51
|
Bharathan NK, Dickinson AJG. Desmoplakin is required for epidermal integrity and morphogenesis in the Xenopus laevis embryo. Dev Biol 2019; 450:115-131. [PMID: 30935896 PMCID: PMC6659752 DOI: 10.1016/j.ydbio.2019.03.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
Abstract
Desmoplakin (Dsp) is a unique and critical desmosomal protein, that is integral to epidermal development. However, it is unclear whether this protein is required specifically for epidermal morphogenesis. Using morpholinos or Crispr/Cas9 mutagenesis we decreased the function of Dsp in frog embryos to better understand its role during epidermal development. Dsp morphant and mutant embryos had developmental defects such as epidermal fragility that mimicked what has been reported in mammals. Most importantly, we also uncovered a novel function for Dsp in the morphogenesis of the epidermis in X. laevis. In particular, Dsp is required during the process of radial intercalation where basally located cells move into the outer epidermal layer. Once inserted these newly intercalated cells expand their apical surface and then they differentiate into specific epidermal cell types. Decreased levels of Dsp resulted in the failure of the radially intercalating cells to expand their apical surface, thereby reducing the number of differentiated multiciliated and secretory cells. Such defects correlate with changes in E-cadherin levels and actin and microtubule localization which could explain the defects in apical expansion. A mutated form of Dsp that maintains cell-cell adhesion but eliminates the connections to the cytoskeleton results in the same epidermal morphogenesis defect. These results suggest a specific role for Dsp in the apical expansion of cells during radial intercalation. We have developed a novel system, in the frog, to demonstrate for the first time that desmosomes not only protect against mechanical stress but are also critical for epidermal morphogenesis.
Collapse
Affiliation(s)
- Navaneetha Krishnan Bharathan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, 1101 East Marshall St., Richmond, VA 23219, United States; Department of Cell Biology, Emory University School of Medicine, 615 Michael Street Atlanta, GA 30322, United States
| | - Amanda J G Dickinson
- Department of Biology, Virginia Commonwealth University, 1000 West Cary St., Richmond, VA 23284, United States.
| |
Collapse
|
52
|
Boutin C, Kodjabachian L. Biology of multiciliated cells. Curr Opin Genet Dev 2019; 56:1-7. [DOI: 10.1016/j.gde.2019.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 04/07/2019] [Accepted: 04/14/2019] [Indexed: 01/09/2023]
|
53
|
Ji Y, Chae S, Lee HK, Park I, Kim C, Ismail T, Kim Y, Park JW, Kwon OS, Kang BS, Lee DS, Bae JS, Kim SH, Moon PG, Baek MC, Park MJ, Kil IS, Rhee SG, Kim J, Huh YH, Shin JY, Min KJ, Kwon TK, Jang DG, Woo HA, Kwon T, Park TJ, Lee HS. Peroxiredoxin5 Controls Vertebrate Ciliogenesis by Modulating Mitochondrial Reactive Oxygen Species. Antioxid Redox Signal 2019; 30:1731-1745. [PMID: 30191719 DOI: 10.1089/ars.2018.7507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AIMS Peroxiredoxin5 (Prdx5), a thioredoxin peroxidase, is an antioxidant enzyme that is widely studied for its antioxidant properties and protective roles in neurological and cardiovascular disorders. This study is aimed at investigating the functional significance of Prdx5 in mitochondria and at analyzing its roles in ciliogenesis during the process of vertebrate development. RESULTS We found that several Prdx genes were strongly expressed in multiciliated cells in developing Xenopus embryos, and their peroxidatic functions were crucial for normal cilia development. Depletion of Prdx5 increased levels of cellular reactive oxygen species (ROS), consequently leading to mitochondrial dysfunction and abnormal cilia formation. Proteomic and transcriptomic approaches revealed that excessive ROS accumulation on Prdx5 depletion subsequently reduced the expression level of pyruvate kinase (PK), a key metabolic enzyme in energy production. We further confirmed that the promotor activity of PK was significantly reduced on Prdx5 depletion and that the reduction in PK expression and its promoter activity led to ciliary defects observed in Prdx5-depleted cells. INNOVATION Our data revealed the novel relationship between ROS and Prdx5 and the consequent effects of this interaction on vertebrate ciliogenesis. The normal process of ciliogenesis is interrupted by the Prdx5 depletion, resulting in excessive ROS levels and suggesting cilia as vulnerable targets of ROS. CONCLUSION Prdx5 plays protective roles in mitochondria and is critical for normal cilia development by regulating the levels of ROS. The loss of Prdx5 is associated with excessive production of ROS, resulting in mitochondrial dysfunction and aberrant ciliogenesis.
Collapse
Affiliation(s)
- Yurim Ji
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Soomin Chae
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Hyun-Kyung Lee
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Inji Park
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Chowon Kim
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Tayaba Ismail
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Youni Kim
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Jeen-Woo Park
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Oh-Shin Kwon
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Beom-Sik Kang
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Dong-Seok Lee
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| | - Jong-Sup Bae
- 2 College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University , Daegu, South Korea
| | - Sang-Hyun Kim
- 3 Department of Pharmacology, College of Medicine, Kyungpook National University , Daegu, South Korea
| | - Pyung-Gon Moon
- 4 Department of Molecular Medicine, College of Medicine, Kyungpook National University , Daegu, South Korea
| | - Moon-Chang Baek
- 4 Department of Molecular Medicine, College of Medicine, Kyungpook National University , Daegu, South Korea
| | - Mae-Ja Park
- 5 Department of Anatomy, College of Medicine, Kyungpook National University , Daegu, South Korea
| | - In Sup Kil
- 6 Yonsei Biomedical Research Institute, Yonsei University College of Medicine , Seoul, South Korea
| | - Sue Goo Rhee
- 6 Yonsei Biomedical Research Institute, Yonsei University College of Medicine , Seoul, South Korea
| | - Joon Kim
- 7 Graduate School of Medical Science and Engineering , Taejon, South Korea
| | - Yang Hoon Huh
- 8 Electron Microscopy Center, Korea Basic Science Institute, Cheongju-si , Chungcheongbuk-do, South Korea
| | - Jong-Yeon Shin
- 9 Genomic Medicine Institute, Medical Research Center, Seoul National University , Macrogen, Inc., Seoul, South Korea
| | - Kyoung-Jin Min
- 10 Department of Immunology, School of Medicine, Keimyung University , Daegu, South Korea
| | - Taeg Kyu Kwon
- 10 Department of Immunology, School of Medicine, Keimyung University , Daegu, South Korea
| | - Dong Gil Jang
- 11 School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, South Korea
| | - Hyun Ae Woo
- 12 College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul, South Korea
| | - Taejoon Kwon
- 11 School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, South Korea
| | - Tae Joo Park
- 11 School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, South Korea
| | - Hyun-Shik Lee
- 1 KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University , Daegu, South Korea
| |
Collapse
|
54
|
Planar cell polarity protein Dishevelled 3 (Dvl3) regulates ectoplasmic specialization (ES) dynamics in the testis through changes in cytoskeletal organization. Cell Death Dis 2019; 10:194. [PMID: 30808893 PMCID: PMC6391420 DOI: 10.1038/s41419-019-1394-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/22/2018] [Accepted: 01/24/2019] [Indexed: 01/31/2023]
Abstract
In the mammalian testes, such as in rats, the directional alignment of polarized elongating/elongated spermatids, in particular step 17–19 spermatids, across the plane of seminiferous epithelium resembles planar cell polarity (PCP) found in hair cells of the cochlea. It is obvious that spermatid PCP is necessary to support the simultaneous development of maximal number of elongating/elongated spermatids to sustain the daily production of > 50 million sperm per adult rat. Studies have shown that the testis indeed expresses multiple PCP proteins necessary to support spermatid PCP. Herein, using physiological and biochemical assays, and morphological analysis, and with the technique of RNA interference (RNAi) to knockdown PCP protein Dishevelled (Dvl) 1 (Dvl1), Dvl2, Dvl3, or Dvl1/2/3, Dvl proteins, in particular Dvl3, it was shown that Dvl3 played a crucial role of support Sertoli cell tight junction (TJ)-permeability barrier function through changes in the organization of actin- and microtubule (MT)-based cytoskeletons. More important, an in vivo knockdown of Dvl1/2/3 in the testis, defects of spermatid polarity were remarkably noted across the seminiferous epithelium, concomitant with defects of spermatid adhesion and spermatid transport, leading to considerably defects in spermatogenesis. More important, Dvl1/2/3 triple knockdown in the testis also impeded the organization of actin- and MT-based cytoskeletons owing to disruptive spatial expression of actin- and MT-regulatory proteins. In summary, PCP Dishevelled proteins, in particular, Dvl3 is a regulator of Sertoli cell blood–testis barrier (BTB) and also spermatid PCP function through its effects on the actin- and MT-based cytoskeletons in Sertoli cells.
Collapse
|
55
|
Roman AC, Garrido-Jimenez S, Diaz-Chamorro S, Centeno F, Carvajal-Gonzalez JM. Centriole Positioning: Not Just a Little Dot in the Cell. Results Probl Cell Differ 2019; 67:201-221. [PMID: 31435796 DOI: 10.1007/978-3-030-23173-6_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Organelle positioning as many other morphological parameters in a cell is not random. Centriole positioning as centrosomes or ciliary basal bodies is not an exception to this rule in cell biology. Indeed, centriole positioning is a tightly regulated process that occurs during development, and it is critical for many organs to function properly, not just during development but also in the adulthood. In this book chapter, we overview our knowledge on centriole positioning in different and highly specialized animal cells like photoreceptor or ependymal cells. We will also discuss recent advances in the discovery of molecular pathways involved in this process, mostly related to the cytoskeleton and the cell polarity pathways. And finally, we present quantitative methods that have been used to assess centriole positioning in different cell types although mostly in epithelial cells.
Collapse
Affiliation(s)
- Angel-Carlos Roman
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Sergio Garrido-Jimenez
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Selene Diaz-Chamorro
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Francisco Centeno
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Jose Maria Carvajal-Gonzalez
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain.
| |
Collapse
|
56
|
Garrido-Jimenez S, Roman AC, Alvarez-Barrientos A, Carvajal-Gonzalez JM. Centriole planar polarity assessment in Drosophila wings. Development 2018; 145:dev.169326. [PMID: 30389850 DOI: 10.1242/dev.169326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/26/2018] [Indexed: 01/28/2023]
Abstract
In vertebrates, planar polarization of ciliary basal bodies has been associated with actin polymerization that occurs downstream of the Frizzled-planar cell polarity (Fz-PCP) pathway. In Drosophila wing epithelial cells, which do not have cilia, centrioles also polarize in a Fz-PCP-dependent manner, although the relationship with actin polymerization remains unknown. By combining existing and new quantitative methods, we unexpectedly found that known PCP effectors linked to actin polymerization phenotypes affect neither final centriole polarization nor apical centriole distribution. But actin polymerization is required upstream of Fz-PCP to maintain the centrioles in restricted areas in the apical-most planes of those epithelial cells before and after the actin-based hair is formed. Furthermore, in the absence of proper core Fz-PCP signalling, actin polymerization is insufficient to drive this off-centred centriole migration. Altogether, the results reveal that there are at least two pathways controlling centriole positioning in Drosophila pupal wings - an upstream actin-dependent mechanism involved in centriole distribution that is PCP independent, and an unknown mechanism that links core Fz-PCP and centriole polarization.
Collapse
Affiliation(s)
- Sergio Garrido-Jimenez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Angel-Carlos Roman
- Champalimaud Neuroscience Programme, Avenida de Brasilia, Lisbon 1400-038, Portugal
| | - Alberto Alvarez-Barrientos
- Servicio de Técnicas Aplicadas a las Biociencias (STAB), Universidad de Extremadura, 06071 Badajoz, Spain
| | - Jose Maria Carvajal-Gonzalez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| |
Collapse
|
57
|
Avenarius MR, Jung JY, Askew C, Jones SM, Hunker KL, Azaiez H, Rehman AU, Schraders M, Najmabadi H, Kremer H, Smith RJH, Géléoc GSG, Dolan DF, Raphael Y, Kohrman DC. Grxcr2 is required for stereocilia morphogenesis in the cochlea. PLoS One 2018; 13:e0201713. [PMID: 30157177 PMCID: PMC6114524 DOI: 10.1371/journal.pone.0201713] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/22/2018] [Indexed: 11/18/2022] Open
Abstract
Hearing and balance depend upon the precise morphogenesis and mechanosensory function of stereocilia, the specialized structures on the apical surface of sensory hair cells in the inner ear. Previous studies of Grxcr1 mutant mice indicated a critical role for this gene in control of stereocilia dimensions during development. In this study, we analyzed expression of the paralog Grxcr2 in the mouse and evaluated auditory and vestibular function of strains carrying targeted mutations of the gene. Peak expression of Grxcr2 occurs during early postnatal development of the inner ear and GRXCR2 is localized to stereocilia in both the cochlea and in vestibular organs. Homozygous Grxcr2 deletion mutants exhibit significant hearing loss by 3 weeks of age that is associated with developmental defects in stereocilia bundle orientation and organization. Despite these bundle defects, the mechanotransduction apparatus assembles in relatively normal fashion as determined by whole cell electrophysiological evaluation and FM1-43 uptake. Although Grxcr2 mutants do not exhibit overt vestibular dysfunction, evaluation of vestibular evoked potentials revealed subtle defects of the mutants in response to linear accelerations. In addition, reduced Grxcr2 expression in a hypomorphic mutant strain is associated with progressive hearing loss and bundle defects. The stereocilia localization of GRXCR2, together with the bundle pathologies observed in the mutants, indicate that GRXCR2 plays an intrinsic role in bundle orientation, organization, and sensory function in the inner ear during development and at maturity.
Collapse
Affiliation(s)
- Matthew R. Avenarius
- Department of Otolaryngology/Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Jae-Yun Jung
- Department of Otolaryngology/Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Charles Askew
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, United States of America
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sherri M. Jones
- Department of Communication Sciences and Disorders, East Carolina University, Greenville, North Carolina, United States of America
| | - Kristina L. Hunker
- Department of Otolaryngology/Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Hela Azaiez
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Atteeq U. Rehman
- Section on Human Genetics, Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Margit Schraders
- Hearing & Genes Division, Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hannie Kremer
- Hearing & Genes Division, Department of Otorhinolaryngology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Richard J. H. Smith
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Gwenaëlle S. G. Géléoc
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David F. Dolan
- Department of Otolaryngology/Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Yehoash Raphael
- Department of Otolaryngology/Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - David C. Kohrman
- Department of Otolaryngology/Kresge Hearing Research Institute, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
58
|
Hua K, Ferland RJ. Primary Cilia Reconsidered in the Context of Ciliopathies: Extraciliary and Ciliary Functions of Cilia Proteins Converge on a Polarity theme? Bioessays 2018; 40:e1700132. [PMID: 29882973 PMCID: PMC6239423 DOI: 10.1002/bies.201700132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Once dismissed as vestigial organelles, primary cilia have garnered the interest of scientists, given their importance in development/signaling, and for their implication in a new disease category known as ciliopathies. However, many, if not all, "cilia" proteins also have locations/functions outside of the primary cilium. These extraciliary functions can complicate the interpretation of a particular ciliopathy phenotype: it may be a result of defects at the cilium and/or at extraciliary locations, and it could be broadly related to a unifying cellular process for these proteins, such as polarity. Assembly of a cilium has many similarities to the development of other polarized structures. This evolutionarily preserved process for the assembly of polarized cell structures offers a perspective on how the cilium may have evolved. We hypothesize that cilia proteins are critical for cell polarity, and that core polarity proteins may have been specialized to form various cellular protrusions, including primary cilia.
Collapse
Affiliation(s)
- Kiet Hua
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
- Department of Neurology, Albany Medical College, Albany, New York, USA, 12208
| |
Collapse
|
59
|
Sheng X, Sheng Y, Liu Y, Li X, Shu B, Li D. Effects of FSS on the expression and localization of the core proteins in two Wnt signaling pathways, and their association with ciliogenesis. Int J Mol Med 2018; 42:1809-1818. [PMID: 30015823 PMCID: PMC6108851 DOI: 10.3892/ijmm.2018.3758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 06/08/2018] [Indexed: 12/31/2022] Open
Abstract
Fluid shear stress (FSS) may alter ciliary structures and ciliogenesis, and it has been reported that the Wnt signaling pathway may regulate cilia assembly and disas-sembly. The present study aimed to investigate the effects of FSS on primary cilia, the Wnt/β-catenin and Wnt/PCP signaling pathways, and the association among them. In the present study, human umbilical vein endothelial cells were subjected to FSS of differing velocities for various periods of time using a shear stress device. Subsequently, immunofluorescence and quantitative polymerase chain reaction were used to detect the expression and localization of the following core proteins: β-catenin in the Wnt/β-catenin signaling pathway; and dishevelled segment polarity protein 2 (Dvl2), fuzzy planar cell polarity protein (Fuz) and VANGL planar cell polarity protein 2 (Vangl2) in the Wnt/planar cell polarity (PCP) signaling pathway. Furthermore, the colocalization of Dvl2 with the basal body was analyzed under low FSS and laminar FSS. The results demonstrated that low FSS promoted the expression of Dvl2 and its colocalization with the basal body. Although Fuz expression was decreased with increasing duration of FSS, no visible alterations were detected in its localization, it was ubiquitously localized in the ciliated region. Conversely, the expression of Vangl2 was increased by laminar FSS, and β-catenin was translocated into the nucleus at the early stage of low FSS. These findings suggested that Dvl2 may participate in low FSS-induced ciliogenesis and β-catenin may participate at the early stage, whereas Vangl2 may be associated with laminar FSS-induced cilia disassembly.
Collapse
Affiliation(s)
- Xin Sheng
- Department of Biochemistry, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yan Sheng
- Laboratory of Basic Medical Morphology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yuehua Liu
- Department of Biochemistry, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xiaoqiong Li
- Department of Biochemistry, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Bo Shu
- Department of Biochemistry, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Dayu Li
- Department of Biochemistry, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
60
|
Viau A, Bienaimé F, Lukas K, Todkar AP, Knoll M, Yakulov TA, Hofherr A, Kretz O, Helmstädter M, Reichardt W, Braeg S, Aschman T, Merkle A, Pfeifer D, Dumit VI, Gubler MC, Nitschke R, Huber TB, Terzi F, Dengjel J, Grahammer F, Köttgen M, Busch H, Boerries M, Walz G, Triantafyllopoulou A, Kuehn EW. Cilia-localized LKB1 regulates chemokine signaling, macrophage recruitment, and tissue homeostasis in the kidney. EMBO J 2018; 37:embj.201798615. [PMID: 29925518 PMCID: PMC6068446 DOI: 10.15252/embj.201798615] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 05/13/2018] [Accepted: 05/22/2018] [Indexed: 12/24/2022] Open
Abstract
Polycystic kidney disease (PKD) and other renal ciliopathies are characterized by cysts, inflammation, and fibrosis. Cilia function as signaling centers, but a molecular link to inflammation in the kidney has not been established. Here, we show that cilia in renal epithelia activate chemokine signaling to recruit inflammatory cells. We identify a complex of the ciliary kinase LKB1 and several ciliopathy‐related proteins including NPHP1 and PKD1. At homeostasis, this ciliary module suppresses expression of the chemokine CCL2 in tubular epithelial cells. Deletion of LKB1 or PKD1 in mouse renal tubules elevates CCL2 expression in a cell‐autonomous manner and results in peritubular accumulation of CCR2+ mononuclear phagocytes, promoting a ciliopathy phenotype. Our findings establish an epithelial organelle, the cilium, as a gatekeeper of tissue immune cell numbers. This represents an unexpected disease mechanism for renal ciliopathies and establishes a new model for how epithelial cells regulate immune cells to affect tissue homeostasis.
Collapse
Affiliation(s)
- Amandine Viau
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,INSERM U1151, Institut Necker Enfants Malades, Department of Growth and Signaling, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Frank Bienaimé
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,INSERM U1151, Institut Necker Enfants Malades, Department of Growth and Signaling, Université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Service d'Explorations Fonctionnelles, Hôpital Necker-Enfants Malades, Paris, France
| | - Kamile Lukas
- Renal Department, University Medical Center, Freiburg, Germany
| | | | - Manuel Knoll
- Department of Rheumatology and Clinical Immunology, University Medical Center, Freiburg, Germany
| | - Toma A Yakulov
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexis Hofherr
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Kretz
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Neuroanatomy, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Helmstädter
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wilfried Reichardt
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Medical Physics, Department of Radiology, and Comprehensive Cancer Center, University Medical Center, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simone Braeg
- Renal Department, University Medical Center, Freiburg, Germany
| | - Tom Aschman
- Department of Rheumatology and Clinical Immunology, University Medical Center, Freiburg, Germany
| | - Annette Merkle
- Medical Physics, Department of Radiology, and Comprehensive Cancer Center, University Medical Center, Freiburg, Germany
| | - Dietmar Pfeifer
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Freiburg, Germany
| | - Verónica I Dumit
- Center for Biological Systems Analysis (ZBSA), Core Facility Proteomics, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Marie-Claire Gubler
- INSERM UMR1163, Laboratory of Inherited Kidney Diseases, Necker-Enfants Malades Hospital, Paris, France.,Imagine Institute, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Roland Nitschke
- Center for Biological Systems Analysis (ZBSA), Life Imaging Center, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Center for Biological Signaling Studies (BIOSS), Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Tobias B Huber
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Center for Biological Signaling Studies (BIOSS), Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Fabiola Terzi
- INSERM U1151, Institut Necker Enfants Malades, Department of Growth and Signaling, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Jörn Dengjel
- Center for Biological Systems Analysis (ZBSA), Core Facility Proteomics, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Florian Grahammer
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Köttgen
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hauke Busch
- German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Melanie Boerries
- German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University, Freiburg, Germany
| | - Gerd Walz
- Renal Department, University Medical Center, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Biological Signaling Studies (BIOSS), Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Antigoni Triantafyllopoulou
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Rheumatology and Clinical Immunology, University Medical Center, Freiburg, Germany.,Department of Rheumatology and Clinical Immunology, Charité - University Medical Centre Berlin, Berlin, Germany
| | - E Wolfgang Kuehn
- Renal Department, University Medical Center, Freiburg, Germany .,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Biological Signaling Studies (BIOSS), Albert-Ludwigs-University Freiburg, Freiburg, Germany
| |
Collapse
|
61
|
Concepcion D, Hamada H, Papaioannou VE. Tbx6 controls left-right asymmetry through regulation of Gdf1. Biol Open 2018; 7:bio.032565. [PMID: 29650695 PMCID: PMC5992533 DOI: 10.1242/bio.032565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The Tbx6 transcription factor plays multiple roles during gastrulation, somite formation and body axis determination. One of the notable features of the Tbx6 homozygous mutant phenotype is randomization of left/right axis determination. Cilia of the node are morphologically abnormal, leading to the hypothesis that disrupted nodal flow is the cause of the laterality defect. However, Tbx6 is expressed around but not in the node, leading to uncertainty as to the mechanism of this effect. In this study, we have examined the molecular characteristics of the node and the genetic cascade determining left/right axis determination. We found evidence that a leftward nodal flow is generated in Tbx6 homozygous mutants despite the cilia defect, establishing the initial asymmetric gene expression in Dand5 around the node, but that the transduction of the signal from the node to the left lateral plate mesoderm is disrupted due to lack of expression of the Nodal coligand Gdf1 around the node. Gdf1 was shown to be a downstream target of Tbx6 and a Gdf1 transgene partially rescues the laterality defect. Summary: Tbx6 affects morphology of the cilia of the node, but a leftward nodal flow is still generated. Downstream of nodal flow, Tbx6 regulates the Nodal coligand Gdf1 leading to disruption of left/right axis determination.
Collapse
Affiliation(s)
- Daniel Concepcion
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Hiroshi Hamada
- RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Virginia E Papaioannou
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
62
|
Patnaik SR, Zhang X, Biswas L, Akhtar S, Zhou X, Kusuluri DK, Reilly J, May-Simera H, Chalmers S, McCarron JG, Shu X. RPGR protein complex regulates proteasome activity and mediates store-operated calcium entry. Oncotarget 2018; 9:23183-23197. [PMID: 29796181 PMCID: PMC5955404 DOI: 10.18632/oncotarget.25259] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/07/2018] [Indexed: 11/25/2022] Open
Abstract
Ciliopathies are a group of genetically heterogeneous disorders, characterized by defects in cilia genesis or maintenance. Mutations in the RPGR gene and its interacting partners, RPGRIP1 and RPGRIP1L, cause ciliopathies, but the function of their proteins remains unclear. Here we show that knockdown (KD) of RPGR, RPGRIP1 or RPGRIP1L in hTERT-RPE1 cells results in abnormal actin cytoskeleton organization. The actin cytoskeleton rearrangement is regulated by the small GTPase RhoA via the planar cell polarity (PCP) pathway. RhoA activity was upregulated in the absence of RPGR, RPGRIP1 or RPGRIP1L proteins. In RPGR, RPGRIP1 or RPGRIP1L KD cells, we observed increased levels of DVl2 and DVl3 proteins, the core components of the PCP pathway, due to impaired proteasomal activity. RPGR, RPGRIP1 or RPGRIP1L KD cells treated with thapsigargin (TG), an inhibitor of sarcoendoplasmic reticulum Ca2+- ATPases, showed impaired store-operated Ca2+ entry (SOCE), which is mediated by STIM1 and Orai1 proteins. STIM1 was not localized to the ER-PM junction upon ER store depletion in RPGR, RPGRIP1 or RPGRIP1L KD cells. Our results demonstrate that the RPGR protein complex is required for regulating proteasomal activity and for modulating SOCE, which may contribute to the ciliopathy phenotype.
Collapse
Affiliation(s)
- Sarita Rani Patnaik
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, Scotland
- Institute of Molecular Physiology, Johannes Gutenberg-Universität Mainz, D-55128 Mainz, Germany
| | - Xun Zhang
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, Scotland
| | - Lincoln Biswas
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, Scotland
| | - Saeed Akhtar
- Cornea Research Chair, Department of Optometry, King Saud University, Riyadh 11433, Kingdom of Saudi Arabia
| | - Xinzhi Zhou
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, Scotland
| | - Deva Krupakar Kusuluri
- Institute of Molecular Physiology, Johannes Gutenberg-Universität Mainz, D-55128 Mainz, Germany
| | - James Reilly
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, Scotland
| | - Helen May-Simera
- Institute of Molecular Physiology, Johannes Gutenberg-Universität Mainz, D-55128 Mainz, Germany
| | - Susan Chalmers
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - John G. McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Xinhua Shu
- Department of Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, Scotland
| |
Collapse
|
63
|
Wang S, Liu A, Wu G, Ding HF, Huang S, Nahman S, Dong Z. The CPLANE protein Intu protects kidneys from ischemia-reperfusion injury by targeting STAT1 for degradation. Nat Commun 2018; 9:1234. [PMID: 29581513 PMCID: PMC5964315 DOI: 10.1038/s41467-018-03628-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 03/01/2018] [Indexed: 12/29/2022] Open
Abstract
Intu is known as a ciliogenesis and planar polarity effector (CPLANE) protein. Although roles for Intu have been reported during embryonic development and in the context of developmental disorders, its function and regulation in adult tissues remain poorly understood. Here we show that ablation of Intu specifically in kidney proximal tubules aggravates renal ischemia-reperfusion injury, and leads to defective post-injury ciliogenesis. We identify signal transducer and activator of transcription 1 (STAT1) as a novel interacting partner of Intu. In vitro, Intu and STAT1 colocalize at the centriole/basal body area, and Intu promotes proteasomal degradation of STAT1. During cell stress, Intu expression preserves cilia length and cell viability, and these actions are antagonized by STAT1 expression. Thus, we propose a role for Intu in protecting cells and tissues after injury by targeting STAT1 for degradation and maintaining primary cilia.
Collapse
Affiliation(s)
- Shixuan Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Aimin Liu
- Department of Biology, Eberly College of Sciences, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Han-Fei Ding
- Cancer Center and Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Shuang Huang
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Stanley Nahman
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA. .,Department of Nephrology and Institute of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
64
|
Sim HJ, Kim SH, Myung KJ, Kwon T, Lee HS, Park TJ. Xenopus: An alternative model system for identifying muco-active agents. PLoS One 2018; 13:e0193310. [PMID: 29470529 PMCID: PMC5823443 DOI: 10.1371/journal.pone.0193310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/08/2018] [Indexed: 12/03/2022] Open
Abstract
The airway epithelium in human plays a central role as the first line of defense against environmental contaminants. Most respiratory diseases such as chronic obstructive pulmonary disease (COPD), asthma, and respiratory infections, disturb normal muco-ciliary functions by stimulating the hypersecretion of mucus. Several muco-active agents have been used to treat hypersecretion symptoms in patients. Current muco-active reagents control mucus secretion by modulating either airway inflammation, cholinergic parasympathetic nerve activities or by reducing the viscosity by cleaving crosslinking in mucin and digesting DNAs in mucus. However, none of the current medication regulates mucus secretion by directly targeting airway goblet cells. The major hurdle for screening potential muco-active agents that directly affect the goblet cells, is the unavailability of in vivo model systems suitable for high-throughput screening. In this study, we developed a high-throughput in vivo model system for identifying muco-active reagents using Xenopus laevis embryos. We tested mucus secretion under various conditions and developed a screening strategy to identify potential muco-regulators. Using this novel screening technique, we identified narasin as a potential muco-regulator. Narasin treatment of developing Xenopus embryos significantly reduced mucus secretion. Furthermore, the human lung epithelial cell line, Calu-3, responded similarly to narasin treatment, validating our technique for discovering muco-active reagents.
Collapse
Affiliation(s)
- Hyo Jung Sim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
| | - Sang-Hyun Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung-Jae Myung
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Taejoon Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
| | - Hyun-Shik Lee
- College of Natural Sciences, Kyungpook National University, Daegu, South Korea
- * E-mail: (TJP); (HSL)
| | - Tae Joo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
- * E-mail: (TJP); (HSL)
| |
Collapse
|
65
|
In vitro study of FUZ as a novel potential therapeutic target in non-small-cell lung cancer. Life Sci 2018; 197:91-100. [PMID: 29421438 DOI: 10.1016/j.lfs.2018.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/01/2018] [Accepted: 02/05/2018] [Indexed: 12/15/2022]
Abstract
FUZ is regarded as a planar cell polarity effector that controls multiple cellular processes during vertebrate development. However, the role of FUZ in tumor biology remains poorly studied. Our purpose of this study is to discover the physiological effects and mechanism of FUZ in non-small-cell lung cancer (NSCLC) in vitro. With the help of bioinformatics analysis, we noticed that the expression level of FUZ negatively correlates with prognosis of NSCLC patients. Exogenous FUZ expression markedly promoted cell proliferation of NSCLC cells. The phosphorylation of Erk1/2, STAT3 and related signaling molecules were induced activated after FUZ over-expression. FUZ also plays an important role in cell motility by regulating cell signaling pathways and inducing epithelial to mesenchymal transition (EMT). FUZ promotes EMT along with the up-regulation of N-cadherin, vimentin, Zeb1, Twist1 and decreased level of E-cadherin. Furthermore, we also carried out FUZ directed siRNA treatments to prove the above observations. Knockdown of FUZ resulted in delayed cell growth as well as impaired cell migration and reversed EMT phonotype. Importantly, we reported for the first time that FUZ is a BNIP3-interacting protein. Loss of FUZ resulted in decreased BNIP3 protein level, but no influence on BNIP3 mRNA level, suggesting weakened stability of BNIP3 protein. Overall, our results in vitro show that FUZ is responsible for NSCLC progression and metastasis, suggesting that FUZ can be a potential therapeutic target for NSCLC.
Collapse
|
66
|
Tu F, Sedzinski J, Ma Y, Marcotte EM, Wallingford JB. Protein localization screening in vivo reveals novel regulators of multiciliated cell development and function. J Cell Sci 2018; 131:jcs.206565. [PMID: 29180514 DOI: 10.1242/jcs.206565] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/20/2017] [Indexed: 12/23/2022] Open
Abstract
Multiciliated cells (MCCs) drive fluid flow in diverse tubular organs and are essential for the development and homeostasis of the vertebrate central nervous system, airway and reproductive tracts. These cells are characterized by dozens or hundreds of motile cilia that beat in a coordinated and polarized manner. In recent years, genomic studies have not only elucidated the transcriptional hierarchy for MCC specification but also identified myriad new proteins that govern MCC ciliogenesis, cilia beating and cilia polarization. Interestingly, this burst of genomic data has also highlighted that proteins with no obvious role in cilia do, in fact, have important ciliary functions. Understanding the function of proteins with little prior history of study presents a special challenge, especially when faced with large numbers of such proteins. Here, we define the subcellular localization in MCCs of ∼200 proteins not previously implicated in cilia biology. Functional analyses arising from the screen provide novel links between actin cytoskeleton and MCC ciliogenesis.
Collapse
Affiliation(s)
- Fan Tu
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jakub Sedzinski
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.,The Danish Stem Cell Centre (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yun Ma
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.,The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen University, SunYat-sen University, Guangzhou, P.R. China
| | - Edward M Marcotte
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - John B Wallingford
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
67
|
Burga A, Wang W, Ben-David E, Wolf PC, Ramey AM, Verdugo C, Lyons K, Parker PG, Kruglyak L. A genetic signature of the evolution of loss of flight in the Galapagos cormorant. Science 2018; 356:356/6341/eaal3345. [PMID: 28572335 PMCID: PMC5567675 DOI: 10.1126/science.aal3345] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/13/2017] [Indexed: 01/19/2023]
Abstract
We have a limited understanding of the genetic and molecular basis of evolutionary changes in the size and proportion of limbs. We studied wing and pectoral skeleton reduction leading to flightlessness in the Galapagos cormorant (Phalacrocorax harrisi). We sequenced and de novo assembled the genomes of four cormorant species and applied a predictive and comparative genomics approach to find candidate variants that may have contributed to the evolution of flightlessness. These analyses and cross-species experiments in Caenorhabditis elegans and in chondrogenic cell lines implicated variants in genes necessary for transcriptional regulation and function of the primary cilium. Cilia are essential for Hedgehog signaling, and humans affected by skeletal ciliopathies suffer from premature bone growth arrest, mirroring skeletal features associated with loss of flight.
Collapse
Affiliation(s)
- Alejandro Burga
- Department of Human Genetics, Department of Biological Chemistry, and Howard Hughes Medical Institute, University of California, Los Angeles, CA, USA.
| | - Weiguang Wang
- Departments of Molecular, Cell and Developmental Biology and Orthopaedic Surgery, University of California and Orthopaedic Institute for Children, Los Angeles, CA, USA
| | - Eyal Ben-David
- Department of Human Genetics, Department of Biological Chemistry, and Howard Hughes Medical Institute, University of California, Los Angeles, CA, USA
| | - Paul C Wolf
- Wildlife Services, U.S. Department of Agriculture, Roseburg, OR, USA
| | - Andrew M Ramey
- U.S. Geological Survey Alaska Science Center, Anchorage, AK, USA
| | - Claudio Verdugo
- Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Karen Lyons
- Departments of Molecular, Cell and Developmental Biology and Orthopaedic Surgery, University of California and Orthopaedic Institute for Children, Los Angeles, CA, USA
| | - Patricia G Parker
- Department of Biology and Whitney Harris World Ecology Center, University of Missouri, St. Louis, MO, USA.,WildCare Institute, Saint Louis Zoo, St. Louis, MO, USA
| | - Leonid Kruglyak
- Department of Human Genetics, Department of Biological Chemistry, and Howard Hughes Medical Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
68
|
Apodaca G. Role of Polarity Proteins in the Generation and Organization of Apical Surface Protrusions. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027813. [PMID: 28264821 DOI: 10.1101/cshperspect.a027813] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protruding from the apical surfaces of epithelial cells are specialized structures, including cilia, microplicae, microvilli, and stereocilia. These contribute to epithelial function by cushioning the apical surface, by amplifying its surface area to facilitate nutrient absorption, and by promoting sensory transduction and barrier function. Despite these important roles, and the diseases that result when their formation is perturbed, there remain significant gaps in our understanding of the biogenesis of apical protrusions, or the pathways that promote their organization and orientation once at the apical surface. Here, I review some general aspects of these apical structures, and then discuss our current understanding of their formation and organization with respect to proteins that specify apicobasolateral polarity and planar cell polarity.
Collapse
Affiliation(s)
- Gerard Apodaca
- Department of Medicine Renal-Electrolyte Division and the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
69
|
Zhang W, Taylor SP, Ennis HA, Forlenza KN, Duran I, Li B, Sanchez JAO, Nevarez L, Nickerson DA, Bamshad M, Lachman RS, Krakow D, Cohn DH. Expanding the genetic architecture and phenotypic spectrum in the skeletal ciliopathies. Hum Mutat 2018; 39:152-166. [PMID: 29068549 PMCID: PMC6198324 DOI: 10.1002/humu.23362] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/12/2017] [Accepted: 10/14/2017] [Indexed: 01/26/2023]
Abstract
Defects in the biosynthesis and/or function of primary cilia cause a spectrum of disorders collectively referred to as ciliopathies. A subset of these disorders is distinguished by profound abnormalities of the skeleton that include a long narrow chest with markedly short ribs, extremely short limbs, and polydactyly. These include the perinatal lethal short-rib polydactyly syndromes (SRPS) and the less severe asphyxiating thoracic dystrophy (ATD), Ellis-van Creveld (EVC) syndrome, and cranioectodermal dysplasia (CED) phenotypes. To identify new genes and define the spectrum of mutations in the skeletal ciliopathies, we analyzed 152 unrelated families with SRPS, ATD, and EVC. Causal variants were discovered in 14 genes in 120 families, including one newly associated gene and two genes previously associated with other ciliopathies. These three genes encode components of three different ciliary complexes; FUZ, which encodes a planar cell polarity complex molecule; TRAF3IP1, which encodes an anterograde ciliary transport protein; and LBR, which encodes a nuclear membrane protein with sterol reductase activity. The results established the molecular basis of SRPS type IV, in which mutations were identified in four different ciliary genes. The data provide systematic insight regarding the genotypes associated with a large cohort of these genetically heterogeneous phenotypes and identified new ciliary components required for normal skeletal development.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - S Paige Taylor
- Department of Human Genetics, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Hayley A Ennis
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Kimberly N Forlenza
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Ivan Duran
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), University of Malaga, Malaga, Spain
| | - Bing Li
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Jorge A Ortiz Sanchez
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Lisette Nevarez
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington
- University of Washington Center for Mendelian Genomics, University of Washington, Seattle, Washington
| | - Michael Bamshad
- Department of Genome Sciences, University of Washington, Seattle, Washington
- University of Washington Center for Mendelian Genomics, University of Washington, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, Washington
| | - Ralph S Lachman
- International Skeletal Dysplasia Registry at UCLA, Los Angeles, California
| | - Deborah Krakow
- Department of Human Genetics, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- International Skeletal Dysplasia Registry at UCLA, Los Angeles, California
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Daniel H Cohn
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- International Skeletal Dysplasia Registry at UCLA, Los Angeles, California
| |
Collapse
|
70
|
Ma Y, Sun Y, Jiang L, Zuo K, Chen H, Guo J, Chen F, Lai Y, Shi J. WDPCP regulates the ciliogenesis of human sinonasal epithelial cells in chronic rhinosinusitis. Cytoskeleton (Hoboken) 2017; 74:82-90. [PMID: 28001338 DOI: 10.1002/cm.21351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 11/06/2022]
Abstract
Damage to the mucociliary clearance system is a typical change in the pathogenesis in chronic rhinosinusitis. However, the mechanisms underlying cilia loss remain unclear. WDPCP is a key protein essential for ciliogenesis, and is also an effector of the planar cell polarity signaling system. In this study, we sought to determine the role of WDPCP in cilia loss in patients with chronic rhinosinusitis. We demonstrated the expression of WDPCP in human sinonasal epithelium from patients with chronic rhinosinusitis and control subjects. We also used air-liquid interface to culture primary human sinonasal epithelial cells in-vitro model and to investigate WDPCP function. We then explored links between rhinosinusitis, WDPCP and inflammation. Accompanied with cilia loss, expression of WDPCP in human sinonasal epithelium from patients with chronic rhinosinusitis was decreased significantly compared with control subjects. In vitro study, we found that WDPCP level increased at first, and then decreased. Inhibiting WDPCP expression could lead to the poor quantity and length of cilia with reduced expression of Septin7. Also, Th1 type inflammatory mediators could decrease the expression of WDPCP. In conclusion, inflammatory cytokines cause reduced WDPCP expression, which contributes to impaired ciliogenesis in human rhinosinusitis.
Collapse
Affiliation(s)
- Yun Ma
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Yueqi Sun
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Lijie Jiang
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Kejun Zuo
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Hexin Chen
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Jiebo Guo
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Fenghong Chen
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Yinyan Lai
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Jianbo Shi
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| |
Collapse
|
71
|
Agbu SO, Liang Y, Liu A, Anderson KV. The small GTPase RSG1 controls a final step in primary cilia initiation. J Cell Biol 2017; 217:413-427. [PMID: 29038301 PMCID: PMC5748968 DOI: 10.1083/jcb.201604048] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/18/2016] [Accepted: 09/21/2017] [Indexed: 12/11/2022] Open
Abstract
Primary cilia are essential for normal development and tissue homeostasis, but the mechanisms that remodel the centriole to promote cilia initiation are not well understood. Agbu et al. report that mouse RSG1, a small GTPase, regulates a late step in cilia initiation, downstream of TTBK2 and the CPLANE protein INTU. Primary cilia, which are essential for normal development and tissue homeostasis, are extensions of the mother centriole, but the mechanisms that remodel the centriole to promote cilia initiation are poorly understood. Here we show that mouse embryos that lack the small guanosine triphosphatase RSG1 die at embryonic day 12.5, with developmental abnormalities characteristic of decreased cilia-dependent Hedgehog signaling. Rsg1 mutant embryos have fewer primary cilia than wild-type embryos, but the cilia that form are of normal length and traffic Hedgehog pathway proteins within the cilium correctly. Rsg1 mother centrioles recruit proteins required for cilia initiation and dock onto ciliary vesicles, but axonemal microtubules fail to elongate normally. RSG1 localizes to the mother centriole in a process that depends on tau tubulin kinase 2 (TTBK2), the CPLANE complex protein Inturned (INTU), and its own GTPase activity. The data suggest a specific role for RSG1 in the final maturation of the mother centriole and ciliary vesicle that allows extension of the ciliary axoneme.
Collapse
Affiliation(s)
- Stephanie O Agbu
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.,Biochemistry, Cell and Molecular Biology Program, Weill Graduate School of Medical Sciences of Cornell University, New York, NY
| | - Yinwen Liang
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Aimin Liu
- Department of Biology, Eberly College of Science, The Pennsylvania State University, University Park, PA
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
72
|
hmmr mediates anterior neural tube closure and morphogenesis in the frog Xenopus. Dev Biol 2017; 430:188-201. [PMID: 28778799 DOI: 10.1016/j.ydbio.2017.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 12/20/2022]
Abstract
Development of the central nervous system requires orchestration of morphogenetic processes which drive elevation and apposition of the neural folds and their fusion into a neural tube. The newly formed tube gives rise to the brain in anterior regions and continues to develop into the spinal cord posteriorly. Conspicuous differences between the anterior and posterior neural tube become visible already during neural tube closure (NTC). Planar cell polarity (PCP)-mediated convergent extension (CE) movements are restricted to the posterior neural plate, i.e. hindbrain and spinal cord, where they propagate neural fold apposition. The lack of CE in the anterior neural plate correlates with a much slower mode of neural fold apposition anteriorly. The morphogenetic processes driving anterior NTC have not been addressed in detail. Here, we report a novel role for the breast cancer susceptibility gene and microtubule (MT) binding protein Hmmr (Hyaluronan-mediated motility receptor, RHAMM) in anterior neurulation and forebrain development in Xenopus laevis. Loss of hmmr function resulted in a lack of telencephalic hemisphere separation, arising from defective roof plate formation, which in turn was caused by impaired neural tissue narrowing. hmmr regulated polarization of neural cells, a function which was dependent on the MT binding domains. hmmr cooperated with the core PCP component vangl2 in regulating cell polarity and neural morphogenesis. Disrupted cell polarization and elongation in hmmr and vangl2 morphants prevented radial intercalation (RI), a cell behavior essential for neural morphogenesis. Our results pinpoint a novel role of hmmr in anterior neural development and support the notion that RI is a major driving force for anterior neurulation and forebrain morphogenesis.
Collapse
|
73
|
Bryja V, Červenka I, Čajánek L. The connections of Wnt pathway components with cell cycle and centrosome: side effects or a hidden logic? Crit Rev Biochem Mol Biol 2017; 52:614-637. [PMID: 28741966 DOI: 10.1080/10409238.2017.1350135] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Wnt signaling cascade has developed together with multicellularity to orchestrate the development and homeostasis of complex structures. Wnt pathway components - such as β-catenin, Dishevelled (DVL), Lrp6, and Axin-- are often dedicated proteins that emerged in evolution together with the Wnt signaling cascade and are believed to function primarily in the Wnt cascade. It is interesting to see that in recent literature many of these proteins are connected with cellular functions that are more ancient and not limited to multicellular organisms - such as cell cycle regulation, centrosome biology, or cell division. In this review, we summarize the recent literature describing this crosstalk. Specifically, we attempt to find the answers to the following questions: Is the response to Wnt ligands regulated by the cell cycle? Is the centrosome and/or cilium required to activate the Wnt pathway? How do Wnt pathway components regulate the centrosomal cycle and cilia formation and function? We critically review the evidence that describes how these connections are regulated and how they help to integrate cell-to-cell communication with the cell and the centrosomal cycle in order to achieve a fine-tuned, physiological response.
Collapse
Affiliation(s)
- Vítězslav Bryja
- a Department of Experimental Biology, Faculty of Science , Masaryk University , Brno , Czech Republic
| | - Igor Červenka
- b Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology , Karolinska Institutet , Stockholm , Sweden
| | - Lukáš Čajánek
- c Department of Histology and Embryology, Faculty of Medicine , Masaryk University , Brno , Czech Republic
| |
Collapse
|
74
|
Seven pass Cadherins CELSR1-3. Semin Cell Dev Biol 2017; 69:102-110. [PMID: 28716607 DOI: 10.1016/j.semcdb.2017.07.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 11/20/2022]
Abstract
Cadherin EGF LAG seven-pass G-type receptors 1, 2 and 3 (CELSR1-3) form a family of three atypical cadherins with multiple functions in epithelia and in the nervous system. During the past decade, evidence has accumulated for a key role of CELSR1 in epithelial planar cell polarity (PCP), and for CELSR2 and CELSR3 in ciliogenesis and neural development, especially neuron migration and axon guidance in the central, peripheral and enteric nervous systems. Phenotypes in mutant mice indicate that CELSR proteins work in concert with FZD3 and FZD6, but several questions remain. Apart from PCP signaling pathways implicating CELSR1 that begin to be unraveled, little is known about other signals generated by CELSR2 and CELSR3. A crucial question concerns the putative ligands that trigger signaling, in particular what is the role of WNT factors. Another critical issue is the identification of novel intracellular pathways and effectors that relay and transmit signals in receptive cells? Answers to those questions should further our understanding of the role of those important molecules not only in development but also in regeneration and disease.
Collapse
|
75
|
Lee U, Kim SO, Hwang JA, Jang JH, Son S, Ryoo IJ, Ahn JS, Kim BY, Lee KH. The Fungal Metabolite Brefeldin A Inhibits Dvl2-Plk1-Dependent Primary Cilium Disassembly. Mol Cells 2017; 40:401-409. [PMID: 28614913 PMCID: PMC5523016 DOI: 10.14348/molcells.2017.0032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/28/2017] [Accepted: 05/11/2017] [Indexed: 11/27/2022] Open
Abstract
The primary cilium is a non-motile microtubule-based organelle that protrudes from the surface of most human cells and works as a cellular antenna to accept extracellular signals. Primary cilia assemble from the basal body during the resting stage (G0 phase) and simultaneously disassemble with cell cycle re-entry. Defective control of assembly or disassembly causes diverse human diseases including ciliopathy and cancer. To identify the effective compounds for studying primary cilium disassembly, we have screened 297 natural compounds and identified 18 and 17 primary cilium assembly and disassembly inhibitors, respectively. Among them, the application of KY-0120, identified as Brefeldin A, disturbed Dvl2-Plk1-mediated cilium disassembly via repression of the interaction of CK1ɛ-Dvl2 and the expression of Plk1 mRNA. Therefore, our study may suggest useful compounds for studying the cellular mechanism of primary cilium disassembly to prevent ciliopathy and cancer.
Collapse
Affiliation(s)
- Uijeong Lee
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113,
Korea
| | - Sun-Ok Kim
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
| | - Jeong-Ah Hwang
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
- Research Institute of Medical Sciences, Department of Physiology, College of Medicine, Chungnam National University, Daejeon 34134,
Korea
| | - Jae-Hyuk Jang
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113,
Korea
| | - Sangkeun Son
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113,
Korea
| | - In-Ja Ryoo
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
| | - Jong Seog Ahn
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113,
Korea
| | - Bo Yeon Kim
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113,
Korea
| | - Kyung Ho Lee
- World Class Institute (WCI), Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungbuk 28116,
Korea
| |
Collapse
|
76
|
Kumar D, Strenkert D, Patel-King RS, Leonard MT, Merchant SS, Mains RE, King SM, Eipper BA. A bioactive peptide amidating enzyme is required for ciliogenesis. eLife 2017; 6. [PMID: 28513435 PMCID: PMC5461114 DOI: 10.7554/elife.25728] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/15/2017] [Indexed: 02/06/2023] Open
Abstract
The pathways controlling cilium biogenesis in different cell types have not been fully elucidated. We recently identified peptidylglycine α-amidating monooxygenase (PAM), an enzyme required for generating amidated bioactive signaling peptides, in Chlamydomonas and mammalian cilia. Here, we show that PAM is required for the normal assembly of motile and primary cilia in Chlamydomonas, planaria and mice. Chlamydomonas PAM knockdown lines failed to assemble cilia beyond the transition zone, had abnormal Golgi architecture and altered levels of cilia assembly components. Decreased PAM gene expression reduced motile ciliary density on the ventral surface of planaria and resulted in the appearance of cytosolic axonemes lacking a ciliary membrane. The architecture of primary cilia on neuroepithelial cells in Pam-/- mouse embryos was also aberrant. Our data suggest that PAM activity and alterations in post-Golgi trafficking contribute to the observed ciliogenesis defects and provide an unanticipated, highly conserved link between PAM, amidation and ciliary assembly.
Collapse
Affiliation(s)
- Dhivya Kumar
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, United States
| | - Daniela Strenkert
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, United States
| | - Ramila S Patel-King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, United States
| | - Michael T Leonard
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, United States
| | - Sabeeha S Merchant
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, United States.,Institute for Genomics and Proteomics, University of California, Los Angeles, Los Angeles, United States
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, United States
| | - Stephen M King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, United States
| | - Betty A Eipper
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, United States.,Department of Neuroscience, University of Connecticut Health Center, Farmington, United States
| |
Collapse
|
77
|
Yang N, Leung ELH, Liu C, Li L, Eguether T, Jun Yao XJ, Jones EC, Norris DA, Liu A, Clark RA, Roop DR, Pazour GJ, Shroyer KR, Chen J. INTU is essential for oncogenic Hh signaling through regulating primary cilia formation in basal cell carcinoma. Oncogene 2017; 36:4997-5005. [PMID: 28459465 PMCID: PMC5578876 DOI: 10.1038/onc.2017.117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/29/2016] [Accepted: 03/23/2017] [Indexed: 12/30/2022]
Abstract
Inturned (INTU), a cilia and planar polarity effector (CPLANE), performs prominent ciliogenic functions during morphogenesis, such as in the skin. INTU is expressed in adult tissues but its role in tissue maintenance is unknown. Here, we report that the expression of the INTU gene is aberrantly elevated in human basal cell carcinoma (BCC), coinciding with increased primary cilia formation and activated hedgehog (Hh) signaling. Disrupting Intu in an oncogenic mutant Smo (SmoM2)-driven BCC mouse model prevented the formation of BCC through suppressing primary cilia formation and Hh signaling, suggesting that Intu performs a permissive role during BCC formation. INTU is essential for IFT-A complex assembly during ciliogenesis. To further determine whether Intu is directly involved in the activation of Hh signaling downstream of ciliogenesis, we examined the Hh signaling pathway in mouse embryonic fibroblasts, which readily respond to Hh pathway activation. Depleting Intu blocked SAG-induced Hh pathway activation, whereas the expression of Gli2ΔN, a constitutively active Gli2, restored Hh pathway activation in Intu-deficient cells, suggesting that INTU functions upstream of Gli2 activation. In contrast, overexpressing Intu did not promote ciliogenesis or Hh signaling. Taken together, data obtained from this study suggest that INTU is indispensable during BCC tumorigenesis and that its aberrant upregulation is likely a prerequisite for primary cilia formation during Hh-dependent tumorigenesis.
Collapse
Affiliation(s)
- N Yang
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - E L-H Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - C Liu
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - L Li
- Department of Dermatology, Peking Union Medical College Hospital, Beijing, China
| | - T Eguether
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - X-J Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - E C Jones
- Department of Dermatology, Stony Brook University, Stony Brook, NY, USA
| | - D A Norris
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Denver, Aurora, CO, USA
| | - A Liu
- Department of Biology, Eberly College of Science, Pennsylvania State University, University Park, PA, USA
| | - R A Clark
- Department of Dermatology, Stony Brook University, Stony Brook, NY, USA
| | - D R Roop
- Charles C. Gates Center for Regenerative Medicine, University of Colorado Denver, Aurora, CO, USA.,Department of Dermatology, University of Colorado Denver, Aurora, CO, USA
| | - G J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - K R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - J Chen
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.,Department of Dermatology, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
78
|
Abstract
Multiciliated cells are epithelial cells that are in contact with bodily fluids and are required for the proper function of major organs including the brain, the respiratory system and the reproductive tracts. Their multiple motile cilia beat unidirectionally to remove particles of external origin from their surface and/or drive cells or fluids into the lumen of the organs. Multiciliated cells in the brain are produced once, almost exclusively during embryonic development, whereas in respiratory tracts and oviducts they regenerate throughout life. In this Review, we provide a cell-to-organ overview of multiciliated cells and highlight recent studies that have greatly increased our understanding of the mechanisms driving the development and function of these cells in vertebrates. We discuss cell fate determination and differentiation of multiciliated cells, and provide a comprehensive account of their locations and functions in mammals.
Collapse
|
79
|
Jiang T, Kindt K, Wu DK. Transcription factor Emx2 controls stereociliary bundle orientation of sensory hair cells. eLife 2017; 6. [PMID: 28266911 PMCID: PMC5388538 DOI: 10.7554/elife.23661] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 03/04/2017] [Indexed: 12/13/2022] Open
Abstract
The asymmetric location of stereociliary bundle (hair bundle) on the apical surface of mechanosensory hair cells (HCs) dictates the direction in which a given HC can respond to cues such as sound, head movements, and water pressure. Notably, vestibular sensory organs of the inner ear, the maculae, exhibit a line of polarity reversal (LPR) across which, hair bundles are polarized in a mirror-image pattern. Similarly, HCs in neuromasts of the zebrafish lateral line system are generated as pairs, and two sibling HCs develop opposite hair bundle orientations. Within these sensory organs, expression of the transcription factor Emx2 is restricted to only one side of the LPR in the maculae or one of the two sibling HCs in neuromasts. Emx2 mediates hair bundle polarity reversal in these restricted subsets of HCs and generates the mirror-image pattern of the sensory organs. Downstream effectors of Emx2 control bundle polarity cell-autonomously via heterotrimeric G proteins.
Collapse
Affiliation(s)
- Tao Jiang
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, United States.,Laboratory of Molecular Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, United States
| | - Katie Kindt
- Section on Sensory Cell Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, United States
| | - Doris K Wu
- Laboratory of Molecular Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, United States
| |
Collapse
|
80
|
Ulmer B, Tingler M, Kurz S, Maerker M, Andre P, Mönch D, Campione M, Deißler K, Lewandoski M, Thumberger T, Schweickert A, Fainsod A, Steinbeißer H, Blum M. A novel role of the organizer gene Goosecoid as an inhibitor of Wnt/PCP-mediated convergent extension in Xenopus and mouse. Sci Rep 2017; 7:43010. [PMID: 28220837 PMCID: PMC5318956 DOI: 10.1038/srep43010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/18/2017] [Indexed: 12/12/2022] Open
Abstract
Goosecoid (Gsc) expression marks the primary embryonic organizer in vertebrates and beyond. While functions have been assigned during later embryogenesis, the role of Gsc in the organizer has remained enigmatic. Using conditional gain-of-function approaches in Xenopus and mouse to maintain Gsc expression in the organizer and along the axial midline, neural tube closure defects (NTDs) arose and dorsal extension was compromised. Both phenotypes represent convergent extension (CE) defects, arising from impaired Wnt/planar cell polarity (PCP) signaling. Dvl2 recruitment to the cell membrane was inhibited by Gsc in Xenopus animal cap assays and key Wnt/PCP factors (RhoA, Vangl2, Prickle, Wnt11) rescued Gsc-mediated NTDs. Re-evaluation of endogenous Gsc functions in MO-mediated gene knockdown frog and knockout mouse embryos unearthed PCP/CE-related phenotypes as well, including cartilage defects in Xenopus and misalignment of inner ear hair cells in mouse. Our results assign a novel function to Gsc as an inhibitor of Wnt/PCP-mediated CE. We propose that in the organizer Gsc represses CE as well: Gsc-expressing prechordal cells, which leave the organizer first, migrate and do not undergo CE like the Gsc-negative notochordal cells, which subsequently emerge from the organizer. In this model, Gsc provides a switch between cell migration and CE, i.e. cell intercalation.
Collapse
Affiliation(s)
- Bärbel Ulmer
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Melanie Tingler
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Sabrina Kurz
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Markus Maerker
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Philipp Andre
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Dina Mönch
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Marina Campione
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Kirsten Deißler
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Mark Lewandoski
- Genetics of Vertebrate Development Section, Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | | | - Axel Schweickert
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University, Jerusalem 9112102, Israel
| | - Herbert Steinbeißer
- Institute of Human Genetics, University Hospital Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Martin Blum
- University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
81
|
Tabler JM, Rigney MM, Berman GJ, Gopalakrishnan S, Heude E, Al-Lami HA, Yannakoudakis BZ, Fitch RD, Carter C, Vokes S, Liu KJ, Tajbakhsh S, Egnor SR, Wallingford JB. Cilia-mediated Hedgehog signaling controls form and function in the mammalian larynx. eLife 2017; 6. [PMID: 28177282 PMCID: PMC5358977 DOI: 10.7554/elife.19153] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 02/06/2017] [Indexed: 12/30/2022] Open
Abstract
Acoustic communication is fundamental to social interactions among animals, including humans. In fact, deficits in voice impair the quality of life for a large and diverse population of patients. Understanding the molecular genetic mechanisms of development and function in the vocal apparatus is thus an important challenge with relevance both to the basic biology of animal communication and to biomedicine. However, surprisingly little is known about the developmental biology of the mammalian larynx. Here, we used genetic fate mapping to chart the embryological origins of the tissues in the mouse larynx, and we describe the developmental etiology of laryngeal defects in mice with disruptions in cilia-mediated Hedgehog signaling. In addition, we show that mild laryngeal defects correlate with changes in the acoustic structure of vocalizations. Together, these data provide key new insights into the molecular genetics of form and function in the mammalian vocal apparatus. DOI:http://dx.doi.org/10.7554/eLife.19153.001 Nearly all animals communicate using sound. In many cases these sounds are in the form of a voice, which in mammals is generated by a specialized organ in the throat called the larynx. Millions of people throughout the world have voice defects that make it difficult for them to communicate. Such defects are distinct from speech defects such as stuttering, and instead result from an inability to control the pitch or volume of the voice. This has a huge impact because our voice is so central to our quality of life. A wide range of human birth defects that are caused by genetic mutations are known to result in voice problems. These include disorders in which the Hedgehog signaling pathway, which allows cells to exchange information, is defective. Projections called cilia that are found on the outside of many cells transmit Hedgehog signals, and birth defects that affect the cilia (called ciliopathies) also often result in voice problems. Although the shape of the larynx has a crucial effect on voice, relatively little is known about how it develops in embryos. Mice are often studied to investigate how human embryos develop. By studying mouse embryos that had genetic mutations similar to those seen in humans with ciliopathies, Tabler, Rigney et al. now show that many different tissues interact in complex ways to form the larynx. A specific group of cells known as the neural crest was particularly important. The neural crest helps to form the face and skull and an excess of these cells causes face and skull defects in individuals with ciliopathies. Tabler, Rigney et al. show that having too many neural crest cells can also contribute towards defects in the larynx of mice with ciliopathies, despite the larynx being in the neck. Further investigation showed that the Hedgehog signaling pathway was required for the larynx to develop properly. Furthermore, recordings of the vocalizations of the mutant mice showed that they had defective voices, thus linking the defects in the shape of the larynx with changes in the vocalizations that the mice made. Overall, Tabler, Rigney et al. show that mice can be used to investigate how the genes that control the shape of the larynx affect the voice. The next step will be to use mice to investigate other genetic defects that cause voice defects in humans. Further research in other animals could also help us to understand how the larynx has evolved. DOI:http://dx.doi.org/10.7554/eLife.19153.002
Collapse
Affiliation(s)
- Jacqueline M Tabler
- Department of Molecular Biosciences, University of Texas at Austin, Austin, United States
| | - Maggie M Rigney
- Department of Molecular Biosciences, University of Texas at Austin, Austin, United States
| | - Gordon J Berman
- Department of Biology, Emory University, Atlanta, United States
| | - Swetha Gopalakrishnan
- Stem Cells and Development, CNRS UMR3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France
| | - Eglantine Heude
- Stem Cells and Development, CNRS UMR3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France
| | - Hadeel Adel Al-Lami
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London, United Kingdom
| | - Basil Z Yannakoudakis
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London, United Kingdom
| | - Rebecca D Fitch
- Department of Molecular Biosciences, University of Texas at Austin, Austin, United States
| | - Christopher Carter
- Department of Molecular Biosciences, University of Texas at Austin, Austin, United States
| | - Steven Vokes
- Department of Molecular Biosciences, University of Texas at Austin, Austin, United States
| | - Karen J Liu
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London, United Kingdom
| | - Shahragim Tajbakhsh
- Stem Cells and Development, CNRS UMR3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France
| | - Se Roian Egnor
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United states
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, United States
| |
Collapse
|
82
|
Adler PN, Wallingford JB. From Planar Cell Polarity to Ciliogenesis and Back: The Curious Tale of the PPE and CPLANE proteins. Trends Cell Biol 2017; 27:379-390. [PMID: 28153580 DOI: 10.1016/j.tcb.2016.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/28/2016] [Accepted: 12/23/2016] [Indexed: 12/29/2022]
Abstract
Why some genes are more popular than others remains an open question, but one example of this phenomenon involves the genes controlling planar cell polarity (PCP), the polarization of cells within a plane of a tissue. Indeed, the so-called 'core' PCP genes such as dishevelled, frizzled, and prickle have been extensively studied both in animal models and by human genetics. By contrast, other genes that influence PCP signaling have received far less attention. Among the latter are inturned, fuzzy, and fritz, but recent work should bring these once obscure regulators into the limelight. We provide here a brief history of planar polarity effector (PPE) and CPLANE (ciliogenesis and planar polarity effector) proteins, discuss recent advances in understanding their molecular mechanisms of action, and describe their roles in human disease.
Collapse
Affiliation(s)
- Paul N Adler
- Departments of Biology and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
83
|
Sedzinski J, Hannezo E, Tu F, Biro M, Wallingford JB. RhoA regulates actin network dynamics during apical surface emergence in multiciliated epithelial cells. J Cell Sci 2017; 130:420-428. [PMID: 28089989 PMCID: PMC5278671 DOI: 10.1242/jcs.194704] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/10/2016] [Indexed: 12/18/2022] Open
Abstract
Homeostatic replacement of epithelial cells from basal precursors is a multistep process involving progenitor cell specification, radial intercalation and, finally, apical surface emergence. Recent data demonstrate that actin-based pushing under the control of the formin protein Fmn1 drives apical emergence in nascent multiciliated epithelial cells (MCCs), but little else is known about this actin network or the control of Fmn1. Here, we explore the role of the small GTPase RhoA in MCC apical emergence. Disruption of RhoA function reduced the rate of apical surface expansion and decreased the final size of the apical domain. Analysis of cell shapes suggests that RhoA alters the balance of forces exerted on the MCC apical surface. Finally, quantitative time-lapse imaging and fluorescence recovery after photobleaching studies argue that RhoA works in concert with Fmn1 to control assembly of the specialized apical actin network in MCCs. These data provide new molecular insights into epithelial apical surface assembly and could also shed light on mechanisms of apical lumen formation.
Collapse
Affiliation(s)
- Jakub Sedzinski
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Edouard Hannezo
- Cavendish Laboratory, Department of Physics, J.J. Thomson Avenue, University of Cambridge, Cambridge CB3 0HE, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Fan Tu
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Maté Biro
- Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag 6, Newtown, New South Wales 2042, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
84
|
Luxenburg C, Geiger B. Multiscale View of Cytoskeletal Mechanoregulation of Cell and Tissue Polarity. Handb Exp Pharmacol 2017; 235:263-284. [PMID: 27807694 DOI: 10.1007/164_2016_34] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The ability of cells to generate, maintain, and repair tissues with complex architecture, in which distinct cells function as coherent units, relies on polarity cues. Polarity can be described as an asymmetry along a defined axis, manifested at the molecular, structural, and functional levels. Several types of cell and tissue polarities were described in the literature, including front-back, apical-basal, anterior-posterior, and left-right polarity. Extensive research provided insights into the specific regulators of each polarization process, as well as into generic elements that affect all types of polarities. The actin cytoskeleton and the associated adhesion structures are major regulators of most, if not all, known forms of polarity. Actin filaments exhibit intrinsic polarity and their ability to bind many proteins including the mechanosensitive adhesion and motor proteins, such as myosins, play key roles in cell polarization. The actin cytoskeleton can generate mechanical forces and together with the associated adhesions, probe the mechanical, structural, and chemical properties of the environment, and transmit signals that impact numerous biological processes, including cell polarity. In this article we highlight novel mechanisms whereby the mechanical forces and actin-adhesion complexes regulate cell and tissue polarity in a variety of natural and experimental systems.
Collapse
Affiliation(s)
- Chen Luxenburg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
85
|
Abstract
Many animal cells assemble single cilia involved in motile and/or sensory functions. In contrast, multiciliated cells (MCCs) assemble up to 300 motile cilia that beat in a coordinate fashion to generate a directional fluid flow. In the human airways, the brain, and the oviduct, MCCs allow mucus clearance, cerebrospinal fluid circulation, and egg transportation, respectively. Impairment of MCC function leads to chronic respiratory infections and increased risks of hydrocephalus and female infertility. MCC differentiation during development or repair involves the activation of a regulatory cascade triggered by the inhibition of Notch activity in MCC progenitors. The downstream events include the simultaneous assembly of a large number of basal bodies (BBs)-from which cilia are nucleated-in the cytoplasm of the differentiating MCCs, their migration and docking at the plasma membrane associated to an important remodeling of the actin cytoskeleton, and the assembly and polarization of motile cilia. The direction of ciliary beating is coordinated both within cells and at the tissue level by a combination of planar polarity cues affecting BB position and hydrodynamic forces that are both generated and sensed by the cilia. Herein, we review the mechanisms controlling the specification and differentiation of MCCs and BB assembly and organization at the apical surface, as well as ciliary assembly and coordination in MCCs.
Collapse
Affiliation(s)
- Alice Meunier
- Institut de Biologie de l'Ecole Normale Supérieure, Institut National de la Santé et de la Recherche Médicale U1024, Centre National de la Recherche Scientifique UMR8197, 75005 Paris, France
| | - Juliette Azimzadeh
- Institut Jacques Monod, Centre National de la Recherche Scientifique UMR7592, Université Paris-Diderot, 75013 Paris, France
| |
Collapse
|
86
|
Carvajal-Gonzalez JM, Mulero-Navarro S, Mlodzik M. Centriole positioning in epithelial cells and its intimate relationship with planar cell polarity. Bioessays 2016; 38:1234-1245. [PMID: 27774671 DOI: 10.1002/bies.201600154] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Planar cell polarity (PCP)-signaling and associated tissue polarization are evolutionarily conserved. A well documented feature of PCP-signaling in vertebrates is its link to centriole/cilia positioning, although the relationship of PCP and ciliogenesis is still debated. A recent report in Drosophila established that Frizzled (Fz)-PCP core signaling has an instructive input to polarized centriole positioning in non-ciliated Drosophila wing epithelia as a PCP read-out. Here, we review the impact of this observation in the context of recent descriptions of the relationship(s) of core Fz-PCP signaling and cilia/centriole positioning in epithelial and non-epithelial cells. All existing data are consistent with a model where Fz-PCP signaling functions upstream of centriole/cilia positioning, independent of ciliogenesis. The combined data sets indicate that the Fz-Dsh PCP complex is instructive for centriole/ciliary positioning via an actin-based mechanism. Thereby, centriole/cilia/centrosome positioning can be considered an evolutionarily conserved readout and common downstream effect of PCP-signaling from flies to mammals.
Collapse
Affiliation(s)
- Jose Maria Carvajal-Gonzalez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Sonia Mulero-Navarro
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Marek Mlodzik
- Department of Developmental and Regenerative Biology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
87
|
Xu W, Jin M, Hu R, Wang H, Zhang F, Yuan S, Cao Y. The Joubert Syndrome Protein Inpp5e Controls Ciliogenesis by Regulating Phosphoinositides at the Apical Membrane. J Am Soc Nephrol 2016; 28:118-129. [PMID: 27401686 DOI: 10.1681/asn.2015080906] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 05/30/2016] [Indexed: 12/23/2022] Open
Abstract
Phosphoinositides, a family of phosphorylated derivatives of phosphatidylinositol (PtdIns), are tightly regulated both temporally and spatially by PtdIns phosphatases and kinases. Mutations in inositol polyphosphate 5-phosphatase E (INPP5E) cause Joubert syndrome, a human disorder associated with numerous ciliopathic defects, including renal cyst formation, linking phosphoinositides to ciliopathies. However, the molecular mechanism by which INPP5E-mediated PtdIns signaling regulates ciliogenesis and cystogenesis is unclear. Here, we utilized an in vivo vertebrate model of renal cystogenesis to show that Inpp5e enzymatic activity at the apical membrane directs apical docking of basal bodies in renal epithelia. Knockdown or knockout of inpp5e led to ciliogenesis defects and cystic kidneys in zebrafish. Furthermore, knockdown of inpp5e in embryos led to defects in cell polarity, cortical organization of F-actin, and apical segregation of PtdIns(4,5)P2 and PtdIns(3,4,5)P3 Knockdown of the ezrin gene, which encodes an ezrin/radixin/moesin (ERM) protein that crosslinks PtdIns(4,5)P2 and F-actin, phenocopied inpp5e knockdowns. Notably, overexpression of the ezrin gene rescued inpp5e morphants. Finally, treatment with the PI 3-kinase inhibitor LY294002, which decreases PtdIns(3,4,5)P3 levels, rescued the cellular, phenotypic, and renal functional defects in inpp5e-knockdown embryos. Together, our data indicate that Inpp5e functions as a key regulator of cell polarity in the renal epithelia, by inhibiting PtdIns(3,4,5)P3 and subsequently stabilizing PtdIns(4,5)P2 and recruiting Ezrin, F-actin, and basal bodies to the apical membrane, and suggest a possible novel approach for treating human ciliopathies.
Collapse
Affiliation(s)
- Wenyan Xu
- Department of Molecular and Cell Biology, Tongji University School of Life Sciences and Technology, Shanghai, China
| | - Miaomiao Jin
- Department of Molecular and Cell Biology, Tongji University School of Life Sciences and Technology, Shanghai, China
| | - Ruikun Hu
- Department of Molecular and Cell Biology, Tongji University School of Life Sciences and Technology, Shanghai, China
| | - Hong Wang
- Department of Molecular and Cell Biology, Tongji University School of Life Sciences and Technology, Shanghai, China
| | - Fan Zhang
- Department of Molecular and Cell Biology, Tongji University School of Life Sciences and Technology, Shanghai, China
| | - Shiaulou Yuan
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut; and
| | - Ying Cao
- Department of Molecular and Cell Biology, Tongji University School of Life Sciences and Technology, Shanghai, China; .,Tongji University and Shanghai Changzheng Hospital Joint Research Center for Translational Medicine, Changzheng Hospital, Shanghai, China
| |
Collapse
|
88
|
Tabler JM, Rice CP, Liu KJ, Wallingford JB. A novel ciliopathic skull defect arising from excess neural crest. Dev Biol 2016; 417:4-10. [PMID: 27395007 DOI: 10.1016/j.ydbio.2016.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/01/2016] [Indexed: 01/17/2023]
Abstract
The skull is essential for protecting the brain from damage, and birth defects involving disorganization of skull bones are common. However, the developmental trajectories and molecular etiologies by which many craniofacial phenotypes arise remain poorly understood. Here, we report a novel skull defect in ciliopathic Fuz mutant mice in which only a single bone pair encases the forebrain, instead of the usual paired frontal and parietal bones. Through genetic lineage analysis, we show that this defect stems from a massive expansion of the neural crest-derived frontal bone. This expansion occurs at the expense of the mesodermally-derived parietal bones, which are either severely reduced or absent. A similar, though less severe, phenotype was observed in Gli3 mutant mice, consistent with a role for Gli3 in cilia-mediated signaling. Excess crest has also been shown to drive defective palate morphogenesis in ciliopathic mice, and that defect is ameliorated by reduction of Fgf8 gene dosage. Strikingly, skull defects in Fuz mutant mice are also rescued by loss of one allele of fgf8, suggesting a potential route to therapy. In sum, this work is significant for revealing a novel skull defect with a previously un-described developmental etiology and for suggesting a common developmental origin for skull and palate defects in ciliopathies.
Collapse
Affiliation(s)
- Jacqueline M Tabler
- Department of Molecular Biosciences, University of Texas at Austin, United States
| | - Christopher P Rice
- Department of Molecular Biosciences, University of Texas at Austin, United States
| | - Karen J Liu
- Department of Craniofacial Development and Stem Cell Biology, King's College London, UK.
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, United States.
| |
Collapse
|
89
|
Barker AR, McIntosh KV, Dawe HR. Centrosome positioning in non-dividing cells. PROTOPLASMA 2016; 253:1007-1021. [PMID: 26319517 DOI: 10.1007/s00709-015-0883-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 08/22/2015] [Indexed: 06/04/2023]
Abstract
Centrioles and centrosomes are found in almost all eukaryotic cells, where they are important for organising the microtubule cytoskeleton in both dividing and non-dividing cells. The spatial location of centrioles and centrosomes is tightly controlled and, in non-dividing cells, plays an important part in cell migration, ciliogenesis and immune cell functions. Here, we examine some of the ways that centrosomes are connected to other organelles and how this impacts on cilium formation, cell migration and immune cell function in metazoan cells.
Collapse
Affiliation(s)
- Amy R Barker
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, London
| | - Kate V McIntosh
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Helen R Dawe
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
90
|
Park I, Lee HK, Kim C, Ismail T, Kim YK, Park JW, Kwon OS, Kang BS, Lee DS, Park TJ, Park MJ, Choi SC, Lee HS. IFT46 plays crucial roles in craniofacial and cilia development. Biochem Biophys Res Commun 2016; 477:419-25. [PMID: 27320864 DOI: 10.1016/j.bbrc.2016.06.083] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/15/2016] [Indexed: 10/21/2022]
Abstract
The intraflagellar transport (IFT) system is essential for bidirectional movement of ciliary components from the basal body to the tip beneath the ciliary sheath and is conserved for cilia and flagella formation in most vertebrates. IFT complex A is involved in anterograde trafficking, whereas complex B is involved in retrograde trafficking. IFT46 is well known as a crucial component of IFT complex B, however, its developmental functions are poorly understood. In this study, we investigated the novel functions of IFT46 during vertebrate development, especially, ciliogenesis and neurogenesis, because IFT46 is strongly expressed in both multiciliated cells of epithelial and neural tissues. Knockdown of IFT46 using morpholino microinjections caused shortening of the body axis as well as the formation of fewer and shorter cilia. Furthermore, loss of IFT46 down-regulated the expression of the neural plate and neural tube markers, thus may influence Wnt/planar cell polarity and the sonic hedgehog signaling pathway during neurogenesis. In addition, loss of IFT46 caused craniofacial defects by interfering with cartilage formation. In conclusion, our results depict that IFT46 plays important roles in cilia as well as in neural and craniofacial development.
Collapse
Affiliation(s)
- Inji Park
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Hyun-Kyung Lee
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Chowon Kim
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Tayaba Ismail
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Yoo-Kyung Kim
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Jeen-Woo Park
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Oh-Shin Kwon
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Beom Sik Kang
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Dong-Seok Lee
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea
| | - Tae-Joo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea
| | - Mae-Ja Park
- Department of Anatomy, College of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Sun-Cheol Choi
- Department of Biomedical Sciences, University of Ulsan, College of Medicine, Seoul, 05505, South Korea.
| | - Hyun-Shik Lee
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
91
|
Toriyama M, Lee C, Taylor SP, Duran I, Cohn DH, Bruel AL, Tabler JM, Drew K, Kelley MR, Kim S, Park TJ, Braun D, Pierquin G, Biver A, Wagner K, Malfroot A, Panigrahi I, Franco B, Al-lami HA, Yeung Y, Choi YJ, Duffourd Y, Faivre L, Rivière JB, Chen J, Liu KJ, Marcotte EM, Hildebrandt F, Thauvin-Robinet C, Krakow D, Jackson PK, Wallingford JB. The ciliopathy-associated CPLANE proteins direct basal body recruitment of intraflagellar transport machinery. Nat Genet 2016; 48:648-56. [PMID: 27158779 PMCID: PMC4978421 DOI: 10.1038/ng.3558] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/01/2016] [Indexed: 12/21/2022]
Abstract
Cilia use microtubule-based intraflagellar transport (IFT) to organize intercellular signaling. Ciliopathies are a spectrum of human diseases resulting from defects in cilia structure or function. The mechanisms regulating the assembly of ciliary multiprotein complexes and the transport of these complexes to the base of cilia remain largely unknown. Combining proteomics, in vivo imaging and genetic analysis of proteins linked to planar cell polarity (Inturned, Fuzzy and Wdpcp), we identified and characterized a new genetic module, which we term CPLANE (ciliogenesis and planar polarity effector), and an extensive associated protein network. CPLANE proteins physically and functionally interact with the poorly understood ciliopathy-associated protein Jbts17 at basal bodies, where they act to recruit a specific subset of IFT-A proteins. In the absence of CPLANE, defective IFT-A particles enter the axoneme and IFT-B trafficking is severely perturbed. Accordingly, mutation of CPLANE genes elicits specific ciliopathy phenotypes in mouse models and is associated with ciliopathies in human patients.
Collapse
Affiliation(s)
| | - Chanjae Lee
- Dept. of Molecular Biosciences, University of Texas at Austin
| | - S. Paige Taylor
- Departments of Orthopaedic Surgery, Human Genetics and Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ivan Duran
- Departments of Orthopaedic Surgery, Human Genetics and Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Daniel H. Cohn
- Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, California, USA, 90095
| | - Ange-Line Bruel
- EA4271GAD Genetics of Developmental Anomalies, FHU-TRANSLAD, Medecine Faculty, Burgundy University, F-21079 Dijon, France
| | | | - Kevin Drew
- Dept. of Molecular Biosciences, University of Texas at Austin
| | - Marcus R. Kelley
- Stanford University School of Medicine, Baxter Laboratory, Department of Microbiology & Immunology, Stanford, California 94305
| | - Sukyoung Kim
- Dept. of Molecular Biosciences, University of Texas at Austin
| | - Tae Joo Park
- Dept. of Molecular Biosciences, University of Texas at Austin
| | - Daniella Braun
- HHMI and Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Kerstin Wagner
- Cardiological Pediatric unit, Hospital Center, Luxemburg
| | - Anne Malfroot
- Clinic of Pediatric Respiratory Diseases, Infectious Diseases, Travel Clinic and Cystic Fibrosis Clinic at the Universitair Ziekenhuis UZ Brussel, Belgium
| | - Inusha Panigrahi
- Department of Pediatrics Advanced, Pediatric Centre Pigmer, Chandigarh, India
| | - Brunella Franco
- Department of Medical Translational Sciences, Division of Pediatrics, Federico II University of Naples, Italy
- Telethon Institute of Genetics and Medicine-TIGEM , Naples Italy
| | - Hadeel Adel Al-lami
- Dept. of Craniofacial and Stem Cell Biology, Dental Institute, King's College London
| | - Yvonne Yeung
- Dept. of Craniofacial and Stem Cell Biology, Dental Institute, King's College London
| | - Yeon Ja Choi
- Departments of Pathology and Dermatology, Stony Brook University, Stony Brook, NY 11794
| | | | - Yannis Duffourd
- EA4271GAD Genetics of Developmental Anomalies, FHU-TRANSLAD, Medecine Faculty, Burgundy University, F-21079 Dijon, France
| | - Laurence Faivre
- EA4271GAD Genetics of Developmental Anomalies, FHU-TRANSLAD, Medecine Faculty, Burgundy University, F-21079 Dijon, France
- Clinical genetics centre and Eastern referral centre for developmental anomalies and malformative syndromes, FHU-TRANSLAD, Children Hospital, CHU Dijon, F-21079 Dijon, France
| | - Jean-Baptiste Rivière
- EA4271GAD Genetics of Developmental Anomalies, FHU-TRANSLAD, Medecine Faculty, Burgundy University, F-21079 Dijon, France
- Laboratory of Molecular Genetics, FHU-TRANSLAD, PTB, CHU Dijon, F-21079 Dijon, France
| | - Jiang Chen
- Departments of Pathology and Dermatology, Stony Brook University, Stony Brook, NY 11794
| | - Karen J. Liu
- Dept. of Craniofacial and Stem Cell Biology, Dental Institute, King's College London
| | | | - Friedhelm Hildebrandt
- HHMI and Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christel Thauvin-Robinet
- EA4271GAD Genetics of Developmental Anomalies, FHU-TRANSLAD, Medecine Faculty, Burgundy University, F-21079 Dijon, France
- Laboratory of Molecular Genetics, FHU-TRANSLAD, PTB, CHU Dijon, F-21079 Dijon, France
| | - Deborah Krakow
- Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, California, USA, 90095
| | - Peter K. Jackson
- Stanford University School of Medicine, Baxter Laboratory, Department of Microbiology & Immunology, Stanford, California 94305
| | | |
Collapse
|
92
|
Yasunaga T, Hoff S, Schell C, Helmstädter M, Kretz O, Kuechlin S, Yakulov TA, Engel C, Müller B, Bensch R, Ronneberger O, Huber TB, Lienkamp SS, Walz G. The polarity protein Inturned links NPHP4 to Daam1 to control the subapical actin network in multiciliated cells. J Cell Biol 2016; 211:963-73. [PMID: 26644512 PMCID: PMC4674276 DOI: 10.1083/jcb.201502043] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Inturned-mediated complex formation of NPHP4 and DAAM1 is important for ciliogenesis and ciliary function in multiciliated cells, presumably because of its requirement for the local rearrangement of actin cytoskeleton. Motile cilia polarization requires intracellular anchorage to the cytoskeleton; however, the molecular machinery that supports this process remains elusive. We report that Inturned plays a central role in coordinating the interaction between cilia-associated proteins and actin-nucleation factors. We observed that knockdown of nphp4 in multiciliated cells of the Xenopus laevis epidermis compromised ciliogenesis and directional fluid flow. Depletion of nphp4 disrupted the subapical actin layer. Comparison to the structural defects caused by inturned depletion revealed striking similarities. Furthermore, coimmunoprecipitation assays demonstrated that the two proteins interact with each other and that Inturned mediates the formation of ternary protein complexes between NPHP4 and DAAM1. Knockdown of daam1, but not formin-2, resulted in similar disruption of the subapical actin web, whereas nphp4 depletion prevented the association of Inturned with the basal bodies. Thus, Inturned appears to function as an adaptor protein that couples cilia-associated molecules to actin-modifying proteins to rearrange the local actin cytoskeleton.
Collapse
Affiliation(s)
- Takayuki Yasunaga
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Sylvia Hoff
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Christoph Schell
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Martin Helmstädter
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Oliver Kretz
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Neuroanatomy, University of Freiburg, 79104 Freiburg, Germany
| | - Sebastian Kuechlin
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Toma A Yakulov
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Christina Engel
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Barbara Müller
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Robert Bensch
- Department of Computer Science, University of Freiburg, 79110 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Olaf Ronneberger
- Department of Computer Science, University of Freiburg, 79110 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Tobias B Huber
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Soeren S Lienkamp
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| |
Collapse
|
93
|
Schenkelaars Q, Fierro-Constain L, Renard E, Borchiellini C. Retracing the path of planar cell polarity. BMC Evol Biol 2016; 16:69. [PMID: 27039172 PMCID: PMC4818920 DOI: 10.1186/s12862-016-0641-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/22/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The Planar Cell Polarity pathway (PCP) has been described as the main feature involved in patterning cell orientation in bilaterian tissues. Recently, a similar phenomenon was revealed in cnidarians, in which the inhibition of this pathway results in the absence of cilia orientation in larvae, consequently proving the functional conservation of PCP signaling between Cnidaria and Bilateria. Nevertheless, despite the growing accumulation of databases concerning basal lineages of metazoans, very few information concerning the existence of PCP components have been gathered outside of Bilateria and Cnidaria. Thus, the origin of this module or its prevalence in early emerging metazoans has yet to be elucidated. RESULTS The present study addresses this question by investigating the genomes and transcriptomes from all poriferan lineages in addition to Trichoplax (Placozoa) and Mnemiopsis (Ctenophora) genomes for the presence of the core components of this pathway. Our results confirm that several PCP components are metazoan innovations. In addition, we show that all members of the PCP pathway, including a bona fide Strabismus ortholog (Van gogh), are retrieved only in one sponge lineage (Homoscleromorpha) out of four. This highly suggests that the full PCP pathway dates back at least to the emergence of homoscleromorph sponges. Consequently, several secondary gene losses would have occurred in the three other poriferan lineages including Amphimedon queenslandica (Demospongiae). Several proteins were not retrieved either in placozoans or ctenophores leading us to discuss the difficulties to predict orthologous proteins in basally branching animals. Finally, we reveal how the study of multigene families may be helpful to unravel the relationships at the base of the metazoan tree. CONCLUSION The PCP pathway antedates the radiation of Porifera and may have arisen in the last common ancestor of animals. Oscarella species now appear as key organisms to understand the ancestral function of PCP signaling and its potential links with Wnt pathways.
Collapse
Affiliation(s)
- Quentin Schenkelaars
- />Institut Méditerranéen de Biodiversité et d’Ecologie marine et continentale (IMBE UMR 7263), Aix Marseille Université, CNRS, IRD, Avignon Université, Station marine d’Endoume, Batterie des Lions, 13007 Marseille, France
- />Department of Genetics and Evolution, Institute of Genetics and Genomics in Geneva (IGe3), Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Laura Fierro-Constain
- />Institut Méditerranéen de Biodiversité et d’Ecologie marine et continentale (IMBE UMR 7263), Aix Marseille Université, CNRS, IRD, Avignon Université, Station marine d’Endoume, Batterie des Lions, 13007 Marseille, France
| | - Emmanuelle Renard
- />Institut Méditerranéen de Biodiversité et d’Ecologie marine et continentale (IMBE UMR 7263), Aix Marseille Université, CNRS, IRD, Avignon Université, Station marine d’Endoume, Batterie des Lions, 13007 Marseille, France
| | - Carole Borchiellini
- />Institut Méditerranéen de Biodiversité et d’Ecologie marine et continentale (IMBE UMR 7263), Aix Marseille Université, CNRS, IRD, Avignon Université, Station marine d’Endoume, Batterie des Lions, 13007 Marseille, France
- />Department of Genetics and Evolution, Institute of Genetics and Genomics in Geneva (IGe3), Faculty of Sciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
94
|
Positioning of centrioles is a conserved readout of Frizzled planar cell polarity signalling. Nat Commun 2016; 7:11135. [PMID: 27021213 PMCID: PMC4820615 DOI: 10.1038/ncomms11135] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/22/2016] [Indexed: 12/28/2022] Open
Abstract
Planar cell polarity (PCP) signalling is a well-conserved developmental pathway regulating cellular orientation during development. An evolutionarily conserved pathway readout is not established and, moreover, it is thought that PCP mediated cellular responses are tissue-specific. A key PCP function in vertebrates is to regulate coordinated centriole/cilia positioning, a function that has not been associated with PCP in Drosophila. Here we report instructive input of Frizzled-PCP (Fz/PCP) signalling into polarized centriole positioning in Drosophila wings. We show that centrioles are polarized in pupal wing cells as a readout of PCP signalling, with both gain and loss-of-function Fz/PCP signalling affecting centriole polarization. Importantly, loss or gain of centrioles does not affect Fz/PCP establishment, implicating centriolar positioning as a conserved PCP-readout, likely downstream of PCP-regulated actin polymerization. Together with vertebrate data, these results suggest a unifying model of centriole/cilia positioning as a common downstream effect of PCP signalling from flies to mammals. Planar cell polarity (PCP) contributes to cellular orientation during development but how this is regulated in Drosophila is unclear. Here, the authors identify Frizzled-PCP signalling as regulating polarised centriole positioning in the wing disc.
Collapse
|
95
|
Wang S, Livingston MJ, Su Y, Dong Z. Reciprocal regulation of cilia and autophagy via the MTOR and proteasome pathways. Autophagy 2016; 11:607-16. [PMID: 25906314 DOI: 10.1080/15548627.2015.1023983] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Primary cilium is an organelle that plays significant roles in a number of cellular functions ranging from cell mechanosensation, proliferation, and differentiation to apoptosis. Autophagy is an evolutionarily conserved cellular function in biology and indispensable for cellular homeostasis. Both cilia and autophagy have been linked to different types of genetic and acquired human diseases. Their interaction has been suggested very recently, but the underlying mechanisms are still not fully understood. We examined autophagy in cells with suppressed cilia and measured cilium length in autophagy-activated or -suppressed cells. It was found that autophagy was repressed in cells with short cilia. Further investigation showed that MTOR activation was enhanced in cilia-suppressed cells and the MTOR inhibitor rapamycin could largely reverse autophagy suppression. In human kidney proximal tubular cells (HK2), autophagy induction was associated with cilium elongation. Conversely, autophagy inhibition by 3-methyladenine (3-MA) and chloroquine (CQ) as well as bafilomycin A1 (Baf) led to short cilia. Cilia were also shorter in cultured atg5-knockout (KO) cells and in atg7-KO kidney proximal tubular cells in mice. MG132, an inhibitor of the proteasome, could significantly restore cilium length in atg5-KO cells, being concomitant with the proteasome activity. Together, the results suggest that cilia and autophagy regulate reciprocally through the MTOR signaling pathway and ubiquitin-proteasome system.
Collapse
Key Words
- 3-MA, 3-methyladenine
- 70kDa, polypeptide 1
- ANKS6, ankyrin repeat and sterile α motif domain containing 6
- ATG/atg, autophagy-related
- Ac-TUBA, acetylated-tubulin α
- Baf, bafilomycin A1
- CF, confluence
- CQ, chloroquine
- DAPI, 4′-6-diamidino-2-phenylindole
- FBS, fetal bovine serum
- HK2, human kidney proximal tubular cells
- IFT, intraflagellar transport
- KAP3, kinesin family-associated protein 3
- KD, knockdown
- KIF3A/3B, kinesin family member 3A/3B
- KO, knockout
- LTA, lotus tetragonolobus agglutinin
- MAP1LC3B/LC3B, microtubule-associated protein 1 light chain 3 β
- MEF, mouse embryonic fibroblast
- MTOR
- MTOR, mechanistic target of rapamycin
- OFD1, oral-ficial-digital syndrome 1
- PBS, phosphate-buffered saline
- PKD, polycystic kidney disease
- RKRB, Krebs-Henseleit saline containing 25 mM NaHCO3
- RPS6KB1, ribosomal protein S6 kinase
- Rapa, rapamycin
- SD, standard deviation
- autophagy
- cilia
- polycystic kidney disease
- proteasome
Collapse
Affiliation(s)
- Shixuan Wang
- a Department of Cellular Biology and Anatomy ; Medical College of Georgia; Georgia Reagents University and Charlie Norwood VA Medical Center ; Augusta , GA USA
| | | | | | | |
Collapse
|
96
|
Mutations in the Dynein1 Complex are Permissible for Basal Body Migration in Photoreceptors but Alter Rab6 Localization. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 854:209-15. [PMID: 26427413 DOI: 10.1007/978-3-319-17121-0_28] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The photoreceptor outer segment is a specialized primary cilium, and anchoring of the basal body at the apical membrane is required for outer segment formation. We hypothesized that basal body localization and outer segment formation would require the microtubule motor dynein 1 and analyzed the zebrafish cannonball and mike oko mutants, which carry mutations in the heavy chain subunit of cytoplasmic dynein 1 (dync1h1) and the p150(Glued) subunit of Dynactin (dctn1a). The distribution of Rab6, a player in the post-Golgi trafficking of rhodopsin, was also examined. Basal body docking was unaffected in both mutants, but Rab6 expression was reduced. The results suggest that dynein 1 is dispensable for basal body docking but that outer segment defects may be due to defects in post-Golgi trafficking.
Collapse
|
97
|
Ossipova O, Chu CW, Fillatre J, Brott BK, Itoh K, Sokol SY. The involvement of PCP proteins in radial cell intercalations during Xenopus embryonic development. Dev Biol 2015; 408:316-27. [PMID: 26079437 PMCID: PMC4810801 DOI: 10.1016/j.ydbio.2015.06.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 06/10/2015] [Accepted: 06/11/2015] [Indexed: 11/19/2022]
Abstract
The planar cell polarity (PCP) pathway orients cells in diverse epithelial tissues in Drosophila and vertebrate embryos and has been implicated in many human congenital defects and diseases, such as ciliopathies, polycystic kidney disease and malignant cancers. During vertebrate gastrulation and neurulation, PCP signaling is required for convergent extension movements, which are primarily driven by mediolateral cell intercalations, whereas the role for PCP signaling in radial cell intercalations has been unclear. In this study, we examine the function of the core PCP proteins Vangl2, Prickle3 (Pk3) and Disheveled in the ectodermal cells, which undergo radial intercalations during Xenopus gastrulation and neurulation. In the epidermis, multiciliated cell (MCC) progenitors originate in the inner layer, but subsequently migrate to the embryo surface during neurulation. We find that the Vangl2/Pk protein complexes are enriched at the apical domain of intercalating MCCs and are essential for the MCC intercalatory behavior. Addressing the underlying mechanism, we identified KIF13B, as a motor protein that binds Disheveled. KIF13B is required for MCC intercalation and acts synergistically with Vangl2 and Disheveled, indicating that it may mediate microtubule-dependent trafficking of PCP proteins necessary for cell shape regulation. In the neural plate, the Vangl2/Pk complexes were also concentrated near the outermost surface of deep layer cells, suggesting a general role for PCP in radial intercalation. Consistent with this hypothesis, the ectodermal tissues deficient in Vangl2 or Disheveled functions contained more cell layers than normal tissues. We propose that PCP signaling is essential for both mediolateral and radial cell intercalations during vertebrate morphogenesis. These expanded roles underscore the significance of vertebrate PCP proteins as factors contributing to a number of diseases, including neural tube defects, tumor metastases, and various genetic syndromes characterized by abnormal migratory cell behaviors.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chih-Wen Chu
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan Fillatre
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Barbara K Brott
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keiji Itoh
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
98
|
Butler MT, Wallingford JB. Control of vertebrate core planar cell polarity protein localization and dynamics by Prickle 2. Development 2015; 142:3429-39. [PMID: 26293301 PMCID: PMC4631750 DOI: 10.1242/dev.121384] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 08/12/2015] [Indexed: 01/21/2023]
Abstract
Planar cell polarity (PCP) is a ubiquitous property of animal tissues and is essential for morphogenesis and homeostasis. In most cases, this fundamental property is governed by a deeply conserved set of 'core PCP' proteins, which includes the transmembrane proteins Van Gogh-like (Vangl) and Frizzled (Fzd), as well as the cytoplasmic effectors Prickle (Pk) and Dishevelled (Dvl). Asymmetric localization of these proteins is thought to be central to their function, and understanding the dynamics of these proteins is an important challenge in developmental biology. Among the processes that are organized by the core PCP proteins is the directional beating of cilia, such as those in the vertebrate node, airway and brain. Here, we exploit the live imaging capabilities of Xenopus to chart the progressive asymmetric localization of fluorescent reporters of Dvl1, Pk2 and Vangl1 in a planar polarized ciliated epithelium. Using this system, we also characterize the influence of Pk2 on the asymmetric dynamics of Vangl1 at the cell cortex, and we define regions of Pk2 that control its own localization and those impacting Vangl1. Finally, our data reveal a striking uncoupling of Vangl1 and Dvl1 asymmetry. This study advances our understanding of conserved PCP protein functions and also establishes a rapid, tractable platform to facilitate future in vivo studies of vertebrate PCP protein dynamics.
Collapse
Affiliation(s)
- Mitchell T Butler
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas 78712, USA
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas 78712, USA Howard Hughes Medical Institute, University of Texas at Austin, Austin, Texas 78712, USA
| |
Collapse
|
99
|
Vertii A, Bright A, Delaval B, Hehnly H, Doxsey S. New frontiers: discovering cilia-independent functions of cilia proteins. EMBO Rep 2015; 16:1275-87. [PMID: 26358956 DOI: 10.15252/embr.201540632] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022] Open
Abstract
In most vertebrates, mitotic spindles and primary cilia arise from a common origin, the centrosome. In non-cycling cells, the centrosome is the template for primary cilia assembly and, thus, is crucial for their associated sensory and signaling functions. During mitosis, the duplicated centrosomes mature into spindle poles, which orchestrate mitotic spindle assembly, chromosome segregation, and orientation of the cell division axis. Intriguingly, both cilia and spindle poles are centrosome-based, functionally distinct structures that require the action of microtubule-mediated, motor-driven transport for their assembly. Cilia proteins have been found at non-cilia sites, where they have distinct functions, illustrating a diverse and growing list of cellular processes and structures that utilize cilia proteins for crucial functions. In this review, we discuss cilia-independent functions of cilia proteins and re-evaluate their potential contributions to "cilia" disorders.
Collapse
Affiliation(s)
- Anastassiia Vertii
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alison Bright
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Heidi Hehnly
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Stephen Doxsey
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
100
|
Schou KB, Pedersen LB, Christensen ST. Ins and outs of GPCR signaling in primary cilia. EMBO Rep 2015; 16:1099-113. [PMID: 26297609 DOI: 10.15252/embr.201540530] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/01/2015] [Indexed: 12/17/2022] Open
Abstract
Primary cilia are specialized microtubule-based signaling organelles that convey extracellular signals into a cellular response in most vertebrate cell types. The physiological significance of primary cilia is underscored by the fact that defects in assembly or function of these organelles lead to a range of severe diseases and developmental disorders. In most cell types of the human body, signaling by primary cilia involves different G protein-coupled receptors (GPCRs), which transmit specific signals to the cell through G proteins to regulate diverse cellular and physiological events. Here, we provide an overview of GPCR signaling in primary cilia, with main focus on the rhodopsin-like (class A) and the smoothened/frizzled (class F) GPCRs. We describe how such receptors dynamically traffic into and out of the ciliary compartment and how they interact with other classes of ciliary GPCRs, such as class B receptors, to control ciliary function and various physiological and behavioral processes. Finally, we discuss future avenues for developing GPCR-targeted drug strategies for the treatment of ciliopathies.
Collapse
|