of polymerization and depolymerization ensures proper cellular function. Disruption of this balance results in various abnormalities and defects or even embryonic lethality. Here, we reviewed recent findings on the structure of G-actin and F-actin and the polymerization of G-actin to F-actin. We also focused on the functions of actin isoforms and the underlying mechanisms of actin polymerization/depolymerization in cellular and organic morphogenesis during development. This information will extend our understanding of the role of actin polymerization in the physiologic or pathologic processes during development and may open new avenues for developing therapeutics for embryonic developmental abnormalities or tissue regeneration.
Collapse
Affiliation(s)
- Yang Bai
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Feng Zhao
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingting Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Fangchun Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xiaoxiao Pang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
14
|
Messer CL, McDonald JA. Expect the unexpected: conventional and unconventional roles for cadherins in collective cell migration. Biochem Soc Trans 2023; 51:1495-1504. [PMID: 37387360 DOI: 10.1042/bst20221202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/25/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023]
Abstract
Migrating cell collectives navigate complex tissue environments both during normal development and in pathological contexts such as tumor invasion and metastasis. To do this, cells in collectives must stay together but also communicate information across the group. The cadherin superfamily of proteins mediates junctional adhesions between cells, but also serve many essential functions in collective cell migration. Besides keeping migrating cell collectives cohesive, cadherins help follower cells maintain their attachment to leader cells, transfer information about front-rear polarity among the cohort, sense and respond to changes in the tissue environment, and promote intracellular signaling, in addition to other cellular behaviors. In this review, we highlight recent studies that reveal diverse but critical roles for both classical and atypical cadherins in collective cell migration, specifically focusing on four in vivo model systems in development: the Drosophila border cells, zebrafish mesendodermal cells, Drosophila follicle rotation, and Xenopus neural crest cells.
Collapse
Affiliation(s)
- C Luke Messer
- Division of Biology, Kansas State University, Manhattan, KS 66502, U.S.A
| | - Jocelyn A McDonald
- Division of Biology, Kansas State University, Manhattan, KS 66502, U.S.A
| |
Collapse
|
15
|
Menin L, Weber J, Villa S, Martini E, Maspero E, Niño CA, Cancila V, Poli A, Maiuri P, Palamidessi A, Frittoli E, Bianchi F, Tripodo C, Walters KJ, Giavazzi F, Scita G, Polo S. A planar polarized MYO6-DOCK7-RAC1 axis promotes tissue fluidification in mammary epithelia. Cell Rep 2023; 42:113001. [PMID: 37590133 PMCID: PMC10530600 DOI: 10.1016/j.celrep.2023.113001] [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: 03/17/2023] [Revised: 04/24/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
Tissue fluidification and collective motility are pivotal in regulating embryonic morphogenesis, wound healing, and tumor metastasis. These processes frequently require that each cell constituent of a tissue coordinates its migration activity and directed motion through the oriented extension of lamellipodium cell protrusions, promoted by RAC1 activity. While the upstream RAC1 regulators in individual migratory cells or leader cells during invasion or wound healing are well characterized, how RAC1 is controlled in follower cells remains unknown. Here, we identify a MYO6-DOCK7 axis essential for spatially restricting RAC1 activity in a planar polarized fashion in model tissue monolayers. The MYO6-DOCK7 axis specifically controls the extension of cryptic lamellipodia required to drive tissue fluidification and cooperative-mode motion in otherwise solid and static carcinoma cell collectives.
Collapse
Affiliation(s)
- Luca Menin
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Janine Weber
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Stefano Villa
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Segrate, Italy
| | - Emanuele Martini
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Elena Maspero
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Carlos A Niño
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Valeria Cancila
- Human Pathology Section, Department of Health Sciences, University of Palermo School of Medicine, Palermo, Italy
| | - Alessandro Poli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Paolo Maiuri
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | | | - Fabrizio Bianchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Claudio Tripodo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy; Human Pathology Section, Department of Health Sciences, University of Palermo School of Medicine, Palermo, Italy
| | - Kylie J Walters
- Protein Processing Section, Center for Structural Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Fabio Giavazzi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Segrate, Italy
| | - Giorgio Scita
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy; Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy.
| | - Simona Polo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy; Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
16
|
Campanale JP, Montell DJ. Who's really in charge: Diverse follower cell behaviors in collective cell migration. Curr Opin Cell Biol 2023; 81:102160. [PMID: 37019053 PMCID: PMC10744998 DOI: 10.1016/j.ceb.2023.102160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 04/05/2023]
Abstract
Collective cell migrations drive morphogenesis, wound healing, and cancer dissemination. Cells located at the front are considered leaders while those behind them are defined topologically as followers. Leader cell behaviors, including chemotaxis and their coupling to followers, have been well-studied and reviewed. However, the contributions of follower cells to collective cell migration represent an emerging area of interest. In this perspective, we highlight recent research into the broadening array of follower cell behaviors found in moving collectives. We describe examples of follower cells that possess cryptic leadership potential and followers that lack that potential but contribute in diverse and sometimes surprising ways to collective movement, even steering from behind. We highlight collectives in which all cells both lead and follow, and a few passive passengers. The molecular mechanisms controlling follower cell function and behavior are just emerging and represent an exciting frontier in collective cell migration research.
Collapse
Affiliation(s)
- Joseph P Campanale
- Molecular, Cellular and Developmental Biology, University of California Santa Barbara
| | - Denise J Montell
- Molecular, Cellular and Developmental Biology, University of California Santa Barbara.
| |
Collapse
|
17
|
López CH, Puliafito A, Xu Y, Lu Z, Di Talia S, Vergassola M. Two-fluid dynamics and micron-thin boundary layers shape cytoplasmic flows in early Drosophila embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532979. [PMID: 36993669 PMCID: PMC10055070 DOI: 10.1101/2023.03.16.532979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Cytoplasmic flows are widely emerging as key functional players in development. In early Drosophila embryos, flows drive the spreading of nuclei across the embryo. Here, we combine hydrodynamic modeling with quantitative imaging to develop a two-fluid model that features an active actomyosin gel and a passive viscous cytosol. Gel contractility is controlled by the cell cycle oscillator, the two fluids being coupled by friction. In addition to recapitulating experimental flow patterns, our model explains observations that remained elusive, and makes a series of new predictions. First, the model captures the vorticity of cytosolic flows, which highlights deviations from Stokes' flow that were observed experimentally but remained unexplained. Second, the model reveals strong differences in the gel and cytosol motion. In particular, a micron-sized boundary layer is predicted close to the cortex, where the gel slides tangentially whilst the cytosolic flow cannot slip. Third, the model unveils a mechanism that stabilizes the spreading of nuclei with respect to perturbations of their initial positions. This self-correcting mechanism is argued to be functionally important for proper nuclear spreading. Fourth, we use our model to analyze the effects of flows on the transport of the morphogen Bicoid, and the establishment of its gradients. Finally, the model predicts that the flow strength should be reduced if the shape of the domain is more round, which is experimentally confirmed in Drosophila mutants. Thus, our two-fluid model explains flows and nuclear positioning in early Drosophila, while making predictions that suggest novel future experiments.
Collapse
Affiliation(s)
| | - Alberto Puliafito
- Department of Oncology, University of Turin, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Str. Prov. 142, km 3.95, 10060 Candiolo, Italy
| | - Yitong Xu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA and
| | - Ziqi Lu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA and
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA and
| | - Massimo Vergassola
- École Normale Supérieure, 75005 Paris, France
- Department of Physics, University of California San Diego, San Diego, CA 92075, USA
| |
Collapse
|
18
|
Williams AM, Horne-Badovinac S. Fat2 polarizes Lar and Sema5c to coordinate the motility of collectively migrating epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.28.530349. [PMID: 36909523 PMCID: PMC10002635 DOI: 10.1101/2023.02.28.530349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Migrating epithelial cells globally align their migration machinery to achieve tissue-level movement. Biochemical signaling across leading-trailing cell-cell interfaces can promote this alignment by partitioning migratory behaviors like protrusion and retraction to opposite sides of the interface. However, how the necessary signaling proteins become organized at this site is poorly understood. The follicular epithelial cells of Drosophila melanogaster have two signaling modules at their leading-trailing interfaces-one composed of the atypical cadherin Fat2 and the receptor tyrosine phosphatase Lar, and one composed of Semaphorin 5c and its receptor Plexin A. Here we show that these modules form one interface signaling system with Fat2 at its core. Trailing edge-enriched Fat2 concentrates both Lar and Sema5c at cells' leading edges, likely by slowing their turnover at this site. Once localized, Lar and Sema5c act in parallel to promote collective migration. Our data suggest a model in which Fat2 couples and polarizes the distributions of multiple effectors that work together to align the migration machinery of neighboring cells.
Collapse
Affiliation(s)
- Audrey Miller Williams
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
Curvature gradient drives polarized tissue flow in the Drosophila embryo. Proc Natl Acad Sci U S A 2023; 120:e2214205120. [PMID: 36724258 PMCID: PMC9963527 DOI: 10.1073/pnas.2214205120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Tissue flow during morphogenesis is commonly driven by local constriction of cell cortices, which is caused by the activation of actomyosin contractility. This can lead to long-range flows due to tissue viscosity. However, in the absence of cell-intrinsic polarized forces or polarity in forces external to the tissue, these flows must be symmetric and centered around the region of contraction. Polarized tissue flows have been previously demonstrated to arise from the coupling of such contractile flows to points of increased friction or adhesion to external structures. However, we show with experiments and modeling that the onset of polarized tissue flow in early Drosophila morphogenesis occurs independent of adhesion and is instead driven by a geometric coupling of apical actomyosin contractility to tissue curvature. Particularly, the onset of polarized flow is driven by a mismatch between the position of apical myosin activation and the position of peak curvature at the posterior pole of the embryo. Our work demonstrates how genetic and geometric information inherited from the mother interact to create polarized flow during embryo morphogenesis.
Collapse
|
20
|
Ruiz-Whalen DM, Aichele CP, Dyson ER, Gallen KC, Stark JV, Saunders JA, Simonet JC, Ventresca EM, Fuentes IM, Marmol N, Moise E, Neubert BC, Riggs DJ, Self AM, Alexander JI, Boamah E, Browne AJ, Correa I, Foster MJ, Harrington N, Holiday TJ, Henry RA, Lee EH, Longo SM, Lorenz LD, Martinez E, Nikonova A, Radu M, Smith SC, Steele LA, Strochlic TI, Archer NF, Aykit YJ, Bolotsky AJ, Boyle M, Criollo J, Eldor O, Cruz G, Fortuona VN, Gounder SD, Greenwood N, Ji KW, Johnson A, Lara S, Montanez B, Saurman M, Singh T, Smith DR, Stapf CA, Tondapu T, Tsiobikas C, Habas R, O'Reilly AM. Gaining Wings to FLY: Using Drosophila Oogenesis as an Entry Point for Citizen Scientists in Laboratory Research. Methods Mol Biol 2023; 2626:399-444. [PMID: 36715918 DOI: 10.1007/978-1-0716-2970-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Citizen science is a productive approach to include non-scientists in research efforts that impact particular issues or communities. In most cases, scientists at advanced career stages design high-quality, exciting projects that enable citizen contribution, a crowdsourcing process that drives discovery forward and engages communities. The challenges of having citizens design their own research with no or limited training and providing access to laboratory tools, reagents, and supplies have limited citizen science efforts. This leaves the incredible life experiences and immersion of citizens in communities that experience health disparities out of the research equation, thus hampering efforts to address community health needs with a full picture of the challenges that must be addressed. Here, we present a robust and reproducible approach that engages participants from Grade 5 through adult in research focused on defining how diet impacts disease signaling. We leverage the powerful genetics, cell biology, and biochemistry of Drosophila oogenesis to define how nutrients impact phenotypes associated with genetic mutants that are implicated in cancer and diabetes. Participants lead the project design and execution, flipping the top-down hierarchy of the prevailing scientific culture to co-create research projects and infuse the research with cultural and community relevance.
Collapse
Affiliation(s)
- Dara M Ruiz-Whalen
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
- eCLOSE Institute, Huntingdon Valley, PA, USA.
| | - Christopher P Aichele
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Ebony R Dyson
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Katherine C Gallen
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Jennifer V Stark
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jasmine A Saunders
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jacqueline C Simonet
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- Arcadia University, Glenside, PA, USA
| | - Erin M Ventresca
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- Albright College, Reading, PA, USA
| | - Isabela M Fuentes
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Nyellis Marmol
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Emly Moise
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Benjamin C Neubert
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Devon J Riggs
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Ava M Self
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jennifer I Alexander
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Ernest Boamah
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Amanda J Browne
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Iliana Correa
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Maya J Foster
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Nicole Harrington
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Troy J Holiday
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ryan A Henry
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- Wilkes University, Wilkes-Barre, PA, USA
| | - Eric H Lee
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Sheila M Longo
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Laurel D Lorenz
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Esteban Martinez
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Anna Nikonova
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Maria Radu
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Shannon C Smith
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lindsay A Steele
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Todd I Strochlic
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, PA, USA
| | - Nicholas F Archer
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Y James Aykit
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Adam J Bolotsky
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Megan Boyle
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jennifer Criollo
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Oren Eldor
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gabriela Cruz
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Valerie N Fortuona
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Shreeya D Gounder
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Nyim Greenwood
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Kayla W Ji
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aminah Johnson
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- eCLOSE Institute, Huntingdon Valley, PA, USA
| | - Sophie Lara
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Maxwell Saurman
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Tanu Singh
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniel R Smith
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Catherine A Stapf
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Tarang Tondapu
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Raymond Habas
- Department of Biology, Temple University, Philadelphia, PA, USA
| | - Alana M O'Reilly
- Immersion Science Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
- eCLOSE Institute, Huntingdon Valley, PA, USA.
| |
Collapse
|
21
|
Kasiah J, McNeill H. Fat and Dachsous cadherins in mammalian development. Curr Top Dev Biol 2023; 154:223-244. [PMID: 37100519 DOI: 10.1016/bs.ctdb.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Cell growth and patterning are critical for tissue development. Here we discuss the evolutionarily conserved cadherins, Fat and Dachsous, and the roles they play during mammalian tissue development and disease. In Drosophila, Fat and Dachsous regulate tissue growth via the Hippo pathway and planar cell polarity (PCP). The Drosophila wing has been an ideal tissue to observe how mutations in these cadherins affect tissue development. In mammals, there are multiple Fat and Dachsous cadherins, which are expressed in many tissues, but mutations in these cadherins that affect growth and tissue organization are context dependent. Here we examine how mutations in the Fat and Dachsous mammalian genes affect development in mammals and contribute to human disease.
Collapse
Affiliation(s)
- Jennysue Kasiah
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States.
| |
Collapse
|
22
|
Collective cell migration during optic cup formation features changing cell-matrix interactions linked to matrix topology. Curr Biol 2022; 32:4817-4831.e9. [PMID: 36208624 DOI: 10.1016/j.cub.2022.09.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/28/2022] [Accepted: 09/16/2022] [Indexed: 11/22/2022]
Abstract
Cell migration is crucial for organismal development and shapes organisms in health and disease. Although a lot of research has revealed the role of intracellular components and extracellular signaling in driving single and collective cell migration, the influence of physical properties of the tissue and the environment on migration phenomena in vivo remains less explored. In particular, the role of the extracellular matrix (ECM), which many cells move upon, is currently unclear. To overcome this gap, we use zebrafish optic cup formation, and by combining novel transgenic lines and image analysis pipelines, we study how ECM properties influence cell migration in vivo. We show that collectively migrating rim cells actively move over an immobile extracellular matrix. These cell movements require cryptic lamellipodia that are extended in the direction of migration. Quantitative analysis of matrix properties revealed that the topology of the matrix changes along the migration path. These changes in matrix topologies are accompanied by changes in the dynamics of cell-matrix interactions. Experiments and theoretical modeling suggest that matrix porosity could be linked to efficient migration. Indeed, interfering with matrix topology by increasing its porosity results in a loss of cryptic lamellipodia, less-directed cell-matrix interactions, and overall inefficient migration. Thus, matrix topology is linked to the dynamics of cell-matrix interactions and the efficiency of directed collective rim cell migration during vertebrate optic cup morphogenesis.
Collapse
|
23
|
Williams AM, Donoughe S, Munro E, Horne-Badovinac S. Fat2 polarizes the WAVE complex in trans to align cell protrusions for collective migration. eLife 2022; 11:e78343. [PMID: 36154691 PMCID: PMC9576270 DOI: 10.7554/elife.78343] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/11/2022] [Indexed: 11/13/2022] Open
Abstract
For a group of cells to migrate together, each cell must couple the polarity of its migratory machinery with that of the other cells in the cohort. Although collective cell migrations are common in animal development, little is known about how protrusions are coherently polarized among groups of migrating epithelial cells. We address this problem in the collective migration of the follicular epithelial cells in Drosophila melanogaster. In this epithelium, the cadherin Fat2 localizes to the trailing edge of each cell and promotes the formation of F-actin-rich protrusions at the leading edge of the cell behind. We show that Fat2 performs this function by acting in trans to concentrate the activity of the WASP family verprolin homolog regulatory complex (WAVE complex) at one long-lived region along each cell's leading edge. Without Fat2, the WAVE complex distribution expands around the cell perimeter and fluctuates over time, and protrusive activity is reduced and unpolarized. We further show that Fat2's influence is very local, with sub-micron-scale puncta of Fat2 enriching the WAVE complex in corresponding puncta just across the leading-trailing cell-cell interface. These findings demonstrate that a trans interaction between Fat2 and the WAVE complex creates stable regions of protrusive activity in each cell and aligns the cells' protrusions across the epithelium for directionally persistent collective migration.
Collapse
Affiliation(s)
- Audrey Miller Williams
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicagoUnited States
| | - Seth Donoughe
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicagoUnited States
| | - Edwin Munro
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicagoUnited States
- Committee on Development, Regeneration, and Stem Cell Biology, University of ChicagoChicagoUnited States
- Institute for Biophysical Dynamics, University of ChicagoChicagoUnited States
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicagoUnited States
- Committee on Development, Regeneration, and Stem Cell Biology, University of ChicagoChicagoUnited States
| |
Collapse
|
24
|
Gu Z, Yao Y, Yang G, Zhu G, Tian Z, Wang R, Wu Q, Wang Y, Wu Y, Chen L, Wang C, Gao J, Kang X, Zhang J, Wang L, Duan S, Zhao Z, Zhang Z, Sun S. Pharmacogenomic landscape of head and neck squamous cell carcinoma informs precision oncology therapy. Sci Transl Med 2022; 14:eabo5987. [PMID: 36070368 DOI: 10.1126/scitranslmed.abo5987] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common and frequently lethal cancer with few therapeutic options. In particular, there are few effective targeted therapies. Development of highly effective therapeutic strategies tailored to patients with HNSCC remains a pressing challenge. To address this, we present a pharmacogenomic study to facilitate precision treatments for patients with HNSCC. We established a large collection of 56 HNSCC patient-derived cells (PDCs), which recapitulated the molecular features of the original tumors. Pharmacological assessment of HNSCCs was conducted using a three-tiered high-throughput drug screening using 2248 compounds across these PDC models and an additional 18 immortalized cell lines. We integrated genomic, transcriptomic, and pharmacological analysis to predict biomarkers, gene-drug associations, and validated biomarkers. These results supported drug repurposing for multiple HNSCC subtypes, including the JAK2 inhibitor fedratinib, for low KRT18-expressing HNSCC cases, and the topoisomerase inhibitor mitoxantrone, for IL6R-activated HNSCC cases. Our results demonstrated concordance between susceptibility predictions from the PDCs and the matched patients' responses to standard clinical medication. Moreover, we identified and experimentally confirmed that high expression of ITGB1 elicited therapeutic resistance to docetaxel and high SOD1 expression conferred resistance to afatinib. We further validated ITGB1 as a predictive biomarker for the efficacy of docetaxel therapy in a phase 2 clinical trial. In summary, our study shows that this HNSCC cell resource, as well as the resulting pharmacogenomic profiles, is effective for biomarker discovery and for guiding precision oncology therapies in HNSCCs.
Collapse
Affiliation(s)
- Ziyue Gu
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yanli Yao
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Guizhu Yang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Guopei Zhu
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.,Department of Oral and Maxillofacial-Head Neck Oncology, Division of Radiation Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhen Tian
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.,Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Rui Wang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Qi Wu
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yujue Wang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yaping Wu
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lan Chen
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Chong Wang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiamin Gao
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Xindan Kang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jie Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lizhen Wang
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.,Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shengzhong Duan
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.,Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Shuyang Sun
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| |
Collapse
|
25
|
Roberto GM, Emery G. Directing with restraint: Mechanisms of protrusion restriction in collective cell migrations. Semin Cell Dev Biol 2022; 129:75-81. [PMID: 35397972 DOI: 10.1016/j.semcdb.2022.03.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 01/15/2023]
Abstract
Cell migration is necessary for morphogenesis, tissue homeostasis, wound healing and immune response. It is also involved in diseases. In particular, cell migration is inherent in metastasis. Cells can migrate individually or in groups. To migrate efficiently, cells need to be able to organize into a leading front that protrudes by forming membrane extensions and a trailing edge that contracts. This organization is scaled up at the group level during collective cell movements. If a cell or a group of cells is unable to limit its leading edge and hence to restrict the formation of protrusions to the front, directional movements are impaired or abrogated. Here we summarize our current understanding of the mechanisms restricting protrusion formation in collective cell migration. We focus on three in vivo examples: the neural crest cell migration, the rotatory migration of follicle cells around the Drosophila egg chamber and the border cell migration during oogenesis.
Collapse
Affiliation(s)
- Gabriela Molinari Roberto
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Downtown station, Montréal, Québec H3C 3J7, Canada
| | - Gregory Emery
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Downtown station, Montréal, Québec H3C 3J7, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada.
| |
Collapse
|
26
|
Donoughe S. Insect egg morphology: evolution, development, and ecology. CURRENT OPINION IN INSECT SCIENCE 2022; 50:100868. [PMID: 34973433 DOI: 10.1016/j.cois.2021.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
The insect egg can be viewed through many lenses: it is the single-celled developmental stage, a resource investment in the next generation, an unusually large and complex cell type, and the protective vessel for embryonic development. In this review, I describe the morphological diversity of insect eggs and then identify recent advances in understanding the patterns of egg evolution, the cellular mechanisms underlying egg development, and notable aspects of egg ecology. I also suggest areas for particularly promising future research on insect egg morphology; these topics touch upon diverse areas such as tissue morphogenesis, life history evolution, organismal scaling, cellular secretion, and oviposition ecology.
Collapse
Affiliation(s)
- Seth Donoughe
- Department of Molecular Genetics and Cell Biology, University of Chicago, IL, USA.
| |
Collapse
|
27
|
Avilés EC, Krol A, Henle SJ, Burroughs-Garcia J, Deans MR, Goodrich LV. Fat3 acts through independent cytoskeletal effectors to coordinate asymmetric cell behaviors during polarized circuit assembly. Cell Rep 2022; 38:110307. [PMID: 35108541 PMCID: PMC8865054 DOI: 10.1016/j.celrep.2022.110307] [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: 08/31/2021] [Revised: 11/23/2021] [Accepted: 01/06/2022] [Indexed: 02/01/2023] Open
Abstract
The polarized flow of information through neural circuits depends on the orderly arrangement of neurons, their processes, and their synapses. This polarity emerges sequentially in development, starting with the directed migration of neuronal precursors, which subsequently elaborate neurites that form synapses in specific locations. In other organs, Fat cadherins sense the position and then polarize individual cells by inducing localized changes in the cytoskeleton that are coordinated across the tissue. Here, we show that the Fat-related protein Fat3 plays an analogous role during the assembly of polarized circuits in the murine retina. We find that the Fat3 intracellular domain (ICD) binds to cytoskeletal regulators and synaptic proteins, with discrete motifs required for amacrine cell migration and neurite retraction. Moreover, upon ICD deletion, extra neurites form but do not make ectopic synapses, suggesting that Fat3 independently regulates synapse localization. Thus, Fat3 serves as a molecular node to coordinate asymmetric cell behaviors across development.
Collapse
Affiliation(s)
- Evelyn C Avilés
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra Krol
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven J Henle
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica Burroughs-Garcia
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Michael R Deans
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
28
|
Töpfer U, Guerra Santillán KY, Fischer-Friedrich E. Stiffness Measurement of Drosophila Egg Chambers by Atomic Force Microscopy. Methods Mol Biol 2022; 2540:301-315. [PMID: 35980585 DOI: 10.1007/978-1-0716-2541-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drosophila egg chamber development requires cellular and molecular mechanisms controlling morphogenesis. Previous research has shown that the mechanical properties of the basement membrane contribute to tissue elongation of the egg chamber. Here, we discuss how indentation with the microindenter of an atomic force microscope can be used to determine an effective stiffness value of a Drosophila egg chamber. We provide information on the preparation of egg chambers prior to the measurement, dish coating, the actual atomic force microscope measurement process, and data analysis. Furthermore, we discuss how to interpret acquired data and which mechanical components are expected to influence measured stiffness values.
Collapse
Affiliation(s)
- Uwe Töpfer
- Institute of Genetics, Technische Universität Dresden, Dresden, Germany
| | - Karla Yanín Guerra Santillán
- Institute of Genetics, Technische Universität Dresden, Dresden, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Elisabeth Fischer-Friedrich
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany.
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
29
|
In search of conserved principles of planar cell polarization. Curr Opin Genet Dev 2021; 72:69-81. [PMID: 34871922 DOI: 10.1016/j.gde.2021.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 11/06/2021] [Accepted: 11/10/2021] [Indexed: 01/26/2023]
Abstract
The making of an embryo and its internal organs entails the spatial coordination of cellular activities. This manifests during tissue morphogenesis as cells change shape, rearrange and divide along preferential axis and during cell differentiation. Cells live in a polarized field and respond to it by polarizing their cellular activities in the plane of the tissue by a phenomenon called planar cell polarization. This phenomenon is ubiquitous in animals and depends on a few conserved planar cell polarity (PCP) pathways. All PCP pathways share two essential characteristics: the existence of local interactions between protein complexes present at the cell surface leading to their asymmetric distribution within cells; a supracellular graded cue that aligns these cellular asymmetries at the tissue level. Here, we discuss the potential common principles of planar cell polarization by comparing the local and global mechanisms employed by the different PCP pathways identified so far. The focus of the review is on the logic of the system rather than the molecules per se.
Collapse
|
30
|
Sherrard KM, Cetera M, Horne-Badovinac S. DAAM mediates the assembly of long-lived, treadmilling stress fibers in collectively migrating epithelial cells in Drosophila. eLife 2021; 10:e72881. [PMID: 34812144 PMCID: PMC8610420 DOI: 10.7554/elife.72881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Stress fibers (SFs) are actomyosin bundles commonly found in individually migrating cells in culture. However, whether and how cells use SFs to migrate in vivo or collectively is largely unknown. Studying the collective migration of the follicular epithelial cells in Drosophila, we found that the SFs in these cells show a novel treadmilling behavior that allows them to persist as the cells migrate over multiple cell lengths. Treadmilling SFs grow at their fronts by adding new integrin-based adhesions and actomyosin segments over time. This causes the SFs to have many internal adhesions along their lengths, instead of adhesions only at the ends. The front-forming adhesions remain stationary relative to the substrate and typically disassemble as the cell rear approaches. By contrast, a different type of adhesion forms at the SF's terminus that slides with the cell's trailing edge as the actomyosin ahead of it shortens. We further show that SF treadmilling depends on cell movement and identify a developmental switch in the formins that mediate SF assembly, with Dishevelled-associated activator of morphogenesis acting during migratory stages and Diaphanous acting during postmigratory stages. We propose that treadmilling SFs keep each cell on a linear trajectory, thereby promoting the collective motility required for epithelial migration.
Collapse
Affiliation(s)
- Kristin M Sherrard
- Department of Molecular Genetics and Cell Biology, The University of ChicagoChicagoUnited States
| | - Maureen Cetera
- Committee on Development, Regeneration, and Stem Cell Biology, The University of ChicagoChicagoUnited States
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, The University of ChicagoChicagoUnited States
- Committee on Development, Regeneration, and Stem Cell Biology, The University of ChicagoChicagoUnited States
| |
Collapse
|
31
|
Jędrzejowska I, Christophoryová J, Garbiec A. Small body size of pseudoscorpions and a distinct architecture of the ovary: A step to miniaturization? J Anat 2021; 239:1182-1195. [PMID: 34131910 PMCID: PMC8546526 DOI: 10.1111/joa.13485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/18/2021] [Accepted: 05/27/2021] [Indexed: 11/29/2022] Open
Abstract
Chelicerata, the second largest subphylum of Arthropoda, includes invertebrates with a wide range of body size. Pseudoscorpions are among small or miniature chelicerates which exhibit several morphological, anatomical, and developmental features related to miniaturization, e.g., replacement of book lungs by tracheae, unpaired gonads, and matrotrophic development of the embryos outside the female body, in the brood sac. In this paper, we show the ovary structure of two pseudoscorpion species, Cheiridium museorum and Apocheiridium ferum (Cheiridiidae). Both cheiridiids are one of the smallest pseudoscorpions. The results of our observations conducted in light, transmission electron, and confocal microscopy demonstrate that the ovary of C. museorum and A. ferum, displays a significant structural difference that is unusual for chelicerates. The difference concerns the spatially restricted position of the germarium. We show that such ovary architecture results in a significantly reduced number of growing oocytes and in consequence a reduced number of deposited eggs. A centrally located germarium implies also a modified pattern of ovary development during oocyte growth due to long distance migration of the germline and the accompanying somatic cells. Herein, we postulate that such an ovary structure is related to the pseudoscorpion's small body size and it is a step towards miniaturization in the smaller pseudoscorpions species.
Collapse
Affiliation(s)
- Izabela Jędrzejowska
- Department of Animal Developmental BiologyFaculty of Biological SciencesUniversity of WrocławWrocławPoland
| | - Jana Christophoryová
- Department of ZoologyFaculty of Natural SciencesComenius UniversityBratislavaSlovakia
| | - Arnold Garbiec
- Department of Animal Developmental BiologyFaculty of Biological SciencesUniversity of WrocławWrocławPoland
| |
Collapse
|
32
|
Physics of liquid crystals in cell biology. Trends Cell Biol 2021; 32:140-150. [PMID: 34756501 DOI: 10.1016/j.tcb.2021.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022]
Abstract
The past decade has witnessed a rapid growth in understanding of the pivotal roles of mechanical stresses and physical forces in cell biology. As a result, an integrated view of cell biology is evolving, where genetic and molecular features are scrutinised hand in hand with physical and mechanical characteristics of cells. Physics of liquid crystals has emerged as a burgeoning new frontier in cell biology over the past few years, fuelled by an increasing identification of orientational order and topological defects in cell biology, spanning scales from subcellular filaments to individual cells and multicellular tissues. Here, we provide an account of the most recent findings and developments, together with future promises and challenges in this rapidly evolving interdisciplinary research direction.
Collapse
|
33
|
Tiwari P, Rengarajan H, Saunders TE. Scaling of internal organs during Drosophila embryonic development. Biophys J 2021; 120:4264-4276. [PMID: 34087212 PMCID: PMC8516638 DOI: 10.1016/j.bpj.2021.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 11/20/2022] Open
Abstract
Many species show a diverse range of sizes; for example, domestic dogs have large variation in body mass. Yet, the internal structure of the organism remains similar, i.e., the system scales to organism size. Drosophila melanogaster has been a powerful model system for exploring scaling mechanisms. In the early embryo, gene expression boundaries scale very precisely to embryo length. Later in development, the adult wings grow with remarkable symmetry and scale well with animal size. Yet, our knowledge of whether internal organs initially scale to embryo size remains largely unknown. Here, we utilize artificially small Drosophila embryos to explore how three critical internal organs-the heart, hindgut, and ventral nerve cord (VNC)-adapt to changes in embryo morphology. We find that the heart scales precisely with embryo length. Intriguingly, reduction in cardiac cell length, rather than number, appears to be important in controlling heart length. The hindgut, which is the first chiral organ to form, displays scaling with embryo size under large-scale changes in the artificially smaller embryos but shows few hallmarks of scaling within wild-type size variation. Finally, the VNC only displays weak scaling behavior; even large changes in embryo geometry result in only small shifts in VNC length. This suggests that the VNC may have an intrinsic minimal length that is largely independent of embryo length. Overall, our work shows that internal organs can adapt to embryo size changes in Drosophila, but the extent to which they scale varies significantly between organs.
Collapse
Affiliation(s)
- Prabhat Tiwari
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | | | - Timothy E Saunders
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore, Singapore; Institute of Molecular and Cell Biology, A(∗)Star, Singapore, Singapore; Warwick Medical School, University of Warwick, Coventry, United Kingdom.
| |
Collapse
|
34
|
Basta LP, Hill-Oliva M, Paramore SV, Sharan R, Goh A, Biswas A, Cortez M, Little KA, Posfai E, Devenport D. New mouse models for high resolution and live imaging of planar cell polarity proteins in vivo. Development 2021; 148:271988. [PMID: 34463728 DOI: 10.1242/dev.199695] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/24/2021] [Indexed: 01/10/2023]
Abstract
The collective polarization of cellular structures and behaviors across a tissue plane is a near universal feature of epithelia known as planar cell polarity (PCP). This property is controlled by the core PCP pathway, which consists of highly conserved membrane-associated protein complexes that localize asymmetrically at cell junctions. Here, we introduce three new mouse models for investigating the localization and dynamics of transmembrane PCP proteins: Celsr1, Fz6 and Vangl2. Using the skin epidermis as a model, we characterize and verify the expression, localization and function of endogenously tagged Celsr1-3xGFP, Fz6-3xGFP and tdTomato-Vangl2 fusion proteins. Live imaging of Fz6-3xGFP in basal epidermal progenitors reveals that the polarity of the tissue is not fixed through time. Rather, asymmetry dynamically shifts during cell rearrangements and divisions, while global, average polarity of the tissue is preserved. We show using super-resolution STED imaging that Fz6-3xGFP and tdTomato-Vangl2 can be resolved, enabling us to observe their complex localization along junctions. We further explore PCP fusion protein localization in the trachea and neural tube, and discover new patterns of PCP expression and localization throughout the mouse embryo.
Collapse
Affiliation(s)
- Lena P Basta
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Michael Hill-Oliva
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA.,Department of Medicine, Columbia University, New York, NY 10032USA
| | - Sarah V Paramore
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Rishabh Sharan
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Audrey Goh
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Abhishek Biswas
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA.,Research Computing, Office of Information Technology, Princeton University, Princeton, NJ 08544, USA
| | - Marvin Cortez
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Katherine A Little
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544USA
| |
Collapse
|
35
|
Bischoff MC, Bogdan S. Collective cell migration driven by filopodia-New insights from the social behavior of myotubes. Bioessays 2021; 43:e2100124. [PMID: 34480489 DOI: 10.1002/bies.202100124] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 01/12/2023]
Abstract
Collective migration is a key process that is critical during development, as well as in physiological and pathophysiological processes including tissue repair, wound healing and cancer. Studies in genetic model organisms have made important contributions to our current understanding of the mechanisms that shape cells into different tissues during morphogenesis. Recent advances in high-resolution and live-cell-imaging techniques provided new insights into the social behavior of cells based on careful visual observations within the context of a living tissue. In this review, we will compare Drosophila testis nascent myotube migration with established in vivo model systems, elucidate similarities, new features and principles in collective cell migration.
Collapse
Affiliation(s)
- Maik C Bischoff
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University Marburg, Marburg, Germany
| | - Sven Bogdan
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
36
|
Jain S, Ladoux B, Mège RM. Mechanical plasticity in collective cell migration. Curr Opin Cell Biol 2021; 72:54-62. [PMID: 34134013 DOI: 10.1016/j.ceb.2021.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/19/2023]
Abstract
Collective cell migration is crucial to maintain epithelium integrity during developmental and repair processes. It requires a tight regulation of mechanical coordination between neighboring cells. This coordination embraces different features including mechanical self-propulsion of individual cells within cellular colonies and large-scale force transmission through cell-cell junctions. This review discusses how the plasticity of biomechanical interactions at cell-cell contacts could help cellular systems to perform coordinated motions and adapt to the properties of the external environment.
Collapse
Affiliation(s)
- Shreyansh Jain
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Benoit Ladoux
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France.
| | - René-Marc Mège
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France.
| |
Collapse
|
37
|
Torban E, Sokol SY. Planar cell polarity pathway in kidney development, function and disease. Nat Rev Nephrol 2021; 17:369-385. [PMID: 33547419 PMCID: PMC8967065 DOI: 10.1038/s41581-021-00395-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Planar cell polarity (PCP) refers to the coordinated orientation of cells in the tissue plane. Originally discovered and studied in Drosophila melanogaster, PCP is now widely recognized in vertebrates, where it is implicated in organogenesis. Specific sets of PCP genes have been identified. The proteins encoded by these genes become asymmetrically distributed to opposite sides of cells within a tissue plane and guide many processes that include changes in cell shape and polarity, collective cell movements or the uniform distribution of cell appendages. A unifying characteristic of these processes is that they often involve rearrangement of actomyosin. Mutations in PCP genes can cause malformations in organs of many animals, including humans. In the past decade, strong evidence has accumulated for a role of the PCP pathway in kidney development including outgrowth and branching morphogenesis of ureteric bud and podocyte development. Defective PCP signalling has been implicated in the pathogenesis of developmental kidney disorders of the congenital anomalies of the kidney and urinary tract spectrum. Understanding the origins, molecular constituents and cellular targets of PCP provides insights into the involvement of PCP molecules in normal kidney development and how dysfunction of PCP components may lead to kidney disease.
Collapse
Affiliation(s)
- Elena Torban
- McGill University and McGill University Health Center Research Institute, 1001 Boulevard Decarie, Block E, Montreal, Quebec, Canada, H4A3J1.,Corresponding authors: Elena Torban (); Sergei Sokol ()
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, USA,Corresponding authors: Elena Torban (); Sergei Sokol ()
| |
Collapse
|
38
|
A junctional PACSIN2/EHD4/MICAL-L1 complex coordinates VE-cadherin trafficking for endothelial migration and angiogenesis. Nat Commun 2021; 12:2610. [PMID: 33972531 PMCID: PMC8110786 DOI: 10.1038/s41467-021-22873-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022] Open
Abstract
Angiogenic sprouting relies on collective migration and coordinated rearrangements of endothelial leader and follower cells. VE-cadherin-based adherens junctions have emerged as key cell-cell contacts that transmit forces between cells and trigger signals during collective cell migration in angiogenesis. However, the underlying molecular mechanisms that govern these processes and their functional importance for vascular development still remain unknown. We previously showed that the F-BAR protein PACSIN2 is recruited to tensile asymmetric adherens junctions between leader and follower cells. Here we report that PACSIN2 mediates the formation of endothelial sprouts during angiogenesis by coordinating collective migration. We show that PACSIN2 recruits the trafficking regulators EHD4 and MICAL-L1 to the rear end of asymmetric adherens junctions to form a recycling endosome-like tubular structure. The junctional PACSIN2/EHD4/MICAL-L1 complex controls local VE-cadherin trafficking and thereby coordinates polarized endothelial migration and angiogenesis. Our findings reveal a molecular event at force-dependent asymmetric adherens junctions that occurs during the tug-of-war between endothelial leader and follower cells, and allows for junction-based guidance during collective migration in angiogenesis. Communication between endothelial leader and follower cells during collective cell migration is crucial for vascular development. Here, the authors show that PACSIN2 guides collective cell migration and angiogenesis by recruiting a protein trafficking complex to asymmetric cell-cell junctions, controlling local junction plasticity.
Collapse
|
39
|
Stock J, Pauli A. Self-organized cell migration across scales - from single cell movement to tissue formation. Development 2021; 148:148/7/dev191767. [PMID: 33824176 DOI: 10.1242/dev.191767] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-organization is a key feature of many biological and developmental processes, including cell migration. Although cell migration has traditionally been viewed as a biological response to extrinsic signals, advances within the past two decades have highlighted the importance of intrinsic self-organizing properties to direct cell migration on multiple scales. In this Review, we will explore self-organizing mechanisms that lay the foundation for both single and collective cell migration. Based on in vitro and in vivo examples, we will discuss theoretical concepts that underlie the persistent migration of single cells in the absence of directional guidance cues, and the formation of an autonomous cell collective that drives coordinated migration. Finally, we highlight the general implications of self-organizing principles guiding cell migration for biological and medical research.
Collapse
Affiliation(s)
- Jessica Stock
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC) Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Andrea Pauli
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC) Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| |
Collapse
|
40
|
Ozawa M, Hiver S, Yamamoto T, Shibata T, Upadhyayula S, Mimori-Kiyosue Y, Takeichi M. Adherens junction regulates cryptic lamellipodia formation for epithelial cell migration. J Cell Biol 2021; 219:152072. [PMID: 32886101 PMCID: PMC7659716 DOI: 10.1083/jcb.202006196] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Collective migration of epithelial cells plays crucial roles in various biological processes such as cancer invasion. In migrating epithelial sheets, leader cells form lamellipodia to advance, and follower cells also form similar motile apparatus at cell-cell boundaries, which are called cryptic lamellipodia (c-lamellipodia). Using adenocarcinoma-derived epithelial cells, we investigated how c-lamellipodia form and found that they sporadically grew from around E-cadherin-based adherens junctions (AJs). WAVE and Arp2/3 complexes were localized along the AJs, and silencing them not only interfered with c-lamellipodia formation but also prevented follower cells from trailing the leaders. Disruption of AJs by removing αE-catenin resulted in uncontrolled c-lamellipodia growth, and this was brought about by myosin II activation and the resultant contraction of AJ-associated actomyosin cables. Additional observations indicated that c-lamellipodia tended to grow at mechanically weak sites of the junction. We conclude that AJs not only tie cells together but also support c-lamellipodia formation by recruiting actin regulators, enabling epithelial cells to undergo ordered collective migration.
Collapse
Affiliation(s)
- Masayuki Ozawa
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Sylvain Hiver
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takaki Yamamoto
- Laboratory for Physical Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Tatsuo Shibata
- Laboratory for Physical Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Srigokul Upadhyayula
- Advanced Bioimaging Center, Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA
| | - Yuko Mimori-Kiyosue
- Laboratory for Molecular and Cellular Dynamics, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Masatoshi Takeichi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
41
|
Vafa F, Bowick MJ, Shraiman BI, Marchetti MC. Fluctuations can induce local nematic order and extensile stress in monolayers of motile cells. SOFT MATTER 2021; 17:3068-3073. [PMID: 33596291 DOI: 10.1039/d0sm02027c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Recent experiments in various cell types have shown that two-dimensional tissues often display local nematic order, with evidence of extensile stresses manifest in the dynamics of topological defects. Using a mesoscopic model where tissue flow is generated by fluctuating traction forces coupled to the nematic order parameter, we show that the resulting tissue dynamics can spontaneously produce local nematic order and an extensile internal stress. A key element of the model is the assumption that in the presence of local nematic alignment, cells preferentially crawl along the nematic axis, resulting in anisotropy of fluctuations. Our work shows that activity can drive either extensile or contractile stresses in tissue, depending on the relative strength of the contractility of the cortical cytoskeleton and tractions by cells on the extracellular matrix.
Collapse
Affiliation(s)
- Farzan Vafa
- Department of Physics, University of California Santa Barbara, Santa Barbara, CA 93106, USA.
| | | | | | | |
Collapse
|
42
|
Popkova A, Rauzi M, Wang X. Cellular and Supracellular Planar Polarity: A Multiscale Cue to Elongate the Drosophila Egg Chamber. Front Cell Dev Biol 2021; 9:645235. [PMID: 33738289 PMCID: PMC7961075 DOI: 10.3389/fcell.2021.645235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/02/2021] [Indexed: 01/10/2023] Open
Abstract
Tissue elongation is known to be controlled by oriented cell division, elongation, migration and rearrangement. While these cellular processes have been extensively studied, new emerging supracellular mechanisms driving tissue extension have recently been unveiled. Tissue rotation and actomyosin contractions have been shown to be key processes driving Drosophila egg chamber elongation. First, egg chamber rotation facilitates the dorsal-ventral alignment of the extracellular matrix and of the cell basal actin fibers. Both fiber-like structures form supracellular networks constraining the egg growth in a polarized fashion thus working as 'molecular corsets'. Second, the supracellular actin fiber network, powered by myosin periodic oscillation, contracts anisotropically driving tissue extension along the egg anterior-posterior axis. During both processes, cellular and supracellular planar polarity provide a critical cue to control Drosophila egg chamber elongation. Here we review how different planar polarized networks are built, maintained and function at both cellular and supracellular levels in the Drosophila ovarian epithelium.
Collapse
Affiliation(s)
- Anna Popkova
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Nice, France
| | - Matteo Rauzi
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Nice, France
| | - Xiaobo Wang
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
43
|
Olson HM, Nechiporuk AV. Lamellipodia-like protrusions and focal adhesions contribute to collective cell migration in zebrafish. Dev Biol 2020; 469:125-134. [PMID: 33096063 DOI: 10.1016/j.ydbio.2020.10.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 02/09/2023]
Abstract
Collective cell migration is a process where cohorts of cells exhibit coordinated migratory behavior. During individual and collective cellular migration, cells must extend protrusions to interact with the extracellular environment, sense chemotactic cues, and act as points of attachment. The mechanisms and regulators of protrusive behavior have been widely studied in individually migrating cells; however, how this behavior is regulated throughout collectives is not well understood. To address this, we used the zebrafish posterior lateral line primordium (pLLP) as a model. The pLLP is a cluster of ~150 cells that migrates along the zebrafish trunk, depositing groups of cells that will become sensory organs. To define protrusive behavior, we performed mosaic analysis to sparsely label pLLP cells with a transgene marking filamentous actin. This approach revealed an abundance of brush-like protrusions throughout the pLLP that orient in the direction of migration. Formation of these protrusions depends on the Arp2/3 complex, a regulator of dendritic actin. This argues that these brush-like protrusions are an in vivo example of lamellipodia. Mosaic analysis demonstrated that these lamellipodia-like protrusions are located in a close proximity to the overlying skin. Immunostaining revealed an abundance of focal adhesion complexes surrounding the pLLP. Disruption of these complexes specifically in pLLP cells led to impaired pLLP migration. Finally, we show that Erk signaling, a known regulator of focal adhesions, is required for proper formation of lamellipodia-like protrusions and pLLP migration. Altogether, our results suggest a model where the coordinated dynamics of lamellipodia-like protrusions, making contact with either the overlying skin or the extracellular matrix through focal adhesions, promotes migration of pLLP cells.
Collapse
Affiliation(s)
- Hannah M Olson
- Department Cell, Developmental & Cancer Biology, Oregon Health & Science University, The Knight Cancer Institute, Portland, OR, USA; Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, USA
| | - Alex V Nechiporuk
- Department Cell, Developmental & Cancer Biology, Oregon Health & Science University, The Knight Cancer Institute, Portland, OR, USA.
| |
Collapse
|
44
|
Bostock MP, Prasad AR, Chaouni R, Yuen AC, Sousa-Nunes R, Amoyel M, Fernandes VM. An Immobilization Technique for Long-Term Time-Lapse Imaging of Explanted Drosophila Tissues. Front Cell Dev Biol 2020; 8:590094. [PMID: 33117817 PMCID: PMC7576353 DOI: 10.3389/fcell.2020.590094] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/15/2020] [Indexed: 01/19/2023] Open
Abstract
Time-lapse imaging is an essential tool to study dynamic biological processes that cannot be discerned from fixed samples alone. However, imaging cell- and tissue-level processes in intact animals poses numerous challenges if the organism is opaque and/or motile. Explant cultures of intact tissues circumvent some of these challenges, but sample drift remains a considerable obstacle. We employed a simple yet effective technique to immobilize tissues in medium-bathed agarose. We applied this technique to study multiple Drosophila tissues from first-instar larvae to adult stages in various orientations and with no evidence of anisotropic pressure or stress damage. Using this method, we were able to image fine features for up to 18 h and make novel observations. Specifically, we report that fibers characteristic of quiescent neuroblasts are inherited by their basal daughters during reactivation; that the lamina in the developing visual system is assembled roughly 2-3 columns at a time; that lamina glia positions are dynamic during development; and that the nuclear envelopes of adult testis cyst stem cells do not break down completely during mitosis. In all, we demonstrate that our protocol is well-suited for tissue immobilization and long-term live imaging, enabling new insights into tissue and cell dynamics in Drosophila.
Collapse
Affiliation(s)
- Matthew P. Bostock
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Anadika R. Prasad
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Rita Chaouni
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
| | - Alice C. Yuen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Rita Sousa-Nunes
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
| | - Marc Amoyel
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Vilaiwan M. Fernandes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
45
|
Inhibition of protein tyrosine phosphatase receptor type F suppresses Wnt signaling in colorectal cancer. Oncogene 2020; 39:6789-6801. [PMID: 32973331 PMCID: PMC7606795 DOI: 10.1038/s41388-020-01472-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 09/15/2020] [Indexed: 02/05/2023]
Abstract
Wnt signaling dysregulation promotes tumorigenesis in colorectal cancer (CRC). We investigated the role of PTPRF, a receptor-type tyrosine phosphatase, in regulating Wnt signaling in CRC. Knockdown of PTPRF decreased cell proliferation in patient-derived primary colon cancer cells and established CRC cell lines. In addition, the rate of proliferation as well as colony formation ability were significantly decreased cells in tumor organoids grown in 3D, whereas the number of differentiated tumor organoids were markedly increased. Consistently, knockdown of PTPRF resulted in a decrease in the expression of genes associated cancer stem cells downstream of Wnt/β-catenin signaling. Treating PTPRF knockdown cells with GSK3 inhibitor rescued the expression of Wnt target genes suggesting that PTPRF functions upstream of the β-catenin destruction complex. PTPRF was found to interact with LRP6 and silencing PTPRF largely decreased the activation of LRP6. Interestingly, this PTPRF-mediated activation of Wnt signaling was blocked in cells treated with clathrin endocytosis inhibitor. Furthermore, knockdown of PTPRF inhibited xenograft tumor growth in vivo and decreased the expression of Wnt target genes. Taken together, our studies identify a novel role of PTPRF as an oncogenic protein phosphatase in supporting the activation of Wnt signaling in CRC.
Collapse
|
46
|
Mechanical induction and competence in epithelial morphogenesis. Curr Opin Genet Dev 2020; 63:36-44. [DOI: 10.1016/j.gde.2020.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 11/18/2022]
|
47
|
Abstract
Planar cell polarization, PCP, describes a form of organization where every cell within a group acquires the same planar characteristics, whether it is orientation of cell division, direction of migration, or localization of a cellular structure. PCP is essential for correct organization of cells into tissues and building a proper body plan. Here we use Hydra, an organism with a single axis of symmetry and a very simple body plan to investigate the function of the cell adhesion molecules Fat-like and Dachsous. We show that Hydra Fat-like and Dachsous are planar polarized, providing a demonstration of planar polarization of proteins in a nonbilaterian organism. We also discover roles for Hydra Fat-like in cell adhesion, spindle orientation, and tissue organization. Fat, Fat-like, and Dachsous family cadherins are giant proteins that regulate planar cell polarity (PCP) and cell adhesion in bilaterians. Their evolutionary origin can be traced back to prebilaterian species, but their ancestral function(s) are unknown. We identified Fat-like and Dachsous cadherins in Hydra, a member of phylum Cnidaria a sister group of bilaterian. We found Hydra does not possess a true Fat homolog, but has homologs of Fat-like (HyFatl) and Dachsous (HyDs) that localize at the apical membrane of ectodermal epithelial cells and are planar polarized perpendicular to the oral–aboral axis of the animal. Using a knockdown approach we found that HyFatl is involved in local cell alignment and cell–cell adhesion, and that reduction of HyFatl leads to defects in tissue organization in the body column. Overexpression and knockdown experiments indicate that the intracellular domain (ICD) of HyFatl affects actin organization through proline-rich repeats. Thus, planar polarization of Fat-like and Dachsous cadherins has ancient, prebilaterian origins, and Fat-like cadherins have ancient roles in cell adhesion, spindle orientation, and tissue organization.
Collapse
|
48
|
Shellard A, Mayor R. Rules of collective migration: from the wildebeest to the neural crest. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190387. [PMID: 32713298 PMCID: PMC7423382 DOI: 10.1098/rstb.2019.0387] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Collective migration, the movement of groups in which individuals affect the behaviour of one another, occurs at practically every scale, from bacteria up to whole species' populations. Universal principles of collective movement can be applied at all levels. In this review, we will describe the rules governing collective motility, with a specific focus on the neural crest, an embryonic stem cell population that undergoes extensive collective migration during development. We will discuss how the underlying principles of individual cell behaviour, and those that emerge from a supracellular scale, can explain collective migration. This article is part of the theme issue 'Multi-scale analysis and modelling of collective migration in biological systems'.
Collapse
Affiliation(s)
- Adam Shellard
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
49
|
Alexandrova AY, Chikina AS, Svitkina TM. Actin cytoskeleton in mesenchymal-to-amoeboid transition of cancer cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:197-256. [PMID: 33066874 DOI: 10.1016/bs.ircmb.2020.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During development of metastasis, tumor cells migrate through different tissues and encounter different extracellular matrices. An ability of cells to adapt mechanisms of their migration to these diverse environmental conditions, called migration plasticity, gives tumor cells an advantage over normal cells for long distant dissemination. Different modes of individual cell motility-mesenchymal and amoeboid-are driven by different molecular mechanisms, which largely depend on functions of the actin cytoskeleton that can be modulated in a wide range by cellular signaling mechanisms in response to environmental conditions. Various triggers can switch one motility mode to another, but regulations of these transitions are incompletely understood. However, understanding of the mechanisms driving migration plasticity is instrumental for finding anti-cancer treatment capable to stop cancer metastasis. In this review, we discuss cytoskeletal features, which allow the individually migrating cells to switch between mesenchymal and amoeboid migrating modes, called mesenchymal-to-amoeboid transition (MAT). We briefly describe main characteristics of different cell migration modes, and then discuss the triggering factors that initiate MAT with special attention to cytoskeletal features essential for migration plasticity.
Collapse
Affiliation(s)
- Antonina Y Alexandrova
- Laboratory of Mechanisms of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.
| | - Aleksandra S Chikina
- Cell Migration and Invasion and Spatio-Temporal Regulation of Antigen Presentation teams, UMR144/U932 Institut Curie, Paris, France
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
50
|
Cerqueira Campos F, Dennis C, Alégot H, Fritsch C, Isabella A, Pouchin P, Bardot O, Horne-Badovinac S, Mirouse V. Oriented basement membrane fibrils provide a memory for F-actin planar polarization via the Dystrophin-Dystroglycan complex during tissue elongation. Development 2020; 147:dev.186957. [PMID: 32156755 DOI: 10.1242/dev.186957] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/24/2020] [Indexed: 12/31/2022]
Abstract
How extracellular matrix contributes to tissue morphogenesis is still an open question. In the Drosophila ovarian follicle, it has been proposed that after Fat2-dependent planar polarization of the follicle cell basal domain, oriented basement membrane (BM) fibrils and F-actin stress fibers constrain follicle growth, promoting its axial elongation. However, the relationship between BM fibrils and stress fibers and their respective impact on elongation are unclear. We found that Dystroglycan (Dg) and Dystrophin (Dys) are involved in BM fibril deposition. Moreover, they also orient stress fibers, by acting locally and in parallel to Fat2. Importantly, Dg-Dys complex-mediated cell-autonomous control of F-actin fiber orientation relies on the preceding BM fibril deposition, indicating two distinct but interdependent functions. Thus, the Dg-Dys complex works as a crucial organizer of the epithelial basal domain, regulating both F-actin and BM. Furthermore, BM fibrils act as a persistent cue for the orientation of stress fibers that are the main effector of elongation.
Collapse
Affiliation(s)
- Fabiana Cerqueira Campos
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Cynthia Dennis
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Hervé Alégot
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Cornelia Fritsch
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Adam Isabella
- Committee on Development, Regeneration and Stem Cell Biology, and Department of Molecular Genetics and Cell Biology - The University of Chicago, 920 East 58th Street, Chicago IL 60653, USA
| | - Pierre Pouchin
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Olivier Bardot
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Sally Horne-Badovinac
- Committee on Development, Regeneration and Stem Cell Biology, and Department of Molecular Genetics and Cell Biology - The University of Chicago, 920 East 58th Street, Chicago IL 60653, USA
| | - Vincent Mirouse
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|