1
|
Groh AMR, Song YL, Tea F, Lu B, Huynh S, Afanasiev E, Bigotte M, Del Bigio MR, Stratton JA. Multiciliated ependymal cells: an update on biology and pathology in the adult brain. Acta Neuropathol 2024; 148:39. [PMID: 39254862 DOI: 10.1007/s00401-024-02784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
Mature multiciliated ependymal cells line the cerebral ventricles where they form a partial barrier between the cerebrospinal fluid (CSF) and brain parenchyma and regulate local CSF microcirculation through coordinated ciliary beating. Although the ependyma is a highly specialized brain interface with barrier, trophic, and perhaps even regenerative capacity, it remains a misfit in the canon of glial neurobiology. We provide an update to seminal reviews in the field by conducting a scoping review of the post-2010 mature multiciliated ependymal cell literature. We delineate how recent findings have either called into question or substantiated classical views of the ependymal cell. Beyond this synthesis, we document the basic methodologies and study characteristics used to describe multiciliated ependymal cells since 1980. Our review serves as a comprehensive resource for future investigations of mature multiciliated ependymal cells.
Collapse
Affiliation(s)
- Adam M R Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Yeji Lori Song
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Fiona Tea
- Department of Neuroscience, University of Montreal, Montréal, QC, Canada
| | - Brianna Lu
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Stephanie Huynh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Elia Afanasiev
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Maxime Bigotte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Marc R Del Bigio
- Department of Pathology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada.
| |
Collapse
|
2
|
Flax RG, Rosston P, Rocha C, Anderson B, Capener JL, Durcan TM, Drewry DH, Prinos P, Axtman AD. Illumination of understudied ciliary kinases. Front Mol Biosci 2024; 11:1352781. [PMID: 38523660 PMCID: PMC10958382 DOI: 10.3389/fmolb.2024.1352781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/29/2024] [Indexed: 03/26/2024] Open
Abstract
Cilia are cellular signaling hubs. Given that human kinases are central regulators of signaling, it is not surprising that kinases are key players in cilia biology. In fact, many kinases modulate ciliogenesis, which is the generation of cilia, and distinct ciliary pathways. Several of these kinases are understudied with few publications dedicated to the interrogation of their function. Recent efforts to develop chemical probes for members of the cyclin-dependent kinase like (CDKL), never in mitosis gene A (NIMA) related kinase (NEK), and tau tubulin kinase (TTBK) families either have delivered or are working toward delivery of high-quality chemical tools to characterize the roles that specific kinases play in ciliary processes. A better understanding of ciliary kinases may shed light on whether modulation of these targets will slow or halt disease onset or progression. For example, both understudied human kinases and some that are more well-studied play important ciliary roles in neurons and have been implicated in neurodevelopmental, neurodegenerative, and other neurological diseases. Similarly, subsets of human ciliary kinases are associated with cancer and oncological pathways. Finally, a group of genetic disorders characterized by defects in cilia called ciliopathies have associated gene mutations that impact kinase activity and function. This review highlights both progress related to the understanding of ciliary kinases as well as in chemical inhibitor development for a subset of these kinases. We emphasize known roles of ciliary kinases in diseases of the brain and malignancies and focus on a subset of poorly characterized kinases that regulate ciliary biology.
Collapse
Affiliation(s)
- Raymond G. Flax
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Peter Rosston
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cecilia Rocha
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Brian Anderson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jacob L. Capener
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Thomas M. Durcan
- The Neuro’s Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Panagiotis Prinos
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Alison D. Axtman
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
3
|
Lee C, Maier W, Jiang YY, Nakano K, Lechtreck KF, Gaertig J. Global and local functions of the Fused kinase ortholog CdaH in intracellular patterning in Tetrahymena. J Cell Sci 2024; 137:jcs261256. [PMID: 37667859 PMCID: PMC10565251 DOI: 10.1242/jcs.261256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023] Open
Abstract
Ciliates assemble numerous microtubular structures into complex cortical patterns. During ciliate division, the pattern is duplicated by intracellular segmentation that produces a tandem of daughter cells. In Tetrahymena thermophila, the induction and positioning of the division boundary involves two mutually antagonistic factors: posterior CdaA (cyclin E) and anterior CdaI (Hippo kinase). Here, we characterized the related cdaH-1 allele, which confers a pleiotropic patterning phenotype including an absence of the division boundary and an anterior-posterior mispositioning of the new oral apparatus. CdaH is a Fused or Stk36 kinase ortholog that localizes to multiple sites that correlate with the effects of its loss, including the division boundary and the new oral apparatus. CdaH acts downstream of CdaA to induce the division boundary and drives asymmetric cytokinesis at the tip of the posterior daughter. CdaH both maintains the anterior-posterior position of the new oral apparatus and interacts with CdaI to pattern ciliary rows within the oral apparatus. Thus, CdaH acts at multiple scales, from induction and positioning of structures on the cell-wide polarity axis to local organelle-level patterning.
Collapse
Affiliation(s)
- Chinkyu Lee
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Wolfgang Maier
- Bioinformatics, University of Freiburg, 79110 Freiburg, Germany
| | - Yu-Yang Jiang
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Kentaro Nakano
- Degree Programs in Biology, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Karl F Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Jacek Gaertig
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
4
|
Zhou M, Han Y, Jiang J. Ulk4 promotes Shh signaling by regulating Stk36 ciliary localization and Gli2 phosphorylation. eLife 2023; 12:RP88637. [PMID: 38096226 PMCID: PMC10721220 DOI: 10.7554/elife.88637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
The Hedgehog (Hh) family of secreted proteins governs embryonic development and adult tissue homeostasis through the Gli family of transcription factors. Gli is thought to be activated at the tip of primary cilium, but the underlying mechanism has remained poorly understood. Here, we show that Unc-51-like kinase 4 (Ulk4), a pseudokinase and a member of the Ulk kinase family, acts in conjunction with another Ulk family member Stk36 to promote Gli2 phosphorylation and Hh pathway activation. Ulk4 interacts with Stk36 through its N-terminal region containing the pseudokinase domain and with Gli2 via its regulatory domain to bridge the kinase and substrate. Although dispensable for Hh-induced Stk36 kinase activation, Ulk4 is essential for Stk36 ciliary tip localization, Gli2 phosphorylation, and activation. In response to Hh, both Ulk4 and Stk36 colocalize with Gli2 at ciliary tip, and Ulk4 and Stk36 depend on each other for their ciliary tip accumulation. We further show that ciliary localization of Ulk4 depends on Stk36 kinase activity and phosphorylation of Ulk4 on Thr1023, and that ciliary tip accumulation of Ulk4 is essential for its function in the Hh pathway. Taken together, our results suggest that Ulk4 regulates Hh signaling by promoting Stk36-mediated Gli2 phosphorylation and activation at ciliary tip.
Collapse
Affiliation(s)
- Mengmeng Zhou
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pharmacology, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
5
|
Zhang H, Yang M, Zhang J, Li L, Guan T, Liu J, Gong X, Yang F, Shen S, Liu M, Han Y. The putative protein kinase Stk36 is essential for ciliogenesis and CSF flow by associating with Ulk4. FASEB J 2023; 37:e23138. [PMID: 37584603 DOI: 10.1096/fj.202300481r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/28/2023] [Accepted: 07/31/2023] [Indexed: 08/17/2023]
Abstract
Motile cilia lining on the ependymal cells are crucial for cerebrospinal fluid (CSF) flow and its dysfunction is often associated with hydrocephalus. Unc51-like-kinase 4 (Ulk4) was previously linked to CSF flow and motile ciliogenesis in mice, as the hypomorph mutant of Ulk4 (Ulk4tm1a/tm1a ) developed hydrocephalic phenotype resulted from defective ciliogenesis and disturbed ciliary motility, while the underling mechanism is largely obscure. Here, we report that serine/threonine kinase 36 (STK36), a paralog of ULK4, directly interacts with ULK4 and this was demonstrated by yeast two-hybrid (Y2H) in yeast and coimmunoprecipitation (co-IP) assays in HEK293T cells, respectively. The interaction region was confined to their respective N-terminal kinase domain. The hypomorph mutant of Stk36 (Stk36tmE4-/- ) also developed progressive hydrocephalus postnatally and dysfunctional CSF flow, with multiple defects of motile cilia, including reduced ciliary number, disorganized ciliary orientation, defected axonemal structure and inconsistent base body (BB) orientation. Stk36tmE4-/- also disturbed the expression of Foxj1 transcription factor and a range of other ciliogenesis-related genes. All these morphological changes, motile cilia defects and transcriptional dysregulation in the Stk36tmE4-/- are practically copied from that in Ulk4tm1a/tm1a mice. Taken together, we conclude that both Stk36 and Ulk4 are crucial for CSF flow, they cooperate by direct binding with their kinase domain to regulate the Foxj1 transcription factor pathways for ciliogenesis and cilia function, not limited to CSF flow. The underlying molecular mechanism probably conserved in evolution and could be extended to other metazoans.
Collapse
Affiliation(s)
- Hongye Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Key Laboratory of Molecular and Cellular Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Meimei Yang
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Jianhua Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Key Laboratory of Molecular and Cellular Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Li Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Key Laboratory of Molecular and Cellular Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Tianyuan Guan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Jiaxin Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Key Laboratory of Molecular and Cellular Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Xuanwei Gong
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Key Laboratory of Molecular and Cellular Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Fan Yang
- Department of Neurology, Hebei Children's Hospital, Shijiazhuang, China
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Min Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Key Laboratory of Molecular and Cellular Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Yongfeng Han
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Key Laboratory of Molecular and Cellular Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| |
Collapse
|
6
|
McCoy CJ, Paupelin-Vaucelle H, Gorilak P, Beneke T, Varga V, Gluenz E. ULK4 and Fused/STK36 interact to mediate assembly of a motile flagellum. Mol Biol Cell 2023; 34:ar66. [PMID: 36989043 PMCID: PMC10295485 DOI: 10.1091/mbc.e22-06-0222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Unc-51-like kinase (ULK) family serine-threonine protein kinase homologues have been linked to the function of motile cilia in diverse species. Mutations in Fused/STK36 and ULK4 in mice resulted in hydrocephalus and other phenotypes consistent with ciliary defects. How either protein contributes to the assembly and function of motile cilia is not well understood. Here we studied the phenotypes of ULK4 and Fused gene knockout (KO) mutants in the flagellated protist Leishmania mexicana. Both KO mutants exhibited a variety of structural defects of the flagellum cytoskeleton. Biochemical approaches indicate spatial proximity of these proteins and indicate a direct interaction between the N-terminus of LmxULK4 and LmxFused. Both proteins display a dispersed localization throughout the cell body and flagellum, with enrichment near the flagellar base and tip. The stable expression of LmxULK4 was dependent on the presence of LmxFused. Fused/STK36 was previously shown to localize to mammalian motile cilia, and we demonstrate here that ULK4 also localizes to the motile cilia in mouse ependymal cells. Taken together these data suggest a model where the pseudokinase ULK4 is a positive regulator of the kinase Fused/ STK36 in a pathway required for stable assembly of motile cilia.
Collapse
Affiliation(s)
- Ciaran J. McCoy
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | | | - Peter Gorilak
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, 142 20 Prague 4, Czech Republic
| | - Tom Beneke
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Vladimir Varga
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, 142 20 Prague 4, Czech Republic
| | - Eva Gluenz
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, United Kingdom
| |
Collapse
|
7
|
Rethinking the cilia hypothesis of hydrocephalus. Neurobiol Dis 2022; 175:105913. [DOI: 10.1016/j.nbd.2022.105913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
|
8
|
Luo S, Zheng N, Lang B. ULK4 in Neurodevelopmental and Neuropsychiatric Disorders. Front Cell Dev Biol 2022; 10:873706. [PMID: 35493088 PMCID: PMC9039724 DOI: 10.3389/fcell.2022.873706] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/29/2022] [Indexed: 11/21/2022] Open
Abstract
The gene Unc51-like kinase 4 (ULK4) belongs to the Unc-51-like serine/threonine kinase family and is assumed to encode a pseudokinase with unclear function. Recently, emerging evidence has suggested that ULK4 may be etiologically involved in a spectrum of neuropsychiatric disorders including schizophrenia, but the underlying mechanism remains unaddressed. Here, we summarize the key findings of the structure and function of the ULK4 protein to provide comprehensive insights to better understand ULK4-related neurodevelopmental and neuropsychiatric disorders and to aid in the development of a ULK4-based therapeutic strategy.
Collapse
Affiliation(s)
- Shilin Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, China
| | - Nanxi Zheng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Nanxi Zheng, ; Bing Lang,
| | - Bing Lang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Nanxi Zheng, ; Bing Lang,
| |
Collapse
|
9
|
Luettich K, Sharma M, Yepiskoposyan H, Breheny D, Lowe FJ. An Adverse Outcome Pathway for Decreased Lung Function Focusing on Mechanisms of Impaired Mucociliary Clearance Following Inhalation Exposure. FRONTIERS IN TOXICOLOGY 2022; 3:750254. [PMID: 35295103 PMCID: PMC8915806 DOI: 10.3389/ftox.2021.750254] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/11/2021] [Indexed: 01/23/2023] Open
Abstract
Adverse outcome pathways (AOPs) help to organize available mechanistic information related to an adverse outcome into key events (KEs) spanning all organizational levels of a biological system(s). AOPs, therefore, aid in the biological understanding of a particular pathogenesis and also help with linking exposures to eventual toxic effects. In the regulatory context, knowledge of disease mechanisms can help design testing strategies using in vitro methods that can measure or predict KEs relevant to the biological effect of interest. The AOP described here evaluates the major processes known to be involved in regulating efficient mucociliary clearance (MCC) following exposures causing oxidative stress. MCC is a key aspect of the innate immune defense against airborne pathogens and inhaled chemicals and is governed by the concerted action of its functional components, the cilia and airway surface liquid (ASL). The AOP network described here consists of sequences of KEs that culminate in the modulation of ciliary beat frequency and ASL height as well as mucus viscosity and hence, impairment of MCC, which in turn leads to decreased lung function.
Collapse
Affiliation(s)
- Karsta Luettich
- Philip Morris International R&D, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Monita Sharma
- PETA Science Consortium International e.V., Stuttgart, Germany
| | - Hasmik Yepiskoposyan
- Philip Morris International R&D, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Damien Breheny
- British American Tobacco (Investments) Ltd., Group Research and Development, Southampton, United Kingdom
| | - Frazer J Lowe
- Broughton Nicotine Services, Earby, Lancashire, United Kingdom
| |
Collapse
|
10
|
Avazzadeh S, Quinlan LR, Reilly J, McDonagh K, Jalali A, Wang Y, McInerney V, Krawczyk J, Ding Y, Fitzgerald J, O'Sullivan M, Forman EB, Lynch SA, Ennis S, Feerick N, Reilly R, Li W, Shen X, Yang G, Lu Y, Peeters H, Dockery P, O'Brien T, Shen S, Gallagher L. NRXN1α +/- is associated with increased excitability in ASD iPSC-derived neurons. BMC Neurosci 2021; 22:56. [PMID: 34525970 PMCID: PMC8442436 DOI: 10.1186/s12868-021-00661-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Background NRXN1 deletions are identified as one of major rare risk factors for autism spectrum disorder (ASD) and other neurodevelopmental disorders. ASD has 30% co-morbidity with epilepsy, and the latter is associated with excessive neuronal firing. NRXN1 encodes hundreds of presynaptic neuro-adhesion proteins categorized as NRXN1α/β/γ. Previous studies on cultured cells show that the short NRXN1β primarily exerts excitation effect, whereas the long NRXN1α which is more commonly deleted in patients involves in both excitation and inhibition. However, patient-derived models are essential for understanding functional consequences of NRXN1α deletions in human neurons. We recently derived induced pluripotent stem cells (iPSCs) from five controls and three ASD patients carrying NRXN1α+/- and showed increased calcium transients in patient neurons. Methods In this study we investigated the electrophysiological properties of iPSC-derived cortical neurons in control and ASD patients carrying NRXN1α+/- using patch clamping. Whole genome RNA sequencing was carried out to further understand the potential underlying molecular mechanism. Results NRXN1α+/- cortical neurons were shown to display larger sodium currents, higher AP amplitude and accelerated depolarization time. RNASeq analyses revealed transcriptomic changes with significant upregulation glutamatergic synapse and ion channels/transporter activity including voltage-gated potassium channels (GRIN1, GRIN3B, SLC17A6, CACNG3, CACNA1A, SHANK1), which are likely to couple with the increased excitability in NRXN1α+/- cortical neurons. Conclusions Together with recent evidence of increased calcium transients, our results showed that human NRXN1α+/- isoform deletions altered neuronal excitability and non-synaptic function, and NRXN1α+/- patient iPSCs may be used as an ASD model for therapeutic development with calcium transients and excitability as readouts. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-021-00661-0.
Collapse
Affiliation(s)
- Sahar Avazzadeh
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | - Leo R Quinlan
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, CÚRAM SFI Centre for Research in Medical Devices, National University of Ireland (NUI), Galway, Ireland
| | - Jamie Reilly
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | - Katya McDonagh
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | | | - Yanqin Wang
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland.,Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Veronica McInerney
- HRB Clinical Research Facility, National University of Ireland (NUI), Galway, Ireland
| | - Janusz Krawczyk
- Department of Haematology, Galway University Hospital, Galway, Ireland
| | - Yicheng Ding
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | | | - Matthew O'Sullivan
- Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Eva B Forman
- Children's University Hospital, Temple Street, Dublin, Ireland
| | - Sally A Lynch
- Children's University Hospital, Temple Street, Dublin, Ireland.,Department of Clinical Genetics, OLCHC, Dublin 12, Ireland
| | - Sean Ennis
- School of Medicine and Medical Science, UCD Academic Centre On Rare Diseases, University College Dublin, Dublin, Ireland
| | - Niamh Feerick
- Centre for Bioengineering, Trinity College Institute of Neuroscience, School of Medicine, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Richard Reilly
- Centre for Bioengineering, Trinity College Institute of Neuroscience, School of Medicine, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Shen
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Guangming Yang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yin Lu
- College of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hilde Peeters
- Centre for Human Genetics, University Hospital Leuven, KU Leuven, 3000, Leuven, Belgium
| | - Peter Dockery
- Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland (NUI), Galway, Ireland
| | - Timothy O'Brien
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland
| | - Sanbing Shen
- School of Medicine, Regenerative Medicine Institute, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Ireland. .,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, D02, Ireland.
| | - Louise Gallagher
- Trinity Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
11
|
Mecklenburg N, Kowalczyk I, Witte F, Görne J, Laier A, Mamo TM, Gonschior H, Lehmann M, Richter M, Sporbert A, Purfürst B, Hübner N, Hammes A. Identification of disease-relevant modulators of the SHH pathway in the developing brain. Development 2021; 148:272000. [PMID: 34463328 DOI: 10.1242/dev.199307] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
Pathogenic gene variants in humans that affect the sonic hedgehog (SHH) pathway lead to severe brain malformations with variable penetrance due to unknown modifier genes. To identify such modifiers, we established novel congenic mouse models. LRP2-deficient C57BL/6N mice suffer from heart outflow tract defects and holoprosencephaly caused by impaired SHH activity. These defects are fully rescued on a FVB/N background, indicating a strong influence of modifier genes. Applying comparative transcriptomics, we identified Pttg1 and Ulk4 as candidate modifiers upregulated in the rescue strain. Functional analyses showed that ULK4 and PTTG1, both microtubule-associated proteins, are positive regulators of SHH signaling, rendering the pathway more resilient to disturbances. In addition, we characterized ULK4 and PTTG1 as previously unidentified components of primary cilia in the neuroepithelium. The identification of genes that powerfully modulate the penetrance of genetic disturbances affecting the brain and heart is likely relevant to understanding the variability in human congenital disorders.
Collapse
Affiliation(s)
- Nora Mecklenburg
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Izabela Kowalczyk
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Franziska Witte
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Jessica Görne
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Alena Laier
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Tamrat M Mamo
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Hannes Gonschior
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Martin Lehmann
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Matthias Richter
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Bettina Purfürst
- Electron microscopy technology platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany.,Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Annette Hammes
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| |
Collapse
|
12
|
Kumar V, Umair Z, Kumar S, Goutam RS, Park S, Kim J. The regulatory roles of motile cilia in CSF circulation and hydrocephalus. Fluids Barriers CNS 2021; 18:31. [PMID: 34233705 PMCID: PMC8261947 DOI: 10.1186/s12987-021-00265-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background Cerebrospinal fluid (CSF) is an ultra-filtrated colorless brain fluid that circulates within brain spaces like the ventricular cavities, subarachnoid space, and the spine. Its continuous flow serves many primary functions, including nourishment, brain protection, and waste removal. Main body The abnormal accumulation of CSF in brain cavities triggers severe hydrocephalus. Accumulating evidence had indicated that synchronized beats of motile cilia (cilia from multiciliated cells or the ependymal lining in brain ventricles) provide forceful pressure to generate and restrain CSF flow and maintain overall CSF circulation within brain spaces. In humans, the disorders caused by defective primary and/or motile cilia are generally referred to as ciliopathies. The key role of CSF circulation in brain development and its functioning has not been fully elucidated. Conclusions In this review, we briefly discuss the underlying role of motile cilia in CSF circulation and hydrocephalus. We have reviewed cilia and ciliated cells in the brain and the existing evidence for the regulatory role of functional cilia in CSF circulation in the brain. We further discuss the findings obtained for defective cilia and their potential involvement in hydrocephalus. Furthermore, this review will reinforce the idea of motile cilia as master regulators of CSF movements, brain development, and neuronal diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Shiv Kumar
- School of Psychology and Neuroscience, University of St. Andrews, St. Mary's Quad, South Street. St. Andrews, Fife, KY16 9JP, UK
| | - Ravi Shankar Goutam
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Soochul Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
13
|
Hu L, Chen Y, Yang CP, Huang Y, Song NN, Chen JY, Sun YL, Ding YQ, Lang B. Ulk4, a Newly Discovered Susceptibility Gene for Schizophrenia, Regulates Corticogenesis in Mice. Front Cell Dev Biol 2021; 9:645368. [PMID: 34235142 PMCID: PMC8255617 DOI: 10.3389/fcell.2021.645368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/23/2021] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia (SCZ) is a chronic and severe mental disease that affects around 1% of the population. The precise etiology of SCZ still remains largely unknown, and no conclusive mechanisms are firmly established. Recent advances in epidemiological and clinical investigation support an overwhelmingly strong neurodevelopmental origin for SCZ. Here, we demonstrated that Unc-51-like kinase 4 (Ulk4), a novel risk factor for major mental disorders including schizophrenia, is involved in the corticogenesis. Deletion of Ulk4 in mice led to significantly thinner layers of II–III, and V in the cerebral cortex, which was confirmed in conditional Ulk4 deletion mice achieved by Cre-loxp strategy. This abnormality might be caused by decreased intermediate neural progenitors and increased apoptosis. Thus, our data suggest that Ulk4 manipulates the behaviors of neural progenitors during brain development and, when functionally defective, leads to the reduction of specific cortical layers. This anomaly may increase predisposition to a range of neurodevelopmental disorders, including SCZ.
Collapse
Affiliation(s)
- Ling Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yi Chen
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Cui-Ping Yang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Ying Huang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Ning-Ning Song
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jia-Yin Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Ling Sun
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.,Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China.,Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Bing Lang
- National Clinical Research Centre for Mental Health, Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
McKnight I, Hart C, Park IH, Shim JW. Genes causing congenital hydrocephalus: Their chromosomal characteristics of telomere proximity and DNA compositions. Exp Neurol 2021; 335:113523. [PMID: 33157092 PMCID: PMC7750280 DOI: 10.1016/j.expneurol.2020.113523] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/10/2020] [Accepted: 10/30/2020] [Indexed: 01/06/2023]
Abstract
Congenital hydrocephalus (CH) is caused by genetic mutations, but whether factors impacting human genetic mutations are disease-specific remains elusive. Given two factors associated with high mutation rates, we reviewed how many disease-susceptible genes match with (i) proximity to telomeres or (ii) high adenine and thymine (A + T) content in human CH as compared to other disorders of the central nervous system (CNS). We extracted genomic information using a genome data viewer. Importantly, 98 of 108 genes causing CH satisfied (i) or (ii), resulting in >90% matching rate. However, such a high accordance no longer sustained as we checked two factors in Alzheimer's disease (AD) and/or familial Parkinson's disease (fPD), resulting in 84% and 59% matching, respectively. A disease-specific matching of telomere proximity or high A + T content predicts causative genes of CH much better than neurodegenerative diseases and other CNS conditions, likely due to sufficient number of known causative genes (n = 108) and precise determination and classification of the genotype and phenotype. Our analysis suggests a need for identifying genetic basis of both factors before human clinical studies, to prioritize putative genes found in preclinical models into the likely (meeting at least one) and more likely candidate (meeting both), which predisposes human genes to mutations.
Collapse
Affiliation(s)
- Ian McKnight
- Department of Biomedical Engineering, Marshall University, Huntington, WV 25755, USA
| | - Christoph Hart
- Department of Biomedical Engineering, Marshall University, Huntington, WV 25755, USA
| | - In-Hyun Park
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Joon W Shim
- Department of Biomedical Engineering, Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
15
|
Ependymal cells-CSF flow regulates stress-induced depression. Mol Psychiatry 2021; 26:7308-7315. [PMID: 34234280 PMCID: PMC8873010 DOI: 10.1038/s41380-021-01202-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 11/09/2022]
Abstract
Major depressive disorder (MDD) is a severe, common mood disorder. While reduced cerebrospinal fluid (CSF) flow adversely affects brain metabolism and fluid balance in the aging population and during development, only indirect evidence links aberrant CSF circulation with many diseases including neurological, neurodegenerative, and psychiatric disorders, such as anxiety and depression. Here we show a very high concentration of p11 as a key molecular determinant for depression in ependymal cells, which is significantly decreased in patients with MDD, and in two mouse models of depression induced by chronic stress, such as restraint and social isolation. The loss of p11 in ependymal cells causes disoriented ependymal planar cell polarity (PCP), reduced CSF flow, and depression-like and anxiety-like behaviors. p11 intrinsically controls PCP core genes, which mediates CSF flow. Viral expression of p11 in ependymal cells specifically rescues the pathophysiological and behavioral deficits caused by loss of p11. Taken together, our results identify a new role and a key molecular determinant for ependymal cell-driven CSF flow in mood disorders and suggest a novel strategy for development of treatments for stress-associated neurological, neurodegenerative, and psychiatric disorders.
Collapse
|
16
|
Gòdia M, Reverter A, González-Prendes R, Ramayo-Caldas Y, Castelló A, Rodríguez-Gil JE, Sánchez A, Clop A. A systems biology framework integrating GWAS and RNA-seq to shed light on the molecular basis of sperm quality in swine. Genet Sel Evol 2020; 52:72. [PMID: 33292187 PMCID: PMC7724732 DOI: 10.1186/s12711-020-00592-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Genetic pressure in animal breeding is sparking the interest of breeders for selecting elite boars with higher sperm quality to optimize ejaculate doses and fertility rates. However, the molecular basis of sperm quality is not yet fully understood. Our aim was to identify candidate genes, pathways and DNA variants associated to sperm quality in swine by analysing 25 sperm-related phenotypes and integrating genome-wide association studies (GWAS) and RNA-seq under a systems biology framework. RESULTS By GWAS, we identified 12 quantitative trait loci (QTL) associated to the percentage of head and neck abnormalities, abnormal acrosomes and motile spermatozoa. Candidate genes included CHD2, KATNAL2, SLC14A2 and ABCA1. By RNA-seq, we identified a wide repertoire of mRNAs (e.g. PRM1, OAZ3, DNAJB8, TPPP2 and TNP1) and miRNAs (e.g. ssc-miR-30d, ssc-miR-34c, ssc-miR-30c-5p, ssc-miR-191, members of the let-7 family and ssc-miR-425-5p) with functions related to sperm biology. We detected 6128 significant correlations (P-value ≤ 0.05) between sperm traits and mRNA abundances. By expression (e)GWAS, we identified three trans-expression QTL involving the genes IQCJ, ACTR2 and HARS. Using the GWAS and RNA-seq data, we built a gene interaction network. We considered that the genes and interactions that were present in both the GWAS and RNA-seq networks had a higher probability of being actually involved in sperm quality and used them to build a robust gene interaction network. In addition, in the final network we included genes with RNA abundances correlated with more than four semen traits and miRNAs interacting with the genes on the network. The final network was enriched for genes involved in gamete generation and development, meiotic cell cycle, DNA repair or embryo implantation. Finally, we designed a panel of 73 SNPs based on the GWAS, eGWAS and final network data, that explains between 5% (for sperm cell concentration) and 36% (for percentage of neck abnormalities) of the phenotypic variance of the sperm traits. CONCLUSIONS By applying a systems biology approach, we identified genes that potentially affect sperm quality and constructed a SNP panel that explains a substantial part of the phenotypic variance for semen quality in our study and that should be tested in other swine populations to evaluate its relevance for the pig breeding sector.
Collapse
Affiliation(s)
- Marta Gòdia
- Animal Genomics Group, Centre for Research in Agricultural Genomics (CRAG) CSIC-IRTA-UAB-UB, Campus UAB, Cerdanyola del Vallès, 08193, Barcelona, Catalonia, Spain
| | - Antonio Reverter
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, 306 Carmody Rd., St. Lucia, Brisbane, QLD, 4067, Australia
| | - Rayner González-Prendes
- Animal Breeding and Genomics, Wageningen University & Research, 6708PB, Wageningen, The Netherlands
| | - Yuliaxis Ramayo-Caldas
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140, Caldes de Montbui, Catalonia, Spain
| | - Anna Castelló
- Animal Genomics Group, Centre for Research in Agricultural Genomics (CRAG) CSIC-IRTA-UAB-UB, Campus UAB, Cerdanyola del Vallès, 08193, Barcelona, Catalonia, Spain.,Unit of Animal Science, Department of Animal and Food Science, Autonomous University of Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Catalonia, Spain
| | - Joan-Enric Rodríguez-Gil
- Unit of Animal Reproduction, Department of Animal Medicine and Surgery, Autonomous University of Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Catalonia, Spain
| | - Armand Sánchez
- Unit of Animal Science, Department of Animal and Food Science, Autonomous University of Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Catalonia, Spain
| | - Alex Clop
- Animal Genomics Group, Centre for Research in Agricultural Genomics (CRAG) CSIC-IRTA-UAB-UB, Campus UAB, Cerdanyola del Vallès, 08193, Barcelona, Catalonia, Spain. .,Consejo Superior de Investigaciones Científicas (CSIC), 08003, Barcelona, Catalonia, Spain.
| |
Collapse
|
17
|
Fraser L, Paukszto Ł, Mańkowska A, Brym P, Gilun P, Jastrzębski JP, Pareek CS, Kumar D, Pierzchała M. Regulatory Potential of Long Non-Coding RNAs (lncRNAs) in Boar Spermatozoa with Good and Poor Freezability. Life (Basel) 2020; 10:life10110300. [PMID: 33233438 PMCID: PMC7700223 DOI: 10.3390/life10110300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/14/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are suggested to play an important role in the sperm biological processes. We performed de novo transcriptome assembly to characterize lncRNAs in spermatozoa, and to investigate the role of the potential target genes of the differentially expressed lncRNAs (DElncRNAs) in sperm freezability. We detected approximately 4007 DElncRNAs, which were differentially expressed in spermatozoa from boars classified as having good and poor semen freezability (GSF and PSF, respectively). Most of the DElncRNAs were upregulated in boars of the PSF group and appeared to significantly affect the sperm's response to the cryopreservation conditions. Furthermore, we predicted that the potential target genes were regulated by DElncRNAs in cis or trans. It was found that DElncRNAs of both freezability groups had potential cis- and trans-regulatory effects on different protein-coding genes, such as COX7A2L, TXNDC8 and SOX-7. Gene Ontology (GO) enrichment revealed that the DElncRNA target genes are associated with numerous biological processes, including signal transduction, response to stress, cell death (apoptosis), motility and embryo development. Significant differences in the de novo assembled transcriptome expression profiles of the DElncRNAs between the freezability groups were confirmed by quantitative real-time PCR analysis. This study reveals the potential effects of protein-coding genes of DElncRNAs on sperm functions, which could contribute to further research on their relevance in semen freezability.
Collapse
Affiliation(s)
- Leyland Fraser
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
- Correspondence:
| | - Łukasz Paukszto
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Anna Mańkowska
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Paweł Brym
- Department of Animal Genetics, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Przemysław Gilun
- Department of Local Physiological Regulations, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Bydgoska 7, 10-243 Olsztyn, Poland;
| | - Jan P. Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Chandra S. Pareek
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus, University, 87-100 Toruń, Poland;
| | - Dibyendu Kumar
- Waksman Institute of Microbiology, Rutgers, The State University of New Jersey, Piscataway, NJ 08554, USA;
| | - Mariusz Pierzchała
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland;
| |
Collapse
|
18
|
Lee L, Ostrowski LE. Motile cilia genetics and cell biology: big results from little mice. Cell Mol Life Sci 2020; 78:769-797. [PMID: 32915243 DOI: 10.1007/s00018-020-03633-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/11/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
Our understanding of motile cilia and their role in disease has increased tremendously over the last two decades, with critical information and insight coming from the analysis of mouse models. Motile cilia form on specific epithelial cell types and typically beat in a coordinated, whip-like manner to facilitate the flow and clearance of fluids along the cell surface. Defects in formation and function of motile cilia result in primary ciliary dyskinesia (PCD), a genetically heterogeneous disorder with a well-characterized phenotype but no effective treatment. A number of model systems, ranging from unicellular eukaryotes to mammals, have provided information about the genetics, biochemistry, and structure of motile cilia. However, with remarkable resources available for genetic manipulation and developmental, pathological, and physiological analysis of phenotype, the mouse has risen to the forefront of understanding mammalian motile cilia and modeling PCD. This is evidenced by a large number of relevant mouse lines and an extensive body of genetic and phenotypic data. More recently, application of innovative cell biological techniques to these models has enabled substantial advancement in elucidating the molecular and cellular mechanisms underlying the biogenesis and function of mammalian motile cilia. In this article, we will review genetic and cell biological studies of motile cilia in mouse models and their contributions to our understanding of motile cilia and PCD pathogenesis.
Collapse
Affiliation(s)
- Lance Lee
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA. .,Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD, USA.
| | - Lawrence E Ostrowski
- Marsico Lung Institute/Cystic Fibrosis Center and Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
19
|
Zou W, Lv Y, Liu ZI, Xia P, Li H, Jiao J. Loss of Rsph9 causes neonatal hydrocephalus with abnormal development of motile cilia in mice. Sci Rep 2020; 10:12435. [PMID: 32709945 PMCID: PMC7382491 DOI: 10.1038/s41598-020-69447-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Hydrocephalus is a brain disorder triggered by cerebrospinal fluid accumulation in brain cavities. Even though cerebrospinal fluid flow is known to be driven by the orchestrated beating of the bundled motile cilia of ependymal cells, little is known about the mechanism of ciliary motility. RSPH9 is increasingly becoming recognized as a vital component of radial spokes in ciliary "9 + 2" ultrastructure organization. Here, we show that deletion of the Rsph9 gene leads to the development of hydrocephalus in the early postnatal period. However, the neurodevelopment and astrocyte development are normal in embryonic Rsph9-/- mice. The tubular structure of the central aqueduct was comparable in Rsph9-/- mice. Using high-speed video microscopy, we visualized lower beating amplitude and irregular rotation beating pattern of cilia bundles in Rsph9-/- mice compared with that of wild-type mice. And the centriolar patch size was significantly increased in Rsph9-/- cells. TEM results showed that deletion of Rsph9 causes little impact in ciliary axonemal organization but the Rsph9-/- cilia frequently had abnormal ectopic ciliary membrane inclusions. In addition, hydrocephalus in Rsph9-/- mice results in the development of astrogliosis, microgliosis and cerebrovascular abnormalities. Eventually, the ependymal cells sloughed off of the lateral wall. Our results collectively suggested that RSPH9 is essential for ciliary structure and motility of mouse ependymal cilia, and its deletion causes the pathogenesis of hydrocephalus.
Collapse
Affiliation(s)
- Wenzheng Zou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqing Lv
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zux Iang Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China.,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, The Innovation Center of Excellence on Brain Science, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pengyan Xia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Beijing, 100101, China
| | - Hong Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China. .,Innovertion Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China. .,Group of Neural Stem Cell and Neurogenesis, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Chaoyang District, Beijing, 100101, China.
| |
Collapse
|
20
|
Jiang Z, Zhou J, Qin X, Zheng H, Gao B, Liu X, Jin G, Zhou Z. MT1-MMP deficiency leads to defective ependymal cell maturation, impaired ciliogenesis, and hydrocephalus. JCI Insight 2020; 5:132782. [PMID: 32229724 DOI: 10.1172/jci.insight.132782] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 03/26/2020] [Indexed: 01/02/2023] Open
Abstract
Hydrocephalus is characterized by abnormal accumulation of cerebrospinal fluid (CSF) in the ventricular cavity. The circulation of CSF in brain ventricles is controlled by the coordinated beating of motile cilia at the surface of ependymal cells (ECs). Here, we show that MT1-MMP is highly expressed in olfactory bulb, rostral migratory stream, and the ventricular system. Mice deficient for membrane-type 1-MMP (MT1-MMP) developed typical phenotypes observed in hydrocephalus, such as dome-shaped skulls, dilated ventricles, corpus callosum agenesis, and astrocyte hypertrophy, during the first 2 weeks of postnatal development. MT1-MMP-deficient mice exhibited reduced and disorganized motile cilia with the impaired maturation of ECs, leading to abnormal CSF flow. Consistent with the defects in motile cilia morphogenesis, the expression of promulticiliogenic genes was significantly decreased, with a concomitant hyperactivation of Notch signaling in the walls of lateral ventricles in Mmp14-/- brains. Inhibition of Notch signaling by γ-secretase inhibitor restored ciliogenesis in Mmp14-/- ECs. Taken together, these data suggest that MT1-MMP is required for ciliogenesis and EC maturation through suppression of Notch signaling during early brain development. Our findings indicate that MT1-MMP is critical for early brain development and loss of MT1-MMP activity gives rise to hydrocephalus.
Collapse
Affiliation(s)
- Zhixin Jiang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.,Shenzhen Institute of Innovation and Research, University of Hong Kong, Shenzhen, China
| | - Jin Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.,Shenzhen Institute of Innovation and Research, University of Hong Kong, Shenzhen, China
| | - Xin Qin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.,Shenzhen Institute of Innovation and Research, University of Hong Kong, Shenzhen, China
| | - Huiling Zheng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.,Institute for Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Bo Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Xinguang Liu
- Institute for Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Guoxiang Jin
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.,Shenzhen Institute of Innovation and Research, University of Hong Kong, Shenzhen, China
| |
Collapse
|
21
|
Avazzadeh S, McDonagh K, Reilly J, Wang Y, Boomkamp SD, McInerney V, Krawczyk J, Fitzgerald J, Feerick N, O'Sullivan M, Jalali A, Forman EB, Lynch SA, Ennis S, Cosemans N, Peeters H, Dockery P, O'Brien T, Quinlan LR, Gallagher L, Shen S. Increased Ca 2+ signaling in NRXN1α +/- neurons derived from ASD induced pluripotent stem cells. Mol Autism 2019; 10:52. [PMID: 31893021 PMCID: PMC6937972 DOI: 10.1186/s13229-019-0303-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/05/2019] [Indexed: 12/28/2022] Open
Abstract
Background Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a high co-morbidity of epilepsy and associated with hundreds of rare risk factors. NRXN1 deletion is among the commonest rare genetic factors shared by ASD, schizophrenia, intellectual disability, epilepsy, and developmental delay. However, how NRXN1 deletions lead to different clinical symptoms is unknown. Patient-derived cells are essential to investigate the functional consequences of NRXN1 lesions to human neurons in different diseases. Methods Skin biopsies were donated by five healthy donors and three ASD patients carrying NRXN1α+/− deletions. Seven control and six NRXN1α+/− iPSC lines were derived and differentiated into day 100 cortical excitatory neurons using dual SMAD inhibition. Calcium (Ca2+) imaging was performed using Fluo4-AM, and the properties of Ca2+ transients were compared between two groups of neurons. Transcriptome analysis was carried out to undercover molecular pathways associated with NRXN1α+/− neurons. Results NRXN1α+/− neurons were found to display altered calcium dynamics, with significantly increased frequency, duration, and amplitude of Ca2+ transients. Whole genome RNA sequencing also revealed altered ion transport and transporter activity, with upregulated voltage-gated calcium channels as one of the most significant pathways in NRXN1α+/− neurons identified by STRING and GSEA analyses. Conclusions This is the first report to show that human NRXN1α+/− neurons derived from ASD patients’ iPSCs present novel phenotypes of upregulated VGCCs and increased Ca2+ transients, which may facilitate the development of drug screening assays for the treatment of ASD.
Collapse
Affiliation(s)
- Sahar Avazzadeh
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Katya McDonagh
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Jamie Reilly
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Yanqin Wang
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland.,2Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Stephanie D Boomkamp
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Veronica McInerney
- 3HRB Clinical Research Facility, National University of Ireland (NUI), Galway, Ireland
| | - Janusz Krawczyk
- 4Department of Haematology, Galway University Hospital, Galway, Ireland
| | | | - Niamh Feerick
- 5School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | - Amirhossein Jalali
- 6School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eva B Forman
- 7Children's University Hospital, Temple Street, Dublin, Ireland
| | - Sally A Lynch
- Department of Clinical Genetics, OLCHC, Dublin 12, Ireland.,9Children's University Hospital, Temple St, Dublin, Ireland.,10Academic Center on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Sean Ennis
- 11UCD Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Nele Cosemans
- 12Centre for Human Genetics, University Hospital Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Hilde Peeters
- 10Academic Center on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Peter Dockery
- 13Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland (NUI), Galway, Ireland
| | - Timothy O'Brien
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| | - Leo R Quinlan
- 14Physiology and Human Movement Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, National University of Ireland (NUI), Galway, Ireland
| | | | - Sanbing Shen
- 1Regenerative Medicine Institute, School of Medicine, Biomedical Science Building BMS-1021, National University of Ireland Galway, Dangan, Upper Newcastle, Galway, Ireland
| |
Collapse
|
22
|
Tassano E, Uccella S, Giacomini T, Striano P, Severino M, Porta S, Gimelli G, Ronchetto P. Intragenic Microdeletion of ULK4 and Partial Microduplication of BRWD3 in Siblings with Neuropsychiatric Features and Obesity. Cytogenet Genome Res 2018; 156:14-21. [PMID: 30086552 DOI: 10.1159/000491871] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 12/13/2022] Open
Abstract
ULK4 and BRWD3 deletions have been identified in patients with developmental/language delay and intellectual disability. Both genes play pivotal roles in brain development. In particular, ULK4 encodes serine/threonine kinases that are critical for the development and function of the nervous system, while BRWD3 plays a crucial role in ubiquitination, as part of the ubiquitin/proteasome system. We report on 2 brothers, aged 7.6 and 20 years, presenting with cognitive impairment, epilepsy, autistic features, hearing loss, and obesity. Array-CGH analysis demonstrated 2 rare CNVs in both siblings: a paternally inherited microdeletion of ∼145 kb at 3p22.1, disrupting the ULK4 gene, and a maternally inherited microduplication of ∼117 kb at Xq21.1 including only the BRWD3 gene. As already described for other recurrent syndromes with variable phenotype, these findings are challenging in genetic counseling because of an evident variable penetrance. We discuss the possible correlations between the clinical phenotype of our patients and the function of the genes involved in these microrearrangements.
Collapse
|
23
|
Liu M, Fitzgibbon M, Wang Y, Reilly J, Qian X, O'Brien T, Clapcote S, Shen S, Roche M. Ulk4 regulates GABAergic signaling and anxiety-related behavior. Transl Psychiatry 2018; 8:43. [PMID: 29391390 PMCID: PMC5804027 DOI: 10.1038/s41398-017-0091-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/09/2017] [Accepted: 11/30/2017] [Indexed: 01/15/2023] Open
Abstract
Excitation/inhibition imbalance has been proposed as a fundamental mechanism in the pathogenesis of neuropsychiatric and neurodevelopmental disorders, in which copy number variations of the Unc-51 like kinase 4 (ULK4) gene encoding a putative Serine/Threonine kinase have been reported in approximately 1/1000 of patients suffering pleiotropic clinical conditions of schizophrenia, depression, autistic spectrum disorder (ASD), developmental delay, language delay, intellectual disability, or behavioral disorder. The current study characterized behavior of heterozygous Ulk4 +/tm1a mice, demonstrating that Ulk4 +/tm1a mice displayed no schizophrenia-like behavior in acoustic startle reactivity and prepulse inhibition tests or depressive-like behavior in the Porsolt swim or tail suspension tests. However, Ulk4 +/tm1a mice exhibited an anxiety-like behavioral phenotype in several tests. Previously identified hypo-anxious (Atp1a2, Ptn, and Mdk) and hyper-anxious (Gria1, Syngap1, and Npy2r) genes were found to be dysregulated accordingly in Ulk4 mutants. Ulk4 was found to be expressed in GABAergic neurons and the Gad67+ interneurons were significantly reduced in the hippocampus and basolateral amygdala of Ulk4 +/tm1a mice. Transcriptome analyses revealed a marked reduction of GABAergic neuronal subtypes, including Pvalb, Sst, Cck, Npy, and Nos3, as well as significant upregulation of GABA receptors, including Gabra1, Gabra3, Gabra4, Gabra5, and Gabrb3. This is the first evidence that Ulk4 plays a major role in regulating GABAergic signaling and anxiety-like behavior, which may have implications for the development of novel anxiolytic treatments.
Collapse
Affiliation(s)
- Min Liu
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Marie Fitzgibbon
- Physiology, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, National University of Ireland Galway, Galway, Ireland
| | - Yanqin Wang
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Jamie Reilly
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Xiaohong Qian
- National Center for Protein Sciences, Beijing Proteome Research Center, National Engineering Research Center for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing, China
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Steve Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland.
| | - Michelle Roche
- Physiology, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
24
|
Abdelhamed Z, Vuong SM, Hill L, Shula C, Timms A, Beier D, Campbell K, Mangano FT, Stottmann RW, Goto J. A mutation in Ccdc39 causes neonatal hydrocephalus with abnormal motile cilia development in mice. Development 2018; 145:145/1/dev154500. [PMID: 29317443 DOI: 10.1242/dev.154500] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/16/2017] [Indexed: 12/24/2022]
Abstract
Pediatric hydrocephalus is characterized by an abnormal accumulation of cerebrospinal fluid (CSF) and is one of the most common congenital brain abnormalities. However, little is known about the molecular and cellular mechanisms regulating CSF flow in the developing brain. Through whole-genome sequencing analysis, we report that a homozygous splice site mutation in coiled-coil domain containing 39 (Ccdc39) is responsible for early postnatal hydrocephalus in the progressive hydrocephalus (prh) mouse mutant. Ccdc39 is selectively expressed in embryonic choroid plexus and ependymal cells on the medial wall of the forebrain ventricle, and the protein is localized to the axoneme of motile cilia. The Ccdc39prh/prh ependymal cells develop shorter cilia with disorganized microtubules lacking the axonemal inner arm dynein. Using high-speed video microscopy, we show that an orchestrated ependymal ciliary beating pattern controls unidirectional CSF flow on the ventricular surface, which generates bulk CSF flow in the developing brain. Collectively, our data provide the first evidence for involvement of Ccdc39 in hydrocephalus and suggest that the proper development of medial wall ependymal cilia is crucial for normal mouse brain development.
Collapse
Affiliation(s)
- Zakia Abdelhamed
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242, USA.,Department of Anatomy and Embryology, Faculty of Medicine (Girls' Section), Al-Azhar University, Cairo 11651, Egypt
| | - Shawn M Vuong
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242, USA
| | - Lauren Hill
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242, USA
| | - Crystal Shula
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242, USA
| | - Andrew Timms
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| | - David Beier
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| | - Kenneth Campbell
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242, USA.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242 USA
| | - Francesco T Mangano
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242, USA
| | - Rolf W Stottmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242 USA .,Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242 USA
| | - June Goto
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45242, USA
| |
Collapse
|
25
|
Liu M, Xu P, Guan Z, Qian X, Dockery P, Fitzgerald U, O'Brien T, Shen S. Ulk4 deficiency leads to hypomyelination in mice. Glia 2017; 66:175-190. [PMID: 29034508 DOI: 10.1002/glia.23236] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/07/2017] [Accepted: 09/11/2017] [Indexed: 12/14/2022]
Abstract
Brain nerve fibers are insulated by myelin which is produced by oligodendrocytes. Defects in myelination are increasingly recognized as a common pathology underlying neuropsychiatric and neurodevelopmental disorders, which are associated with deletions of the Unc-51-like kinase 4 (ULK4) gene. Key transcription factors have been identified for oligodendrogenesis, but little is known about their associated regulators. Here we report that Ulk4 acts as a key regulator of myelination. Myelination is reduced by half in the Ulk4tm1a/tm1a hypomorph brain, whereas expression of axonal marker genes Tubb3, Nefh, Nefl and Nefm remains unaltered. Transcriptome analyses reveal that 8 (Gfap, Mbp, Mobp, Plp1, Slc1a2, Ttr, Cnp, Scd2) of the 10 most significantly altered genes in the Ulk4tm1a/tm1a brain are myelination-related. Ulk4 is co-expressed in Olig2+ (pan-oligodendrocyte marker) and CC1+ (mature myelinated oligodendrocyte marker) cells during postnatal development. Major oligodendrogeneic transcription factors, including Olig2, Olig1, Myrf, Sox10, Sox8, Sox6, Sox17, Nkx2-2, Nkx6-2 and Carhsp1, are significantly downregulated in the mutants. mRNA transcripts enriched in oligodendrocyte progenitor cells (OPCs), the newly formed oligodendrocytes (NFOs) and myelinating oligodendrocytes (MOs), are significantly attenuated. Expression of stage-specific oligodendrocyte factors including Cspg4, Sox17, Nfasc, Enpp6, Sirt2, Cnp, Plp1, Mbp, Ugt8, Mag and Mog are markedly decreased. Indirect effects of axon caliber and neuroinflammation may also contribute to the hypomyelination, as Ulk4 mutants display smaller axons and increased neuroinflammation. This is the first evidence demonstrating that ULK4 is a crucial regulator of myelination, and ULK4 may therefore become a novel therapeutic target for hypomyelination diseases.
Collapse
Affiliation(s)
- Min Liu
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Ping Xu
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Proteome Research Center, National Engineering Research Center for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Zhenlong Guan
- Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Xiaohong Qian
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Proteome Research Center, National Engineering Research Center for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing, 102206, China
| | - Peter Dockery
- Anatomy, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Una Fitzgerald
- National Centre for Biomedical Engineering Science, Galway Neuroscience Centre, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland
| |
Collapse
|
26
|
Fukuda T, Yanagi S. Psychiatric behaviors associated with cytoskeletal defects in radial neuronal migration. Cell Mol Life Sci 2017; 74:3533-3552. [PMID: 28516224 PMCID: PMC11107632 DOI: 10.1007/s00018-017-2539-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/21/2017] [Accepted: 05/11/2017] [Indexed: 12/17/2022]
Abstract
Normal development of the cerebral cortex is an important process for higher brain functions, such as language, and cognitive and social functions. Psychiatric disorders, such as schizophrenia and autism, are thought to develop owing to various dysfunctions occurring during the development of the cerebral cortex. Radial neuronal migration in the embryonic cerebral cortex is a complex process, which is achieved by strict control of cytoskeletal dynamics, and impairments in this process are suggested to cause various psychiatric disorders. Our recent findings indicate that radial neuronal migration as well as psychiatric behaviors is rescued by controlling microtubule stability during the embryonic stage. In this review, we outline the relationship between psychiatric disorders, such as schizophrenia and autism, and radial neuronal migration in the cerebral cortex by focusing on the cytoskeleton and centrosomes. New treatment strategies for psychiatric disorders will be discussed.
Collapse
Affiliation(s)
- Toshifumi Fukuda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
27
|
The secret life of kinases: insights into non-catalytic signalling functions from pseudokinases. Biochem Soc Trans 2017; 45:665-681. [PMID: 28620028 DOI: 10.1042/bst20160331] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 12/31/2022]
Abstract
Over the past decade, our understanding of the mechanisms by which pseudokinases, which comprise ∼10% of the human and mouse kinomes, mediate signal transduction has advanced rapidly with increasing structural, biochemical, cellular and genetic studies. Pseudokinases are the catalytically defective counterparts of conventional, active protein kinases and have been attributed functions as protein interaction domains acting variously as allosteric modulators of conventional protein kinases and other enzymes, as regulators of protein trafficking or localisation, as hubs to nucleate assembly of signalling complexes, and as transmembrane effectors of such functions. Here, by categorising mammalian pseudokinases based on their known functions, we illustrate the mechanistic diversity among these proteins, which can be viewed as a window into understanding the non-catalytic functions that can be exerted by conventional protein kinases.
Collapse
|
28
|
Protein diversity in discrete structures at the distal tip of the trypanosome flagellum. Proc Natl Acad Sci U S A 2017; 114:E6546-E6555. [PMID: 28724725 DOI: 10.1073/pnas.1703553114] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The distal end of the eukaryotic flagellum/cilium is important for axonemal growth and signaling and has distinct biomechanical properties. Specific flagellum tip structures exist, yet their composition, dynamics, and functions are largely unknown. We used biochemical approaches to identify seven constituents of the flagella connector at the tip of an assembling trypanosome flagellum and three constituents of the axonemal capping structure at the tips of both assembling and mature flagella. Both tip structures contain evolutionarily conserved as well as kinetoplastid-specific proteins, and component assembly into the structures occurs very early during flagellum extension. Localization and functional studies reveal that the flagella connector membrane junction is attached to the tips of extending microtubules of the assembling flagellum by a kinesin-15 family member. On the opposite side, a kinetoplastid-specific kinesin facilitates attachment of the junction to the microtubules in the mature flagellum. Functional studies also suggest roles of several other components and the definition of subdomains in the tip structures.
Collapse
|
29
|
Shimada IS, Acar M, Burgess RJ, Zhao Z, Morrison SJ. Prdm16 is required for the maintenance of neural stem cells in the postnatal forebrain and their differentiation into ependymal cells. Genes Dev 2017; 31:1134-1146. [PMID: 28698301 PMCID: PMC5538436 DOI: 10.1101/gad.291773.116] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 06/12/2017] [Indexed: 11/24/2022]
Abstract
Shimada et al. demonstrate that Prdm16 is required for neural stem cell maintenance and neurogenesis in the adult lateral ventricle subventricular zone and dentate gyrus. Prdm16 is also required for the formation of ciliated ependymal cells in the lateral ventricle. We and others showed previously that PR domain-containing 16 (Prdm16) is a transcriptional regulator required for stem cell function in multiple fetal and neonatal tissues, including the nervous system. However, Prdm16 germline knockout mice died neonatally, preventing us from testing whether Prdm16 is also required for adult stem cell function. Here we demonstrate that Prdm16 is required for neural stem cell maintenance and neurogenesis in the adult lateral ventricle subventricular zone and dentate gyrus. We also discovered that Prdm16 is required for the formation of ciliated ependymal cells in the lateral ventricle. Conditional Prdm16 deletion during fetal development using Nestin-Cre prevented the formation of ependymal cells, disrupting cerebrospinal fluid flow and causing hydrocephalus. Postnatal Prdm16 deletion using Nestin-CreERT2 did not cause hydrocephalus or prevent the formation of ciliated ependymal cells but caused defects in their differentiation. Prdm16 was required in neural stem/progenitor cells for the expression of Foxj1, a transcription factor that promotes ependymal cell differentiation. These studies show that Prdm16 is required for adult neural stem cell maintenance and neurogenesis as well as the formation of ependymal cells.
Collapse
Affiliation(s)
- Issei S Shimada
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Melih Acar
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Bahcesehir University, School of Medicine, Istanbul 34734, Turkey
| | - Rebecca J Burgess
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Zhiyu Zhao
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Sean J Morrison
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
30
|
Liu M, Xu P, O'Brien T, Shen S. Multiple roles of Ulk4 in neurogenesis and brain function. NEUROGENESIS 2017; 4:e1313646. [PMID: 28596978 DOI: 10.1080/23262133.2017.1313646] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/27/2016] [Accepted: 01/18/2017] [Indexed: 10/19/2022]
Abstract
Neurogenesis is essential for proper brain formation and function, and abnormal neural proliferation is an underlying neuropathology of many brain disorders. Recent advances on adult neurogenesis demonstrate that neural stem cells (NSCs) at the subventricular zone (SVZ) are largely derived during mid-embryonic neurogenesis from a subset of cells, which slow down in their pace of cell division,1 become quiescent cells and can be reactivated in need.2 The NSCs at birth constitute the stem cell pool for both postnatal oligodendrogenesis3 and adult neurogenesis.1,2 However, little is known about factors that control the size of NSC pool. The article published in Stem Cells on Jun 14, 2016 by Liu and colleagues described a member of the Unc-51-like serine/threonine kinase family, Ulk4, which plays a critical role in regulating the NSC pool size.4 Authors presented evidence of cell cycle-dependent Ulk4 expression in vitro and in vivo, and reduced NSC pool in targetedly disrupted Ulk4 newborn mice, with disturbed pathways of cell cycle regulation and WNT signaling (Fig. 1), suggesting that ULK4 may be associated with neurodevelopmental, neuropsychiatric as well as neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Liu
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Ping Xu
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Proteome Research Center, National Engineering Research Center for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing, China
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI) Galway, Galway, Ireland
| |
Collapse
|