51
|
Zhang X, Jia S, Chen Z, Chong YL, Xie H, Feng D, Wu X, Song DZ, Roy S, Zhao C. Cilia-driven cerebrospinal fluid flow directs expression of urotensin neuropeptides to straighten the vertebrate body axis. Nat Genet 2018; 50:1666-1673. [DOI: 10.1038/s41588-018-0260-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 09/21/2018] [Indexed: 01/27/2023]
|
52
|
Ferreira RR, Pakula G, Klaeyle L, Fukui H, Vilfan A, Supatto W, Vermot J. Chiral Cilia Orientation in the Left-Right Organizer. Cell Rep 2018; 25:2008-2016.e4. [DOI: 10.1016/j.celrep.2018.10.069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 09/13/2018] [Accepted: 10/18/2018] [Indexed: 01/28/2023] Open
|
53
|
Braun E, Keren K. HydraRegeneration: Closing the Loop with Mechanical Processes in Morphogenesis. Bioessays 2018; 40:e1700204. [DOI: 10.1002/bies.201700204] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/29/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Erez Braun
- Department of Physics & Network Biology Research LaboratoriesTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Kinneret Keren
- Department of Physics & Network Biology Research LaboratoriesTechnion – Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
54
|
Boselli F, Steed E, Freund JB, Vermot J. Anisotropic shear stress patterns predict the orientation of convergent tissue movements in the embryonic heart. Development 2017; 144:4322-4327. [PMID: 29183943 PMCID: PMC5769631 DOI: 10.1242/dev.152124] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/31/2017] [Indexed: 12/28/2022]
Abstract
Myocardial contractility and blood flow provide essential mechanical cues for the morphogenesis of the heart. In general, endothelial cells change their migratory behavior in response to shear stress patterns, according to flow directionality. Here, we assessed the impact of shear stress patterns and flow directionality on the behavior of endocardial cells, the specialized endothelial cells of the heart. At the early stages of zebrafish heart valve formation, we show that endocardial cells are converging to the valve-forming area and that this behavior depends upon mechanical forces. Quantitative live imaging and mathematical modeling allow us to correlate this tissue convergence with the underlying flow forces. We predict that tissue convergence is associated with the direction of the mean wall shear stress and of the gradient of harmonic phase-averaged shear stresses, which surprisingly do not match the overall direction of the flow. This contrasts with the usual role of flow directionality in vascular development and suggests that the full spatial and temporal complexity of the wall shear stress should be taken into account when studying endothelial cell responses to flow in vivo. Summary: Blood flow modeling shows that dynamic shear stress patterns, rather than mean flow direction, predict the stereotypical behavior of endocardial cells during the early steps of heart valve formation.
Collapse
Affiliation(s)
- Francesco Boselli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France
| | - Emily Steed
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France
| | - Jonathan B Freund
- Mechanical Science & Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France .,Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France
| |
Collapse
|
55
|
Kumar A, Placone JK, Engler AJ. Understanding the extracellular forces that determine cell fate and maintenance. Development 2017; 144:4261-4270. [PMID: 29183939 DOI: 10.1242/dev.158469] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cells interpret signals from their microenvironment while simultaneously modifying the niche through secreting factors and exerting mechanical forces. Many soluble stem cell cues have been determined over the past century, but in the past decade, our molecular understanding of mechanobiology has advanced to explain how passive and active forces induce similar signaling cascades that drive self-renewal, migration, differentiation or a combination of these outcomes. Improvements in stem cell culture methods, materials and biophysical tools that assess function have improved our understanding of these cascades. Here, we summarize these advances and offer perspective on ongoing challenges.
Collapse
Affiliation(s)
- Aditya Kumar
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Jesse K Placone
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA .,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| |
Collapse
|
56
|
Kunnen SJ, Malas TB, Semeins CM, Bakker AD, Peters DJM. Comprehensive transcriptome analysis of fluid shear stress altered gene expression in renal epithelial cells. J Cell Physiol 2017; 233:3615-3628. [PMID: 29044509 PMCID: PMC5765508 DOI: 10.1002/jcp.26222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/05/2017] [Indexed: 12/21/2022]
Abstract
Renal epithelial cells are exposed to mechanical forces due to flow‐induced shear stress within the nephrons. Shear stress is altered in renal diseases caused by tubular dilation, obstruction, and hyperfiltration, which occur to compensate for lost nephrons. Fundamental in regulation of shear stress are primary cilia and other mechano‐sensors, and defects in cilia formation and function have profound effects on development and physiology of kidneys and other organs. We applied RNA sequencing to get a comprehensive overview of fluid‐shear regulated genes and pathways in renal epithelial cells. Functional enrichment‐analysis revealed TGF‐β, MAPK, and Wnt signaling as core signaling pathways up‐regulated by shear. Inhibitors of TGF‐β and MAPK/ERK signaling modulate a wide range of mechanosensitive genes, identifying these pathways as master regulators of shear‐induced gene expression. However, the main down‐regulated pathway, that is, JAK/STAT, is independent of TGF‐β and MAPK/ERK. Other up‐regulated cytokine pathways include FGF, HB‐EGF, PDGF, and CXC. Cellular responses to shear are modified at several levels, indicated by altered expression of genes involved in cell‐matrix, cytoskeleton, and glycocalyx remodeling, as well as glycolysis and cholesterol metabolism. Cilia ablation abolished shear induced expression of a subset of genes, but genes involved in TGF‐β, MAPK, and Wnt signaling were hardly affected, suggesting that other mechano‐sensors play a prominent role in the shear stress response of renal epithelial cells. Modulations in signaling due to variations in fluid shear stress are relevant for renal physiology and pathology, as suggested by elevated gene expression at pathological levels of shear stress compared to physiological shear.
Collapse
Affiliation(s)
- Steven J Kunnen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Tareq B Malas
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis M Semeins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
57
|
Harlepp S, Thalmann F, Follain G, Goetz JG. Hemodynamic forces can be accurately measured in vivo with optical tweezers. Mol Biol Cell 2017; 28:3252-3260. [PMID: 28904205 PMCID: PMC5687027 DOI: 10.1091/mbc.e17-06-0382] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/15/2022] Open
Abstract
Force sensing and generation at the tissue and cellular scale is central to many biological events. There is a growing interest in modern cell biology for methods enabling force measurements in vivo. Optical trapping allows noninvasive probing of piconewton forces and thus emerged as a promising mean for assessing biomechanics in vivo. Nevertheless, the main obstacles lie in the accurate determination of the trap stiffness in heterogeneous living organisms, at any position where the trap is used. A proper calibration of the trap stiffness is thus required for performing accurate and reliable force measurements in vivo. Here we introduce a method that overcomes these difficulties by accurately measuring hemodynamic profiles in order to calibrate the trap stiffness. Doing so, and using numerical methods to assess the accuracy of the experimental data, we measured flow profiles and drag forces imposed to trapped red blood cells of living zebrafish embryos. Using treatments enabling blood flow tuning, we demonstrated that such a method is powerful in measuring hemodynamic forces in vivo with accuracy and confidence. Altogether this study demonstrates the power of optical tweezing in measuring low range hemodynamic forces in vivo and offers an unprecedented tool in both cell and developmental biology.
Collapse
Affiliation(s)
- Sébastien Harlepp
- Université de Strasbourg, 67000 Strasbourg, France .,IPCMS, UMR7504, 67200 Strasbourg, France.,LabEx NIE, Université de Strasbourg, 67000 Strasbourg, France
| | - Fabrice Thalmann
- Université de Strasbourg, 67000 Strasbourg, France.,ICS, UPR22, 67034 Strasbourg, France
| | - Gautier Follain
- Université de Strasbourg, 67000 Strasbourg, France.,Inserm UMR_S1109, MN3T, 67200 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, 67000 Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Jacky G Goetz
- Université de Strasbourg, 67000 Strasbourg, France .,Inserm UMR_S1109, MN3T, 67200 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, 67000 Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| |
Collapse
|
58
|
Taormina MJ, Hay EA, Parthasarathy R. Passive and Active Microrheology of the Intestinal Fluid of the Larval Zebrafish. Biophys J 2017; 113:957-965. [PMID: 28834731 PMCID: PMC5567605 DOI: 10.1016/j.bpj.2017.06.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/12/2017] [Accepted: 06/26/2017] [Indexed: 12/24/2022] Open
Abstract
The fluids of the intestine serve as a physical barrier to pathogens, a medium for the diffusion of nutrients and metabolites, and an environment for commensal microbes. The rheological properties of intestinal mucus have therefore been the subject of many investigations, thus far limited to in vitro studies due to the difficulty of measurement in the natural context of the gut. This limitation especially hinders our understanding of how the gut microbiota interact with the intestinal space, since examination of this calls not only for in vivo measurement techniques, but for techniques that can be applied to model organisms in which the microbial state of the gut can be controlled. We have addressed this challenge with two complementary approaches. We performed passive microrheological measurements using thermally driven nanoparticles and active microrheology using micron-scale ellipsoidal magnetic microparticles, in both cases using light-sheet fluorescence microscopy to optically access the intestinal bulb of the larval zebrafish, a model vertebrate. We present viscosity measurements in germ-free animals (devoid of gut microbes), animals colonized by a single bacterial species, and conventionally reared animals, and find that in all cases, the mucin-rich intestinal liquid is well described as a Newtonian fluid. Surprisingly, despite known differences in the number of secretory cells in germ-free zebrafish and their conventional counterparts, the fluid viscosity for these two groups is very similar, as measured with either technique. Our study provides, to our knowledge, the first in vivo microrheological measurements of the intestinal space in living animals, and we comment on its implications for timescales of host-microbe interactions in the gut.
Collapse
Affiliation(s)
- Michael J Taormina
- Department of Physics, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, Oregon
| | - Edouard A Hay
- Department of Physics, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, Oregon
| | - Raghuveer Parthasarathy
- Department of Physics, Institute of Molecular Biology, Materials Science Institute, University of Oregon, Eugene, Oregon.
| |
Collapse
|
59
|
Nakajima H, Mochizuki N. Flow pattern-dependent endothelial cell responses through transcriptional regulation. Cell Cycle 2017; 16:1893-1901. [PMID: 28820314 DOI: 10.1080/15384101.2017.1364324] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Blood flow provides endothelial cells (ECs) lining the inside of blood vessels with mechanical stimuli as well as humoral stimuli. Fluid shear stress, the frictional force between flowing blood and ECs, is recognized as an essential mechanical cue for vascular growth, remodeling, and homeostasis. ECs differentially respond to distinct flow patterns. High laminar shear flow leads to inhibition of cell cycle progression and stabilizes vessels, whereas low shear flow or disturbed flow leads to increased turnover of ECs and inflammatory responses of ECs prone to atherogenic. These differences of EC responses dependent on flow pattern are mainly ascribed to distinct patterns of gene expression. In this review, we highlight flow pattern-dependent transcriptional regulation in ECs by focusing on KLF2 and NFκB, major transcription factors responding to laminar flow and disturbed flow, respectively. Moreover, we introduce roles of a new flow-responsive transcriptional co-regulator, YAP, in blood vessel maintenance and discuss how these transcriptional regulators are spatiotemporally regulated by flow and then regulate EC functions in normal and pathological conditions.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- a Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute , Suita , Osaka , Japan
| | - Naoki Mochizuki
- a Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute , Suita , Osaka , Japan.,b AMED-CREST. National Cerebral and Cardiovascular Center , Suita , Osaka , Japan
| |
Collapse
|
60
|
Nakajima H, Yamamoto K, Agarwala S, Terai K, Fukui H, Fukuhara S, Ando K, Miyazaki T, Yokota Y, Schmelzer E, Belting HG, Affolter M, Lecaudey V, Mochizuki N. Flow-Dependent Endothelial YAP Regulation Contributes to Vessel Maintenance. Dev Cell 2017; 40:523-536.e6. [PMID: 28350986 DOI: 10.1016/j.devcel.2017.02.019] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/16/2017] [Accepted: 02/24/2017] [Indexed: 11/16/2022]
Abstract
Endothelial cells (ECs) line the inside of blood vessels and respond to mechanical cues generated by blood flow. Mechanical stimuli regulate the localization of YAP by reorganizing the actin cytoskeleton. Here we demonstrate blood-flow-mediated regulation of endothelial YAP in vivo. We indirectly monitored transcriptional activity of Yap1 (zebrafish YAP) and its spatiotemporal localization in living zebrafish and found that Yap1 entered the nucleus and promoted transcription in response to blood flow. In cultured human ECs, laminar shear stress induced nuclear import of YAP and its transcriptional activity in a manner independent of Hippo signaling. We uncovered a molecular mechanism by which flow induced the nuclear translocation of YAP through the regulation of filamentous actin and angiomotin. Yap1 mutant zebrafish showed a defect in vascular stability, indicating an essential role for Yap1 in blood vessels. Our data imply that endothelial Yap1 functions in response to flow to maintain blood vessels.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Sobhika Agarwala
- Developmental Biology, SFB850, Institute for Biology I, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Kenta Terai
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8315, Japan
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Nippon Medical School, Kawasaki, Kanagawa 211-8533, Japan
| | - Koji Ando
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Takahiro Miyazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Yasuhiro Yokota
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Etienne Schmelzer
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Markus Affolter
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Virginie Lecaudey
- Department of Developmental Biology of Vertebrates, Institute for Cell Biology and Neurosciences, Goethe University of Frankfurt, 60438 Frankfurt, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan; AMED-CREST, National Cerebral and Cardiovascular Center, 5-7-1, Suita, Osaka 565-8565, Japan.
| |
Collapse
|
61
|
Abstract
In vivo, cells of the vascular system are subjected to various mechanical stimuli and have demonstrated the ability to adapt their behavior via mechanotransduction. Recent advances in microfluidic and "on-chip" techniques have provided the technology to study these alterations in cell behavior. Contrary to traditional in vitro assays such as transwell plates and parallel plate flow chambers, these microfluidic devices (MFDs) provide the opportunity to integrate multiple mechanical cues (e.g. shear stress, confinement, substrate stiffness, vessel geometry and topography) with in situ quantification capabilities. As such, MFDs can be used to recapitulate the in vivo mechanical setting and systematically vary microenvironmental conditions for improved mechanobiological studies of the endothelium. Additionally, adequate modelling provides for enhanced understanding of disease progression, design of cell separation and drug delivery systems, and the development of biomaterials for tissue engineering applications. Here, we will discuss the advances in knowledge about endothelial cell mechanosensing resulting from the design and application of biomimetic on-chip and microfluidic platforms.
Collapse
|
62
|
Ferreira RR, Vilfan A, Jülicher F, Supatto W, Vermot J. Physical limits of flow sensing in the left-right organizer. eLife 2017; 6. [PMID: 28613157 PMCID: PMC5544429 DOI: 10.7554/elife.25078] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/13/2017] [Indexed: 12/20/2022] Open
Abstract
Fluid flows generated by motile cilia are guiding the establishment of the left-right asymmetry of the body in the vertebrate left-right organizer. Competing hypotheses have been proposed: the direction of flow is sensed either through mechanosensation, or via the detection of chemical signals transported in the flow. We investigated the physical limits of flow detection to clarify which mechanisms could be reliably used for symmetry breaking. We integrated parameters describing cilia distribution and orientation obtained in vivo in zebrafish into a multiscale physical study of flow generation and detection. Our results show that the number of immotile cilia is too small to ensure robust left and right determination by mechanosensing, given the large spatial variability of the flow. However, motile cilia could sense their own motion by a yet unknown mechanism. Finally, transport of chemical signals by the flow can provide a simple and reliable mechanism of asymmetry establishment. DOI:http://dx.doi.org/10.7554/eLife.25078.001
Collapse
Affiliation(s)
- Rita R Ferreira
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | | | - Frank Jülicher
- Max-Planck-Institute for the Physics of Complex Systems, Dresden, Germany
| | - Willy Supatto
- Laboratory for Optics and Biosciences, Ecole Polytechnique, Centre National de la Recherche Scientifique (UMR7645), Institut National de la Santé et de la Recherche Médicale (U1182) and Paris Saclay University, Palaiseau, France
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Université de Strasbourg, Illkirch, France
| |
Collapse
|
63
|
Ferreira RR, Vermot J. The balancing roles of mechanical forces during left-right patterning and asymmetric morphogenesis. Mech Dev 2017; 144:71-80. [DOI: 10.1016/j.mod.2016.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/03/2016] [Indexed: 11/17/2022]
|
64
|
Walentek P, Quigley IK. What we can learn from a tadpole about ciliopathies and airway diseases: Using systems biology in Xenopus to study cilia and mucociliary epithelia. Genesis 2017; 55:10.1002/dvg.23001. [PMID: 28095645 PMCID: PMC5276738 DOI: 10.1002/dvg.23001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/11/2022]
Abstract
Over the past years, the Xenopus embryo has emerged as an incredibly useful model organism for studying the formation and function of cilia and ciliated epithelia in vivo. This has led to a variety of findings elucidating the molecular mechanisms of ciliated cell specification, basal body biogenesis, cilia assembly, and ciliary motility. These findings also revealed the deep functional conservation of signaling, transcriptional, post-transcriptional, and protein networks employed in the formation and function of vertebrate ciliated cells. Therefore, Xenopus research can contribute crucial insights not only into developmental and cell biology, but also into the molecular mechanisms underlying cilia related diseases (ciliopathies) as well as diseases affecting the ciliated epithelium of the respiratory tract in humans (e.g., chronic lung diseases). Additionally, systems biology approaches including transcriptomics, genomics, and proteomics have been rapidly adapted for use in Xenopus, and broaden the applications for current and future translational biomedical research. This review aims to present the advantages of using Xenopus for cilia research, highlight some of the evolutionarily conserved key concepts and mechanisms of ciliated cell biology that were elucidated using the Xenopus model, and describe the potential for Xenopus research to address unresolved questions regarding the molecular mechanisms of ciliopathies and airway diseases.
Collapse
Affiliation(s)
- Peter Walentek
- Department of Molecular and Cell Biology; Genetics, Genomics and Development Division; Developmental and Regenerative Biology Group; University of California, Berkeley, CA 94720, USA
| | - Ian K. Quigley
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
65
|
Wittkowske C, Reilly GC, Lacroix D, Perrault CM. In Vitro Bone Cell Models: Impact of Fluid Shear Stress on Bone Formation. Front Bioeng Biotechnol 2016; 4:87. [PMID: 27896266 PMCID: PMC5108781 DOI: 10.3389/fbioe.2016.00087] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 10/25/2016] [Indexed: 01/06/2023] Open
Abstract
This review describes the role of bone cells and their surrounding matrix in maintaining bone strength through the process of bone remodeling. Subsequently, this work focusses on how bone formation is guided by mechanical forces and fluid shear stress in particular. It has been demonstrated that mechanical stimulation is an important regulator of bone metabolism. Shear stress generated by interstitial fluid flow in the lacunar-canalicular network influences maintenance and healing of bone tissue. Fluid flow is primarily caused by compressive loading of bone as a result of physical activity. Changes in loading, e.g., due to extended periods of bed rest or microgravity in space are associated with altered bone remodeling and formation in vivo. In vitro, it has been reported that bone cells respond to fluid shear stress by releasing osteogenic signaling factors, such as nitric oxide, and prostaglandins. This work focusses on the application of in vitro models to study the effects of fluid flow on bone cell signaling, collagen deposition, and matrix mineralization. Particular attention is given to in vitro set-ups, which allow long-term cell culture and the application of low fluid shear stress. In addition, this review explores what mechanisms influence the orientation of collagen fibers, which determine the anisotropic properties of bone. A better understanding of these mechanisms could facilitate the design of improved tissue-engineered bone implants or more effective bone disease models.
Collapse
Affiliation(s)
- Claudia Wittkowske
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK; INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Gwendolen C Reilly
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK; Department of Material Science, University of Sheffield, Sheffield, UK
| | - Damien Lacroix
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK; INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Cecile M Perrault
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK; INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
66
|
Faubel R, Westendorf C, Bodenschatz E, Eichele G. Cilia-based flow network in the brain ventricles. Science 2016; 353:176-8. [PMID: 27387952 DOI: 10.1126/science.aae0450] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/10/2016] [Indexed: 12/24/2022]
Abstract
Cerebrospinal fluid conveys many physiologically important signaling factors through the ventricular cavities of the brain. We investigated the transport of cerebrospinal fluid in the third ventricle of the mouse brain and discovered a highly organized pattern of cilia modules, which collectively give rise to a network of fluid flows that allows for precise transport within this ventricle. We also discovered a cilia-based switch that reliably and periodically alters the flow pattern so as to create a dynamic subdivision that may control substance distribution in the third ventricle. Complex flow patterns were also present in the third ventricles of rats and pigs. Our work suggests that ciliated epithelia can generate and maintain complex, spatiotemporally regulated flow networks.
Collapse
Affiliation(s)
- Regina Faubel
- Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Christian Westendorf
- Max Planck Institute for Dynamics and Self-Organization, Am Fassberg 17, 37077 Göttingen, Germany
| | - Eberhard Bodenschatz
- Max Planck Institute for Dynamics and Self-Organization, Am Fassberg 17, 37077 Göttingen, Germany
| | - Gregor Eichele
- Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany.
| |
Collapse
|
67
|
Warren KM, Islam MM, LeDuc PR, Steward R. 2D and 3D Mechanobiology in Human and Nonhuman Systems. ACS APPLIED MATERIALS & INTERFACES 2016; 8:21869-21882. [PMID: 27214883 DOI: 10.1021/acsami.5b12064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Mechanobiology involves the investigation of mechanical forces and their effect on the development, physiology, and pathology of biological systems. The human body has garnered much attention from many groups in the field, as mechanical forces have been shown to influence almost all aspects of human life ranging from breathing to cancer metastasis. Beyond being influential in human systems, mechanical forces have also been shown to impact nonhuman systems such as algae and zebrafish. Studies of nonhuman and human systems at the cellular level have primarily been done in two-dimensional (2D) environments, but most of these systems reside in three-dimensional (3D) environments. Furthermore, outcomes obtained from 3D studies are often quite different than those from 2D studies. We present here an overview of a select group of human and nonhuman systems in 2D and 3D environments. We also highlight mechanobiological approaches and their respective implications for human and nonhuman physiology.
Collapse
Affiliation(s)
- Kristin M Warren
- Departments of Mechanical Engineering, Biomedical Engineering, Computational Biology, and Biological Sciences, Carnegie Mellon University , Pittsburgh, Pennsylvania 15213, United States
| | - Md Mydul Islam
- Department of Mechanical and Aerospace Engineering and Burnett School of Biomedical Sciences, University of Central Florida , Orlando, Florida 32827, United States
| | - Philip R LeDuc
- Departments of Mechanical Engineering, Biomedical Engineering, Computational Biology, and Biological Sciences, Carnegie Mellon University , Pittsburgh, Pennsylvania 15213, United States
| | - Robert Steward
- Department of Mechanical and Aerospace Engineering and Burnett School of Biomedical Sciences, University of Central Florida , Orlando, Florida 32827, United States
| |
Collapse
|
68
|
Wang S, Lakomy DS, Garcia MD, Lopez AL, Larin KV, Larina IV. Four-dimensional live imaging of hemodynamics in mammalian embryonic heart with Doppler optical coherence tomography. JOURNAL OF BIOPHOTONICS 2016; 9:837-47. [PMID: 26996292 PMCID: PMC5152918 DOI: 10.1002/jbio.201500314] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/03/2016] [Accepted: 03/01/2016] [Indexed: 05/19/2023]
Abstract
Hemodynamic analysis of the mouse embryonic heart is essential for understanding the functional aspects of early cardiogenesis and advancing the research in congenital heart defects. However, high-resolution imaging of cardiac hemodynamics in mammalian models remains challenging, primarily due to the dynamic nature and deep location of the embryonic heart. Here we report four-dimensional micro-scale imaging of blood flow in the early mouse embryonic heart, enabling time-resolved measurement and analysis of flow velocity throughout the heart tube. Our method uses Doppler optical coherence tomography in live mouse embryo culture, and employs a post-processing synchronization approach to reconstruct three-dimensional data over time at a 100 Hz volume rate. Experiments were performed on live mouse embryos at embryonic day 9.0. Our results show blood flow dynamics inside the beating heart, with the capability for quantitative flow velocity assessment in the primitive atrium, atrioventricular and bulboventricular regions, and bulbus cordis. Combined cardiodynamic and hemodynamic analysis indicates this functional imaging method can be utilized to further investigate the mechanical relationship between blood flow dynamics and cardiac wall movement, bringing new possibilities to study biomechanics in early mammalian cardiogenesis. Four-dimensional live hemodynamic imaging of the mouse embryonic heart at embryonic day 9.0 using Doppler optical coherence tomography, showing directional blood flows in the sinus venosus, primitive atrium, atrioventricular region and vitelline vein.
Collapse
Affiliation(s)
- Shang Wang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - David S Lakomy
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - Monica D Garcia
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - Andrew L Lopez
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - Kirill V Larin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd., 77204, Houston, TX 77204, U.S
- Interdisciplinary Laboratory of Biophotonics, Tomsk State University, Tomsk, 634050, Russia
| | - Irina V Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S..
| |
Collapse
|
69
|
Wang S, Lakomy DS, Garcia MD, Lopez AL, Larin KV, Larina IV. Four-dimensional live imaging of hemodynamics in mammalian embryonic heart with Doppler optical coherence tomography. JOURNAL OF BIOPHOTONICS 2016. [PMID: 26996292 DOI: 10.1002/jbio.v9.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Hemodynamic analysis of the mouse embryonic heart is essential for understanding the functional aspects of early cardiogenesis and advancing the research in congenital heart defects. However, high-resolution imaging of cardiac hemodynamics in mammalian models remains challenging, primarily due to the dynamic nature and deep location of the embryonic heart. Here we report four-dimensional micro-scale imaging of blood flow in the early mouse embryonic heart, enabling time-resolved measurement and analysis of flow velocity throughout the heart tube. Our method uses Doppler optical coherence tomography in live mouse embryo culture, and employs a post-processing synchronization approach to reconstruct three-dimensional data over time at a 100 Hz volume rate. Experiments were performed on live mouse embryos at embryonic day 9.0. Our results show blood flow dynamics inside the beating heart, with the capability for quantitative flow velocity assessment in the primitive atrium, atrioventricular and bulboventricular regions, and bulbus cordis. Combined cardiodynamic and hemodynamic analysis indicates this functional imaging method can be utilized to further investigate the mechanical relationship between blood flow dynamics and cardiac wall movement, bringing new possibilities to study biomechanics in early mammalian cardiogenesis. Four-dimensional live hemodynamic imaging of the mouse embryonic heart at embryonic day 9.0 using Doppler optical coherence tomography, showing directional blood flows in the sinus venosus, primitive atrium, atrioventricular region and vitelline vein.
Collapse
Affiliation(s)
- Shang Wang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - David S Lakomy
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - Monica D Garcia
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - Andrew L Lopez
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
| | - Kirill V Larin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S
- Department of Biomedical Engineering, University of Houston, 3605 Cullen Blvd., 77204, Houston, TX 77204, U.S
- Interdisciplinary Laboratory of Biophotonics, Tomsk State University, Tomsk, 634050, Russia
| | - Irina V Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, U.S..
| |
Collapse
|
70
|
Rashidi H, Alhaque S, Szkolnicka D, Flint O, Hay DC. Fluid shear stress modulation of hepatocyte-like cell function. Arch Toxicol 2016; 90:1757-61. [PMID: 26979076 PMCID: PMC4894932 DOI: 10.1007/s00204-016-1689-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 02/25/2016] [Indexed: 12/19/2022]
Abstract
Freshly isolated human adult hepatocytes are considered to be the gold standard tool for in vitro studies. However, primary hepatocyte scarcity, cell cycle arrest and the rapid loss of cell phenotype limit their widespread deployment. Human embryonic stem cells and induced pluripotent stem cells provide renewable sources of hepatocyte-like cells (HLCs). Despite the use of various differentiation methodologies, HLCs like primary human hepatocytes exhibit unstable phenotype in culture. It has been shown that the functional capacity can be improved by adding back elements of human physiology, such as cell co-culture or through the use of natural and/or synthetic surfaces. In this study, the effect of fluid shear stress on HLC performance was investigated. We studied two important liver functions, cytochrome P450 drug metabolism and serum protein secretion, in static cultures and those exposed to fluid shear stress. Our study demonstrates that fluid shear stress improved Cyp1A2 activity by approximately fivefold. This was paralleled by an approximate ninefold increase in sensitivity to a drug, primarily metabolised by Cyp2D6. In addition to metabolic capacity, fluid shear stress also improved hepatocyte phenotype with an approximate fourfold reduction in the secretion of a foetal marker, alpha-fetoprotein. We believe these studies highlight the importance of introducing physiologic cues in cell-based models to improve somatic cell phenotype.
Collapse
Affiliation(s)
- Hassan Rashidi
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Sharmin Alhaque
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Dagmara Szkolnicka
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Oliver Flint
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - David C Hay
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
71
|
Steed E, Faggianelli N, Roth S, Ramspacher C, Concordet JP, Vermot J. klf2a couples mechanotransduction and zebrafish valve morphogenesis through fibronectin synthesis. Nat Commun 2016; 7:11646. [PMID: 27221222 PMCID: PMC4894956 DOI: 10.1038/ncomms11646] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 04/15/2016] [Indexed: 12/23/2022] Open
Abstract
The heartbeat and blood flow signal to endocardial cell progenitors through mechanosensitive proteins that modulate the genetic program controlling heart valve morphogenesis. To date, the mechanism by which mechanical forces coordinate tissue morphogenesis is poorly understood. Here we use high-resolution imaging to uncover the coordinated cell behaviours leading to heart valve formation. We find that heart valves originate from progenitors located in the ventricle and atrium that generate the valve leaflets through a coordinated set of endocardial tissue movements. Gene profiling analyses and live imaging reveal that this reorganization is dependent on extracellular matrix proteins, in particular on the expression of fibronectin1b. We show that blood flow and klf2a, a major endocardial flow-responsive gene, control these cell behaviours and fibronectin1b synthesis. Our results uncover a unique multicellular layering process leading to leaflet formation and demonstrate that endocardial mechanotransduction and valve morphogenesis are coupled via cellular rearrangements mediated by fibronectin synthesis.
Collapse
Affiliation(s)
- Emily Steed
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Nathalie Faggianelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Stéphane Roth
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Caroline Ramspacher
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| | - Jean-Paul Concordet
- Muséum National d'Histoire Naturelle, 75231 Paris Cedex 05, France
- CNRS UMR 7196, 75231 Paris Cedex 05, France
- INSERM U1154, 75231 Paris Cedex 05, France
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France
- Université de Strasbourg, Illkirch 67404, France
| |
Collapse
|
72
|
Abstract
The molecular mechanisms underlying cardiogenesis are of critical biomedical importance due to the high prevalence of cardiac birth defects. Over the past two decades, the zebrafish has served as a powerful model organism for investigating heart development, facilitated by its powerful combination of optical access to the embryonic heart and plentiful opportunities for genetic analysis. Work in zebrafish has identified numerous factors that are required for various aspects of heart formation, including the specification and differentiation of cardiac progenitor cells, the morphogenesis of the heart tube, cardiac chambers, and atrioventricular canal, and the establishment of proper cardiac function. However, our current roster of regulators of cardiogenesis is by no means complete. It is therefore valuable for ongoing studies to continue pursuit of additional genes and pathways that control the size, shape, and function of the zebrafish heart. An extensive arsenal of techniques is available to distinguish whether particular mutations, morpholinos, or small molecules disrupt specific processes during heart development. In this chapter, we provide a guide to the experimental strategies that are especially effective for the characterization of cardiac phenotypes in the zebrafish embryo.
Collapse
Affiliation(s)
- A R Houk
- University of California, San Diego, CA, United States
| | - D Yelon
- University of California, San Diego, CA, United States
| |
Collapse
|
73
|
Pedrizzetti G, Martiniello AR, Bianchi V, D'Onofrio A, Caso P, Tonti G. Changes in electrical activation modify the orientation of left ventricular flow momentum: novel observations using echocardiographic particle image velocimetry. Eur Heart J Cardiovasc Imaging 2016; 17:203-9. [PMID: 26060201 PMCID: PMC4882880 DOI: 10.1093/ehjci/jev137] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/03/2015] [Indexed: 02/06/2023] Open
Abstract
AIMS Changes in electrical activation sequence are known to affect the timing of cardiac mechanical events. We aim to demonstrate that these also modify global properties of the intraventricular blood flow pattern. We also explore whether such global changes present a relationship with clinical outcome. METHODS AND RESULTS We investigated 30 heart failure patients followed up after cardiac resynchronization therapy (CRT). All subjects underwent echocardiography before implant and at follow-up after 6+ months. Left ventricular mechanics was investigated at follow-up during active CRT and was repeated after a temporary interruption <5 min later. Strain analysis, performed by speckle tracking, was used to assess the entity of contraction (global longitudinal strain) and its synchronicity (standard deviation of time to peak of radial strain). Intraventricular fluid dynamics, by echographic particle image velocimetry, was used to evaluate the directional distribution of global momentum associated with blood motion. The discontinuation of CRT pacing reflects into a reduction of deformation synchrony and into the deviation of blood flow momentum from the base-apex orientation with the development of transversal flow-mediated haemodynamic forces. The deviation of flow momentum presents a significant correlation with the degree of volumetric reduction after CRT. CONCLUSION Changes in electrical activation alter the orientation of blood flow momentum. The long-term CRT outcome correlates with the degree of re-alignment of haemodynamic forces. These preliminary results suggest that flow orientation could be used for optimizing the biventricular pacing setting. However, larger prospective studies are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Gianni Pedrizzetti
- Department of Engineering and Architecture, University of Trieste, P.le Europa 1., Trieste 34127, Italy
| | | | - Valter Bianchi
- Department of Cardiology, Monaldi Hospital, AORN Ospedali dei Colli, Napoli, Italy
| | - Antonio D'Onofrio
- Department of Cardiology, Monaldi Hospital, AORN Ospedali dei Colli, Napoli, Italy
| | - Pio Caso
- Department of Cardiology, Monaldi Hospital, AORN Ospedali dei Colli, Napoli, Italy
| | - Giovanni Tonti
- Cardiology Division, 'G. d'Annunzio' University, Chieti, Italy
| |
Collapse
|
74
|
Hemodynamics driven cardiac valve morphogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1760-6. [PMID: 26608609 DOI: 10.1016/j.bbamcr.2015.11.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 11/22/2022]
Abstract
Mechanical forces are instrumental to cardiovascular development and physiology. The heart beats approximately 2.6 billion times in a human lifetime and heart valves ensure that these contractions result in an efficient, unidirectional flow of the blood. Composed of endocardial cells (EdCs) and extracellular matrix (ECM), cardiac valves are among the most mechanically challenged structures of the body both during and after their development. Understanding how hemodynamic forces modulate cardiovascular function and morphogenesis is key to unraveling the relationship between normal and pathological cardiovascular development and physiology. Most valve diseases have their origins in embryogenesis, either as signs of abnormal developmental processes or the aberrant re-expression of fetal gene programs normally quiescent in adulthood. Here we review recent discoveries in the mechanobiology of cardiac valve development and introduce the latest technologies being developed in the zebrafish, including live cell imaging and optical technologies, as well as modeling approaches that are currently transforming this field. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
|
75
|
Boselli F, Vermot J. Live imaging and modeling for shear stress quantification in the embryonic zebrafish heart. Methods 2015; 94:129-34. [PMID: 26390811 DOI: 10.1016/j.ymeth.2015.09.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/07/2015] [Accepted: 09/17/2015] [Indexed: 01/17/2023] Open
Abstract
Hemodynamic shear stress is sensed by the endocardial cells composing the inner cell layer of the heart, and plays a major role in cardiac morphogenesis. Yet, the underlying hemodynamics and the associated mechanical stimuli experienced by endocardial cells remains poorly understood. Progress in the field has been hampered by the need for high temporal resolution imaging allowing the flow profiles generated in the beating heart to be resolved. To fill this gap, we propose a method to analyze the wall dynamics, the flow field, and the wall shear stress of the developing zebrafish heart. This method combines live confocal imaging and computational fluid dynamics to overcome difficulties related to live imaging of blood flow in the developing heart. To provide an example of the applicability of the method, we discuss the hemodynamic frequency content sensed by endocardial cells at the onset of valve formation, and how the fundamental frequency of the wall shear stress represents a unique mechanical cue to endocardial, heart-valve precursors.
Collapse
Affiliation(s)
- Francesco Boselli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France; Université de Strasbourg, Illkirch, France.
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France; Université de Strasbourg, Illkirch, France.
| |
Collapse
|
76
|
Goenezen S, Chivukula VK, Midgett M, Phan L, Rugonyi S. 4D subject-specific inverse modeling of the chick embryonic heart outflow tract hemodynamics. Biomech Model Mechanobiol 2015; 15:723-43. [PMID: 26361767 DOI: 10.1007/s10237-015-0720-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 08/17/2015] [Indexed: 01/10/2023]
Abstract
Blood flow plays a critical role in regulating embryonic cardiac growth and development, with altered flow leading to congenital heart disease. Progress in the field, however, is hindered by a lack of quantification of hemodynamic conditions in the developing heart. In this study, we present a methodology to quantify blood flow dynamics in the embryonic heart using subject-specific computational fluid dynamics (CFD) models. While the methodology is general, we focused on a model of the chick embryonic heart outflow tract (OFT), which distally connects the heart to the arterial system, and is the region of origin of many congenital cardiac defects. Using structural and Doppler velocity data collected from optical coherence tomography, we generated 4D ([Formula: see text]) embryo-specific CFD models of the heart OFT. To replicate the blood flow dynamics over time during the cardiac cycle, we developed an iterative inverse-method optimization algorithm, which determines the CFD model boundary conditions such that differences between computed velocities and measured velocities at one point within the OFT lumen are minimized. Results from our developed CFD model agree with previously measured hemodynamics in the OFT. Further, computed velocities and measured velocities differ by [Formula: see text]15 % at locations that were not used in the optimization, validating the model. The presented methodology can be used in quantifications of embryonic cardiac hemodynamics under normal and altered blood flow conditions, enabling an in-depth quantitative study of how blood flow influences cardiac development.
Collapse
Affiliation(s)
- Sevan Goenezen
- Department of Mechanical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Venkat Keshav Chivukula
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Madeline Midgett
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Ly Phan
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Sandra Rugonyi
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
77
|
Bokka KK, Jesudason EC, Lozoya OA, Guilak F, Warburton D, Lubkin SR. Morphogenetic Implications of Peristalsis-Driven Fluid Flow in the Embryonic Lung. PLoS One 2015; 10:e0132015. [PMID: 26147967 PMCID: PMC4493131 DOI: 10.1371/journal.pone.0132015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 05/15/2015] [Indexed: 12/14/2022] Open
Abstract
Epithelial organs are almost universally secretory. The lung secretes mucus of extremely variable consistency. In the early prenatal period, the secretions are of largely unknown composition, consistency, and flow rates. In addition to net outflow from secretion, the embryonic lung exhibits transient reversing flows from peristalsis. Airway peristalsis (AP) begins as soon as the smooth muscle forms, and persists until birth. Since the prenatal lung is liquid-filled, smooth muscle action can transport fluid far from the immediately adjacent tissues. The sensation of internal fluid flows has been shown to have potent morphogenetic effects, as has the transport of morphogens. We hypothesize that these effects play an important role in lung morphogenesis. To test these hypotheses in a quantitative framework, we analyzed the fluid-structure interactions between embryonic tissues and lumen fluid resulting from peristaltic waves that partially occlude the airway. We found that if the airway is closed, fluid transport is minimal; by contrast, if the trachea is open, shear rates can be very high, particularly at the stenosis. We performed a parametric analysis of flow characteristics' dependence on tissue stiffnesses, smooth muscle force, geometry, and fluid viscosity, and found that most of these relationships are governed by simple ratios. We measured the viscosity of prenatal lung fluid with passive bead microrheology. This paper reports the first measurements of the viscosity of embryonic lung lumen fluid. In the range tested, lumen fluid can be considered Newtonian, with a viscosity of 0.016 ± 0.008 Pa-s. We analyzed the interaction between the internal flows and diffusion and conclude that AP has a strong effect on flow sensing away from the tip and on transport of morphogens. These effects may be the intermediate mechanisms for the enhancement of branching seen in occluded embryonic lungs.
Collapse
Affiliation(s)
- Kishore K. Bokka
- Department of Mechanical Engineering, North Carolina State University, Raleigh, North Carolina, United States of America
| | | | - Oswaldo A. Lozoya
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Farshid Guilak
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David Warburton
- The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Sharon R. Lubkin
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
78
|
Smith DJ, Montenegro-Johnson TD, Lopes SS. Organized chaos in Kupffer's vesicle: how a heterogeneous structure achieves consistent left-right patterning. BIOARCHITECTURE 2015; 4:119-25. [PMID: 25454897 DOI: 10.4161/19490992.2014.956593] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Successful establishment of left-right asymmetry is crucial to healthy vertebrate development. In many species this process is initiated in a ciliated, enclosed cavity, for example Kupffer's vesicle (KV) in zebrafish. The microarchitecture of KV is more complex than that present in the left-right organizer of many other species. While swirling flow in KV is recognized as essential for left-right patterning, its generation, nature and conversion to asymmetric gene expression are only beginning to be fully understood. We recently [Sampaio, P et al. Dev Cell 29:716-728] combined imaging, genetics and fluid dynamics simulation to characterize normal and perturbed ciliary activity, and their correlation to asymmetric charon expression and embryonic organ fate. Randomness in cilia number and length have major implications for robust flow generation; even a modest change in mean cilia length has a major effect on flow speed to due to nonlinear scaling arising from fluid mechanics. Wildtype, and mutant embryos with normal liver laterality, exhibit stronger flow on the left prior to asymmetric inhibition of charon. Our discovery of immotile cilia, taken with data on morphant embryos with very few cilia, further support the role of mechanosensing in initiating and/or enhancing flow conversion into gene expression.
Collapse
Key Words
- DA, dorsal roof-anterior
- DC, dorsal roof-central
- DP, dorsal roof-posterior
- EQ, equatorial region of Kupffer's vesicle separating dorsal roof and ventral floor
- KV, Kupffer's vesicle
- Kupffer's vesicle
- MO-control, embryo treated with mismatch control morpholino
- VA, ventral floor-anterior
- VC, ventral floor-central
- VP, ventral floor-posterior
- WT, wildtype
- cilia
- dld-/-, homozygous deltaD null mutant
- dnah7-MO, dnah7-morpholino knockdown embryo
- heterotaxia
- left-right asymmetry
- situs inversus
- zebrafish
Collapse
Affiliation(s)
- D J Smith
- a School of Mathematics ; University of Birmingham ; Birmingham , UK
| | | | | |
Collapse
|
79
|
Dynamic reciprocity revisited. J Theor Biol 2015; 370:205-8. [DOI: 10.1016/j.jtbi.2015.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 01/12/2015] [Accepted: 01/15/2015] [Indexed: 11/20/2022]
|
80
|
Boselli F, Freund JB, Vermot J. Blood flow mechanics in cardiovascular development. Cell Mol Life Sci 2015; 72:2545-59. [PMID: 25801176 PMCID: PMC4457920 DOI: 10.1007/s00018-015-1885-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/25/2015] [Accepted: 03/12/2015] [Indexed: 11/29/2022]
Abstract
Hemodynamic forces are fundamental to development. Indeed, much of cardiovascular morphogenesis reflects a two-way interaction between mechanical forces and the gene network activated in endothelial cells via mechanotransduction feedback loops. As these interactions are becoming better understood in different model organisms, it is possible to identify common mechanogenetic rules, which are strikingly conserved and shared in many tissues and species. Here, we discuss recent findings showing how hemodynamic forces potentially modulate cardiovascular development as well as the underlying fluid and tissue mechanics, with special attention given to the flow characteristics that are unique to the small scales of embryos.
Collapse
Affiliation(s)
- Francesco Boselli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France,
| | | | | |
Collapse
|
81
|
Abstract
Motile cilia generate directional flows that move mucus through airways, cerebrospinal fluid through brain ventricles, and oocytes through fallopian tubes. In addition, specialized monocilia beat in a rotational pattern to create asymmetric flows that are involved in establishing the left-right (LR) body axis during embryogenesis. These monocilia, which we refer to as "left-right cilia," produce a leftward flow of extraembryonic fluid in a transient "organ of asymmetry" that directs asymmetric signaling and development of LR asymmetries in the cardiovascular system and gastrointestinal tract. The asymmetric flows are thought to establish a chemical gradient and/or activate mechanosensitive cilia to initiate calcium ion signals and a conserved Nodal (TGFβ) pathway on the left side of the embryo, but the mechanisms underlying this process remain unclear. The zebrafish organ of asymmetry, called Kupffer's vesicle, provides a useful model system for investigating LR cilia and cilia-powered fluid flows. Here, we describe methods to visualize flows in Kupffer's vesicle using fluorescent microspheres and introduce a new and freely available MATLAB particle tracking code to quantitatively describe these flows. Analysis of normal and aberrant flows indicates this approach is useful for characterizing flow properties that impact LR asymmetry and may be more broadly applicable for quantifying other cilia flows.
Collapse
|
82
|
Renz M, Otten C, Faurobert E, Rudolph F, Zhu Y, Boulday G, Duchene J, Mickoleit M, Dietrich AC, Ramspacher C, Steed E, Manet-Dupé S, Benz A, Hassel D, Vermot J, Huisken J, Tournier-Lasserve E, Felbor U, Sure U, Albiges-Rizo C, Abdelilah-Seyfried S. Regulation of β1 Integrin-Klf2-Mediated Angiogenesis by CCM Proteins. Dev Cell 2015; 32:181-90. [DOI: 10.1016/j.devcel.2014.12.016] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 11/03/2014] [Accepted: 12/19/2014] [Indexed: 12/01/2022]
|
83
|
A quantitative approach to study endothelial cilia bending stiffness during blood flow mechanodetection in vivo. Methods Cell Biol 2015; 127:161-73. [DOI: 10.1016/bs.mcb.2015.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
84
|
Pedrizzetti G, Martiniello AR, Bianchi V, D’Onofrio A, Caso P, Tonti G. Cardiac fluid dynamics anticipates heart adaptation. J Biomech 2015; 48:388-91. [PMID: 25529139 DOI: 10.1016/j.jbiomech.2014.11.049] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/07/2014] [Accepted: 11/26/2014] [Indexed: 10/24/2022]
|
85
|
Seki A, Nishii K, Hagiwara N. Gap junctional regulation of pressure, fluid force, and electrical fields in the epigenetics of cardiac morphogenesis and remodeling. Life Sci 2014; 129:27-34. [PMID: 25447447 DOI: 10.1016/j.lfs.2014.10.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/16/2014] [Accepted: 10/29/2014] [Indexed: 01/25/2023]
Abstract
Epigenetic factors of pressure load, fluid force, and electrical fields that occur during cardiac contraction affect cardiac development, morphology, function, and pathogenesis. These factors are orchestrated by intercellular communication mediated by gap junctions, which synchronize action potentials and second messengers. Misregulation of the gap junction protein connexin (Cx) alters cardiogenesis, and can be a pathogenic factor causing cardiac conduction disturbance, fatal arrhythmia, and cardiac remodeling in disease states such as hypertension and ischemia. Changes in Cx expression can occur even when the DNA sequence of the Cx gene itself is unaltered. Posttranslational modifications might reduce arrhythmogenic substrates, improve cardiac function, and promote remodeling in a diseased heart. In this review, we discuss the epigenetic features of gap junctions that regulate cardiac morphology and remodeling. We further discuss potential clinical applications of current knowledge of the structure and function of gap junctions.
Collapse
Affiliation(s)
- Akiko Seki
- Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan; Support Center for Women Health Care Professionals and Researchers, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan.
| | - Kiyomasa Nishii
- Department of Anatomy and Neurobiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Nobuhisa Hagiwara
- Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
86
|
Abstract
The precision of axonal pathfinding and the accurate formation of functional neural circuitry are crucial for an organism during development as well as during adult central and peripheral nerve regeneration. While chemical cues are believed to be primarily responsible for axonal pathfinding, we hypothesize that forces due to localized fluid flow may directly affect neuronal guidance during early organ development. Here, we report direct evidence of fluid flow influencing axonal migration, producing turning angles of up to 90°. Microfluidic flow simulations indicate that an axon may experience significant bending force due to cross-flow, which may contribute to the observed axonal turning. This method of flow-based guidance was successfully used to fasciculate one advancing axon onto another, showcasing the potential of this technique to be used for the formation of in vitro neuronal circuits.
Collapse
|
87
|
Freund JB, Vermot J. The wall-stress footprint of blood cells flowing in microvessels. Biophys J 2014; 106:752-62. [PMID: 24507616 DOI: 10.1016/j.bpj.2013.12.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 12/03/2013] [Accepted: 12/11/2013] [Indexed: 02/09/2023] Open
Abstract
It is well known that mechanotransduction of hemodynamic forces mediates cellular processes, particularly those that lead to vascular development and maintenance. Both the strength and space-time character of these forces have been shown to affect remodeling and morphogenesis. However, the role of blood cells in the process remains unclear. We investigate the possibility that in the smallest vessels blood's cellular character of itself will lead to forces fundamentally different than the time-averaged forces usually considered, with fluctuations that may significantly exceed their mean values. This is quantitated through the use of a detailed simulation model of microvessel flow in two principal configurations: a diameter D=6.5 μm tube-a model for small capillaries through which red blood cells flow in single-file-and a D=12 μm tube-a model for a nascent vein or artery through which the cells flow in a confined yet chaotic fashion. Results in both cases show strong sensitivity to the mean flow speed U. Peak stresses exceed their means by greater than a factor of 10 when U/D≲10 s(-1), which corresponds to the inverse relaxation time of a healthy red blood cell. This effect is more significant for smaller D cases. At faster flow rates, including those more commonly observed under normal, nominally static physiological conditions, the peak fluctuations are more comparable with the mean shear stress. Implications for mechanotransduction of hemodynamic forces are discussed.
Collapse
Affiliation(s)
- Jonathan B Freund
- Mechanical Science & Engineering and Aerospace Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801.
| | - Julien Vermot
- IGBMC, CNRS/INSERM/UdS, BP.10142, F-67404 Illkirch, France
| |
Collapse
|
88
|
Kalogirou S, Malissovas N, Moro E, Argenton F, Stainier DYR, Beis D. Intracardiac flow dynamics regulate atrioventricular valve morphogenesis. Cardiovasc Res 2014; 104:49-60. [PMID: 25100766 DOI: 10.1093/cvr/cvu186] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Valvular heart disease is responsible for considerable morbidity and mortality. Cardiac valves develop as the heart contracts, and they function throughout the lifetime of the organism to prevent retrograde blood flow. Their precise morphogenesis is crucial for cardiac function. Zebrafish is an ideal model to investigate cardiac valve development as it allows these studies to be carried out in vivo through non-invasive imaging. Accumulating evidence suggests a role for contractility and intracardiac flow dynamics in cardiac valve development. However, these two factors have proved difficult to uncouple, especially since altering myocardial function affects the intracardiac flow pattern. METHODS AND RESULTS Here, we describe novel zebrafish models of developmental valve defects. We identified two mutant alleles of myosin heavy chain 6 that can be raised to adulthood despite having only one functional chamber-the ventricle. The adult mutant ventricle undergoes remodelling, and the atrioventricular (AV) valves fail to form four cuspids. In parallel, we characterized a novel mutant allele of southpaw, a nodal-related gene involved in the establishment of left-right asymmetry, which exhibits randomized heart and endoderm positioning. We first observed that in southpaw mutants the relative position of the two cardiac chambers is altered, affecting the geometry of the heart, while myocardial function appears unaffected. Mutant hearts that loop properly or exhibit situs inversus develop normally, whereas midline, unlooped hearts exhibit defects in their transvalvular flow pattern during AV valve development as well as defects in valve morphogenesis. CONCLUSION Our data indicate that intracardiac flow dynamics regulate valve morphogenesis independently of myocardial contractility.
Collapse
Affiliation(s)
- Stamatia Kalogirou
- Developmental Biology, Biomedical Research Foundation Academy of Athens, Soranou Ephessiou 4, 11527 Athens, Greece
| | - Nikos Malissovas
- Developmental Biology, Biomedical Research Foundation Academy of Athens, Soranou Ephessiou 4, 11527 Athens, Greece
| | - Enrico Moro
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Didier Y R Stainier
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology and Cardiovascular Research Institute, University of San Francisco, Sans Francisco, CA, USA Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Dimitris Beis
- Developmental Biology, Biomedical Research Foundation Academy of Athens, Soranou Ephessiou 4, 11527 Athens, Greece
| |
Collapse
|
89
|
Biechler SV, Junor L, Evans AN, Eberth JF, Price RL, Potts JD, Yost MJ, Goodwin RL. The impact of flow-induced forces on the morphogenesis of the outflow tract. Front Physiol 2014; 5:225. [PMID: 24987377 PMCID: PMC4060072 DOI: 10.3389/fphys.2014.00225] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/30/2014] [Indexed: 11/20/2022] Open
Abstract
One percent of infants are born with congenital heart disease (CHD), which commonly involves outflow tract (OFT) defects. These infants often require complex surgeries, which are associated with long term adverse remodeling effects, and receive replacement valves with limited strength, biocompatibility, and growth capability. To address these problematic issues, researchers have carried out investigations in valve development and valve mechanics. A longstanding hypothesis is that flow-induced forces regulate fibrous valve development, however, the specific mechanisms behind this mechanotransduction remain unclear. The purpose of this study was to implement an in vitro system of outflow tract development to test the response of embryonic OFT tissues to fluid flow. A dynamic, three-dimensional bioreactor system was used to culture embryonic OFT tissue under different levels of flow as well as the absence of flow. In the absence of flow, OFT tissues took on a more primitive phenotype that is characteristic of early OFT cushion development where widely dispersed mesenchymal cells are surrounded by a sparse, disorganized extracellular matrix (ECM). Whereas OFT tissues subjected to physiologically matched flow formed compact mounds of cells, initated, fibrous ECM development, while prolonged supraphysiological flow resulted in abnormal tissue remodeling. This study indicates that both the timing and magnitude of flow alter cellular processes that determine if OFT precursor tissue undergoes normal or pathological development. Specifically, these experiments showed that flow-generated forces regulate the deposition and localization of fibrous ECM proteins, indicating that mechanosensitive signaling pathways are capable of driving pathological OFT development if flows are not ideal.
Collapse
Affiliation(s)
- Stefanie V Biechler
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Lorain Junor
- Instrumentation Resource Facility, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Ashlie N Evans
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA
| | - John F Eberth
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Robert L Price
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA ; Instrumentation Resource Facility, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Jay D Potts
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA ; Instrumentation Resource Facility, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Michael J Yost
- Department of Surgery, Medical University of South Carolina Columbia, SC, USA
| | - Richard L Goodwin
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA
| |
Collapse
|
90
|
Pozzi P, Sironi L, D'Alfonso L, Bouzin M, Collini M, Chirico G, Pallavicini P, Cotelli F, Foglia EA. Electron multiplying charge-coupled device-based fluorescence cross-correlation spectroscopy for blood velocimetry on zebrafish embryos. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:067007. [PMID: 24946713 DOI: 10.1117/1.jbo.19.6.067007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/19/2014] [Indexed: 05/28/2023]
Abstract
Biomedical issues in vasculogenesis and cardiogenesis require methods to follow hemodynamics with high spatial (micrometers) and time (milliseconds) resolution. At the same time, we need to follow relevant morphogenetic processes on large fields of view. Fluorescence cross-correlation spectroscopy coupled to scanning or wide-field microscopy meets these needs but has limited flexibility in the excitation pattern. To overcome this limitation, we develop here a two-photon two-spots setup coupled to an all-reflective near-infrared (NIR) optimized scanning system and to an electron multiplying charge-coupled device. Two NIR laser spots are spaced at adjustable micron-size distances (1 to 50 μm) by means of a Twyman-Green interferometer and repeatedly scanned on the sample, allowing acquisition of information on flows at 4 ms-3 μm time-space resolution in parallel on an extended field of view. We analyze the effect of nonhomogeneous and variable flow on the cross-correlation function by numerical simulations and show exemplary application of this setup in studies of blood flow in zebrafish embryos in vivo. By coupling the interferometer with the scanning mirrors and by computing the cross-correlation function of fluorescent red blood cells, we are able to map speed patterns in embryos' vessels.
Collapse
Affiliation(s)
- Paolo Pozzi
- Università degli Studi di Milano-Bicocca, Dipartimento di Fisica, Piazza della Scienza 3, I-20126, Milano, Italy
| | - Laura Sironi
- Università degli Studi di Milano-Bicocca, Dipartimento di Fisica, Piazza della Scienza 3, I-20126, Milano, Italy
| | - Laura D'Alfonso
- Università degli Studi di Milano-Bicocca, Dipartimento di Fisica, Piazza della Scienza 3, I-20126, Milano, Italy
| | - Margaux Bouzin
- Università degli Studi di Milano-Bicocca, Dipartimento di Fisica, Piazza della Scienza 3, I-20126, Milano, Italy
| | - Maddalena Collini
- Università degli Studi di Milano-Bicocca, Dipartimento di Fisica, Piazza della Scienza 3, I-20126, Milano, Italy
| | - Giuseppe Chirico
- Università degli Studi di Milano-Bicocca, Dipartimento di Fisica, Piazza della Scienza 3, I-20126, Milano, Italy
| | | | - Franco Cotelli
- Università degli Studi di Milano, Dipartimento di Bioscienze, Via Celoria 26, I-20133, Milano, Italy
| | - Efrem A Foglia
- Università degli Studi di Milano, Dipartimento di Bioscienze, Via Celoria 26, I-20133, Milano, Italy
| |
Collapse
|
91
|
Goetz JG, Steed E, Ferreira RR, Roth S, Ramspacher C, Boselli F, Charvin G, Liebling M, Wyart C, Schwab Y, Vermot J. Endothelial cilia mediate low flow sensing during zebrafish vascular development. Cell Rep 2014; 6:799-808. [PMID: 24561257 DOI: 10.1016/j.celrep.2014.01.032] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 12/20/2013] [Accepted: 01/23/2014] [Indexed: 11/16/2022] Open
Abstract
VIDEO ABSTRACT The pattern of blood flow has long been thought to play a significant role in vascular morphogenesis, yet the flow-sensing mechanism that is involved at early embryonic stages, when flow forces are low, remains unclear. It has been proposed that endothelial cells use primary cilia to sense flow, but this has never been tested in vivo. Here we show, by noninvasive, high-resolution imaging of live zebrafish embryos, that endothelial cilia progressively deflect at the onset of blood flow and that the deflection angle correlates with calcium levels in endothelial cells. We demonstrate that alterations in shear stress, ciliogenesis, or expression of the calcium channel PKD2 impair the endothelial calcium level and both increase and perturb vascular morphogenesis. Altogether, these results demonstrate that endothelial cilia constitute a highly sensitive structure that permits the detection of low shear forces during vascular morphogenesis.
Collapse
Affiliation(s)
- Jacky G Goetz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Emily Steed
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Rita R Ferreira
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Stéphane Roth
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Caroline Ramspacher
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Francesco Boselli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Gilles Charvin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Michael Liebling
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Claire Wyart
- Institut du Cerveau et de la Moelle Épinière (ICM), Hôpital de la Pitié-Salpêtrière, 75013 Paris, France; Inserm UMRS 1127, 75013 Paris, France; CNRS UMR 7225, 75013 Paris, France; UPMC University of Paris 06, 75005 Paris, France
| | - Yannick Schwab
- Electron Microscopy Core Facility, Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France.
| |
Collapse
|
92
|
Anton H, Harlepp S, Ramspacher C, Wu D, Monduc F, Bhat S, Liebling M, Paoletti C, Charvin G, Freund JB, Vermot J. Pulse propagation by a capacitive mechanism drives embryonic blood flow. Development 2013; 140:4426-34. [DOI: 10.1242/dev.096768] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pulsatile flow is a universal feature of the blood circulatory system in vertebrates and can lead to diseases when abnormal. In the embryo, blood flow forces stimulate vessel remodeling and stem cell proliferation. At these early stages, when vessels lack muscle cells, the heart is valveless and the Reynolds number (Re) is low, few details are available regarding the mechanisms controlling pulses propagation in the developing vascular network. Making use of the recent advances in optical-tweezing flow probing approaches, fast imaging and elastic-network viscous flow modeling, we investigated the blood-flow mechanics in the zebrafish main artery and show how it modifies the heart pumping input to the network. The movement of blood cells in the embryonic artery suggests that elasticity of the network is an essential factor mediating the flow. Based on these observations, we propose a model for embryonic blood flow where arteries act like a capacitor in a way that reduces heart effort. These results demonstrate that biomechanics is key in controlling early flow propagation and argue that intravascular elasticity has a role in determining embryonic vascular function.
Collapse
Affiliation(s)
- Halina Anton
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Sebastien Harlepp
- Institut de Physique et de Chimie des Matériaux de Strasbourg, Université de Strasbourg, UMR 7504, 23 rue du Loess, 67034 Strasbourg, France
| | - Caroline Ramspacher
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Dave Wu
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Fabien Monduc
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Sandeep Bhat
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Michael Liebling
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Camille Paoletti
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Gilles Charvin
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | | | - Julien Vermot
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| |
Collapse
|
93
|
Cimetta E, Godier-Furnémont A, Vunjak-Novakovic G. Bioengineering heart tissue for in vitro testing. Curr Opin Biotechnol 2013; 24:926-32. [PMID: 23932513 PMCID: PMC3783612 DOI: 10.1016/j.copbio.2013.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/07/2013] [Accepted: 07/08/2013] [Indexed: 02/07/2023]
Abstract
A classical paradigm of tissue engineering is to grow tissues for implantation by using human stem cells in conjunction with biomaterial scaffolds (templates for tissue formation) and bioreactors (culture systems providing environmental control). A reverse paradigm is now emerging through microphysiological platforms for preclinical testing of drugs and modeling of disease that contain large numbers of very small human tissues. We discuss the biomimetic approach as a common underlying principle and some of the specifics related to the design and utilization of platforms with heart micro-tissues for high-throughput screening in vitro.
Collapse
Affiliation(s)
- Elisa Cimetta
- Columbia University, Department of Biomedical Engineering, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | | | | |
Collapse
|
94
|
Peralta M, Steed E, Harlepp S, González-Rosa JM, Monduc F, Ariza-Cosano A, Cortés A, Rayón T, Gómez-Skarmeta JL, Zapata A, Vermot J, Mercader N. Heartbeat-driven pericardiac fluid forces contribute to epicardium morphogenesis. Curr Biol 2013; 23:1726-35. [PMID: 23954432 DOI: 10.1016/j.cub.2013.07.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/14/2013] [Accepted: 07/01/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Hydrodynamic forces play a central role in organ morphogenesis. The role of blood flow in shaping the developing heart is well established, but the role of fluid forces generated in the pericardial cavity surrounding the heart is unknown. Mesothelial cells lining the pericardium generate the proepicardium (PE), the precursor cell population of the epicardium, the outer layer covering the myocardium, which is essential for its maturation and the formation of the heart valves and coronary vasculature. However, there is no evidence from in vivo studies showing how epicardial precursor cells reach and attach to the heart. RESULTS Using optical tools for real-time analysis in the zebrafish, including high-speed imaging and optical tweezing, we show that the heartbeat generates pericardiac fluid advections that drive epicardium formation. These flow forces trigger PE formation and epicardial progenitor cell release and motion. The pericardial flow also influences the site of PE cell adhesion to the myocardium. We additionally identify novel mesothelial sources of epicardial precursors and show that precursor release and adhesion occur both through pericardiac fluid advections and through direct contact with the myocardium. CONCLUSIONS Two hydrodynamic forces couple cardiac development and function: first, blood flow inside the heart, and second, the pericardial fluid advections outside the heart identified here. This pericardiac fluid flow is essential for epicardium formation and represents the first example of hydrodynamic flow forces controlling organogenesis through an action on mesothelial cells.
Collapse
Affiliation(s)
- Marina Peralta
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares Carlos III, calle Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Abstract
During development, mechanical forces cause changes in size, shape, number, position, and gene expression of cells. They are therefore integral to any morphogenetic processes. Force generation by actin-myosin networks and force transmission through adhesive complexes are two self-organizing phenomena driving tissue morphogenesis. Coordination and integration of forces by long-range force transmission and mechanosensing of cells within tissues produce large-scale tissue shape changes. Extrinsic mechanical forces also control tissue patterning by modulating cell fate specification and differentiation. Thus, the interplay between tissue mechanics and biochemical signaling orchestrates tissue morphogenesis and patterning in development.
Collapse
|
96
|
Bruot N, Cicuta P. Emergence of polar order and cooperativity in hydrodynamically coupled model cilia. J R Soc Interface 2013; 10:20130571. [PMID: 23883957 DOI: 10.1098/rsif.2013.0571] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
As a model of ciliary beat, we use two-state oscillators that have a defined direction of oscillation and have strong synchronization properties. By allowing the direction of oscillation to vary according to the interaction with the fluid, with a timescale longer than the timescale of synchronization, we show in simulations that several oscillators can align in a direction set by the geometrical configuration of the system. In this system, the alignment depends on the state of synchronization of the system, and is therefore linked to the beat pattern of the model cilia. By testing various configurations from two to 64 oscillators, we deduce empirically that, when the synchronization state of neighbouring oscillators is in phase, the angles of the oscillators align in a configuration of high hydrodynamic coupling. In arrays of oscillators that break the planar symmetry, a global direction of alignment emerges reflecting this polarity. In symmetric configurations, where several directions are geometrically equivalent, the array still displays strong internal cooperative behaviour. It also appears that the shape of the array is more important than the lattice type and orientation in determining the preferred direction.
Collapse
Affiliation(s)
- Nicolas Bruot
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, UK
| | | |
Collapse
|
97
|
Lee HJ, Li N, Evans SM, Diaz MF, Wenzel PL. Biomechanical force in blood development: extrinsic physical cues drive pro-hematopoietic signaling. Differentiation 2013; 86:92-103. [PMID: 23850217 DOI: 10.1016/j.diff.2013.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 02/07/2023]
Abstract
The hematopoietic system is dynamic during development and in adulthood, undergoing countless spatial and temporal transitions during the course of one's life. Microenvironmental cues in the many unique hematopoietic niches differ, characterized by distinct soluble molecules, membrane-bound factors, and biophysical features that meet the changing needs of the blood system. Research from the last decade has revealed the importance of substrate elasticity and biomechanical force in determination of stem cell fate. Our understanding of the role of these factors in hematopoiesis is still relatively poor; however, the developmental origin of blood cells from the endothelium provides a model for comparison. Many endothelial mechanical sensors and second messenger systems may also determine hematopoietic stem cell fate, self renewal, and homing behaviors. Further, the intimate contact of hematopoietic cells with mechanosensitive cell types, including osteoblasts, endothelial cells, mesenchymal stem cells, and pericytes, places them in close proximity to paracrine signaling downstream of mechanical signals. The objective of this review is to present an overview of the sensors and intracellular signaling pathways activated by mechanical cues and highlight the role of mechanotransductive pathways in hematopoiesis.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
98
|
Buchanan C, Rylander MN. Microfluidic culture models to study the hydrodynamics of tumor progression and therapeutic response. Biotechnol Bioeng 2013; 110:2063-72. [PMID: 23616255 DOI: 10.1002/bit.24944] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 04/12/2013] [Accepted: 04/17/2013] [Indexed: 02/03/2023]
Abstract
The integration of tissue engineering strategies with microfluidic technologies has enabled the design of in vitro microfluidic culture models that better adapt to morphological changes in tissue structure and function over time. These biomimetic microfluidic scaffolds accurately mimic native 3D microenvironments, as well as permit precise and simultaneous control of chemical gradients, hydrodynamic stresses, and cellular niches within the system. The recent application of microfluidic in vitro culture models to cancer research offers enormous potential to aid in the development of improved therapeutic strategies by supporting the investigation of tumor angiogenesis and metastasis under physiologically relevant flow conditions. The intrinsic material properties and fluid mechanics of microfluidic culture models enable high-throughput anti-cancer drug screening, permit well-defined and controllable input parameters to monitor tumor cell response to various hydrodynamic conditions or treatment modalities, as well as provide a platform for elucidating fundamental mechanisms of tumor physiology. This review highlights recent developments and future applications of microfluidic culture models to study tumor progression and therapeutic targeting under conditions of hydrodynamic stress relevant to the complex tumor microenvironment.
Collapse
Affiliation(s)
- Cara Buchanan
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Lab 340 ICTAS Building I, Stanger Street, Blacksburg, Virginia 24061, USA.
| | | |
Collapse
|
99
|
Wang L, Fu C, Fan H, Du T, Dong M, Chen Y, Jin Y, Zhou Y, Deng M, Gu A, Jing Q, Liu T, Zhou Y. miR-34b regulates multiciliogenesis during organ formation in zebrafish. Development 2013; 140:2755-64. [PMID: 23698347 DOI: 10.1242/dev.092825] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiciliated cells (MCCs) possess multiple motile cilia and are distributed throughout the vertebrate body, performing important physiological functions by regulating fluid movement in the intercellular space. Neither their function during organ development nor the molecular mechanisms underlying multiciliogenesis are well understood. Although dysregulation of members of the miR-34 family plays a key role in the progression of various cancers, the physiological function of miR-34b, especially in regulating organ formation, is largely unknown. Here, we demonstrate that miR-34b expression is enriched in kidney MCCs and the olfactory placode in zebrafish. Inhibiting miR-34b function using morpholino antisense oligonucleotides disrupted kidney proximal tubule convolution and the proper distribution of distal transporting cells and MCCs. Microarray analysis of gene expression, cilia immunostaining and a fluid flow assay revealed that miR-34b is functionally required for the multiciliogenesis of MCCs in the kidney and olfactory placode. We hypothesize that miR-34b regulates kidney morphogenesis by controlling the movement and distribution of kidney MCCs and fluid flow. We found that cmyb was genetically downstream of miR-34b and acted as a key regulator of multiciliogenesis. Elevated expression of cmyb blocked membrane docking of centrioles, whereas loss of cmyb impaired centriole multiplication, both of which resulted in defects in the formation of ciliary bundles. Thus, miR-34b serves as a guardian to maintain the proper level of cmyb expression. In summary, our studies have uncovered an essential role for miR-34b-Cmyb signaling during multiciliogenesis and kidney morphogenesis.
Collapse
Affiliation(s)
- Lei Wang
- Key Laboratory of Stem Cell Biology and State Key Laboratory of Medical Genomics and Laboratory of Development and Diseases, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, and Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Atherosclerotic plaques develop in a nonrandom manner along the vasculature following a hemodynamically determined distribution profile. The pathogenesis of shear stress-induced inflammation and atherosclerotic lesion formation has led to discussions about personalized strategies in prevention and treatment. Recent discoveries involving the tyrosine kinase receptor Tie1 in (1) mechanotransduction, (2) inflammation, and (3) neovascularization have invigorated these efforts. In this review, we present the current understanding on Tie1 and its role in these key components of atherogenesis.
Collapse
Affiliation(s)
- Kel Vin Woo
- Department of Pediatrics, Division of Cardiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|