101
|
Solecki DJ. Neuronal Polarity Pathways as Central Integrators of Cell-Extrinsic Information During Interactions of Neural Progenitors With Germinal Niches. Front Mol Neurosci 2022; 15:829666. [PMID: 35600073 PMCID: PMC9116468 DOI: 10.3389/fnmol.2022.829666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Germinal niche interactions and their effect on developing neurons have become the subject of intense investigation. Dissecting the complex interplay of cell-extrinsic and cell-intrinsic factors at the heart of these interactions reveals the critical basic mechanisms of neural development and how it goes awry in pediatric neurologic disorders. A full accounting of how developing neurons navigate their niches to mature and integrate into a developing neural circuit requires a combination of genetic characterization of and physical access to neurons and their supporting cell types plus transformative imaging to determine the cell biological and gene-regulatory responses to niche cues. The mouse cerebellar cortex is a prototypical experimental system meeting all of these criteria. The lessons learned therein have been scaled to other model systems and brain regions to stimulate discoveries of how developing neurons make many developmental decisions. This review focuses on how mouse cerebellar granule neuron progenitors interact with signals in their germinal niche and how that affects the neuronal differentiation and cell polarization programs that underpin lamination of the developing cerebellum. We show how modeling of these mechanisms in other systems has added to the growing evidence of how defective neuronal polarity contributes to developmental disease.
Collapse
|
102
|
Masyuk AI, Masyuk TV, Trussoni CE, Pirius NE, LaRusso NF. Autophagy promotes hepatic cystogenesis in polycystic liver disease by depletion of cholangiocyte ciliogenic proteins. Hepatology 2022; 75:1110-1122. [PMID: 34942041 PMCID: PMC9035076 DOI: 10.1002/hep.32298] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUNDS AND AIMS Polycystic liver disease (PLD) is characterized by defective cholangiocyte cilia that regulate progressive growth of hepatic cysts. Because formation of primary cilia is influenced by autophagy through degradation of proteins involved in ciliogenesis, we hypothesized that ciliary defects in PLD cholangiocytes (PLDCs) originate from autophagy-mediated depletion of ciliogenic proteins ADP-ribosylation factor-like protein 3 (ARL3) and ADP-ribosylation factor-like protein 13B (ARL13B) and ARL-dependent mislocation of a ciliary-localized bile acid receptor, Takeda G-protein-coupled receptor 5 (TGR5), the activation of which enhances hepatic cystogenesis (HCG). The aims here were to determine whether: (1) ciliogenesis is impaired in PLDC, is associated with increased autophagy, and involves autophagy-mediated depletion of ARL3 and ARL13B; (2) depletion of ARL3 and ARL13B in PLDC cilia impacts ciliary localization of TGR5; and (3) pharmacological inhibition of autophagy re-establishes cholangiocyte cilia and ciliary localization of ARL3, ARL3B, and TGR5 and reduces HCG. APPROACH AND RESULTS By using liver tissue from healthy persons and patients with PLD, in vitro and in vivo models of PLD, and in vitro models of ciliogenesis, we demonstrated that, in PLDCs: ciliogenesis is impaired; autophagy is enhanced; ARL3 and ARL13B are ubiquitinated by HDAC6, depleted in cilia, and present in autophagosomes; depletion of ARL3 and ARL13B impacts ciliary localization of TGR5; and pharmacological inhibition of autophagy with mefloquine and verteporfin re-establishes cholangiocyte cilia and ciliary localization of ARL3, ARL13B, and TGR5 and reduces HCG. CONCLUSIONS The intersection between autophagy, defective cholangiocyte cilia, and enhanced HCG contributes to PLD progression and can be considered a target for therapeutic interventions.
Collapse
Affiliation(s)
- Anatoliy I. Masyuk
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Tatyana V. Masyuk
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Christy E. Trussoni
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Nicholas E. Pirius
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Nicholas F. LaRusso
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| |
Collapse
|
103
|
Shimada IS, Kato Y. Ciliary signaling in stem cells in health and disease: Hedgehog pathway and beyond. Semin Cell Dev Biol 2022; 129:115-125. [PMID: 35466055 DOI: 10.1016/j.semcdb.2022.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
The primary cilium is a hair-like sensory compartment that protrudes from the cellular surface. The primary cilium is enriched in a variety of signaling molecules that regulate cellular activities. Stem cells have primary cilia. They reside in a specialized environment, called the stem cell niche. This niche contains a variety of secreted factors, and some of their receptors are localized in the primary cilia of stem cells. Here, we summarize the current understanding of the function of cilia in compartmentalized signaling in stem cells. We describe how ciliary signaling regulates stem cells and progenitor cells during development, tissue homeostasis and tumorigenesis. We summarize our understanding of cilia regulated signaling -primary involving the hedgehog pathway- in stem cells in diverse settings that include neuroepithelial cells, radial glia, cerebellar granule neuron precursors, hematopoietic stem cells, hair follicle stem cells, bone marrow mesenchymal stem cells and mammary gland stem cells. Overall, our review highlights a variety of roles that ciliary signaling plays in regulating stem cells throughout life.
Collapse
Affiliation(s)
- Issei S Shimada
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Azakawasumi, Mizuzho-cho, Mizuho-ku, Nagoya, 467-8601 Aichi, Japan.
| | - Yoichi Kato
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Azakawasumi, Mizuzho-cho, Mizuho-ku, Nagoya, 467-8601 Aichi, Japan.
| |
Collapse
|
104
|
Dewees SI, Vargová R, Hardin KR, Turn RE, Devi S, Linnert J, Wolfrum U, Caspary T, Eliáš M, Kahn RA. Phylogenetic profiling and cellular analyses of ARL16 reveal roles in traffic of IFT140 and INPP5E. Mol Biol Cell 2022; 33:ar33. [PMID: 35196065 PMCID: PMC9250359 DOI: 10.1091/mbc.e21-10-0509-t] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/11/2022] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
The ARF family of regulatory GTPases is ancient, with 16 members predicted to have been present in the last eukaryotic common ancestor. Our phylogenetic profiling of paralogues in diverse species identified four family members whose presence correlates with that of a cilium/flagellum: ARL3, ARL6, ARL13, and ARL16. No prior evidence links ARL16 to cilia or other cell functions, despite its presence throughout eukaryotes. Deletion of ARL16 in mouse embryonic fibroblasts (MEFs) results in decreased ciliogenesis yet increased ciliary length. We also found Arl16 knockout (KO) in MEFs to alter ciliary protein content, including loss of ARL13B, ARL3, INPP5E, and the IFT-A core component IFT140. Instead, both INPP5E and IFT140 accumulate at the Golgi in Arl16 KO lines, while other intraflagellar transport (IFT) proteins do not, suggesting a specific defect in traffic from Golgi to cilia. We propose that ARL16 regulates a Golgi-cilia traffic pathway and is required specifically in the export of IFT140 and INPP5E from the Golgi.
Collapse
Affiliation(s)
- Skylar I. Dewees
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Laney Graduate School, Emory University, Atlanta, GA 30307
| | - Romana Vargová
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, CZ-710 00, Ostrava, Czech Republic
| | - Katherine R. Hardin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Laney Graduate School, Emory University, Atlanta, GA 30307
| | - Rachel E. Turn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Laney Graduate School, Emory University, Atlanta, GA 30307
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA 94305-5124
| | - Saroja Devi
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| | - Joshua Linnert
- Institute of Molecular Physiology, Johannes Gutenberg University, Mainz 55128, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University, Mainz 55128, Germany
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Marek Eliáš
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, CZ-710 00, Ostrava, Czech Republic
| | - Richard A. Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
105
|
Djenoune L, Berg K, Brueckner M, Yuan S. A change of heart: new roles for cilia in cardiac development and disease. Nat Rev Cardiol 2022; 19:211-227. [PMID: 34862511 PMCID: PMC10161238 DOI: 10.1038/s41569-021-00635-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/27/2022]
Abstract
Although cardiac abnormalities have been observed in a growing class of human disorders caused by defective primary cilia, the function of cilia in the heart remains an underexplored area. The primary function of cilia in the heart was long thought to be restricted to left-right axis patterning during embryogenesis. However, new findings have revealed broad roles for cilia in congenital heart disease, valvulogenesis, myocardial fibrosis and regeneration, and mechanosensation. In this Review, we describe advances in our understanding of the mechanisms by which cilia function contributes to cardiac left-right axis development and discuss the latest findings that highlight a broader role for cilia in cardiac development. Specifically, we examine the growing line of evidence connecting cilia function to the pathogenesis of congenital heart disease. Furthermore, we also highlight research from the past 10 years demonstrating the role of cilia function in common cardiac valve disorders, including mitral valve prolapse and aortic valve disease, and describe findings that implicate cardiac cilia in mechanosensation potentially linking haemodynamic and contractile forces with genetic regulation of cardiac development and function. Finally, given the presence of cilia on cardiac fibroblasts, we also explore the potential role of cilia in fibrotic growth and summarize the evidence implicating cardiac cilia in heart regeneration.
Collapse
Affiliation(s)
- Lydia Djenoune
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn Berg
- Department of Paediatrics, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Martina Brueckner
- Department of Paediatrics, Yale University School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| | - Shiaulou Yuan
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
106
|
Binó L, Mikulenková E, Štepánek L, Bernatík O, Vysloužil D, Pejšková P, Gorilák P, Huranová M, Varga V, Čajánek L. A protocol for generation and live-cell imaging analysis of primary cilia reporter cell lines. STAR Protoc 2022; 3:101199. [PMID: 35257113 PMCID: PMC8897589 DOI: 10.1016/j.xpro.2022.101199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Primary cilia are hair-like sensory organelles protruding from the surface of most human cells. As cilia are dynamic, several aspects of their biology can only be revealed by real-time analysis in living cells. Here we describe the generation of primary cilia reporter cell lines. Furthermore, we provide a detailed protocol of how to use the reporter cell lines for live-cell imaging microscopy analysis of primary cilia to study their growth as well as intraciliary transport. For complete details on the use and execution of this protocol, please refer to Bernatik et al. (2020) and Pejskova et al. (2020).
Collapse
Affiliation(s)
- Lucia Binó
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czechia
| | - Erika Mikulenková
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czechia
| | - Luděk Štepánek
- Laboratory of Cell Motility, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czechia
| | - Ondřej Bernatík
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czechia
| | - David Vysloužil
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czechia
| | - Petra Pejšková
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czechia
| | - Peter Gorilák
- Laboratory of Cell Motility, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czechia
- Charles University, Faculty of Science, Albertov 6, 128 00 Prague, Czechia
| | - Martina Huranová
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czechia
| | - Vladimír Varga
- Laboratory of Cell Motility, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czechia
| | - Lukáš Čajánek
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 62500 Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czechia
| |
Collapse
|
107
|
Nandamuri SP, Lusk S, Kwan KM. Loss of zebrafish dzip1 results in inappropriate recruitment of periocular mesenchyme to the optic fissure and ocular coloboma. PLoS One 2022; 17:e0265327. [PMID: 35286359 PMCID: PMC8920261 DOI: 10.1371/journal.pone.0265327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/28/2022] [Indexed: 01/13/2023] Open
Abstract
Cilia are essential for the development and function of many different tissues. Although cilia machinery is crucial in the eye for photoreceptor development and function, a role for cilia in early eye development and morphogenesis is still somewhat unclear: many zebrafish cilia mutants retain cilia at early stages due to maternal deposition of cilia components. An eye phenotype has been described in the mouse Arl13 mutant, however, zebrafish arl13b is maternally deposited, and an early role for cilia proteins has not been tested in zebrafish eye development. Here we use the zebrafish dzip1 mutant, which exhibits a loss of cilia throughout stages of early eye development, to examine eye development and morphogenesis. We find that in dzip1 mutants, initial formation of the optic cup proceeds normally, however, the optic fissure subsequently fails to close and embryos develop the structural eye malformation ocular coloboma. Further, neural crest cells, which are implicated in optic fissure closure, do not populate the optic fissure correctly, suggesting that their inappropriate localization may be the underlying cause of coloboma. Overall, our results indicate a role for dzip1 in proper neural crest localization in the optic fissure and optic fissure closure.
Collapse
Affiliation(s)
- Sri Pratima Nandamuri
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States of America
| | - Sarah Lusk
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States of America
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States of America
| |
Collapse
|
108
|
Turn RE, Hu Y, Dewees SI, Devi N, East MP, Hardin KR, Khatib T, Linnert J, Wolfrum U, Lim MJ, Casanova JE, Caspary T, Kahn RA. The ARF GAPs ELMOD1 and ELMOD3 act at the Golgi and cilia to regulate ciliogenesis and ciliary protein traffic. Mol Biol Cell 2022; 33:ar13. [PMID: 34818063 PMCID: PMC9236152 DOI: 10.1091/mbc.e21-09-0443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/11/2022] Open
Abstract
ELMODs are a family of three mammalian paralogues that display GTPase-activating protein (GAP) activity toward a uniquely broad array of ADP-ribosylation factor (ARF) family GTPases that includes ARF-like (ARL) proteins. ELMODs are ubiquitously expressed in mammalian tissues, highly conserved across eukaryotes, and ancient in origin, being present in the last eukaryotic common ancestor. We described functions of ELMOD2 in immortalized mouse embryonic fibroblasts (MEFs) in the regulation of cell division, microtubules, ciliogenesis, and mitochondrial fusion. Here, using similar strategies with the paralogues ELMOD1 and ELMOD3, we identify novel functions and locations of these cell regulators and compare them to those of ELMOD2, allowing the determination of functional redundancy among the family members. We found strong similarities in phenotypes resulting from deletion of either Elmod1 or Elmod3 and marked differences from those arising in Elmod2 deletion lines. Deletion of either Elmod1 or Elmod3 results in the decreased ability of cells to form primary cilia, loss of a subset of proteins from cilia, and accumulation of some ciliary proteins at the Golgi, predicted to result from compromised traffic from the Golgi to cilia. These phenotypes are reversed upon activating mutant expression of either ARL3 or ARL16, linking their roles to ELMOD1/3 actions.
Collapse
Affiliation(s)
- Rachel E. Turn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA 94305
| | - Yihan Hu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Skylar I. Dewees
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
| | - Narra Devi
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| | - Michael P. East
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Katherine R. Hardin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
| | - Tala Khatib
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
| | - Joshua Linnert
- Institute of Molecular Physiology, Johannes Gutenberg University, Mainz 55128, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University, Mainz 55128, Germany
| | - Michael J. Lim
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - James E. Casanova
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Richard A. Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
109
|
Arjona M, Goshayeshi A, Rodriguez-Mateo C, Brett JO, Both P, Ishak H, Rando TA. Tubastatin A maintains adult skeletal muscle stem cells in a quiescent state ex vivo and improves their engraftment ability in vivo. Stem Cell Reports 2022; 17:82-95. [PMID: 35021050 PMCID: PMC8758944 DOI: 10.1016/j.stemcr.2021.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 01/11/2023] Open
Abstract
Adult skeletal muscle stem cells (MuSCs) are important for muscle regeneration and constitute a potential source of cell therapy. However, upon isolation, MuSCs rapidly exit quiescence and lose transplantation potency. Maintenance of the quiescent state in vitro preserves MuSC transplantation efficiency and provides an opportunity to study the biology of quiescence. Here we show that Tubastatin A (TubA), an Hdac6 inhibitor, prevents primary cilium resorption, maintains quiescence, and enhances MuSC survival ex vivo. Phenotypic characterization and transcriptomic analysis of TubA-treated cells revealed that TubA maintains most of the biological features and molecular signatures of quiescence. Furthermore, TubA-treated MuSCs showed improved engraftment ability upon transplantation. TubA also induced a return to quiescence and improved engraftment of cycling MuSCs, revealing a potentially expanded application for MuSC therapeutics. Altogether, these studies demonstrate the ability of TubA to maintain MuSC quiescence ex vivo and to enhance the therapeutic potential of MuSCs and their progeny.
Collapse
Affiliation(s)
- Marina Arjona
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Armon Goshayeshi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Cristina Rodriguez-Mateo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Jamie O Brett
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Stem Cell Biology and Regenerative Medicine Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Pieter Both
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Stem Cell Biology and Regenerative Medicine Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Heather Ishak
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Neurology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
110
|
Li J, Zhang X, Guo J, Yu C, Yang J. Molecular Mechanisms and Risk Factors for the Pathogenesis of Hydrocephalus. Front Genet 2022; 12:777926. [PMID: 35047005 PMCID: PMC8762052 DOI: 10.3389/fgene.2021.777926] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/03/2021] [Indexed: 12/22/2022] Open
Abstract
Hydrocephalus is a neurological condition due to the aberrant circulation and/or obstruction of cerebrospinal fluid (CSF) flow with consequent enlargement of cerebral ventricular cavities. However, it is noticed that a lot of patients may still go through symptomatic progression despite standard shunting procedures, suggesting that hydrocephalus is far more complicated than a simple CSF circulative/obstructive disorder. Growing evidence indicates that genetic factors play a fundamental role in the pathogenesis of some hydrocephalus. Although the genetic research of hydrocephalus in humans is limited, many genetic loci of hydrocephalus have been defined in animal models. In general, the molecular abnormalities involved in the pathogenesis of hydrocephalus include brain development and ependymal cell dysfunction, apoptosis, inflammation, free radical generation, blood flow, and cerebral metabolism. Moreover, recent studies have indicated that the molecular abnormalities relevant to aberrant cerebral glymphatic drainage turn into an attractive subject in the CSF circulation disorder. Furthermore, the prevalent risk factors could facilitate the development of hydrocephalus. In this review, we elicited some possible fundamental molecular mechanisms and facilitating risk factors involved in the pathogenesis of hydrocephalus, and aimed to widen the diagnosis and therapeutic strategies for hydrocephalus management. Such knowledge could be used to improve patient care in different ways, such as early precise diagnosis and effective therapeutic regimens.
Collapse
Affiliation(s)
- Jingwen Li
- Department of Neurosurgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xinjie Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jian Guo
- Department of Neurosurgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chen Yu
- Department of Neurosurgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jun Yang
- Department of Neurosurgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
111
|
Bashford AL, Subramanian V. OUP accepted manuscript. Hum Mol Genet 2022; 31:3245-3265. [PMID: 35470378 PMCID: PMC9523558 DOI: 10.1093/hmg/ddac095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Andrew L Bashford
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Vasanta Subramanian
- To whom correspondence should be addressed. Tel: +44 1225386315; Fax: +44 1225386779;
| |
Collapse
|
112
|
Oh S, Son M, Jang JT, Park CH, Son KH, Byun K. Pyrogallol-Phloroglucinol-6, 6-Bieckol Restored Primary Cilia Length, Which Was Decreased by High-Fat Diet in Visceral Adipose Tissue, and Decreased Adipogenesis. Int J Endocrinol 2022; 2022:8486965. [PMID: 35469126 PMCID: PMC9034920 DOI: 10.1155/2022/8486965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 01/18/2022] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
Length of primary cilia, which involves cell cycle reentry and disassembly of cilia, promotes cell mitosis. It is known that the cilia length in adipose tissue of the high-fat diet (HFD) animals was shortened and accompanied by increased adipogenesis. Male C57BL/6N mice were randomly divided into groups. The mice group was given the normal fat diet (NFD/saline), HFD mice group for 4 weeks, and then HFD was also treated for the next 4 weeks with saline (HFD/saline), Ecklonia cava extract (HFD/ECE), or pyrogallol-phloroglucinol-6, 6-bieckol, a segment of ECE (HFD/PPB). We evaluated the effect of ECE and PPB on modulating cilia length of visceral adipose tissue and decreasing adipogenesis by decreasing cell cycle reentry using an HFD-fed mouse model. ECE and PPB decreased physiological changes, which increased by HFD, but ECE and PPB decreased the upregulation of the IL-6/STAT3/AURKA signaling pathway, which is involved in cilia disassembly. In addition, ECE or PPB elongated the cilia and decreased cyclin A2 and Cdk2 expression, which promote cell cycle reentry, and decreased the adipogenesis genes. PPB and ECE restored cilia length and decreased adipogenesis through modulating the IL-6/STAT3/AURKA pathway and decreasing cell cycle reentry in the visceral adipose tissue of HFD/saline mice group.
Collapse
Affiliation(s)
- Seyeon Oh
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School and Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Myeongjoo Son
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School and Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Department of Anatomy & Cell Biology, Gachon University College of Medicine, Incheon 21936, Republic of Korea
| | - Ji Tae Jang
- Aqua Green Technology Co., Ltd., Smart Bldg., Jeju 63243, Republic of Korea
| | - Chul Hyun Park
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School and Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Department of Anatomy & Cell Biology, Gachon University College of Medicine, Incheon 21936, Republic of Korea
| |
Collapse
|
113
|
Hyland RM, Brody SL. Impact of Motile Ciliopathies on Human Development and Clinical Consequences in the Newborn. Cells 2021; 11:125. [PMID: 35011687 PMCID: PMC8750550 DOI: 10.3390/cells11010125] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Motile cilia are hairlike organelles that project outward from a tissue-restricted subset of cells to direct fluid flow. During human development motile cilia guide determination of the left-right axis in the embryo, and in the fetal and neonatal periods they have essential roles in airway clearance in the respiratory tract and regulating cerebral spinal fluid flow in the brain. Dysregulation of motile cilia is best understood through the lens of the genetic disorder primary ciliary dyskinesia (PCD). PCD encompasses all genetic motile ciliopathies resulting from over 60 known genetic mutations and has a unique but often underrecognized neonatal presentation. Neonatal respiratory distress is now known to occur in the majority of patients with PCD, laterality defects are common, and very rarely brain ventricle enlargement occurs. The developmental function of motile cilia and the effect and pathophysiology of motile ciliopathies are incompletely understood in humans. In this review, we will examine the current understanding of the role of motile cilia in human development and clinical considerations when assessing the newborn for suspected motile ciliopathies.
Collapse
Affiliation(s)
- Rachael M. Hyland
- Department of Pediatrics, Division of Newborn Medicine, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63110,USA;
| | - Steven L. Brody
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
114
|
Bentley-Ford MR, Andersen RS, Croyle MJ, Haycraft CJ, Clearman KR, Foote JB, Reiter JF, Yoder BK. ATXN10 Is Required for Embryonic Heart Development and Maintenance of Epithelial Cell Phenotypes in the Adult Kidney and Pancreas. Front Cell Dev Biol 2021; 9:705182. [PMID: 34970537 PMCID: PMC8712648 DOI: 10.3389/fcell.2021.705182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Atxn10 is a gene known for its role in cytokinesis and is associated with spinocerebellar ataxia (SCA10), a slowly progressing cerebellar syndrome caused by an intragenic pentanucleotide repeat expansion. Atxn10 is also implicated in the ciliopathy syndromes nephronophthisis (NPHP) and Joubert syndrome (JBTS), which are caused by the disruption of cilia function leading to nephron loss, impaired renal function, and cerebellar hypoplasia. How Atxn10 disruption contributes to these disorders remains unknown. Here, we generated Atxn10 congenital and conditional mutant mouse models. Our data indicate that while ATXN10 protein can be detected around the base of the cilium as well as in the cytosol, its loss does not cause overt changes in cilia formation or morphology. Congenital loss of Atxn10 results in embryonic lethality around E10.5 associated with pericardial effusion and loss of trabeculation. Similarly, tissue-specific loss of ATXN10 in the developing endothelium (Tie2-Cre) and myocardium (cTnT-Cre) also results in embryonic lethality with severe cardiac malformations occurring in the latter. Using an inducible Cagg-CreER to disrupt ATXN10 systemically at postnatal stages, we show that ATXN10 is also required for survival in adult mice. Loss of ATXN10 results in severe pancreatic and renal abnormalities leading to lethality within a few weeks post ATXN10 deletion in adult mice. Evaluation of these phenotypes further identified rapid epithelial-to-mesenchymal transition (EMT) in these tissues. In the pancreas, the phenotype includes signs of both acinar to ductal metaplasia and EMT with aberrant cilia formation and severe defects in glucose homeostasis related to pancreatic insufficiency or defects in feeding or nutrient intake. Collectively, this study identifies ATXN10 as an essential protein for survival.
Collapse
Affiliation(s)
- Melissa R. Bentley-Ford
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Reagan S. Andersen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mandy J. Croyle
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Courtney J. Haycraft
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kelsey R. Clearman
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jeremy B. Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jeremy F. Reiter
- Department of Biochemistry and Biophysics, University of Alabama at Birmingham, San Francisco, CA, United States
- Chan Zuckerberg Biohub, San Francisco, San Francisco, CA, United States
| | - Bradley K. Yoder
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
115
|
Ha K, Delling M. Electrophysiological Recordings of the Polycystin Complex in the Primary Cilium of Cultured Mouse IMCD-3 Cell Line. Bio Protoc 2021; 11:e4196. [PMID: 34761068 PMCID: PMC8554804 DOI: 10.21769/bioprotoc.4196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/12/2021] [Accepted: 07/23/2021] [Indexed: 11/02/2022] Open
Abstract
PC-1 and PC-2 form an ion channel complex called the polycystin complex, which predominantly localizes to a small hair-like organelle called the primary cilium. The polycystin complex permeates cations, K+, Na+, and Ca2+, and has an unusual 1:3 stoichiometry that combines one PC-1 subunit with three PC-2 subunits. However, the small size and shape of primary cilia impose technical challenges to study the polycystin complex in its native environment. In this paper, we describe the methodology to directly record ion channel activity in primary cilia. This method will allow a detailed functional characterization of how mutations within the polycystin complex cause Autosomal Dominant Polycystic Kidney Disease (ADPKD), essential to develop novel therapeutics for this ciliopathy.
Collapse
Affiliation(s)
- Kotdaji Ha
- Department of Physiology, UCSF, San Francisco, USA
| | | |
Collapse
|
116
|
Abstract
TRIP6, a member of the ZYXIN-family of LIM domain proteins, is a focal adhesion component. Trip6 deletion in the mouse, reported here, reveals a function in the brain: ependymal and choroid plexus epithelial cells are carrying, unexpectedly, fewer and shorter cilia, are poorly differentiated, and the mice develop hydrocephalus. TRIP6 carries numerous protein interaction domains and its functions require homodimerization. Indeed, TRIP6 disruption in vitro (in a choroid plexus epithelial cell line), via RNAi or inhibition of its homodimerization, confirms its function in ciliogenesis. Using super-resolution microscopy, we demonstrate TRIP6 localization at the pericentriolar material and along the ciliary axoneme. The requirement for homodimerization which doubles its interaction sites, its punctate localization along the axoneme, and its co-localization with other cilia components suggest a scaffold/co-transporter function for TRIP6 in cilia. Thus, this work uncovers an essential role of a LIM-domain protein assembly factor in mammalian ciliogenesis.
Collapse
|
117
|
Chen S, Alhassen W, Vakil Monfared R, Vachirakorntong B, Nauli SM, Baldi P, Alachkar A. Dynamic Changes of Brain Cilia Transcriptomes across the Human Lifespan. Int J Mol Sci 2021; 22:10387. [PMID: 34638726 PMCID: PMC8509004 DOI: 10.3390/ijms221910387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/27/2022] Open
Abstract
Almost all brain cells contain primary cilia, antennae-like microtubule sensory organelles, on their surface, which play critical roles in brain functions. During neurodevelopmental stages, cilia are essential for brain formation and maturation. In the adult brain, cilia play vital roles as signaling hubs that receive and transduce various signals and regulate cell-to-cell communications. These distinct roles suggest that cilia functions, and probably structures, change throughout the human lifespan. To further understand the age-dependent changes in cilia roles, we identified and analyzed age-dependent patterns of expression of cilia's structural and functional components across the human lifespan. We acquired cilia transcriptomic data for 16 brain regions from the BrainSpan Atlas and analyzed the age-dependent expression patterns using a linear regression model by calculating the regression coefficient. We found that 67% of cilia transcripts were differentially expressed genes with age (DEGAs) in at least one brain region. The age-dependent expression was region-specific, with the highest and lowest numbers of DEGAs expressed in the ventrolateral prefrontal cortex and hippocampus, respectively. The majority of cilia DEGAs displayed upregulation with age in most of the brain regions. The transcripts encoding cilia basal body components formed the majority of cilia DEGAs, and adjacent cerebral cortices exhibited large overlapping pairs of cilia DEGAs. Most remarkably, specific α/β-tubulin subunits (TUBA1A, TUBB2A, and TUBB2B) and SNAP-25 exhibited the highest rates of downregulation and upregulation, respectively, across age in almost all brain regions. α/β-tubulins and SNAP-25 expressions are known to be dysregulated in age-related neurodevelopmental and neurodegenerative disorders. Our results support a role for the high dynamics of cilia structural and functional components across the lifespan in the normal physiology of brain circuits. Furthermore, they suggest a crucial role for cilia signaling in the pathophysiological mechanisms of age-related psychiatric/neurological disorders.
Collapse
Affiliation(s)
- Siwei Chen
- Department of Computer Science, School of Information and Computer Sciences, University of California-Irvine, Irvine, CA 92617, USA; (S.C.); (P.B.)
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California-Irvine, Irvine, CA 92617, USA
| | - Wedad Alhassen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92617, USA; (W.A.); (R.V.M.); (B.V.)
| | - Roudabeh Vakil Monfared
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92617, USA; (W.A.); (R.V.M.); (B.V.)
| | - Benjamin Vachirakorntong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92617, USA; (W.A.); (R.V.M.); (B.V.)
| | - Surya M. Nauli
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University Rinker Health Science Campus, Irvine, CA 92618, USA;
| | - Pierre Baldi
- Department of Computer Science, School of Information and Computer Sciences, University of California-Irvine, Irvine, CA 92617, USA; (S.C.); (P.B.)
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California-Irvine, Irvine, CA 92617, USA
| | - Amal Alachkar
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California-Irvine, Irvine, CA 92617, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92617, USA; (W.A.); (R.V.M.); (B.V.)
| |
Collapse
|
118
|
Xu L, Nagai Y, Kajihara Y, Ito G, Tomita T. The Regulation of Rab GTPases by Phosphorylation. Biomolecules 2021; 11:biom11091340. [PMID: 34572553 PMCID: PMC8469595 DOI: 10.3390/biom11091340] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 01/11/2023] Open
Abstract
Rab proteins are small GTPases that act as molecular switches for intracellular vesicle trafficking. Although their function is mainly regulated by regulatory proteins such as GTPase-activating proteins and guanine nucleotide exchange factors, recent studies have shown that some Rab proteins are physiologically phosphorylated in the switch II region by Rab kinases. As the switch II region of Rab proteins undergoes a conformational change depending on the bound nucleotide, it plays an essential role in their function as a ‘switch’. Initially, the phosphorylation of Rab proteins in the switch II region was shown to inhibit the association with regulatory proteins. However, recent studies suggest that it also regulates the binding of Rab proteins to effector proteins, determining which pathways to regulate. These findings suggest that the regulation of the Rab function may be more dynamically regulated by phosphorylation than just through the association with regulatory proteins. In this review, we summarize the recent findings and discuss the physiological and pathological roles of Rab phosphorylation.
Collapse
Affiliation(s)
- Lejia Xu
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
| | - Yuki Nagai
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
| | - Yotaro Kajihara
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
| | - Genta Ito
- Department of Biomolecular Chemistry, Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Correspondence: (G.I.); (T.T.)
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; (L.X.); (Y.N.); (Y.K.)
- Social Cooperation Program of Brain and Neurological Disorders, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Correspondence: (G.I.); (T.T.)
| |
Collapse
|
119
|
Zhou K, Zhou Y, Yang D, Chen T, Liu X, Li S, Wang Z. The type 3 adenylyl cyclase is crucial for intestinal mucosal neural network in the gut lamina propria. Neurogastroenterol Motil 2021; 33:e14140. [PMID: 33939232 DOI: 10.1111/nmo.14140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 02/13/2021] [Accepted: 03/02/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND The type 3 adenylyl cyclase (AC3) enzyme is involved in the synthesis of cyclic adenosine monophosphate (cAMP). It is primarily expressed in the central nervous system (CNS) and plays a crucial role in neurogenesis and neural dendritic arborization. However, the AC3's functional role in the gastrointestinal tract remains ambiguous. METHODS AC3 expression in enteric tissue of AC3+/+ mice was investigated using immunohistochemistry and RT-PCR. AC3 knock-out mice (AC3-/- ) were used to examine the effect of AC3 on the enteric nervous system (ENS) function and the number of cilia and apoptotic cells. Additionally, total gastrointestinal transit time and colonic motility were compared between the AC3-/- and AC3+/+ groups of mice. KEY RESULTS AC3 was predominately expressed in the myenteric plexus of the large intestine. Colonic-bead expulsion analysis showed accelerated propulsion in the large intestine of the AC3-/- mice. The AC3-/- mice demonstrated reduced nerve fibers and enteric glial cells count in colonic mucosa compared to the AC3+/+ mice. Furthermore, AC3-/- mice exhibited increased cellular apoptosis and reduced ARL13B+ cilium cells in the colonic lamina propria compared to the AC3+/+ mice. CONCLUSIONS In AC3-/- mice, innervation of the lamina propria in the colonic mucosa was reduced and colonic propulsion was accelerated. AC3 is crucial for the development and function of the adult neural network of ENS. AC3 deficiency caused atrophy in the colonic mucosal neural network of mice.
Collapse
Affiliation(s)
- Kang Zhou
- College of Life Science, Hebei University, Baoding, China
| | - Yanfen Zhou
- College of Life Science, Hebei University, Baoding, China
| | - Dong Yang
- College of Life Science, Hebei University, Baoding, China
| | - Tingrong Chen
- College of Life Science, Hebei University, Baoding, China
| | - Xinxia Liu
- College of Life Science, Hebei University, Baoding, China.,Medical College, Hebei University, Baoding, China
| | - Shujuan Li
- College of Life Science, Hebei University, Baoding, China
| | - Zhenshan Wang
- College of Life Science, Hebei University, Baoding, China
| |
Collapse
|
120
|
From Bipotent Neuromesodermal Progenitors to Neural-Mesodermal Interactions during Embryonic Development. Int J Mol Sci 2021; 22:ijms22179141. [PMID: 34502050 PMCID: PMC8431582 DOI: 10.3390/ijms22179141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
To ensure the formation of a properly patterned embryo, multiple processes must operate harmoniously at sequential phases of development. This is implemented by mutual interactions between cells and tissues that together regulate the segregation and specification of cells, their growth and morphogenesis. The formation of the spinal cord and paraxial mesoderm derivatives exquisitely illustrate these processes. Following early gastrulation, while the vertebrate body elongates, a population of bipotent neuromesodermal progenitors resident in the posterior region of the embryo generate both neural and mesodermal lineages. At later stages, the somitic mesoderm regulates aspects of neural patterning and differentiation of both central and peripheral neural progenitors. Reciprocally, neural precursors influence the paraxial mesoderm to regulate somite-derived myogenesis and additional processes by distinct mechanisms. Central to this crosstalk is the activity of the axial notochord, which, via sonic hedgehog signaling, plays pivotal roles in neural, skeletal muscle and cartilage ontogeny. Here, we discuss the cellular and molecular basis underlying this complex developmental plan, with a focus on the logic of sonic hedgehog activities in the coordination of the neural-mesodermal axis.
Collapse
|
121
|
Pinto AL, Rasteiro M, Bota C, Pestana S, Sampaio P, Hogg C, Burgoyne T, Lopes SS. Zebrafish Motile Cilia as a Model for Primary Ciliary Dyskinesia. Int J Mol Sci 2021; 22:8361. [PMID: 34445067 PMCID: PMC8393663 DOI: 10.3390/ijms22168361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/21/2022] Open
Abstract
Zebrafish is a vertebrate teleost widely used in many areas of research. As embryos, they develop quickly and provide unique opportunities for research studies owing to their transparency for at least 48 h post fertilization. Zebrafish have many ciliated organs that include primary cilia as well as motile cilia. Using zebrafish as an animal model helps to better understand human diseases such as Primary Ciliary Dyskinesia (PCD), an autosomal recessive disorder that affects cilia motility, currently associated with more than 50 genes. The aim of this study was to validate zebrafish motile cilia, both in mono and multiciliated cells, as organelles for PCD research. For this purpose, we obtained systematic high-resolution data in both the olfactory pit (OP) and the left-right organizer (LRO), a superficial organ and a deep organ embedded in the tail of the embryo, respectively. For the analysis of their axonemal ciliary structure, we used conventional transmission electron microscopy (TEM) and electron tomography (ET). We characterised the wild-type OP cilia and showed, for the first time in zebrafish, the presence of motile cilia (9 + 2) in the periphery of the pit and the presence of immotile cilia (still 9 + 2), with absent outer dynein arms, in the centre of the pit. In addition, we reported that a central pair of microtubules in the LRO motile cilia is common in zebrafish, contrary to mouse embryos, but it is not observed in all LRO cilia from the same embryo. We further showed that the outer dynein arms of the microtubular doublet of both the OP and LRO cilia are structurally similar in dimensions to the human respiratory cilia at the resolution of TEM and ET. We conclude that zebrafish is a good model organism for PCD research but investigators need to be aware of the specific physical differences to correctly interpret their results.
Collapse
Affiliation(s)
- Andreia L. Pinto
- Paediatric Respiratory Medicine, Primary Ciliary Dyskinesia Centre, Royal Brompton & Harefield NHS Trust, London SW3 6NP, UK; (A.L.P.); (C.H.); (T.B.)
- CEDOC, NOVA Medical School, Rua Câmara Pestana nº 6, 6-A, Edifício CEDOC II, 1150-082 Lisboa, Portugal; (M.R.); (C.B.); (S.P.); (P.S.)
- Department of Life Sciences, NOVA School of Science and Technology, 2825-149 Caparica, Portugal
| | - Margarida Rasteiro
- CEDOC, NOVA Medical School, Rua Câmara Pestana nº 6, 6-A, Edifício CEDOC II, 1150-082 Lisboa, Portugal; (M.R.); (C.B.); (S.P.); (P.S.)
| | - Catarina Bota
- CEDOC, NOVA Medical School, Rua Câmara Pestana nº 6, 6-A, Edifício CEDOC II, 1150-082 Lisboa, Portugal; (M.R.); (C.B.); (S.P.); (P.S.)
| | - Sara Pestana
- CEDOC, NOVA Medical School, Rua Câmara Pestana nº 6, 6-A, Edifício CEDOC II, 1150-082 Lisboa, Portugal; (M.R.); (C.B.); (S.P.); (P.S.)
| | - Pedro Sampaio
- CEDOC, NOVA Medical School, Rua Câmara Pestana nº 6, 6-A, Edifício CEDOC II, 1150-082 Lisboa, Portugal; (M.R.); (C.B.); (S.P.); (P.S.)
| | - Claire Hogg
- Paediatric Respiratory Medicine, Primary Ciliary Dyskinesia Centre, Royal Brompton & Harefield NHS Trust, London SW3 6NP, UK; (A.L.P.); (C.H.); (T.B.)
- Department of Paediatrics, Imperial College London, London SW3 6LY, UK
| | - Thomas Burgoyne
- Paediatric Respiratory Medicine, Primary Ciliary Dyskinesia Centre, Royal Brompton & Harefield NHS Trust, London SW3 6NP, UK; (A.L.P.); (C.H.); (T.B.)
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Susana S. Lopes
- CEDOC, NOVA Medical School, Rua Câmara Pestana nº 6, 6-A, Edifício CEDOC II, 1150-082 Lisboa, Portugal; (M.R.); (C.B.); (S.P.); (P.S.)
| |
Collapse
|
122
|
Speight P, Rozycki M, Venugopal S, Szászi K, Kofler M, Kapus A. Myocardin-related transcription factor and serum response factor regulate cilium turnover by both transcriptional and local mechanisms. iScience 2021; 24:102739. [PMID: 34278253 PMCID: PMC8261663 DOI: 10.1016/j.isci.2021.102739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/02/2020] [Accepted: 06/15/2021] [Indexed: 12/31/2022] Open
Abstract
Turnover of the primary cilium (PC) is critical for proliferation and tissue homeostasis. Each key component of the PC resorption machinery, the HEF1/Aurora kinase A (AurA)/HDAC6 pathway harbors cis-elements potentially targeted by the transcriptional co-activator myocardin-related transcription factor (MRTF) and/or its partner serum response factor (SRF). Thus we investigated if MRTF and/or SRF regulate PC turnover. Here we show that (1) both MRTF and SRF are indispensable for serum-induced PC resorption, and (2) they act via both transcriptional and local mechanisms. Intriguingly, MRTF and SRF are present in the basal body and/or the PC, and serum facilitates ciliary MRTF recruitment. MRTF promotes the stability and ciliary accumulation of AurA and facilitates SRF phosphorylation. Ciliary SRF interacts with AurA and HDAC6. MRTF also inhibits ciliogenesis. It interacts with and is required for the correct localization of the ciliogenesis modulator CEP290. Thus, MRTF and SRF are critical regulators of PC assembly and/or disassembly, acting both as transcription factors and as PC constituents.
Collapse
Affiliation(s)
- Pam Speight
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - Matthew Rozycki
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - Shruthi Venugopal
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5B 1T8, Canada
| | - Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
123
|
Centrosome regulation and function in mammalian cortical neurogenesis. Curr Opin Neurobiol 2021; 69:256-266. [PMID: 34303132 DOI: 10.1016/j.conb.2021.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023]
Abstract
As the primary microtubule-organizing center in animal cells, centrosomes regulate microtubule cytoskeleton to support various cellular behaviors. They also serve as the base for nucleating primary cilia, the hub of diverse signaling pathways. Cells typically possess one centrosome that contains two inequal centrioles and undergoes semi-conservative duplication during cell division, resulting in two centrosomes with an inherent asymmetry in age and properties. While the centrosome is ubiquitously present, mutations of centrosome proteins are strongly associated with human microcephaly characterized by a small cerebral cortex, underscoring the importance of an intact centrosome in supporting cortical neurogenesis. Here we review recent advances on centrosome regulation and function in mammalian cortical neural progenitors and discuss the implications for a better understanding of cortical neurogenesis and related disease mechanisms.
Collapse
|
124
|
Gilloteaux J, Bouchat J, Bielarz V, Brion JP, Nicaise C. A primary cilium in oligodendrocytes: a fine structure signal of repairs in thalamic Osmotic Demyelination Syndrome (ODS). Ultrastruct Pathol 2021; 45:128-157. [PMID: 34154511 DOI: 10.1080/01913123.2021.1891161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A murine osmotic demyelination syndrome (ODS) model of the central nervous system included the relay thalamic ventral posterolateral (VPL) and ventral posteromedial (VPM) nuclei. Morphologic comparisons between treatments have revealed oligodendrocyte changes and, already 12 hours following the osmolality restoration, some heavily contrasted oligodendrocytes formed a unique intracellular primary cilium. This unique structure, found in vivo, in mature CNS oligodendrocytes, could account for a local awakening of some of the developmental proteome as it can be expressed in oligodendrocyte precursor cells. This resilience accompanied the emergence of arl13b protein expression along with restoration of nerve cell body axon hillocks shown in a previous issue of this journal. Additionally, the return of several thalamic oligodendrocyte fine features (nucleus, organelles) was shown 36 h later, including some mitosis. Those cell restorations and recognized translational activities comforted that local repairs could again take place, due to oligodendrocyte resilience after ODS instead or added to a postulated immigration of oligodendrocyte precursor cells distant from the sites of myelinolysis.
Collapse
Affiliation(s)
- Jacques Gilloteaux
- Unit of Research in Molecular Physiology (Urphym - NARILIS), Départment of Médecine, Université de Namur, Namur, Belgium.,Department of Anatomical Sciences, St George's University School of Medicine, KB Taylor Global Scholar's Program at UNN, School of Health and Life Sciences, Newcastle upon Tyne, UK
| | - Joanna Bouchat
- Unit of Research in Molecular Physiology (Urphym - NARILIS), Départment of Médecine, Université de Namur, Namur, Belgium
| | - Valery Bielarz
- Unit of Research in Molecular Physiology (Urphym - NARILIS), Départment of Médecine, Université de Namur, Namur, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculté de Médecine Université Libre de Bruxelles, Brussels, Belgium
| | - Charles Nicaise
- Unit of Research in Molecular Physiology (Urphym - NARILIS), Départment of Médecine, Université de Namur, Namur, Belgium
| |
Collapse
|
125
|
Suciu SK, Long AB, Caspary T. Smoothened and ARL13B are critical in mouse for superior cerebellar peduncle targeting. Genetics 2021; 218:6300527. [PMID: 34132778 PMCID: PMC8864748 DOI: 10.1093/genetics/iyab084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/15/2021] [Indexed: 01/07/2023] Open
Abstract
Patients with the ciliopathy Joubert syndrome present with physical anomalies, intellectual disability, and a hindbrain malformation described as the "molar tooth sign" due to its appearance on an MRI. This radiological abnormality results from a combination of hypoplasia of the cerebellar vermis and inappropriate targeting of the white matter tracts of the superior cerebellar peduncles. ARL13B is a cilia-enriched regulatory GTPase established to regulate cell fate, cell proliferation, and axon guidance through vertebrate Hedgehog signaling. In patients, mutations in ARL13B cause Joubert syndrome. To understand the etiology of the molar tooth sign, we used mouse models to investigate the role of ARL13B during cerebellar development. We found that ARL13B regulates superior cerebellar peduncle targeting and these fiber tracts require Hedgehog signaling for proper guidance. However, in mouse, the Joubert-causing R79Q mutation in ARL13B does not disrupt Hedgehog signaling nor does it impact tract targeting. We found a small cerebellar vermis in mice lacking ARL13B function but no cerebellar vermis hypoplasia in mice expressing the Joubert-causing R79Q mutation. In addition, mice expressing a cilia-excluded variant of ARL13B that transduces Hedgehog normally showed normal tract targeting and vermis width. Taken together, our data indicate that ARL13B is critical for the control of cerebellar vermis width as well as superior cerebellar peduncle axon guidance, likely via Hedgehog signaling. Thus, our work highlights the complexity of ARL13B in molar tooth sign etiology.
Collapse
Affiliation(s)
- Sarah K Suciu
- Genetics and Molecular Biology Graduate Program, Emory University, Atlanta, GA 30322, USA,Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - Alyssa B Long
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA,Corresponding author: Department of Human Genetics, 615 Michael Street, Suite 301, Atlanta, GA 30322.
| |
Collapse
|
126
|
Yelland T, Garcia E, Samarakoon Y, Ismail S. The Structural and Biochemical Characterization of UNC119B Cargo Binding and Release Mechanisms. Biochemistry 2021; 60:1952-1963. [PMID: 34130453 PMCID: PMC8246649 DOI: 10.1021/acs.biochem.1c00251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/25/2021] [Indexed: 11/29/2022]
Abstract
Two paralogs of the guanine dissociation inhibitor-like solubilizing factors UNC119, UNC119A and UNC119B, are present in the human genome. UNC119 binds to N-myristoylated proteins and masks the hydrophobic lipid from the hydrophilic cytosol, facilitating trafficking between different membranes. Two classes of UNC119 cargo proteins have been classified: low affinity cargoes, released by the Arf-like proteins ARL2 and ARL3, and high affinity cargoes, which are specifically released by ARL3 and trafficked to either the primary cilium or the immunological synapse. The UNC119 homologues have reported differences in functionality, but the structural and biochemical bases for these differences are unknown. Using myristoylated peptide binding and release assays, we show that peptides sharing the previously identified UNC119A high affinity motif show significant variations of binding affinities to UNC119B of up to 427-fold. Furthermore, we solve the first two crystal structures of UNC119B, one in complex with the high affinity cargo peptide of LCK and a second one in complex with the release factor ARL3. Using these novel structures, we identify a stretch of negatively charged amino acids unique to UNC119B that may undergo a conformational change following binding of a release factor which we propose as an additional release mechanism specific to UNC119B.
Collapse
Affiliation(s)
- Tamas Yelland
- Beatson
Cancer Research UK Institute, Glasgow G61 1BD, Scotland
| | - Esther Garcia
- Beatson
Cancer Research UK Institute, Glasgow G61 1BD, Scotland
| | | | - Shehab Ismail
- Beatson
Cancer Research UK Institute, Glasgow G61 1BD, Scotland
- Institute
of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, Scotland
- Department
of Chemistry, KU Leuven, Celestijnenlaan 200G, 3001 Heverlee, Belgium
| |
Collapse
|
127
|
Senatore E, Chiuso F, Rinaldi L, Intartaglia D, Delle Donne R, Pedone E, Catalanotti B, Pirone L, Fiorillo B, Moraca F, Giamundo G, Scala G, Raffeiner A, Torres-Quesada O, Stefan E, Kwiatkowski M, van Pijkeren A, Morleo M, Franco B, Garbi C, Conte I, Feliciello A. The TBC1D31/praja2 complex controls primary ciliogenesis through PKA-directed OFD1 ubiquitylation. EMBO J 2021; 40:e106503. [PMID: 33934390 PMCID: PMC8126939 DOI: 10.15252/embj.2020106503] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
The primary cilium is a microtubule‐based sensory organelle that dynamically links signalling pathways to cell differentiation, growth, and development. Genetic defects of primary cilia are responsible for genetic disorders known as ciliopathies. Orofacial digital type I syndrome (OFDI) is an X‐linked congenital ciliopathy caused by mutations in the OFD1 gene and characterized by malformations of the face, oral cavity, digits and, in the majority of cases, polycystic kidney disease. OFD1 plays a key role in cilium biogenesis. However, the impact of signalling pathways and the role of the ubiquitin‐proteasome system (UPS) in the control of OFD1 stability remain unknown. Here, we identify a novel complex assembled at centrosomes by TBC1D31, including the E3 ubiquitin ligase praja2, protein kinase A (PKA), and OFD1. We show that TBC1D31 is essential for ciliogenesis. Mechanistically, upon G‐protein‐coupled receptor (GPCR)‐cAMP stimulation, PKA phosphorylates OFD1 at ser735, thus promoting OFD1 proteolysis through the praja2‐UPS circuitry. This pathway is essential for ciliogenesis. In addition, a non‐phosphorylatable OFD1 mutant dramatically affects cilium morphology and dynamics. Consistent with a role of the TBC1D31/praja2/OFD1 axis in ciliogenesis, alteration of this molecular network impairs ciliogenesis in vivo in Medaka fish, resulting in developmental defects. Our findings reveal a multifunctional transduction unit at the centrosome that links GPCR signalling to ubiquitylation and proteolysis of the ciliopathy protein OFD1, with important implications on cilium biology and development. Derangement of this control mechanism may underpin human genetic disorders.
Collapse
Affiliation(s)
- Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | | | - Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Emilia Pedone
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy
| | | | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy
| | - Bianca Fiorillo
- Department of Pharmacy, University Federico II, Naples, Italy
| | - Federica Moraca
- Department of Pharmacy, University Federico II, Naples, Italy.,Net4Science srl, University "Magna Graecia", Catanzaro, Italy
| | | | - Giovanni Scala
- Department of Biology, University Federico II, Naples, Italy
| | - Andrea Raffeiner
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Omar Torres-Quesada
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Eduard Stefan
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | | | | | - Manuela Morleo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Science, University Federico II, Naples, Italy
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Biology, University Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| |
Collapse
|
128
|
Ho EK, Stearns T. Hedgehog signaling and the primary cilium: implications for spatial and temporal constraints on signaling. Development 2021; 148:dev195552. [PMID: 33914866 PMCID: PMC8126410 DOI: 10.1242/dev.195552] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mechanisms of vertebrate Hedgehog signaling are linked to the biology of the primary cilium, an antenna-like organelle that projects from the surface of most vertebrate cell types. Although the advantages of restricting signal transduction to cilia are often noted, the constraints imposed are less frequently considered, and yet they are central to how Hedgehog signaling operates in developing tissues. In this Review, we synthesize current understanding of Hedgehog signal transduction, ligand secretion and transport, and cilia dynamics to explore the temporal and spatial constraints imposed by the primary cilium on Hedgehog signaling in vivo.
Collapse
Affiliation(s)
- Emily K. Ho
- Department of Developmental Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Tim Stearns
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
129
|
Cell intercalation driven by SMAD3 underlies secondary neural tube formation. Dev Cell 2021; 56:1147-1163.e6. [PMID: 33878300 DOI: 10.1016/j.devcel.2021.03.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 01/07/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Body axis elongation is a hallmark of the vertebrate embryo, involving the architectural remodeling of the tail bud. Although it is clear how neuromesodermal progenitors (NMPs) contribute to embryo elongation, the dynamic events that lead to de novo lumen formation and that culminate in the formation of a 3-dimensional, neural tube from NMPs, are poorly understood. Here, we used in vivo imaging of the chicken embryo to show that cell intercalation downstream of TGF-β/SMAD3 signaling is required for secondary neural tube formation. Our analysis describes the events in embryo elongation including lineage restriction, the epithelial-to-mesenchymal transition of NMPs, and the initiation of lumen formation. We show that the resolution of a single, centrally positioned lumen, which occurs through the intercalation of central cells, requires SMAD3/Yes-associated protein (YAP) activity. We anticipate that these findings will be relevant to understand caudal, skin-covered neural tube defects, among the most frequent birth defects detected in humans.
Collapse
|
130
|
HDAC6 Signaling at Primary Cilia Promotes Proliferation and Restricts Differentiation of Glioma Cells. Cancers (Basel) 2021; 13:cancers13071644. [PMID: 33915983 PMCID: PMC8036575 DOI: 10.3390/cancers13071644] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Glioblastoma is the most common and lethal brain tumor in adults because it becomes resistant to virtually every treatment. Histone deacetylase 6 (HDAC6), which is located primarily in the cytoplasm, has a unique role in promoting the disassembly of cells’ primary cilium, a non-motile “antenna” that must be broken down before cells can progress through the cell cycle. The role of HDAC6 and its function in gliomas have not been investigated with respect to tumor cell cilia. We have found that inhibitors of HDAC6 cause rapid and specific changes inside glioma cilia, reducing tumor cell proliferative capacity and promoting cell differentiation. Importantly, the HDAC6 inhibitors did not affect the proliferation or differentiation of glioma cells that we genetically modified unable to grow cilia. Our findings reveal a conserved and critical role for HDAC6 in glioma growth that is dependent on cilia. Abstract Histone deacetylase 6 (HDAC6) is an emerging therapeutic target that is overexpressed in glioblastoma when compared to other HDACs. HDAC6 catalyzes the deacetylation of alpha-tubulin and mediates the disassembly of primary cilia, a process required for cell cycle progression. HDAC6 inhibition disrupts glioma proliferation, but whether this effect is dependent on tumor cell primary cilia is unknown. We found that HDAC6 inhibitors ACY-1215 (1215) and ACY-738 (738) inhibited the proliferation of multiple patient-derived and mouse glioma cells. While both inhibitors triggered rapid increases in acetylated alpha-tubulin (aaTub) in the cytosol and led to increased frequencies of primary cilia, they unexpectedly reduced the levels of aaTub in the cilia. To test whether the antiproliferative effects of HDAC6 inhibitors are dependent on tumor cell cilia, we generated patient-derived glioma lines devoid of cilia through depletion of ciliogenesis genes ARL13B or KIF3A. At low concentrations, 1215 or 738 did not decrease the proliferation of cilia-depleted cells. Moreover, the differentiation of glioma cells that was induced by HDAC6 inhibition did not occur after the inhibition of cilia formation. These data suggest HDAC6 signaling at primary cilia promotes the proliferation of glioma cells by restricting their ability to differentiate. Surprisingly, overexpressing HDAC6 did not reduce cilia length or the frequency of ciliated glioma cells, suggesting other factors are required to control HDAC6-mediated cilia disassembly in glioma cells. Collectively, our findings suggest that HDAC6 promotes the proliferation of glioma cells through primary cilia.
Collapse
|
131
|
Yusifov E, Dumoulin A, Stoeckli ET. Investigating Primary Cilia during Peripheral Nervous System Formation. Int J Mol Sci 2021; 22:3176. [PMID: 33804711 PMCID: PMC8003989 DOI: 10.3390/ijms22063176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
The primary cilium plays a pivotal role during the embryonic development of vertebrates. It acts as a somatic signaling hub for specific pathways, such as Sonic Hedgehog signaling. In humans, mutations in genes that cause dysregulation of ciliogenesis or ciliary function lead to severe developmental disorders called ciliopathies. Beyond its role in early morphogenesis, growing evidence points towards an essential function of the primary cilium in neural circuit formation in the central nervous system. However, very little is known about a potential role in the formation of the peripheral nervous system. Here, we investigate the presence of the primary cilium in neural crest cells and their derivatives in the trunk of developing chicken embryos in vivo. We found that neural crest cells, sensory neurons, and boundary cap cells all bear a primary cilium during key stages of early peripheral nervous system formation. Moreover, we describe differences in the ciliation of neuronal cultures of different populations from the peripheral and central nervous systems. Our results offer a framework for further in vivo and in vitro investigations on specific roles that the primary cilium might play during peripheral nervous system formation.
Collapse
Affiliation(s)
| | | | - Esther T. Stoeckli
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; (E.Y.); (A.D.)
| |
Collapse
|
132
|
Kathryn Anderson, grand dame of developmental biology. Proc Natl Acad Sci U S A 2021; 118:2101148118. [PMID: 33597255 DOI: 10.1073/pnas.2101148118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
133
|
Hansen JN, Rassmann S, Stüven B, Jurisch-Yaksi N, Wachten D. CiliaQ: a simple, open-source software for automated quantification of ciliary morphology and fluorescence in 2D, 3D, and 4D images. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2021; 44:18. [PMID: 33683488 PMCID: PMC7940315 DOI: 10.1140/epje/s10189-021-00031-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/01/2021] [Indexed: 05/16/2023]
Abstract
Cilia are hair-like membrane protrusions that emanate from the surface of most vertebrate cells and are classified into motile and primary cilia. Motile cilia move fluid flow or propel cells, while also fulfill sensory functions. Primary cilia are immotile and act as a cellular antenna, translating environmental cues into cellular responses. Ciliary dysfunction leads to severe diseases, commonly termed ciliopathies. The molecular details underlying ciliopathies and ciliary function are, however, not well understood. Since cilia are small subcellular compartments, imaging-based approaches have been used to study them. However, tools to comprehensively analyze images are lacking. Automatic analysis approaches require commercial software and are limited to 2D analysis and only a few parameters. The widely used manual analysis approaches are time consuming, user-biased, and difficult to compare. Here, we present CiliaQ, a package of open-source, freely available, and easy-to-use ImageJ plugins. CiliaQ allows high-throughput analysis of 2D and 3D, static or time-lapse images from fluorescence microscopy of cilia in cell culture or tissues, and outputs a comprehensive list of parameters for ciliary morphology, length, bending, orientation, and fluorescence intensity, making it broadly applicable. We envision CiliaQ as a resource and platform for reproducible and comprehensive analysis of ciliary function in health and disease.
Collapse
Affiliation(s)
- Jan Niklas Hansen
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
| | - Sebastian Rassmann
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Birthe Stüven
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, The Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, The Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs University Hospital, Trondheim, Norway
| | - Dagmar Wachten
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
| |
Collapse
|
134
|
Tereshko L, Gao Y, Cary BA, Turrigiano GG, Sengupta P. Ciliary neuropeptidergic signaling dynamically regulates excitatory synapses in postnatal neocortical pyramidal neurons. eLife 2021; 10:e65427. [PMID: 33650969 PMCID: PMC7952091 DOI: 10.7554/elife.65427] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Primary cilia are compartmentalized sensory organelles present on the majority of neurons in the mammalian brain throughout adulthood. Recent evidence suggests that cilia regulate multiple aspects of neuronal development, including the maintenance of neuronal connectivity. However, whether ciliary signals can dynamically modulate postnatal circuit excitability is unknown. Here we show that acute cell-autonomous knockdown of ciliary signaling rapidly strengthens glutamatergic inputs onto cultured rat neocortical pyramidal neurons and increases spontaneous firing. This increased excitability occurs without changes to passive neuronal properties or intrinsic excitability. Further, the neuropeptide receptor somatostatin receptor 3 (SSTR3) is localized nearly exclusively to excitatory neuron cilia both in vivo and in culture, and pharmacological manipulation of SSTR3 signaling bidirectionally modulates excitatory synaptic inputs onto these neurons. Our results indicate that ciliary neuropeptidergic signaling dynamically modulates excitatory synapses and suggest that defects in this regulation may underlie a subset of behavioral and cognitive disorders associated with ciliopathies.
Collapse
Affiliation(s)
- Lauren Tereshko
- Department of Biology, Brandeis UniversityWalthamUnited States
| | - Ya Gao
- Department of Biology, Brandeis UniversityWalthamUnited States
| | - Brian A Cary
- Department of Biology, Brandeis UniversityWalthamUnited States
| | | | - Piali Sengupta
- Department of Biology, Brandeis UniversityWalthamUnited States
| |
Collapse
|
135
|
Understanding Primary Ciliary Dyskinesia and Other Ciliopathies. J Pediatr 2021; 230:15-22.e1. [PMID: 33242470 PMCID: PMC8690631 DOI: 10.1016/j.jpeds.2020.11.040] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Ciliopathies are a collection of disorders related to cilia dysfunction. Cilia are specialized organelles that project from the surface of most cells. Motile and primary (sensory) cilia are essential structures and have wide ranging functions. Our understanding of the genetics, pathophysiology, and clinical manifestations of motile ciliopathies, including primary ciliary dyskinesia (PCD), has rapidly advanced since the disease was linked to ciliary ultrastructural defects nearly five decades ago. We will provide an overview of different types of cilia, their role in child health and disease, focusing on motile ciliopathies, and describe recent advances that have led to improved diagnostics and may yield therapeutic targets to restore ciliary structure and function.
Collapse
|
136
|
Bernatik O, Paclikova P, Kotrbova A, Bryja V, Cajanek L. Primary Cilia Formation Does Not Rely on WNT/β-Catenin Signaling. Front Cell Dev Biol 2021; 9:623753. [PMID: 33718363 PMCID: PMC7952446 DOI: 10.3389/fcell.2021.623753] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
Primary cilia act as crucial regulators of embryo development and tissue homeostasis. They are instrumental for modulation of several signaling pathways, including Hedgehog, WNT, and TGF-β. However, gaps exist in our understanding of how cilia formation and function is regulated. Recent work has implicated WNT/β-catenin signaling pathway in the regulation of ciliogenesis, yet the results are conflicting. One model suggests that WNT/β-catenin signaling negatively regulates cilia formation, possibly via effects on cell cycle. In contrast, second model proposes a positive role of WNT/β-catenin signaling on cilia formation, mediated by the re-arrangement of centriolar satellites in response to phosphorylation of the key component of WNT/β-catenin pathway, β-catenin. To clarify these discrepancies, we investigated possible regulation of primary cilia by the WNT/β-catenin pathway in cell lines (RPE-1, NIH3T3, and HEK293) commonly used to study ciliogenesis. We used WNT3a to activate or LGK974 to block the pathway, and examined initiation of ciliogenesis, cilium length, and percentage of ciliated cells. We show that the treatment by WNT3a has no- or lesser inhibitory effect on cilia formation. Importantly, the inhibition of secretion of endogenous WNT ligands using LGK974 blocks WNT signaling but does not affect ciliogenesis. Finally, using knock-out cells for key WNT pathway components, namely DVL1/2/3, LRP5/6, or AXIN1/2 we show that neither activation nor deactivation of the WNT/β-catenin pathway affects the process of ciliogenesis. These results suggest that WNT/β-catenin-mediated signaling is not generally required for efficient cilia formation. In fact, activation of the WNT/β-catenin pathway in some systems seems to moderately suppress ciliogenesis.
Collapse
Affiliation(s)
- Ondrej Bernatik
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petra Paclikova
- Section of Animal Physiology and Immunology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Anna Kotrbova
- Section of Animal Physiology and Immunology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Vitezslav Bryja
- Section of Animal Physiology and Immunology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Lukas Cajanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
137
|
Abstract
Ciliogenesis describes the assembly of cilia in interphase cells. Several hundred proteins have been linked to ciliogenesis, which proceeds through a highly coordinated multistage process at the distal end of centrioles requiring membranes. In this short review, we focus on recently reported insights into the biogenesis of the primary cilium membrane and its association with other ciliogenic processes in the intracellular ciliogenesis pathway.
Collapse
Affiliation(s)
- Saurabh Shakya
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Laboratory of Cellular and Developmental Signaling, Frederick, MD 21702, USA
| | - Christopher J Westlake
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Laboratory of Cellular and Developmental Signaling, Frederick, MD 21702, USA
| |
Collapse
|
138
|
Latour BL, Van De Weghe JC, Rusterholz TD, Letteboer SJ, Gomez A, Shaheen R, Gesemann M, Karamzade A, Asadollahi M, Barroso-Gil M, Chitre M, Grout ME, van Reeuwijk J, van Beersum SE, Miller CV, Dempsey JC, Morsy H, Bamshad MJ, Nickerson DA, Neuhauss SC, Boldt K, Ueffing M, Keramatipour M, Sayer JA, Alkuraya FS, Bachmann-Gagescu R, Roepman R, Doherty D. Dysfunction of the ciliary ARMC9/TOGARAM1 protein module causes Joubert syndrome. J Clin Invest 2021; 130:4423-4439. [PMID: 32453716 DOI: 10.1172/jci131656] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Joubert syndrome (JBTS) is a recessive neurodevelopmental ciliopathy characterized by a pathognomonic hindbrain malformation. All known JBTS genes encode proteins involved in the structure or function of primary cilia, ubiquitous antenna-like organelles essential for cellular signal transduction. Here, we used the recently identified JBTS-associated protein armadillo repeat motif-containing 9 (ARMC9) in tandem-affinity purification and yeast 2-hybrid screens to identify a ciliary module whose dysfunction underlies JBTS. In addition to the known JBTS-associated proteins CEP104 and CSPP1, we identified coiled-coil domain containing 66 (CCDC66) and TOG array regulator of axonemal microtubules 1 (TOGARAM1) as ARMC9 interaction partners. We found that TOGARAM1 variants cause JBTS and disrupt TOGARAM1 interaction with ARMC9. Using a combination of protein interaction analyses, characterization of patient-derived fibroblasts, and analysis of CRISPR/Cas9-engineered zebrafish and hTERT-RPE1 cells, we demonstrated that dysfunction of ARMC9 or TOGARAM1 resulted in short cilia with decreased axonemal acetylation and polyglutamylation, but relatively intact transition zone function. Aberrant serum-induced ciliary resorption and cold-induced depolymerization in ARMC9 and TOGARAM1 patient cell lines suggest a role for this new JBTS-associated protein module in ciliary stability.
Collapse
Affiliation(s)
- Brooke L Latour
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Tamara Ds Rusterholz
- Institute of Medical Genetics, and.,Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Stef Jf Letteboer
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Arianna Gomez
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Ranad Shaheen
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Matthias Gesemann
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Arezou Karamzade
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Asadollahi
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Miguel Barroso-Gil
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Manali Chitre
- Department of Paediatric Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Megan E Grout
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jeroen van Reeuwijk
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sylvia Ec van Beersum
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Caitlin V Miller
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jennifer C Dempsey
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Heba Morsy
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, Washington, USA.,The University of Washington Center for Mendelian Genomics is detailed in Supplemental Acknowledgments.,University of Washington Center for Mendelian Genomics, Seattle, Washington, USA.,Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Deborah A Nickerson
- The University of Washington Center for Mendelian Genomics is detailed in Supplemental Acknowledgments.,University of Washington Center for Mendelian Genomics, Seattle, Washington, USA
| | - Stephan Cf Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Karsten Boldt
- Medical Proteome Center, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Marius Ueffing
- Medical Proteome Center, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Mohammad Keramatipour
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ruxandra Bachmann-Gagescu
- Institute of Medical Genetics, and.,Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Ronald Roepman
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Dan Doherty
- Department of Pediatrics, University of Washington, Seattle, Washington, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
139
|
Kopinke D, Norris AM, Mukhopadhyay S. Developmental and regenerative paradigms of cilia regulated hedgehog signaling. Semin Cell Dev Biol 2021; 110:89-103. [PMID: 32540122 PMCID: PMC7736055 DOI: 10.1016/j.semcdb.2020.05.029] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 01/08/2023]
Abstract
Primary cilia are immotile appendages that have evolved to receive and interpret a variety of different extracellular cues. Cilia play crucial roles in intercellular communication during development and defects in cilia affect multiple tissues accounting for a heterogeneous group of human diseases called ciliopathies. The Hedgehog (Hh) signaling pathway is one of these cues and displays a unique and symbiotic relationship with cilia. Not only does Hh signaling require cilia for its function but the majority of the Hh signaling machinery is physically located within the cilium-centrosome complex. More specifically, cilia are required for both repressing and activating Hh signaling by modifying bifunctional Gli transcription factors into repressors or activators. Defects in balancing, interpreting or establishing these repressor/activator gradients in Hh signaling either require cilia or phenocopy disruption of cilia. Here, we will summarize the current knowledge on how spatiotemporal control of the molecular machinery of the cilium allows for a tight control of basal repression and activation states of the Hh pathway. We will then discuss several paradigms on how cilia influence Hh pathway activity in tissue morphogenesis during development. Last, we will touch on how cilia and Hh signaling are being reactivated and repurposed during adult tissue regeneration. More specifically, we will focus on mesenchymal stem cells within the connective tissue and discuss the similarities and differences of how cilia and ciliary Hh signaling control the formation of fibrotic scar and adipose tissue during fatty fibrosis of several tissues.
Collapse
Affiliation(s)
- Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA.
| | - Alessandra M Norris
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
140
|
Liu S, Trupiano MX, Simon J, Guo J, Anton ES. The essential role of primary cilia in cerebral cortical development and disorders. Curr Top Dev Biol 2021; 142:99-146. [PMID: 33706927 DOI: 10.1016/bs.ctdb.2020.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary cilium, first described in the 19th century in different cell types and organisms by Alexander Ecker, Albert Kolliker, Aleksandr Kowalevsky, Paul Langerhans, and Karl Zimmermann (Ecker, 1844; Kolliker, 1854; Kowalevsky, 1867; Langerhans, 1876; Zimmermann, 1898), play an essential modulatory role in diverse aspects of nervous system development and function. The primary cilium, sometimes referred to as the cell's 'antennae', can receive wide ranging inputs from cellular milieu, including morphogens, growth factors, neuromodulators, and neurotransmitters. Its unique structural and functional organization bequeaths it the capacity to hyper-concentrate signaling machinery in a restricted cellular domain approximately one-thousandth the volume of cell soma. Thus enabling it to act as a signaling hub that integrates diverse developmental and homestatic information from cellular milieu to regulate the development and function of neural cells. Dysfunction of primary cilia contributes to the pathophysiology of several brain malformations, intellectual disabilities, epilepsy, and psychiatric disorders. This review focuses on the most essential contributions of primary cilia to cerebral cortical development and function, in the context of neurodevelopmental disorders and malformations. It highlights the recent progress made in identifying the mechanisms underlying primary cilia's role in cortical progenitors, neurons and glia, in health and disease. A future challenge will be to translate these insights and advances into effective clinical treatments for ciliopathies.
Collapse
Affiliation(s)
- Siling Liu
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Mia X Trupiano
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jeremy Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jiami Guo
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States.
| |
Collapse
|
141
|
Nechipurenko I, Lavrentyeva S, Sengupta P. GRDN-1/Girdin regulates dendrite morphogenesis and cilium position in two specialized sensory neuron types in C. elegans. Dev Biol 2021; 472:38-51. [PMID: 33460640 DOI: 10.1016/j.ydbio.2020.12.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
Primary cilia are located at the dendritic tips of sensory neurons and house the molecular machinery necessary for detection and transduction of sensory stimuli. The mechanisms that coordinate dendrite extension with cilium position during sensory neuron development are not well understood. Here, we show that GRDN-1, the Caenorhabditis elegans ortholog of the highly conserved scaffold and signaling protein Girdin/GIV, regulates both cilium position and dendrite extension in the postembryonic AQR and PQR gas-sensing neurons. Mutations in grdn-1 disrupt dendrite outgrowth and mislocalize cilia to the soma or proximal axonal segments in AQR, and to a lesser extent, in PQR. GRDN-1 is localized to the basal body and regulates localization of HMR-1/Cadherin to the distal AQR dendrite. However, knockdown of HMR-1 and/or loss of SAX-7/LICAM, molecules previously implicated in sensory dendrite development in C. elegans, do not alter AQR dendrite morphology or cilium position. We find that GRDN-1 localization in AQR is regulated by UNC-116/Kinesin-1, and that correspondingly, unc-116 mutants exhibit severe AQR dendrite outgrowth and cilium positioning defects. In contrast, GRDN-1 and cilium localization in PQR is modulated by LIN-44/Wnt signaling. Together, these findings identify upstream regulators of GRDN-1, and describe new cell-specific roles for this multifunctional protein in sensory neuron development.
Collapse
Affiliation(s)
- Inna Nechipurenko
- Department of Biology, Brandeis University, Waltham, MA, USA; Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, USA.
| | | | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, MA, USA.
| |
Collapse
|
142
|
ElMaghloob Y, Sot B, McIlwraith MJ, Garcia E, Yelland T, Ismail S. ARL3 activation requires the co-GEF BART and effector-mediated turnover. eLife 2021; 10:e64624. [PMID: 33438581 PMCID: PMC7817177 DOI: 10.7554/elife.64624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/12/2021] [Indexed: 12/28/2022] Open
Abstract
The ADP-ribosylation factor-like 3 (ARL3) is a ciliopathy G-protein which regulates the ciliary trafficking of several lipid-modified proteins. ARL3 is activated by its guanine exchange factor (GEF) ARL13B via an unresolved mechanism. BART is described as an ARL3 effector which has also been implicated in ciliopathies, although the role of its ARL3 interaction is unknown. Here, we show that, at physiological GTP:GDP levels, human ARL3GDP is weakly activated by ARL13B. However, BART interacts with nucleotide-free ARL3 and, in concert with ARL13B, efficiently activates ARL3. In addition, BART binds ARL3GTP and inhibits GTP dissociation, thereby stabilising the active G-protein; the binding of ARL3 effectors then releases BART. Finally, using live cell imaging, we show that BART accesses the primary cilium and colocalises with ARL13B. We propose a model wherein BART functions as a bona fide co-GEF for ARL3 and maintains the active ARL3GTP, until it is recycled by ARL3 effectors.
Collapse
Affiliation(s)
| | - Begoña Sot
- Fundación IMDEA-Nanociencia, Campus de CantoblancoMadridSpain
- Unidad Asociada de Nanobiotecnología (CNB-CSIC e IMDEA Nanociencia), Campus de CantoblancoMadridSpain
| | | | | | | | - Shehab Ismail
- CRUK- Beatson InstituteGlasgowUnited Kingdom
- Institute of Cancer Sciences, University of GlasgowGlasgowUnited Kingdom
- Department of Chemistry, KU Leuven, CelestijnenlaanHeverleeBelgium
| |
Collapse
|
143
|
Andreu-Cervera A, Catala M, Schneider-Maunoury S. Cilia, ciliopathies and hedgehog-related forebrain developmental disorders. Neurobiol Dis 2020; 150:105236. [PMID: 33383187 DOI: 10.1016/j.nbd.2020.105236] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Development of the forebrain critically depends on the Sonic Hedgehog (Shh) signaling pathway, as illustrated in humans by the frequent perturbation of this pathway in holoprosencephaly, a condition defined as a defect in the formation of midline structures of the forebrain and face. The Shh pathway requires functional primary cilia, microtubule-based organelles present on virtually every cell and acting as cellular antennae to receive and transduce diverse chemical, mechanical or light signals. The dysfunction of cilia in humans leads to inherited diseases called ciliopathies, which often affect many organs and show diverse manifestations including forebrain malformations for the most severe forms. The purpose of this review is to provide the reader with a framework to understand the developmental origin of the forebrain defects observed in severe ciliopathies with respect to perturbations of the Shh pathway. We propose that many of these defects can be interpreted as an imbalance in the ratio of activator to repressor forms of the Gli transcription factors, which are effectors of the Shh pathway. We also discuss the complexity of ciliopathies and their relationships with forebrain disorders such as holoprosencephaly or malformations of cortical development, and emphasize the need for a closer examination of forebrain defects in ciliopathies, not only through the lens of animal models but also taking advantage of the increasing potential of the research on human tissues and organoids.
Collapse
Affiliation(s)
- Abraham Andreu-Cervera
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS) UMR7622, Institut national pour la Santé et la Recherche Médicale (Inserm) U1156, Institut de Biologie Paris Seine - Laboratoire de Biologie du Développement (IBPS-LBD), 9 Quai Saint-Bernard, 75005 Paris, France; Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, Campus de San Juan; Avda. Ramón y Cajal s/n, 03550 Alicante, Spain
| | - Martin Catala
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS) UMR7622, Institut national pour la Santé et la Recherche Médicale (Inserm) U1156, Institut de Biologie Paris Seine - Laboratoire de Biologie du Développement (IBPS-LBD), 9 Quai Saint-Bernard, 75005 Paris, France.
| | - Sylvie Schneider-Maunoury
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS) UMR7622, Institut national pour la Santé et la Recherche Médicale (Inserm) U1156, Institut de Biologie Paris Seine - Laboratoire de Biologie du Développement (IBPS-LBD), 9 Quai Saint-Bernard, 75005 Paris, France.
| |
Collapse
|
144
|
Cullen CL, O'Rourke M, Beasley SJ, Auderset L, Zhen Y, Pepper RE, Gasperini R, Young KM. Kif3a deletion prevents primary cilia assembly on oligodendrocyte progenitor cells, reduces oligodendrogenesis and impairs fine motor function. Glia 2020; 69:1184-1203. [PMID: 33368703 PMCID: PMC7986221 DOI: 10.1002/glia.23957] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/06/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022]
Abstract
Primary cilia are small microtubule‐based organelles capable of transducing signals from growth factor receptors embedded in the cilia membrane. Developmentally, oligodendrocyte progenitor cells (OPCs) express genes associated with primary cilia assembly, disassembly, and signaling, however, the importance of primary cilia for adult myelination has not been explored. We show that OPCs are ciliated in vitro and in vivo, and that they disassemble their primary cilia as they progress through the cell cycle. OPC primary cilia are also disassembled as OPCs differentiate into oligodendrocytes. When kinesin family member 3a (Kif3a), a gene critical for primary cilium assembly, was conditionally deleted from adult OPCs in vivo (Pdgfrα‐CreER™:: Kif3afl/fl transgenic mice), OPCs failed to assemble primary cilia. Kif3a‐deletion was also associated with reduced OPC proliferation and oligodendrogenesis in the corpus callosum and motor cortex and a progressive impairment of fine motor coordination.
Collapse
Affiliation(s)
- Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Megan O'Rourke
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Shannon J Beasley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Loic Auderset
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Yilan Zhen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Renee E Pepper
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Robert Gasperini
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.,School of Medicine, University of Tasmania, Hobart, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
145
|
Primary cilia mediate early life programming of adiposity through lysosomal regulation in the developing mouse hypothalamus. Nat Commun 2020; 11:5772. [PMID: 33188191 PMCID: PMC7666216 DOI: 10.1038/s41467-020-19638-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 10/22/2020] [Indexed: 12/21/2022] Open
Abstract
Hypothalamic neurons including proopiomelanocortin (POMC)-producing neurons regulate body weights. The non-motile primary cilium is a critical sensory organelle on the cell surface. An association between ciliary defects and obesity has been suggested, but the underlying mechanisms are not fully understood. Here we show that inhibition of ciliogenesis in POMC-expressing developing hypothalamic neurons, by depleting ciliogenic genes IFT88 and KIF3A, leads to adulthood obesity in mice. In contrast, adult-onset ciliary dysgenesis in POMC neurons causes no significant change in adiposity. In developing POMC neurons, abnormal cilia formation disrupts axonal projections through impaired lysosomal protein degradation. Notably, maternal nutrition and postnatal leptin surge have a profound impact on ciliogenesis in the hypothalamus of neonatal mice; through these effects they critically modulate the organization of hypothalamic feeding circuits. Our findings reveal a mechanism of early life programming of adult adiposity, which is mediated by primary cilia in developing hypothalamic neurons. Ciliary defects and obesity has been associated, but the underlying mechanism is unclear. Here, the authors show that inhibition of ciliogenesis in POMC neurons during development results in lysosomal protein degradation-dependent axonal disruption and adult obesity in mice.
Collapse
|
146
|
Lauter G, Coschiera A, Yoshihara M, Sugiaman-Trapman D, Ezer S, Sethurathinam S, Katayama S, Kere J, Swoboda P. Differentiation of ciliated human midbrain-derived LUHMES neurons. J Cell Sci 2020; 133:jcs249789. [PMID: 33115758 DOI: 10.1242/jcs.249789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Many human cell types are ciliated, including neural progenitors and differentiated neurons. Ciliopathies are characterized by defective cilia and comprise various disease states, including brain phenotypes, where the underlying biological pathways are largely unknown. Our understanding of neuronal cilia is rudimentary, and an easy-to-maintain, ciliated human neuronal cell model is absent. The Lund human mesencephalic (LUHMES) cell line is a ciliated neuronal cell line derived from human fetal mesencephalon. LUHMES cells can easily be maintained and differentiated into mature, functional neurons within one week. They have a single primary cilium as proliferating progenitor cells and as postmitotic, differentiating neurons. These developmental stages are completely separable within one day of culture condition change. The sonic hedgehog (SHH) signaling pathway is active in differentiating LUHMES neurons. RNA-sequencing timecourse analyses reveal molecular pathways and gene-regulatory networks critical for ciliogenesis and axon outgrowth at the interface between progenitor cell proliferation, polarization and neuronal differentiation. Gene expression dynamics of cultured LUHMES neurons faithfully mimic the corresponding in vivo dynamics of human fetal midbrain. In LUHMES cells, neuronal cilia biology can be investigated from proliferation through differentiation to mature neurons.
Collapse
Affiliation(s)
- Gilbert Lauter
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | - Andrea Coschiera
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | - Masahito Yoshihara
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | | | - Sini Ezer
- University of Helsinki, Research Program of Molecular Neurology and Folkhälsan Institute of Genetics, FI-00290 Helsinki, Finland
| | - Shalini Sethurathinam
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | - Shintaro Katayama
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
- University of Helsinki, Stem Cells and Metabolism Research Program and Folkhälsan Research Center, FI-00290 Helsinki, Finland
| | - Juha Kere
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
- University of Helsinki, Research Program of Molecular Neurology and Folkhälsan Institute of Genetics, FI-00290 Helsinki, Finland
| | - Peter Swoboda
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| |
Collapse
|
147
|
Saade M, Ferrero DS, Blanco-Ameijeiras J, Gonzalez-Gobartt E, Flores-Mendez M, Ruiz-Arroyo VM, Martínez-Sáez E, Ramón Y Cajal S, Akizu N, Verdaguer N, Martí E. Multimerization of Zika Virus-NS5 Causes Ciliopathy and Forces Premature Neurogenesis. Cell Stem Cell 2020; 27:920-936.e8. [PMID: 33147489 DOI: 10.1016/j.stem.2020.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/16/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
Zika virus (ZikV) is a flavivirus that infects neural tissues, causing congenital microcephaly. ZikV has evolved multiple mechanisms to restrict proliferation and enhance cell death, although the underlying cellular events involved remain unclear. Here we show that the ZikV-NS5 protein interacts with host proteins at the base of the primary cilia in neural progenitor cells, causing an atypical non-genetic ciliopathy and premature neuron delamination. Furthermore, in human microcephalic fetal brain tissue, ZikV-NS5 persists at the base of the motile cilia in ependymal cells, which also exhibit a severe ciliopathy. Although the enzymatic activity of ZikV-NS5 appears to be dispensable, the amino acids Y25, K28, and K29 that are involved in NS5 oligomerization are essential for localization and interaction with components of the cilium base, promoting ciliopathy and premature neurogenesis. These findings lay the foundation for therapies that target ZikV-NS5 multimerization and prevent the developmental malformations associated with congenital Zika syndrome.
Collapse
Affiliation(s)
- Murielle Saade
- Developmental Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain.
| | - Diego S Ferrero
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain
| | - José Blanco-Ameijeiras
- Developmental Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Elena Gonzalez-Gobartt
- Developmental Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Marco Flores-Mendez
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Victor M Ruiz-Arroyo
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Elena Martínez-Sáez
- Department of Pathology, Vall d'Hebron University Hospital, Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona and Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona 08035, Spain
| | - Santiago Ramón Y Cajal
- Department of Pathology, Vall d'Hebron University Hospital, Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona and Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona 08035, Spain
| | - Naiara Akizu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nuria Verdaguer
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Elisa Martí
- Developmental Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, C/Baldiri i Reixac 20, Barcelona 08028, Spain.
| |
Collapse
|
148
|
Brooks ER, Islam MT, Anderson KV, Zallen JA. Sonic hedgehog signaling directs patterned cell remodeling during cranial neural tube closure. eLife 2020; 9:60234. [PMID: 33103996 PMCID: PMC7655103 DOI: 10.7554/elife.60234] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022] Open
Abstract
Neural tube closure defects are a major cause of infant mortality, with exencephaly accounting for nearly one-third of cases. However, the mechanisms of cranial neural tube closure are not well understood. Here, we show that this process involves a tissue-wide pattern of apical constriction controlled by Sonic hedgehog (Shh) signaling. Midline cells in the mouse midbrain neuroepithelium are flat with large apical surfaces, whereas lateral cells are taller and undergo synchronous apical constriction, driving neural fold elevation. Embryos lacking the Shh effector Gli2 fail to produce appropriate midline cell architecture, whereas embryos with expanded Shh signaling, including the IFT-A complex mutants Ift122 and Ttc21b and embryos expressing activated Smoothened, display apical constriction defects in lateral cells. Disruption of lateral, but not midline, cell remodeling results in exencephaly. These results reveal a morphogenetic program of patterned apical constriction governed by Shh signaling that generates structural changes in the developing mammalian brain.
Collapse
Affiliation(s)
- Eric R Brooks
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Mohammed Tarek Islam
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, United States
| |
Collapse
|
149
|
Yamazoe T, Nagai T, Umeda S, Sugaya Y, Mizuno K. Roles of TOG and jelly-roll domains of centrosomal protein CEP104 in its functions in cilium elongation and Hedgehog signaling. J Biol Chem 2020; 295:14723-14736. [PMID: 32820051 DOI: 10.1074/jbc.ra120.013334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/11/2020] [Indexed: 12/26/2022] Open
Abstract
Primary cilia are generated through the extension of the microtubule-based axoneme. Centrosomal protein 104 (CEP104) localizes to the tip of the elongating axoneme, and CEP104 mutations are linked to a ciliopathy, Joubert syndrome. Thus, CEP104 has been implicated in ciliogenesis. However, the mechanism by which CEP104 regulates ciliogenesis remains elusive. We report here that CEP104 is critical for cilium elongation but not for initiating ciliogenesis. We also demonstrated that the tumor-overexpressed gene (TOG) domain of CEP104 exhibits microtubule-polymerizing activity and that this activity is essential for the cilium-elongating activity of CEP104. Knockdown/rescue experiments showed that the N-terminal jelly-roll (JR) fold partially contributes to cilium-elongating activity of CEP104, but neither the zinc-finger region nor the SXIP motif is required for this activity. CEP104 binds to a centriole-capping protein, CP110, through the zinc-finger region and to a microtubule plus-end-binding protein, EB1, through the SXIP motif, indicating that the binding of CP110 and EB1 is dispensable for the cilium-elongating activity of CEP104. Moreover, CEP104 depletion does not affect CP110 removal from the mother centriole, which suggests that CEP104 functions after the removal of CP110. Last, we also showed that CEP104 is required for the ciliary entry of Smoothened and export of GPR161 upon Hedgehog signal activation and that the TOG domain plays a critical role in this activity. Our results define the roles of the individual domains of CEP104 in its functions in cilium elongation and Hedgehog signaling and should enhance our understanding of the mechanism underlying CEP104 mutation-associated ciliopathies.
Collapse
Affiliation(s)
- Takashi Yamazoe
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Tomoaki Nagai
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan; Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan.
| | - Shinya Umeda
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yuko Sugaya
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Kensaku Mizuno
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan; Institute of Liberal Arts and Sciences, Tohoku University, Kawauchi, Sendai, Miyagi, Japan.
| |
Collapse
|
150
|
Mateska I, Nanda K, Dye NA, Alexaki VI, Eaton S. Range of SHH signaling in adrenal gland is limited by membrane contact to cells with primary cilia. J Biophys Biochem Cytol 2020; 219:211483. [PMID: 33090184 PMCID: PMC7588141 DOI: 10.1083/jcb.201910087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 07/27/2020] [Accepted: 09/15/2020] [Indexed: 01/04/2023] Open
Abstract
The signaling protein Sonic Hedgehog (SHH) is crucial for the development and function of many vertebrate tissues. It remains largely unclear, however, what defines the range and specificity of pathway activation. The adrenal gland represents a useful model to address this question, where the SHH pathway is activated in a very specific subset of cells lying near the SHH-producing cells, even though there is an abundance of lipoproteins that would allow SHH to travel and signal long-range. We determine that, whereas adrenal cells can secrete SHH on lipoproteins, this form of SHH is inactive due to the presence of cosecreted inhibitors, potentially explaining the absence of long-range signaling. Instead, we find that SHH-producing cells signal at short range via membrane-bound SHH, only to receiving cells with primary cilia. Finally, our data from NCI-H295R adrenocortical carcinoma cells suggest that adrenocortical tumors may evade these regulatory control mechanisms by acquiring the ability to activate SHH target genes in response to TGF-β.
Collapse
Affiliation(s)
- Ivona Mateska
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany,Biotechnologisches Zentrum, Technische Universität Dresden, Dresden, Germany,Correspondence to Ivona Mateska:
| | - Kareena Nanda
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Natalie A. Dye
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Suzanne Eaton
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany,Biotechnologisches Zentrum, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|