101
|
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
|
102
|
Olthof AM, Rasmussen JS, Campeau PM, Kanadia RN. Disrupted minor intron splicing is prevalent in Mendelian disorders. Mol Genet Genomic Med 2020; 8:e1374. [PMID: 32573973 PMCID: PMC7507305 DOI: 10.1002/mgg3.1374] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Background Splicing is crucial for proper gene expression, and is predominately executed by the major spliceosome. Conversely, 722 introns in 699 human minor intron‐containing genes (MIGs) are spliced by the minor spliceosome. Splicing of these minor introns is disrupted in diseases caused by pathogenic variants in the minor spliceosome, ultimately leading to the aberrant expression of a subset of these MIGs. However, the effect of variants in minor introns and MIGs on diseases remains unexplored. Methods Variants in MIGs and associated clinical manifestations were identified using ClinVar. The HPO database was then used to curate the related symptoms and affected organ systems. Results: We found pathogenic variants in 211 MIGs, which commonly resulted in intellectual disability, seizures and microcephaly. This revealed a subset of MIGs whose aberrant splicing may contribute to the pathogenesis of minor spliceosome‐related diseases. Moreover, we identified 51 pathogenic variants in minor intron splice sites that reduce the splice site strength and can induce alternative splicing. Conclusion These findings highlight that disrupted minor intron splicing has a broader impact on human diseases than previously appreciated. The hope is that this knowledge will aid in the development of therapeutic strategies that incorporate the minor intron splicing pathway.
Collapse
Affiliation(s)
- Anouk M Olthof
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Jeffrey S Rasmussen
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | | | - Rahul N Kanadia
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.,Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
103
|
Frasca A, Spiombi E, Palmieri M, Albizzati E, Valente MM, Bergo A, Leva B, Kilstrup‐Nielsen C, Bianchi F, Di Carlo V, Di Cunto F, Landsberger N. MECP2 mutations affect ciliogenesis: a novel perspective for Rett syndrome and related disorders. EMBO Mol Med 2020; 12:e10270. [PMID: 32383329 PMCID: PMC7278541 DOI: 10.15252/emmm.201910270] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 12/20/2022] Open
Abstract
Mutations in MECP2 cause several neurological disorders of which Rett syndrome (RTT) represents the best-defined condition. Although mainly working as a transcriptional repressor, MeCP2 is a multifunctional protein revealing several activities, the involvement of which in RTT remains obscure. Besides being mainly localized in the nucleus, MeCP2 associates with the centrosome, an organelle from which primary cilia originate. Primary cilia function as "sensory antennae" protruding from most cells, and a link between primary cilia and mental illness has recently been reported. We herein demonstrate that MeCP2 deficiency affects ciliogenesis in cultured cells, including neurons and RTT fibroblasts, and in the mouse brain. Consequently, the cilium-related Sonic Hedgehog pathway, which is essential for brain development and functioning, is impaired. Microtubule instability participates in these phenotypes that can be rescued by HDAC6 inhibition together with the recovery of RTT-related neuronal defects. Our data indicate defects of primary cilium as a novel pathogenic mechanism that by contributing to the clinical features of RTT might impact on proper cerebellum/brain development and functioning, thus providing a novel therapeutic target.
Collapse
Affiliation(s)
- Angelisa Frasca
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Eleonora Spiombi
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Michela Palmieri
- Neuroscience DivisionIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Elena Albizzati
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Maria Maddalena Valente
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | - Anna Bergo
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | - Barbara Leva
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | - Charlotte Kilstrup‐Nielsen
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | | | - Valerio Di Carlo
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri OttolenghiOrbassanoItaly
- Department of NeuroscienceUniversity of TorinoTorinoItaly
| | - Nicoletta Landsberger
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
- Neuroscience DivisionIRCCS San Raffaele Scientific InstituteMilanItaly
| |
Collapse
|
104
|
Ko D, Kim J, Rhee K, Choi HJ. Identification of a Structurally Dynamic Domain for Oligomer Formation in Rootletin. J Mol Biol 2020; 432:3915-3932. [DOI: 10.1016/j.jmb.2020.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/10/2020] [Accepted: 04/13/2020] [Indexed: 10/24/2022]
|
105
|
Mier P, Andrade-Navarro MA. The features of polyglutamine regions depend on their evolutionary stability. BMC Evol Biol 2020; 20:59. [PMID: 32448113 PMCID: PMC7247214 DOI: 10.1186/s12862-020-01626-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/13/2020] [Indexed: 11/29/2022] Open
Abstract
Background Polyglutamine regions (polyQ) are one of the most studied and prevalent homorepeats in eukaryotes. They have a particular length-dependent codon usage, which relates to a characteristic CAG-slippage mechanism. Pathologically expanded tracts of polyQ are known to form aggregates and are involved in the development of several human neurodegenerative diseases. The non-pathogenic function of polyQ is to mediate protein-protein interactions via a coiled-coil pairing with an interactor. They are usually located in a helical context. Results Here we study the stability of polyQ regions in evolution, using a set of 60 proteomes from four distinct taxonomic groups (Insecta, Teleostei, Sauria and Mammalia). The polyQ regions can be distinctly grouped in three categories based on their evolutionary stability: stable, unstable by length variation (inserted), and unstable by mutations (mutated). PolyQ regions in these categories can be significantly distinguished by their glutamine codon usage, and we show that the CAG-slippage mechanism is predominant in inserted polyQ of Sauria and Mammalia. The polyQ amino acid context is also influenced by the polyQ stability, with a higher proportion of proline residues around inserted polyQ. By studying the secondary structure of the sequences surrounding polyQ regions, we found that regarding the structural conformation around a polyQ, its stability category is more relevant than its taxonomic information. The protein-protein interaction capacity of a polyQ is also affected by its stability, as stable polyQ have more interactors than unstable polyQ. Conclusions Our results show that apart from the sequence of a polyQ, information about its orthologous sequences is needed to assess its function. Codon usage, amino acid context, structural conformation and the protein-protein interaction capacity of polyQ from all studied taxa critically depend on the region stability. There are however some taxa-specific polyQ features that override this importance. We conclude that a taxa-driven evolutionary analysis is of the highest importance for the comprehensive study of any feature of polyglutamine regions.
Collapse
Affiliation(s)
- Pablo Mier
- Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 15, 55128, Mainz, Germany.
| | - Miguel A Andrade-Navarro
- Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 15, 55128, Mainz, Germany
| |
Collapse
|
106
|
Experience Recruits MSK1 to Expand the Dynamic Range of Synapses and Enhance Cognition. J Neurosci 2020; 40:4644-4660. [PMID: 32376781 PMCID: PMC7294801 DOI: 10.1523/jneurosci.2765-19.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
Experience powerfully influences neuronal function and cognitive performance, but the cellular and molecular events underlying the experience-dependent enhancement of mental ability have remained elusive. In particular, the mechanisms that couple the external environment to the genomic changes underpinning this improvement are unknown. To address this, we have used male mice harboring an inactivating mutation of mitogen- and stress-activated protein kinase 1 (MSK1), a brain-derived neurotrophic factor (BDNF)-activated enzyme downstream of the mitogen-activated protein kinase (MAPK) pathway. We show that MSK1 is required for the full extent of experience-induced improvement of spatial memory, for the expansion of the dynamic range of synapses, exemplified by the enhancement of hippocampal long-term potentiation (LTP) and long-term depression (LTD), and for the regulation of the majority of genes influenced by enrichment. In addition, and unexpectedly, we show that experience is associated with an MSK1-dependent downregulation of key MAPK and plasticity-related genes, notably of EGR1/Zif268 and Arc/Arg3.1, suggesting the establishment of a novel genomic landscape adapted to experience. By coupling experience to homeostatic changes in gene expression MSK1, represents a prime mechanism through which the external environment has an enduring influence on gene expression, synaptic function, and cognition. SIGNIFICANCE STATEMENT Our everyday experiences strongly influence the structure and function of the brain. Positive experiences encourage the growth and development of the brain and support enhanced learning and memory and resistance to mood disorders such as anxiety. While this has been known for many years, how this occurs is not clear. Here, we show that many of the positive aspects of experience depend on an enzyme called mitogen- and stress-activated protein kinase 1 (MSK1). Using male mice with a mutation in MSK1, we show that MSK1 is necessary for the majority of gene expression changes associated with experience, extending the range over which the communication between neurons occurs, and for both the persistence of memory and the ability to learn new task rules.
Collapse
|
107
|
Chen JL, Chang CH, Tsai JW. Gli2 Rescues Delays in Brain Development Induced by Kif3a Dysfunction. Cereb Cortex 2020; 29:751-764. [PMID: 29342244 DOI: 10.1093/cercor/bhx356] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 12/23/2017] [Indexed: 01/01/2023] Open
Abstract
The primary cilium in neural stem cells plays distinct roles in different stages during cortical development. Ciliary dysfunctions in human (i.e., ciliopathy) cause developmental defects in multiple organs, including brain developmental delays, which lead to intellectual disabilities and cognitive deficits. However, effective treatment to this devastating developmental disorder is still lacking. Here, we first investigated the effects of ciliopathy on neural stem cells by knocking down Kif3a, a kinesin II motor required for ciliogenesis, in the neurogenic stage of cortical development by in utero electroporation of mouse embryos. Brains electroporated with Kif3a shRNA showed defects in neuronal migration and differentiation, delays in neural stem cell cycle progression, and failures in interkinetic nuclear migration. Interestingly, introduction of Gli1 and Gli2 both can restore the cell cycle progression by elevating cyclin D1 in neural stem cells. Remarkably, enforced Gli2 expression, but not Gli1, partially restored the ability of Kif3a-knockdown neurons to differentiate and move from the germinal ventricular zone to the cortical plate. Moreover, Cyclin D1 knockdown abolished Gli2's rescue effect. These findings suggest Gli2 may rescue neural stem cell proliferation, differentiation and migration through Cyclin D1 pathway and may serve as a potential therapeutic target for human ciliopathy syndromes through modulating the progression of neural stem cell cycle.
Collapse
Affiliation(s)
- Jia-Long Chen
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chia-Hsiang Chang
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Taiwan International Graduate Program (TIGP) in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Jin-Wu Tsai
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center (BRC), Biophotonics and Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
108
|
Sterpka A, Yang J, Strobel M, Zhou Y, Pauplis C, Chen X. Diverged morphology changes of astrocytic and neuronal primary cilia under reactive insults. Mol Brain 2020; 13:28. [PMID: 32122360 PMCID: PMC7053156 DOI: 10.1186/s13041-020-00571-y] [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: 12/06/2019] [Accepted: 02/24/2020] [Indexed: 12/13/2022] Open
Abstract
Primary cilia are centriole-derived sensory organelles that are present in most mammalian cells, including astrocytes and neurons. Evidence is emerging that astrocyte and neuronal primary cilia demonstrate a dichotomy in the mature mouse brain. However, it is unknown how astrocytic and neuronal primary cilia change their morphology and ciliary proteins when exposed to reactive insults including epilepsy and traumatic brain injury. We used a double transgenic mouse strain (Arl13b-mCherry; Centrin2-GFP), in which we found spontaneous seizures, and a cortical injury model to examine the morphological changes of astrocytic and neuronal primary cilia under reactive conditions. Transgenic overexpression of Arl13b drastically increases the length of astrocytic and neuronal primary cilia in the hippocampus, as well as the cilia lengths of cultured astrocytes and neurons. Spontaneous seizures shorten Arl13b-positive astrocytic cilia and AC3-positive neuronal cilia in the hippocampus. In a cortical injury model, Arl13b is not detectable in primary cilia, but Arl13b protein relocates to the cell body and has robust expression in the proximity of injured tissues. In contrast, the number of AC3-positive cilia near injured tissues remains unchanged, but their lengths become shorter. These results on astrocytic cilia implicate Arl13b in regulating astrocyte proliferation and tissue regeneration, while the shortening of AC3-positive cilia suggests adaptive changes of neuronal primary cilia under excitotoxicity.
Collapse
Affiliation(s)
- Ashley Sterpka
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Juan Yang
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Matthew Strobel
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Yuxin Zhou
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Connor Pauplis
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Xuanmao Chen
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA.
| |
Collapse
|
109
|
Wan S, Kim J, Won KJ. SHARP: hyperfast and accurate processing of single-cell RNA-seq data via ensemble random projection. Genome Res 2020; 30:205-213. [PMID: 31992615 PMCID: PMC7050522 DOI: 10.1101/gr.254557.119] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/23/2020] [Indexed: 01/01/2023]
Abstract
To process large-scale single-cell RNA-sequencing (scRNA-seq) data effectively without excessive distortion during dimension reduction, we present SHARP, an ensemble random projection-based algorithm that is scalable to clustering 10 million cells. Comprehensive benchmarking tests on 17 public scRNA-seq data sets show that SHARP outperforms existing methods in terms of speed and accuracy. Particularly, for large-size data sets (more than 40,000 cells), SHARP runs faster than other competitors while maintaining high clustering accuracy and robustness. To the best of our knowledge, SHARP is the only R-based tool that is scalable to clustering scRNA-seq data with 10 million cells.
Collapse
Affiliation(s)
- Shibiao Wan
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Junil Kim
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen North, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen North, Denmark
| | - Kyoung Jae Won
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen North, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen North, Denmark
| |
Collapse
|
110
|
Devlin LA, Ramsbottom SA, Overman LM, Lisgo SN, Clowry G, Molinari E, Powell L, Miles CG, Sayer JA. Embryonic and foetal expression patterns of the ciliopathy gene CEP164. PLoS One 2020; 15:e0221914. [PMID: 31990917 PMCID: PMC6986751 DOI: 10.1371/journal.pone.0221914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/03/2020] [Indexed: 01/20/2023] Open
Abstract
Nephronophthisis-related ciliopathies (NPHP-RC) are a group of inherited genetic disorders that share a defect in the formation, maintenance or functioning of the primary cilium complex, causing progressive cystic kidney disease and other clinical manifestations. Mutations in centrosomal protein 164 kDa (CEP164), also known as NPHP15, have been identified as a cause of NPHP-RC. Here we have utilised the MRC-Wellcome Trust Human Developmental Biology Resource (HDBR) to perform immunohistochemistry studies on human embryonic and foetal tissues to determine the expression patterns of CEP164 during development. Notably expression is widespread, yet defined, in multiple organs including the kidney, retina and cerebellum. Murine studies demonstrated an almost identical Cep164 expression pattern. Taken together, these data support a conserved role for CEP164 throughout the development of numerous organs, which, we suggest, accounts for the multi-system disease phenotype of CEP164-mediated NPHP-RC.
Collapse
Affiliation(s)
- L. A. Devlin
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, England, United Kingdom
| | - S. A. Ramsbottom
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, England, United Kingdom
| | - L. M. Overman
- MRC-Wellcome Trust Human Developmental Biology Resource, Institute of Genetic Medicine, International Centre for Life, Newcastle upon Tyne, England, United Kingdom
| | - S. N. Lisgo
- MRC-Wellcome Trust Human Developmental Biology Resource, Institute of Genetic Medicine, International Centre for Life, Newcastle upon Tyne, England, United Kingdom
| | - G. Clowry
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, England, United Kingdom
| | - E. Molinari
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, England, United Kingdom
| | - L. Powell
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, England, United Kingdom
| | - C. G. Miles
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, England, United Kingdom
| | - J. A. Sayer
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, England, United Kingdom
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Freeman Road, Newcastle upon Tyne, England, United Kingdom
- National Institute for Health Research Newcastle Biomedical Research Centre, Newcastle upon Tyne, England, United Kingdom
- * E-mail:
| |
Collapse
|
111
|
Bowie E, Goetz SC. TTBK2 and primary cilia are essential for the connectivity and survival of cerebellar Purkinje neurons. eLife 2020; 9:51166. [PMID: 31934864 PMCID: PMC7028366 DOI: 10.7554/elife.51166] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Primary cilia are vital signaling organelles that extend from most types of cells, including neurons and glia. These structures are essential for development of many tissues and organs; however, their function in adult tissues, particularly neurons in the brain, remains largely unknown. Tau tubulin kinase 2 (TTBK2) is a critical regulator of ciliogenesis, and is also mutated in a hereditary neurodegenerative disorder, spinocerebellar ataxia type 11 (SCA11). Here, we show that conditional knockout of Ttbk2 in adult mice results in degenerative cerebellar phenotypes that recapitulate aspects of SCA11 including motor coordination deficits and defects to Purkinje cell (PC) integrity. We also find that the Ttbk2 conditional mutant mice quickly lose cilia throughout the brain. We show that conditional knockout of the key ciliary trafficking gene Ift88 in adult mice results in nearly identical cerebellar phenotypes to those of the Ttbk2 knockout, indicating that disruption of ciliary signaling is a key driver of these phenotypes. Our data suggest that primary cilia play an integral role in maintaining the function of PCs in the adult cerebellum and reveal novel insights into mechanisms involved in neurodegeneration.
Collapse
Affiliation(s)
- Emily Bowie
- University Program in Genetics and Genomics, Duke University, Durham, United States
| | - Sarah C Goetz
- Department of Pharmacology and Cancer Biology, Duke University, Durham, United States
| |
Collapse
|
112
|
Intraflagellar transport 20: New target for the treatment of ciliopathies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118641. [PMID: 31893523 DOI: 10.1016/j.bbamcr.2019.118641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 11/22/2022]
Abstract
Cilia are ubiquitous in mammalian cells. The formation and assembly of cilia depend on the normal functioning of the ciliary transport system. In recent years, various proteins involved in the intracellular transport of the cilium have attracted attention, as many diseases are caused by disorders in cilia formation. Intraflagellar transport 20 (IFT20) is a subunit of IFT complex B, which contains approximately 20 protein particles. Studies have shown that defects in IFT20 are associated with numerous system -related diseases, such as those of the urinary system, cardiovascular system, skeletal system, nervous system, immune system, reproductive system, and respiratory system. This review summarizes current research on IFT20.We describe studies related to the role of IFT20 in cilia formation and discuss new targets for treating diseases associated with ciliary dysplasia.
Collapse
|
113
|
Guo J, Otis JM, Suciu SK, Catalano C, Xing L, Constable S, Wachten D, Gupton S, Lee J, Lee A, Blackley KH, Ptacek T, Simon JM, Schurmans S, Stuber GD, Caspary T, Anton ES. Primary Cilia Signaling Promotes Axonal Tract Development and Is Disrupted in Joubert Syndrome-Related Disorders Models. Dev Cell 2019; 51:759-774.e5. [PMID: 31846650 PMCID: PMC6953258 DOI: 10.1016/j.devcel.2019.11.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/08/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022]
Abstract
Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.
Collapse
Affiliation(s)
- Jiami Guo
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA.
| | - James M Otis
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sarah K Suciu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christy Catalano
- Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA
| | - Lei Xing
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sandii Constable
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dagmar Wachten
- Biophysical Imaging, Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Stephanie Gupton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Janice Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Amelia Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Katherine H Blackley
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Travis Ptacek
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Stephane Schurmans
- Laboratory of Functional Genetics, GIGA Research Center, University of Liège, Liège, Belgium
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain and Emotion, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
114
|
Vandervore LV, Schot R, Kasteleijn E, Oegema R, Stouffs K, Gheldof A, Grochowska MM, van der Sterre MLT, van Unen LMA, Wilke M, Elfferich P, van der Spek PJ, Heijsman D, Grandone A, Demmers JAA, Dekkers DHW, Slotman JA, Kremers GJ, Schaaf GJ, Masius RG, van Essen AJ, Rump P, van Haeringen A, Peeters E, Altunoglu U, Kalayci T, Poot RA, Dobyns WB, Bahi-Buisson N, Verheijen FW, Jansen AC, Mancini GMS. Heterogeneous clinical phenotypes and cerebral malformations reflected by rotatin cellular dynamics. Brain 2019; 142:867-884. [PMID: 30879067 PMCID: PMC6439326 DOI: 10.1093/brain/awz045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/26/2018] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
Recessive mutations in RTTN, encoding the protein rotatin, were originally identified as cause of polymicrogyria, a cortical malformation. With time, a wide variety of other brain malformations has been ascribed to RTTN mutations, including primary microcephaly. Rotatin is a centrosomal protein possibly involved in centriolar elongation and ciliogenesis. However, the function of rotatin in brain development is largely unknown and the molecular disease mechanism underlying cortical malformations has not yet been elucidated. We performed both clinical and cell biological studies, aimed at clarifying rotatin function and pathogenesis. Review of the 23 published and five unpublished clinical cases and genomic mutations, including the effect of novel deep intronic pathogenic mutations on RTTN transcripts, allowed us to extrapolate the core phenotype, consisting of intellectual disability, short stature, microcephaly, lissencephaly, periventricular heterotopia, polymicrogyria and other malformations. We show that the severity of the phenotype is related to residual function of the protein, not only the level of mRNA expression. Skin fibroblasts from eight affected individuals were studied by high resolution immunomicroscopy and flow cytometry, in parallel with in vitro expression of RTTN in HEK293T cells. We demonstrate that rotatin regulates different phases of the cell cycle and is mislocalized in affected individuals. Mutant cells showed consistent and severe mitotic failure with centrosome amplification and multipolar spindle formation, leading to aneuploidy and apoptosis, which could relate to depletion of neuronal progenitors often observed in microcephaly. We confirmed the role of rotatin in functional and structural maintenance of primary cilia and determined that the protein localized not only to the basal body, but also to the axoneme, proving the functional interconnectivity between ciliogenesis and cell cycle progression. Proteomics analysis of both native and exogenous rotatin uncovered that rotatin interacts with the neuronal (non-muscle) myosin heavy chain subunits, motors of nucleokinesis during neuronal migration, and in human induced pluripotent stem cell-derived bipolar mature neurons rotatin localizes at the centrosome in the leading edge. This illustrates the role of rotatin in neuronal migration. These different functions of rotatin explain why RTTN mutations can lead to heterogeneous cerebral malformations, both related to proliferation and migration defects.
Collapse
Affiliation(s)
- Laura V Vandervore
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Rachel Schot
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Esmee Kasteleijn
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Renske Oegema
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Department of Pathology, Clinical Bio-informatics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Katrien Stouffs
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Alexander Gheldof
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Martyna M Grochowska
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Marianne L T van der Sterre
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Leontine M A van Unen
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Peter Elfferich
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Peter J van der Spek
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli studi della Campania "L. Vanvitelli", Naples, Italy
| | - Daphne Heijsman
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli studi della Campania "L. Vanvitelli", Naples, Italy
| | - Anna Grandone
- Department of Molecular Genetics, Proteomics Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Jeroen A A Demmers
- Department of Pathology, Optical Imaging Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Dick H W Dekkers
- Department of Pathology, Optical Imaging Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Johan A Slotman
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center (Erasmus MC), 3015 CN Rotterdam, The Netherlands
| | - Gert-Jan Kremers
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center (Erasmus MC), 3015 CN Rotterdam, The Netherlands
| | - Gerben J Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, RB, Groningen, The Netherlands
| | - Roy G Masius
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Anton J van Essen
- Department of Clinical Genetics, LUMC, Leiden University Medical Center, Postzone K-5-R, Postbus 9600, RC Leiden, The Netherlands
| | - Patrick Rump
- Department of Clinical Genetics, LUMC, Leiden University Medical Center, Postzone K-5-R, Postbus 9600, RC Leiden, The Netherlands
| | - Arie van Haeringen
- Department of Pediatric Neurology, Juliana Hospital, Els Borst-Eilersplein 275, 2545 AA Den Haag, The Netherlands
| | - Els Peeters
- Department of Medical genetics, Istanbul Medical Faculty, Istanbul University, Topkapı Mahallesi, Turgut Özal Millet Cd, 34093 Fatih/İstanbul, Turkey
| | - Umut Altunoglu
- Department of Cell biology, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Tugba Kalayci
- Department of Cell biology, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Raymond A Poot
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.,Imagine Institute, INSERM UMR-1163, Laboratory Genetics and Embryology of Congenital Malformations, Paris Descartes University, Institut des Maladies Génétiques 24, Boulevard de Montparnasse, Paris, France
| | - Nadia Bahi-Buisson
- Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel, Brussels, Belgium
| | - Frans W Verheijen
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Anna C Jansen
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| |
Collapse
|
115
|
Shohayeb B, Ho U, Yeap YY, Parton RG, Millard SS, Xu Z, Piper M, Ng DCH. The association of microcephaly protein WDR62 with CPAP/IFT88 is required for cilia formation and neocortical development. Hum Mol Genet 2019; 29:248-263. [DOI: 10.1093/hmg/ddz281] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
WDR62 mutations that result in protein loss, truncation or single amino-acid substitutions are causative for human microcephaly, indicating critical roles in cell expansion required for brain development. WDR62 missense mutations that retain protein expression represent partial loss-of-function mutants that may therefore provide specific insights into radial glial cell processes critical for brain growth. Here we utilized CRISPR/Cas9 approaches to generate three strains of WDR62 mutant mice; WDR62 V66M/V66M and WDR62R439H/R439H mice recapitulate conserved missense mutations found in humans with microcephaly, with the third strain being a null allele (WDR62stop/stop). Each of these mutations resulted in embryonic lethality to varying degrees and gross morphological defects consistent with ciliopathies (dwarfism, anophthalmia and microcephaly). We find that WDR62 mutant proteins (V66M and R439H) localize to the basal body but fail to recruit CPAP. As a consequence, we observe deficient recruitment of IFT88, a protein that is required for cilia formation. This underpins the maintenance of radial glia as WDR62 mutations caused premature differentiation of radial glia resulting in reduced generation of neurons and cortical thinning. These findings highlight the important role of the primary cilium in neocortical expansion and implicate ciliary dysfunction as underlying the pathology of MCPH2 patients.
Collapse
Affiliation(s)
- Belal Shohayeb
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St Lucia 4072, Australia
| | - Uda Ho
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St Lucia 4072, Australia
| | - Yvonne Y Yeap
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St Lucia 4072, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia 4072, Australia
| | - S Sean Millard
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St Lucia 4072, Australia
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Michael Piper
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St Lucia 4072, Australia
| | - Dominic C H Ng
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
116
|
Bayne AN, Trempe JF. Mechanisms of PINK1, ubiquitin and Parkin interactions in mitochondrial quality control and beyond. Cell Mol Life Sci 2019; 76:4589-4611. [PMID: 31254044 PMCID: PMC11105328 DOI: 10.1007/s00018-019-03203-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is a degenerative movement disorder resulting from the loss of specific neuron types in the midbrain. Early environmental and pathophysiological studies implicated mitochondrial damage and protein aggregation as the main causes of PD. These findings are now vindicated by the characterization of more than 20 genes implicated in rare familial forms of the disease. In particular, two proteins encoded by the Parkin and PINK1 genes, whose mutations cause early-onset autosomal recessive PD, function together in a mitochondrial quality control pathway. In this review, we will describe recent development in our understanding of their mechanisms of action, structure, and function. We explain how PINK1 acts as a mitochondrial damage sensor via the regulated proteolysis of its N-terminus and the phosphorylation of ubiquitin tethered to outer mitochondrial membrane proteins. In turn, phospho-ubiquitin recruits and activates Parkin via conformational changes that increase its ubiquitin ligase activity. We then describe how the formation of polyubiquitin chains on mitochondria triggers the recruitment of the autophagy machinery or the formation of mitochondria-derived vesicles. Finally, we discuss the evidence for the involvement of these mechanisms in physiological processes such as immunity and inflammation, as well as the links to other PD genes.
Collapse
Affiliation(s)
- Andrew N Bayne
- Department of Pharmacology and Therapeutics and Centre for Structural Biology, McGill University, 3655 Prom Sir William Osler, Montreal, QC, H3G 1Y6, Canada
| | - Jean-François Trempe
- Department of Pharmacology and Therapeutics and Centre for Structural Biology, McGill University, 3655 Prom Sir William Osler, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
117
|
Abstract
Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical Hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this Review, we highlight central mechanisms by which primary cilia coordinate HH, G protein-coupled receptor, WNT, receptor tyrosine kinase and transforming growth factor-β (TGFβ)/bone morphogenetic protein (BMP) signalling and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.
Collapse
|
118
|
Gpr63 is a modifier of microcephaly in Ttc21b mouse mutants. PLoS Genet 2019; 15:e1008467. [PMID: 31730647 PMCID: PMC6881074 DOI: 10.1371/journal.pgen.1008467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/27/2019] [Accepted: 10/08/2019] [Indexed: 11/19/2022] Open
Abstract
The primary cilium is a signaling center critical for proper embryonic development. Previous studies have demonstrated that mice lacking Ttc21b have impaired retrograde trafficking within the cilium and multiple organogenesis phenotypes, including microcephaly. Interestingly, the severity of the microcephaly in Ttc21baln/aln homozygous null mutants is considerably affected by the genetic background and mutants on an FVB/NJ (FVB) background develop a forebrain significantly smaller than mutants on a C57BL/6J (B6) background. We performed a Quantitative Trait Locus (QTL) analysis to identify potential genetic modifiers and identified two regions linked to differential forebrain size: modifier of alien QTL1 (Moaq1) on chromosome 4 at 27.8 Mb and Moaq2 on chromosome 6 at 93.6 Mb. These QTLs were validated by constructing congenic strains. Further analysis of Moaq1 identified an orphan G-protein coupled receptor (GPCR), Gpr63, as a candidate gene. We identified a SNP that is polymorphic between the FVB and B6 strains in Gpr63 and creates a missense mutation predicted to be deleterious in the FVB protein. We used CRISPR-Cas9 genome editing to create two lines of FVB congenic mice: one with the B6 sequence of Gpr63 and the other with a deletion allele leading to a truncation of the GPR63 C-terminal tail. We then demonstrated that Gpr63 can localize to the cilium in vitro. These alleles affect ciliary localization of GPR63 in vitro and genetically interact with Ttc21baln/aln as Gpr63;Ttc21b double mutants show unique phenotypes including spina bifida aperta and earlier embryonic lethality. This validated Gpr63 as a modifier of multiple Ttc21b neural phenotypes and strongly supports Gpr63 as a causal gene (i.e., a quantitative trait gene, QTG) within the Moaq1 QTL. TTC21B in humans is a known ciliopathy gene and contributes to the pathophysiology of a number of ciliopathies. Mice homozygous for a null allele of Ttc21b also have a spectrum of ciliopathy phenotypes, including microcephaly (small brain). Further work has shown that the severity of the microcephaly significantly depends on the genetic background of the mouse model. The genetic mechanisms of the Ttc21b pathophysiology and the interacting gene network remain far from understood. As an initial attempt to understand the underlying mechanism(s) underlying the variable effects on brain size, we performed a quantitative trait locus (QTL) analysis and found two regions of genomic significance that correlated with smaller brain size. We confirmed both QTLs with congenic lines. One of the two regions was small enough that we considered candidate genes and hypothesized Gpr63 might be a contributing locus for a number of reasons. We evaluated this hypothesis directly with precise variant creation using genome editing and provide evidence that Ttc21b and Gpr63 do indeed genetically interact. Thus, we have been able to combine classical QTL analysis and genome editing to directly test the resulting hypothesis.
Collapse
|
119
|
Dynamic Changes in Ultrastructure of the Primary Cilium in Migrating Neuroblasts in the Postnatal Brain. J Neurosci 2019; 39:9967-9988. [PMID: 31685650 DOI: 10.1523/jneurosci.1503-19.2019] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 10/08/2019] [Accepted: 10/24/2019] [Indexed: 11/21/2022] Open
Abstract
New neurons, referred to as neuroblasts, are continuously generated in the ventricular-subventricular zone of the brain throughout an animal's life. These neuroblasts are characterized by their unique potential for proliferation, formation of chain-like cell aggregates, and long-distance and high-speed migration through the rostral migratory stream (RMS) toward the olfactory bulb (OB), where they decelerate and differentiate into mature interneurons. The dynamic changes of ultrastructural features in postnatal-born neuroblasts during migration are not yet fully understood. Here we report the presence of a primary cilium, and its ultrastructural morphology and spatiotemporal dynamics, in migrating neuroblasts in the postnatal RMS and OB. The primary cilium was observed in migrating neuroblasts in the postnatal RMS and OB in male and female mice and zebrafish, and a male rhesus monkey. Inhibition of intraflagellar transport molecules in migrating neuroblasts impaired their ciliogenesis and rostral migration toward the OB. Serial section transmission electron microscopy revealed that each migrating neuroblast possesses either a pair of centrioles or a basal body with an immature or mature primary cilium. Using immunohistochemistry, live imaging, and serial block-face scanning electron microscopy, we demonstrate that the localization and orientation of the primary cilium are altered depending on the mitotic state, saltatory migration, and deceleration of neuroblasts. Together, our results highlight a close mutual relationship between spatiotemporal regulation of the primary cilium and efficient chain migration of neuroblasts in the postnatal brain.SIGNIFICANCE STATEMENT Immature neurons (neuroblasts) generated in the postnatal brain have a mitotic potential and migrate in chain-like cell aggregates toward the olfactory bulb. Here we report that migrating neuroblasts possess a tiny cellular protrusion called a primary cilium. Immunohistochemical studies with zebrafish, mouse, and monkey brains suggest that the presence of the primary cilium in migrating neuroblasts is evolutionarily conserved. Ciliogenesis in migrating neuroblasts in the rostral migratory stream is suppressed during mitosis and promoted after cell cycle exit. Moreover, live imaging and 3D electron microscopy revealed that ciliary localization and orientation change during saltatory movement of neuroblasts. Our results reveal highly organized dynamics in maturation and positioning of the primary cilium during neuroblast migration that underlie saltatory movement of postnatal-born neuroblasts.
Collapse
|
120
|
Wang G, Hu HB, Chang Y, Huang Y, Song ZQ, Zhou SB, Chen L, Zhang YC, Wu M, Tu HQ, Yuan JF, Wang N, Pan X, Li AL, Zhou T, Zhang XM, He K, Li HY. Rab7 regulates primary cilia disassembly through cilia excision. J Cell Biol 2019; 218:4030-4041. [PMID: 31619485 PMCID: PMC6891077 DOI: 10.1083/jcb.201811136] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 07/07/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Wang et al. identify Rab7 as a novel regulator of primary cilia disassembly. Their findings demonstrate that Rab7 localization to primary cilia is required for intraciliary F-actin polymerization, which is indispensable for the regulation of cilia ectocytosis and disassembly. The primary cilium is a sensory organelle that protrudes from the cell surface. Primary cilia undergo dynamic transitions between assembly and disassembly to exert their function in cell signaling. In this study, we identify the small GTPase Rab7 as a novel regulator of cilia disassembly. Depletion of Rab7 potently induced spontaneous ciliogenesis in proliferating cells and promoted cilia elongation during quiescence. Moreover, Rab7 performs an essential role in cilia disassembly; knockdown of Rab7 blocked serum-induced ciliary resorption, and active Rab7 was required for this process. Further, we demonstrate that Rab7 depletion significantly suppresses cilia tip excision, referred to as cilia ectocytosis, which has been identified as required for cilia disassembly. Mechanically, the failure of F-actin polymerization at the site of excision of cilia tips caused suppression of cilia ectocytosis on Rab7 depletion. Overall, our results suggest a novel function for Rab7 in regulating cilia ectocytosis and cilia disassembly via control of intraciliary F-actin polymerization.
Collapse
Affiliation(s)
- Guang Wang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.,Cancer Institute, Institute of Translational Medicine, The Second Military Medical University, Shanghai, China
| | - Huai-Bin Hu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yan Chang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.,Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yan Huang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Shi-Bo Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Liang Chen
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Hai-Qing Tu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Na Wang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xin Pan
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xue-Min Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Kun He
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Hui-Yan Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China .,Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
121
|
Kim YJ, Kim J. Therapeutic perspectives for structural and functional abnormalities of cilia. Cell Mol Life Sci 2019; 76:3695-3709. [PMID: 31147753 PMCID: PMC11105626 DOI: 10.1007/s00018-019-03158-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022]
Abstract
Ciliopathies are a group of hereditary disorders that result from structural or functional abnormalities of cilia. Recent intense research efforts have uncovered the genetic bases of ciliopathies, and our understanding of the assembly and functions of cilia has been improved significantly. Although mechanism-specific therapies for ciliopathies have not yet received regulatory approval, the use of innovative therapeutic modalities such as oligonucleotide therapy, gene replacement therapy, and gene editing in addition to symptomatic treatments are expected to provide valid treatment options in the near future. Moreover, candidate chemical compounds for developing small molecule drugs to treat ciliopathies have been identified. This review introduces the key features of cilia and ciliopathies, and summarizes the advances as well as the challenges that remain with the development of therapies for treating ciliopathies.
Collapse
Affiliation(s)
- Yong Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
122
|
Reble E, Feng Y, Wigg KG, Barr CL. DNA Variant in the RPGRIP1L Gene Influences Alternative Splicing. MOLECULAR NEUROPSYCHIATRY 2019; 5:97-106. [PMID: 32399473 DOI: 10.1159/000502199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/18/2019] [Indexed: 12/22/2022]
Abstract
The retinitis pigmentosa GTPase regulator interacting protein 1-like (RPGRIP1L) gene encodes a ciliary protein that is critical for processes related to brain development, including development of left-right asymmetry, sonic hedgehog signaling, and neural tube formation. RPGRIP1L is a risk factor for retinal degeneration, and rare, deleterious variants in the RPGRIP1L gene cause Joubert syndrome and Meckel syndrome, both autosomal recessive disorders. These syndromes are characterized by dysfunctional primary cilia that result in abnormal development - and even lethality in the case of Meckel syndrome. Genetic studies have also implicated RPGRIP1L in psychiatric disorders by suggestive findings from genome-wide association studies and findings from rare-variant exome analyses for bipolar disorder and de novo mutations in autism. In this study we identify a common variant in RPGRIP1L, rs7203525, that influences alternative splicing, increasing the inclusion of exon 20 of RPGRIP1L. We detected this alternative splicing association in human postmortem brain tissue samples and, using a minigene assay combined with in vitro mutagenesis, confirmed that the alternative splicing is attributable to the alleles of this variant. The predominate RPGRIP1L isoform expressed in adult brains does not contain exon 20; thus, a shift to include this exon may impact brain function.
Collapse
Affiliation(s)
- Emma Reble
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Yu Feng
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Karen G Wigg
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cathy L Barr
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
123
|
Ubina T, Magallanes M, Srivastava S, Warden CD, Yee JK, Salvaterra PM. A Human Embryonic Stem Cell Model of Aβ-Dependent Chronic Progressive Neurodegeneration. Front Neurosci 2019; 13:1007. [PMID: 31616241 PMCID: PMC6763609 DOI: 10.3389/fnins.2019.01007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/05/2019] [Indexed: 11/13/2022] Open
Abstract
We describe the construction and phenotypic analysis of a human embryonic stem cell model of progressive Aβ-dependent neurodegeneration (ND) with potential relevance to Alzheimer’s disease (AD). We modified one allele of the normal APP locus to directly express a secretory form of Aβ40 or Aβ42, enabling expression from this edited allele to bypass the normal amyloidogenic APP processing pathway. Following neuronal differentiation, edited cell lines specifically accumulate intracellular aggregated/oligomeric Aβ, exhibit a synaptic deficit, and have an abnormal accumulation of endolysosomal vesicles. Edited cultures progress to a stage of overt ND. All phenotypes appear at earlier culture times for Aβ42 relative to Aβ40. Whole transcriptome RNA-Seq analysis identified 23 up and 70 down regulated genes (differentially expressed genes) with similar directional fold change but larger absolute values in the Aβ42 samples suggesting common underlying pathogenic mechanisms. Pathway/annotation analysis suggested that down regulation of extracellular matrix and cilia functions is significantly overrepresented. This cellular model could be useful for uncovering mechanisms directly linking Aβ to neuronal death and as a tool to screen for new therapeutic agents that slow or prevent human ND.
Collapse
Affiliation(s)
- Teresa Ubina
- Department of Developmental and Stem Cell Biology, Beckman Research Institute - City of Hope, Duarte, CA, United States.,Department of Biology, California State University, San Bernardino, San Bernardino, CA, United States
| | - Martha Magallanes
- Department of Developmental and Stem Cell Biology, Beckman Research Institute - City of Hope, Duarte, CA, United States
| | - Saumya Srivastava
- Department of Developmental and Stem Cell Biology, Beckman Research Institute - City of Hope, Duarte, CA, United States
| | - Charles D Warden
- Integrative Genomics Core, Beckman Research Institute - City of Hope, Duarte, CA, United States
| | - Jiing-Kuan Yee
- Department of Diabetes, Beckman Research Institute - City of Hope, Duarte, CA, United States.,Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute - City of Hope, Duarte, CA, United States
| | - Paul M Salvaterra
- Department of Developmental and Stem Cell Biology, Beckman Research Institute - City of Hope, Duarte, CA, United States.,Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute - City of Hope, Duarte, CA, United States
| |
Collapse
|
124
|
He Y, Luo X, Zhou B, Hu T, Meng X, Audano PA, Kronenberg ZN, Eichler EE, Jin J, Guo Y, Yang Y, Qi X, Su B. Long-read assembly of the Chinese rhesus macaque genome and identification of ape-specific structural variants. Nat Commun 2019; 10:4233. [PMID: 31530812 PMCID: PMC6749001 DOI: 10.1038/s41467-019-12174-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/27/2019] [Indexed: 12/20/2022] Open
Abstract
We present a high-quality de novo genome assembly (rheMacS) of the Chinese rhesus macaque (Macaca mulatta) using long-read sequencing and multiplatform scaffolding approaches. Compared to the current Indian rhesus macaque reference genome (rheMac8), rheMacS increases sequence contiguity 75-fold, closing 21,940 of the remaining assembly gaps (60.8 Mbp). We improve gene annotation by generating more than two million full-length transcripts from ten different tissues by long-read RNA sequencing. We sequence resolve 53,916 structural variants (96% novel) and identify 17,000 ape-specific structural variants (ASSVs) based on comparison to ape genomes. Many ASSVs map within ChIP-seq predicted enhancer regions where apes and macaque show diverged enhancer activity and gene expression. We further characterize a subset that may contribute to ape- or great-ape-specific phenotypic traits, including taillessness, brain volume expansion, improved manual dexterity, and large body size. The rheMacS genome assembly serves as an ideal reference for future biomedical and evolutionary studies. Comparative genomic analysis of human and primate relatives can reveal important biological and evolutionary insights. Here, the authors present a long-read assembly of the Chinese rhesus macaque genome and identify ape-specific structural variants.
Collapse
Affiliation(s)
- Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xin Luo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Bin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ting Hu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoyu Meng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Peter A Audano
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Zev N Kronenberg
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA.,Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Jie Jin
- Nextomics Biosciences, Wuhan, 430000, China
| | - Yongbo Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanan Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China. .,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
125
|
Thomas S, Boutaud L, Reilly ML, Benmerah A. Cilia in hereditary cerebral anomalies. Biol Cell 2019; 111:217-231. [DOI: 10.1111/boc.201900012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Sophie Thomas
- Laboratory of Embryology and Genetics of Human MalformationINSERM UMR 1163Paris Descartes UniversityImagine Institute 75015 Paris France
| | - Lucile Boutaud
- Laboratory of Embryology and Genetics of Human MalformationINSERM UMR 1163Paris Descartes UniversityImagine Institute 75015 Paris France
| | - Madeline Louise Reilly
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163Paris Descartes UniversityImagine Institute 75015 Paris France
- Paris Diderot University 75013 Paris France
| | - Alexandre Benmerah
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163Paris Descartes UniversityImagine Institute 75015 Paris France
| |
Collapse
|
126
|
Tajhya R, Delling M. New insights into ion channel-dependent signalling during left-right patterning. J Physiol 2019; 598:1741-1752. [PMID: 31106399 DOI: 10.1113/jp277835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/23/2019] [Indexed: 01/20/2023] Open
Abstract
The left-right organizer (LRO) in the mouse consists of pit cells within the depression, located at the end of the developing notochord, also known as the embryonic node and crown cells lining the outer periphery of the node. Cilia on pit cells are posteriorly tilted, rotate clockwise and generate leftward fluid flow. Primary cilia on crown cells are required to interpret the directionality of fluid movement and initiate flow-dependent gene transcription. Crown cells express PC1-L1 and PC2, which may form a heteromeric polycystin channel complex on primary cilia. It is still only poorly understood how fluid flow activates the ciliary polycystin complex. Besides polycystin channels voltage gated channels like HCN4 and KCNQ1 have been implicated in establishing asymmetry. How this electrical network of ion channels initiates left-sided signalling cascades and differential gene expression is currently only poorly defined.
Collapse
Affiliation(s)
- Rajeev Tajhya
- Department of Physiology, University of California, 1550 4th Street, San Francisco, CA, 94518, USA
| | - Markus Delling
- Department of Physiology, University of California, 1550 4th Street, San Francisco, CA, 94518, USA
| |
Collapse
|
127
|
King CR, A A Quadros AR, Chazeau A, Saarloos I, van der Graaf AJ, Verhage M, Toonen RF. Fbxo41 Promotes Disassembly of Neuronal Primary Cilia. Sci Rep 2019; 9:8179. [PMID: 31160656 PMCID: PMC6546786 DOI: 10.1038/s41598-019-44589-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/20/2019] [Indexed: 01/02/2023] Open
Abstract
Neuronal primary cilia are signaling organelles with crucial roles in brain development and disease. Cilia structure is decisive for their signaling capacities but the mechanisms regulating it are poorly understood. We identify Fbxo41 as a novel Skp1/Cullin1/F-box (SCF) E3-ligase complex subunit that targets to neuronal centrioles where its accumulation promotes disassembly of primary cilia, and affects sonic hedgehog signaling, a canonical ciliary pathway. Fbxo41 targeting to centrioles requires its Coiled-coil and F-box domains. Levels of Fbxo41 at the centrioles inversely correlate with neuronal cilia length, and mutations that disrupt Fbxo41 targeting or assembly into SCF-complexes also disturb its function in cilia disassembly and signaling. Fbxo41 dependent cilia disassembly in mitotic and post-mitotic cells requires rearrangements of the actin-cytoskeleton, but requires Aurora A kinase activation only in mitotic cells, highlighting important mechanistical differences controlling cilia size between mitotic and post-mitotic cells. Phorbol esters induce recruitment of overexpressed Fbxo41 to centrioles and cilia disassembly in neurons, but disassembly can also occur in absence of Fbxo41. We propose that Fbxo41 targeting to centrosomes regulates neuronal cilia structure and signaling capacity in addition to Fbxo41-independent pathways controlling cilia size.
Collapse
Affiliation(s)
- Cillian R King
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Ana R A A Quadros
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Anaël Chazeau
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Ingrid Saarloos
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands.,Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Anne Jolien van der Graaf
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands.,Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
128
|
Ray M, Acharya S, Shambhavi S, Lakhotia SC. Over-expression of Hsp83 in grossly depleted hsrω lncRNA background causes synthetic lethality and l(2)gl phenocopy in Drosophila. J Biosci 2019; 44:36. [PMID: 31180049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We examined interactions between the 83 kDa heat-shock protein (Hsp83) and hsrω long noncoding RNAs (lncRNAs) in hsrω66 Hsp90GFP homozygotes, which almost completely lack hsrω lncRNAs but over-express Hsp83. All +/+; hsrω66 Hsp90GFP progeny died before the third instar. Rare Sp/CyO; hsrω66 Hsp90GFP reached the third instar stage but phenocopied l(2)gl mutants, becoming progressively bulbous and transparent with enlarged brain and died after prolonged larval life. Additionally, ventral ganglia too were elongated. However, hsrω66 Hsp90GFP/TM6B heterozygotes, carrying +/+ or Sp/CyO second chromosomes, developed normally. Total RNA sequencing (+/+, +/+; hsrω66/hsrω66, Sp/CyO; hsrω66/ hsrω66, +/+; Hsp90GFP/Hsp90GFP and Sp/CyO; hsrω66 Hsp90GFP/hsrω66 Hsp90GFP late third instar larvae) revealed similar effects on many genes in hsrω66 and Hsp90GFP homozygotes. Besides additive effect on many of them, numerous additional genes were affected in Sp/CyO; hsrω66 Hsp90GFP larvae, with l(2)gl and several genes regulating the central nervous system being highly down-regulated in surviving Sp/CyO; hsrω66 Hsp90GFP larvae, but not in hsrω66 or Hsp90GFP single mutants. Hsp83 and several omega speckle-associated hnRNPs were bioinformatically found to potentially bind with these gene promoters and transcripts. Since Hsp83 and hnRNPs are also known to interact, elevated Hsp83 in an altered background of hnRNP distribution and dynamics, due to near absence of hsrω lncRNAs and omega speckles, can severely perturb regulatory circuits with unexpected consequences, including down-regulation of tumoursuppressor genes such as l(2)gl.
Collapse
Affiliation(s)
- Mukulika Ray
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | | | | | | |
Collapse
|
129
|
Wang TM, Shen GP, Chen MY, Zhang JB, Sun Y, He J, Xue WQ, Li XZ, Huang SY, Zheng XH, Zhang SD, Hu YZ, Qin HD, Bei JX, Ma J, Mu J, Yao Shugart Y, Jia WH. Genome-Wide Association Study of Susceptibility Loci for Radiation-Induced Brain Injury. J Natl Cancer Inst 2019; 111:620-628. [PMID: 30299488 PMCID: PMC6579742 DOI: 10.1093/jnci/djy150] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/24/2018] [Accepted: 07/29/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Radiation-induced brain injury is a nonnegligible issue in the management of cancer patients treated by partial or whole brain irradiation. In particular, temporal lobe injury (TLI), a deleterious late complication in nasopharyngeal carcinoma, greatly affects the long-term life quality of these patients. Although genome-wide association studies (GWASs) have successfully identified single nucleotide polymorphisms (SNPs) associated with radiation toxicity, genetic variants contributing to the radiation-induced brain injury have not yet been assessed. METHODS We recruited and performed follow-up for a prospective observational cohort, Genetic Architecture of Radiotherapy Toxicity and Prognosis, using magnetic resonance imaging for TLI diagnosis. We conducted genome-wide association analysis in 1082 patients and validated the top associations in two independent cohorts of 1119 and 741 patients, respectively. All statistical tests were two-sided. RESULTS We identified a promoter variant rs17111237 (A > G, minor allele frequency [MAF] = 0.14) in CEP128 associated with TLI risk (hazard ratio = 1.45, 95% confidence interval = 1.26 to 1.66, Pcombined=3.18 × 10-7) which is in moderate linkage disequilibrium (LD) with rs162171 (MAF = 0.18, R2 = 0.69), the top signal in CEP128 (hazard ratio = 1.46, 95% confidence interval = 1.29-1.66, Pcombined= 6.17 × 10-9). Combining the clinical variables with the top SNP, we divided the patients into different subgroups with varying risk with 5-year TLI-free rates ranging from 33.7% to 95.5%. CEP128, a key component of mother centriole, tightly interacts with multiple radiation-resistant genes and plays an important role in maintaining the functional cilia, which otherwise will lead to a malfunction of the neural network. We found that A > G alteration at rs17111237 impaired the promoter activity of CEP128 and knockdown of CEP128 decreased the clonogenic cell survival of U87 cells under radiation. Noteworthy, 12.7% (27/212) of the GWAS-based associated genes (P < .001) were enriched in the neurogenesis pathway. CONCLUSIONS This three-stage study is the first GWAS of radiation-induced brain injury that implicates the genetic susceptibility gene CEP128 involved in TLI development and provides the novel insight into the underlying mechanisms of radiation-induced brain injury.
Collapse
Affiliation(s)
- Tong-Min Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guo-Ping Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming-Yuan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma
| | - Jiang-Bo Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wen-Qiong Xue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xi-Zhao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shao-Yi Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Hui Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shao-Dan Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ye-Zhu Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hai-De Qin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianbing Mu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Yin Yao Shugart
- Unit on Statistical Genomics, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | - Wei-Hua Jia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
130
|
Zhang Y, Williams PR, Jacobi A, Wang C, Goel A, Hirano AA, Brecha NC, Kerschensteiner D, He Z. Elevating Growth Factor Responsiveness and Axon Regeneration by Modulating Presynaptic Inputs. Neuron 2019; 103:39-51.e5. [PMID: 31122676 DOI: 10.1016/j.neuron.2019.04.033] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/01/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022]
Abstract
Despite robust effects on immature neurons, growth factors minimally promote axon regeneration in the adult central nervous system (CNS). Attempting to improve growth-factor responsiveness in mature neurons by dedifferentiation, we overexpressed Lin28 in the retina. Lin28-treated retinas responded to insulin-like growth factor-1 (IGF1) by initiating retinal ganglion cell (RGC) axon regeneration after axotomy. Surprisingly, this effect was cell non-autonomous. Lin28 expression was required only in amacrine cells, inhibitory neurons that innervate RGCs. Ultimately, we found that optic-nerve crush pathologically upregulated activity in amacrine cells, which reduced RGC electrical activity and suppressed growth-factor signaling. Silencing amacrine cells or pharmacologically blocking inhibitory neurotransmission also induced IGF1 competence. Remarkably, RGCs regenerating across these manipulations localized IGF1 receptor to their primary cilia, which maintained their signaling competence and regenerative ability. Thus, our results reveal a circuit-based mechanism that regulates CNS axon regeneration and implicate primary cilia as a regenerative signaling hub.
Collapse
Affiliation(s)
- Yiling Zhang
- F.M. Kirby Neurobiology Center, Children's Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Philip R Williams
- F.M. Kirby Neurobiology Center, Children's Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
| | - Anne Jacobi
- F.M. Kirby Neurobiology Center, Children's Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Chen Wang
- F.M. Kirby Neurobiology Center, Children's Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Anurag Goel
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Arlene A Hirano
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; United States Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Nicholas C Brecha
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; United States Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Children's Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
131
|
Abstract
Primary cilia are singular, sensory organelles that extend from the plasma membrane of most quiescent mammalian cells. These slender, microtubule-based organelles receive and transduce extracellular cues and regulate signaling pathways. Primary cilia are critical to the development and function of many tissue types, and mutation of ciliary genes causes multi-system disorders, termed ciliopathies. Notably, renal cystic disease is one of the most common clinical features of ciliopathies, highlighting a central role for primary cilia in the kidney. Additionally, acute kidney injury and chronic kidney disease are associated with altered primary cilia lengths on renal epithelial cells, suggesting ciliary dynamics and renal physiology are linked. Here we describe methods to examine primary cilia in kidney tissue and in cultured renal cells. We include immunofluorescence and scanning electron microscopy to determine ciliary localization of proteins and cilia structure. Further, we detail cellular assays to measure cilia assembly and disassembly, which regulate cilia length.
Collapse
|
132
|
Bashford AL, Subramanian V. Mice with a conditional deletion of Talpid3 (KIAA0586) - a model for Joubert syndrome. J Pathol 2019; 248:396-408. [PMID: 30924151 PMCID: PMC6767539 DOI: 10.1002/path.5271] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 03/07/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Joubert syndrome (JS) is a ciliopathy associated with mutations in numerous genes encoding cilia components. TALPID3 encoded by KIAA0856 in man (2700049A03Rik in mouse) is a centrosomal protein essential for the assembly of primary cilia. Mutations in KIAA0856 have been recently identified in JS patients. Herein, we describe a novel mouse JS model with a conditional deletion of the conserved exons 11–12 of Talpid3 in the central nervous system which recapitulates the complete cerebellar phenotype seen in JS. Talpid3 mutant mice exhibit key hallmarks of JS including progressive ataxia, severely hypoplastic cerebellar hemispheres and vermis, together with abnormal decussation of the superior cerebellar peduncles. The Purkinje cell layer is disorganised with abnormal dendritic arborisation. The external granule layer (EGL) is thinner, lacks primary cilia, and has a reduced level of proliferation. Furthermore, we describe novel cellular defects including ectopic clusters of mature granule neurons, and abnormal parallel fibre‐derived synapses and disorientation of cells in the EGL. The defective glial scaffold results in abnormal granule cell migration which manifests as ectopic clusters of granule neurons. In addition, we show a reduction in Wnt7a expression suggesting that defects may arise not only from deficiencies in the Hedgehog (Hh) pathway but also due to the additional roles of Talpid3. The Talpid3 conditional knockout mouse is a novel JS model which fully recapitulates the JS cerebellar phenotype. These findings reveal a role for Talpid3 in granule precursor cell migration in the cerebellum (either direct or indirect) which together with defective Hh signalling underlies the JS phenotype. Our findings also illustrate the utility of creating conditional mouse models to assist in unravelling the molecular and cellular mechanisms underlying JS. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Andrew L Bashford
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | | |
Collapse
|
133
|
Huang X, Lin Z, Meng L, Wang K, Liu X, Zhou W, Zheng H, Niu L. Non-invasive Low-Intensity Pulsed Ultrasound Modulates Primary Cilia of Rat Hippocampal Neurons. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:1274-1283. [PMID: 30795858 DOI: 10.1016/j.ultrasmedbio.2018.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 12/18/2018] [Accepted: 12/29/2018] [Indexed: 05/17/2023]
Abstract
Transcranial modulation of primary cilia may provide new opportunities in the treatment of neurodegenerative diseases. This study investigates the effect of non-invasive low-intensity pulsed ultrasound (LIPUS) stimulation on primary cilia of rat hippocampal neurons. Three hours of LIPUS stimulation significantly reduced the incidence rate and length of cilia on cultured neurons (p < 0.01). Similarly, increasing the duration and intensity of LIPUS stimulation decreased the incidence and length of cilia. LIPUS stimulation improved c-fos expression when it was delivered to CA1 of the intact hippocampus of rats. And prolonged LIPUS stimulation (frequency: 0.5 MHz, pulse repetition frequency: 500 Hz, duty cycle: 5%, Ispta: 255 mW/cm2, 10 min/d, 10 d) caused a statistically significant reduction in the incidence rate (p < 0.05) and length of primary cilia (p < 0.01) of neurons in rat CA1 hippocampus. These results indicate the promising potential of LIPUS stimulation in the treatment of primary cilium-related brain diseases.
Collapse
Affiliation(s)
- Xiaowei Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Zhengrong Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Long Meng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kaiyue Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiufang Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
134
|
Over-expression of Hsp83 in grossly depleted hsrω lncRNA background causes synthetic lethality and l(2)gl phenocopy in Drosophila. J Biosci 2019. [DOI: 10.1007/s12038-019-9852-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
135
|
Kakiuchi A, Kohno T, Kakuki T, Kaneko Y, Konno T, Hosaka Y, Hata T, Kikuchi S, Ninomiya T, Himi T, Takano K, Kojima T. Rho-kinase and PKCα Inhibition Induces Primary Cilia Elongation and Alters the Behavior of Undifferentiated and Differentiated Temperature-sensitive Mouse Cochlear Cells. J Histochem Cytochem 2019; 67:523-535. [PMID: 30917058 DOI: 10.1369/0022155419841013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Primary cilia, regulated via distinct signal transduction pathways, play crucial roles in various cellular behaviors. However, the full regulatory mechanism involved in primary cilia development during cellular differentiation is not fully understood, particularly for the sensory hair cells of the mammalian cochlea. In this study, we investigated the effects of the Rho-kinase inhibitor Y27632 and PKCα inhibitor GF109203X on primary cilia-related cell behavior in undifferentiated and differentiated temperature-sensitive mouse cochlear precursor hair cells (the conditionally immortalized US/VOT-E36 cell line). Our results indicate that treatment with Y27632 or GF109203X induced primary cilia elongation and tubulin acetylation in both differentiated and undifferentiated cells. Concomitant with cilia elongation, Y27632 treatment also increased Hook2 and cyclinD1 expression, while only Hook2 expression was increased after treatment with GF109203X. In the undifferentiated cells, we observed an increase in the number of S and G2/M stage cells and a decrease of G1 cells after treatment with Y27632, while the opposite was observed after treatment with GF109203X. Finally, while both treatments decreased oxidative stress, only treatment with Y27632, not GF109203X, induced cell cycle-dependent cell proliferation and cell migration.
Collapse
Affiliation(s)
- Akito Kakiuchi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takuya Kakuki
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yakuto Kaneko
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yukino Hosaka
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomohiro Hata
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shin Kikuchi
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takafumi Ninomiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuo Himi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
136
|
Kalogeropoulou A, Lygerou Z, Taraviras S. Cortical Development and Brain Malformations: Insights From the Differential Regulation of Early Events of DNA Replication. Front Cell Dev Biol 2019; 7:29. [PMID: 30915332 PMCID: PMC6421272 DOI: 10.3389/fcell.2019.00029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/20/2019] [Indexed: 12/27/2022] Open
Abstract
During the development of the cortex distinct populations of Neural Stem Cells (NSCs) are defined by differences in their cell cycle duration, self-renewal capacity and transcriptional profile. A key difference across the distinct populations of NSCs is the length of G1 phase, where the licensing of the DNA replication origins takes place by the assembly of a pre-replicative complex. Licensing of DNA replication is a process that is adapted accordingly to the cell cycle length of NSCs to secure the timed duplication of the genome. Moreover, DNA replication should be efficiently coordinated with ongoing transcription for the prevention of conflicts that would impede the progression of both processes, compromising the normal course of development. In the present review we discuss how the differential regulation of the licensing and initiation of DNA replication in different cortical NSCs populations is integrated with the properties of these stem cells populations. Moreover, we examine the implication of the initial steps of DNA replication in the pathogenetic mechanisms of neurodevelopmental defects and Zika virus-related microcephaly, highlighting the significance of the differential regulation of DNA replication during brain development.
Collapse
Affiliation(s)
| | - Zoi Lygerou
- Department of General Biology, Medical School, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
137
|
Pruski M, Lang B. Primary Cilia-An Underexplored Topic in Major Mental Illness. Front Psychiatry 2019; 10:104. [PMID: 30886591 PMCID: PMC6409319 DOI: 10.3389/fpsyt.2019.00104] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/12/2019] [Indexed: 12/20/2022] Open
Abstract
Though much progress has been made in recent years towards understanding the function and physiology of primary cilia, they remain a somewhat elusive organelle. Some studies have explored the role of primary cilia in the developing nervous system, and their dysfunction has been linked with several neurosensory deficits. Yet, very little has been written on their potential role in psychiatric disorders. This article provides an overview of some of the functions of primary cilia in signalling pathways, and demonstrates that they are a worthy candidate in psychiatric research. The links between primary cilia and major mental illness have been demonstrated to exist at several levels, spanning genetics, signalling pathways, and pharmacology as well as cell division and migration. The primary focus of this review is on the sensory role of the primary cilium and the neurodevelopmental hypothesis of psychiatric disease. As such, the primary cilium is demonstrated to be a key link between the cellular environment and cell behaviour, and hence of key importance in the considerations of the nature and nurture debate in psychiatric research.
Collapse
Affiliation(s)
- Michal Pruski
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Critical Care Laboratory, Critical Care Directorate, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
138
|
Lee J, Yi S, Chang JY, Kim JT, Sul HJ, Park KC, Zhu X, Cheng SY, Kero J, Kim J, Shong M. Loss of Primary Cilia Results in the Development of Cancer in the Murine Thyroid Gland. Mol Cells 2019; 42:113-122. [PMID: 30622229 PMCID: PMC6399002 DOI: 10.14348/molcells.2018.0430] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 01/26/2023] Open
Abstract
Communications at the interface between the apical membrane of follicular cells and the follicular lumen are critical for the homeostasis of thyroid gland. Primary cilia at the apical membrane of thyroid follicular cells may sense follicular luminal environment and regulate follicular homeostasis, although their role in vivo remains to be determined. Here, mice devoid of primary cilia were generated by thyroid follicular epithelial cell-specific deletion of the gene encoding intraflagellar transport protein 88 (Ift88 ). Thyroid follicular cell-specific Ift88-deficient mice showed normal folliculogenesis and hormonogenesis; however, those older than 7 weeks showed irregularly dilated and destroyed follicles in the thyroid gland. With increasing age, follicular cells with malignant properties showing the characteristic nuclear features of human thyroid carcinomas formed papillary and solid proliferative nodules from degenerated thyroid follicles. Furthermore, malignant tumor cells manifested as tumor emboli in thyroid vessels. These findings suggest that loss-of-function of Ift88/primary cilia results in malignant transformation from degenerated thyroid follicles.
Collapse
Affiliation(s)
- Junguee Lee
- Department of Pathology, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943,
Korea
| | - Shinae Yi
- Research Center for Endocrine and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015,
Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015,
Korea
| | - Jung Tae Kim
- Research Center for Endocrine and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015,
Korea
| | - Hae Joung Sul
- Department of Pathology, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943,
Korea
| | - Ki Cheol Park
- Clinical Research Institute, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943,
Korea
| | - Xuguang Zhu
- Laboratory of Molecular Biology, National Cancer Institute, MD 20892-4264,
USA
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, MD 20892-4264,
USA
| | - Jukka Kero
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku,
Finland
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34040,
Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015,
Korea
| |
Collapse
|
139
|
Xiang Y, Tanaka Y, Cakir B, Patterson B, Kim KY, Sun P, Kang YJ, Zhong M, Liu X, Patra P, Lee SH, Weissman SM, Park IH. hESC-Derived Thalamic Organoids Form Reciprocal Projections When Fused with Cortical Organoids. Cell Stem Cell 2019; 24:487-497.e7. [PMID: 30799279 DOI: 10.1016/j.stem.2018.12.015] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/01/2018] [Accepted: 12/20/2018] [Indexed: 12/22/2022]
Abstract
Human brain organoid techniques have rapidly advanced to facilitate investigating human brain development and diseases. These efforts have largely focused on generating telencephalon due to its direct relevance in a variety of forebrain disorders. Despite its importance as a relay hub between cortex and peripheral tissues, the investigation of three-dimensional (3D) organoid models for the human thalamus has not been explored. Here, we describe a method to differentiate human embryonic stem cells (hESCs) to thalamic organoids (hThOs) that specifically recapitulate the development of thalamus. Single-cell RNA sequencing revealed a formation of distinct thalamic lineages, which diverge from telencephalic fate. Importantly, we developed a 3D system to create the reciprocal projections between thalamus and cortex by fusing the two distinct region-specific organoids representing the developing thalamus or cortex. Our study provides a platform for understanding human thalamic development and modeling circuit organizations and related disorders in the brain.
Collapse
Affiliation(s)
- Yangfei Xiang
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yoshiaki Tanaka
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Benjamin Patterson
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Pingnan Sun
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Young-Jin Kang
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Mei Zhong
- Department of Cell Biology, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xinran Liu
- Department of Cell Biology, Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Prabir Patra
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA
| | - Sang-Hun Lee
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sherman M Weissman
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
140
|
Sarkisian MR, Semple-Rowland SL. Emerging Roles of Primary Cilia in Glioma. Front Cell Neurosci 2019; 13:55. [PMID: 30842728 PMCID: PMC6391589 DOI: 10.3389/fncel.2019.00055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/04/2019] [Indexed: 12/12/2022] Open
Abstract
Primary cilia are microtubule-based organelles that are typically present on cells during the G0 or G1-S/G2 phases of the cell cycle. Recent studies of glioblastoma (GBM) biopsies, a brain tumor that is notorious for its aggressive growth and resistance to treatment, show that many cells in the tumor lack cilia. At this point, it remains unclear whether primary cilia promote or suppress glioma tumorigenesis. In this review, we will discuss the different roles that have been proposed for primary cilia in glioma and how cilia may contribute to the resistance of these tumors to current therapies.
Collapse
Affiliation(s)
- Matthew R Sarkisian
- Department of Neuroscience, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, United States.,Preston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, United States
| | - Susan L Semple-Rowland
- Department of Neuroscience, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
141
|
Reilly ML, Benmerah A. Ciliary kinesins beyond IFT: Cilium length, disassembly, cargo transport and signalling. Biol Cell 2019; 111:79-94. [PMID: 30720881 DOI: 10.1111/boc.201800074] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/18/2019] [Indexed: 02/06/2023]
Abstract
Cilia and flagella are microtubule-based antenna which are highly conserved among eukaryotes. In vertebrates, primary and motile cilia have evolved to exert several key functions during development and tissue homoeostasis. Ciliary dysfunction in humans causes a highly heterogeneous group of diseases called ciliopathies, a class of genetic multisystemic disorders primarily affecting kidney, skeleton, retina, lung and the central nervous system. Among key ciliary proteins, kinesin family members (KIF) are microtubule-interacting proteins involved in many diverse cellular functions, including transport of cargo (organelles, proteins and lipids) along microtubules and regulating the dynamics of cytoplasmic and spindle microtubules through their depolymerising activity. Many KIFs are also involved in diverse ciliary functions including assembly/disassembly, motility and signalling. We here review these ciliary kinesins in vertebrates and focus on their involvement in ciliopathy-related disorders.
Collapse
Affiliation(s)
- Madeline Louise Reilly
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Paris Descartes University, Imagine Institute, Paris, 75015, France.,Paris Diderot University, Paris, 75013, France
| | - Alexandre Benmerah
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Paris Descartes University, Imagine Institute, Paris, 75015, France
| |
Collapse
|
142
|
Monaco S, Baur K, Hellwig A, Hölzl-Wenig G, Mandl C, Ciccolini F. A Flow Cytometry-Based Approach for the Isolation and Characterization of Neural Stem Cell Primary Cilia. Front Cell Neurosci 2019; 12:519. [PMID: 30692915 PMCID: PMC6339872 DOI: 10.3389/fncel.2018.00519] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/12/2018] [Indexed: 01/15/2023] Open
Abstract
In the adult mammalian brain, the apical surface of the subependymal zone (SEZ) is covered by many motile ependymal cilia and a few primary cilia originating from rare intermingled neural stem cells (NSCs). In NSCs the primary cilia are key for the transduction of essential extracellular signals such as Sonic hedgehog (SHH) and platelet-derived growth factor (PDGF). Despite their importance, the analysis of NSC primary cilia is greatly hampered by the fact that they are overwhelmingly outnumbered by the motile cilia. We here take advantage of flow cytometry to purify the two cilia types and allow their molecular characterization. Primary cilia were identified based on immunoreactivity to the marker adenylate cyclase type III (AC3) and differential levels of prominin-1 whereas motile cilia displayed immunoreactivity only to the latter. Consistent with the morphological differences between the two classes of cilia, enrichment of motile cilia positively correlated with size. Moreover, we observed age-dependent variations in the abundance of the two groups of ciliary organelles reflecting the changes associated with their development. The two cilia groups also differed with respect to the expression of signaling molecules, since PDGF receptor (PDGFR)α, smoothened (Smo) and CXC chemokine receptor (CXCR)4 were only detected in isolated primary but not motile cilia. Thus, our novel method of cilia isolation and characterization by flow cytometry has the potential to be extended to the study of cilia from different tissues and organs, providing a powerful tool for the investigation of primary cilia in physiological and pathological conditions.
Collapse
Affiliation(s)
- Sara Monaco
- Interdisciplinary Center for Neurosciences (IZN), Department of Neurobiology, University of Heidelberg, Heidelberg, Germany
| | - Katja Baur
- Interdisciplinary Center for Neurosciences (IZN), Department of Neurobiology, University of Heidelberg, Heidelberg, Germany
| | - Andrea Hellwig
- Interdisciplinary Center for Neurosciences (IZN), Department of Neurobiology, University of Heidelberg, Heidelberg, Germany
| | - Gabriele Hölzl-Wenig
- Interdisciplinary Center for Neurosciences (IZN), Department of Neurobiology, University of Heidelberg, Heidelberg, Germany
| | - Claudia Mandl
- Interdisciplinary Center for Neurosciences (IZN), Department of Neurobiology, University of Heidelberg, Heidelberg, Germany
| | - Francesca Ciccolini
- Interdisciplinary Center for Neurosciences (IZN), Department of Neurobiology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
143
|
Kilander MBC, Wang CH, Chang CH, Nestor JE, Herold K, Tsai JW, Nestor MW, Lin YC. A rare human CEP290 variant disrupts the molecular integrity of the primary cilium and impairs Sonic Hedgehog machinery. Sci Rep 2018; 8:17335. [PMID: 30478281 PMCID: PMC6255789 DOI: 10.1038/s41598-018-35614-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 11/07/2018] [Indexed: 12/25/2022] Open
Abstract
The primary cilium is a microtubule-enriched cell-communication organelle that participates in mechanisms controlling tissue development and maintenance, including cerebellar architecture. Centrosomal protein of 290 kDa (CEP290) is a protein important for centrosomal function and ciliogenesis. Mutations in CEP290 have been linked to a group of multi-organ disorders - termed ciliopathies. The neurophysiological deficits observed in ciliopathies are sometimes associated with the progression of autistic traits. Here, the cellular function of two rare variants of CEP290 identified from recent exome sequencing of autistic individuals are investigated. Cells expressing Cep290 carrying the missense mutation R1747Q in mouse exhibited a defective Sonic hedgehog (Shh) signalling response, mislocalisation of the Shh receptor Smoothened (Smo), and dysregulation of ciliary protein mobility, which ultimately disrupted the proliferation of cerebellar granule progenitors (CGPs). This data was furthermore corroborated in an autism patient-derived iPSC line harbouring the R1746Q rare CEP290 variant. Evidence from this study suggests that the R1746Q mutation interferes with the function of CEP290 to maintain the ciliary diffusion barrier and disrupts the integrity of the molecular composition in the primary cilium, which may contribute to alterations in neuroarchitecture.
Collapse
Affiliation(s)
| | - Chun-Hung Wang
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chia-Hsiang Chang
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
- Taiwan International Graduate Program (TIGP) in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Jonathan E Nestor
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, 21201, USA
| | - Kevin Herold
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jin-Wu Tsai
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
- Brain Research Center (BRC), and Biophotonics and Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei, 112, Taiwan
| | - Michael W Nestor
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, 21201, USA
| | - Yu-Chih Lin
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, 21201, USA.
| |
Collapse
|
144
|
Sterpka A, Chen X. Neuronal and astrocytic primary cilia in the mature brain. Pharmacol Res 2018; 137:114-121. [PMID: 30291873 PMCID: PMC6410375 DOI: 10.1016/j.phrs.2018.10.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 12/17/2022]
Abstract
Primary cilia are tiny microtubule-based signaling devices that regulate a variety of physiological functions, including metabolism and cell division. Defects in primary cilia lead to a myriad of diseases in humans such as obesity and cancers. In the mature brain, both neurons and astrocytes contain a single primary cilium. Although neuronal primary cilia are not directly involved in synaptic communication, their pathophysiological impacts on obesity and mental disorders are well recognized. In contrast, research on astrocytic primary cilia lags far behind. Currently, little is known about their functions and molecular pathways in the mature brain. Unlike neurons, postnatal astrocytes retain the capacity of cell division and can become reactive and proliferate in response to various brain insults such as epilepsy, ischemia, traumatic brain injury, and neurodegenerative β-amyloid plaques. Since primary cilia derive from the mother centrioles, astrocyte proliferation must occur in coordination with the dismantling and ciliogenesis of astrocyte cilia. In this regard, the functions, signal pathways, and structural dynamics of neuronal and astrocytic primary cilia are fundamentally different. Here we discuss and compare the current understanding of neuronal and astrocytic primary cilia.
Collapse
Affiliation(s)
- Ashley Sterpka
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, United States
| | - Xuanmao Chen
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, United States.
| |
Collapse
|
145
|
Galati DF, Sullivan KD, Pham AT, Espinosa JM, Pearson CG. Trisomy 21 Represses Cilia Formation and Function. Dev Cell 2018; 46:641-650.e6. [PMID: 30100262 PMCID: PMC6557141 DOI: 10.1016/j.devcel.2018.07.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/15/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022]
Abstract
Trisomy 21 (T21) is the most prevalent human chromosomal disorder, causing a range of cardiovascular, musculoskeletal, and neurological abnormalities. However, the cellular processes disrupted by T21 are poorly understood. Consistent with the clinical overlap between T21 and ciliopathies, we discovered that T21 disrupts cilia formation and signaling. Cilia defects arise from increased expression of Pericentrin, a centrosome scaffold and trafficking protein encoded on chromosome 21. Elevated Pericentrin is necessary and sufficient for T21 cilia defects. Pericentrin accumulates at centrosomes and dramatically in the cytoplasm surrounding centrosomes. Centrosome Pericentrin recruits more γ-tubulin and enhances microtubules, whereas cytoplasmic Pericentrin assembles into large foci that do not efficiently traffic. Moreover, the Pericentrin-associated cilia assembly factor IFT20 and the ciliary signaling molecule Smoothened do not efficiently traffic to centrosomes and cilia. Thus, increased centrosome protein dosage produces ciliopathy-like outcomes in T21 cells by decreasing trafficking between the cytoplasm, centrosomes, and cilia.
Collapse
Affiliation(s)
- Domenico F Galati
- Department of Cell and Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Kelly D Sullivan
- Department of Pharmacology, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Andrew T Pham
- Department of Cell and Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joaquin M Espinosa
- Department of Pharmacology, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Chad G Pearson
- Department of Cell and Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
146
|
McKenzie CW, Preston CC, Finn R, Eyster KM, Faustino RS, Lee L. Strain-specific differences in brain gene expression in a hydrocephalic mouse model with motile cilia dysfunction. Sci Rep 2018; 8:13370. [PMID: 30190587 PMCID: PMC6127338 DOI: 10.1038/s41598-018-31743-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/22/2018] [Indexed: 01/10/2023] Open
Abstract
Congenital hydrocephalus results from cerebrospinal fluid accumulation in the ventricles of the brain and causes severe neurological damage, but the underlying causes are not well understood. It is associated with several syndromes, including primary ciliary dyskinesia (PCD), which is caused by dysfunction of motile cilia. We previously demonstrated that mouse models of PCD lacking ciliary proteins CFAP221, CFAP54 and SPEF2 all have hydrocephalus with a strain-dependent severity. While morphological defects are more severe on the C57BL/6J (B6) background than 129S6/SvEvTac (129), cerebrospinal fluid flow is perturbed on both backgrounds, suggesting that abnormal cilia-driven flow is not the only factor underlying the hydrocephalus phenotype. Here, we performed a microarray analysis on brains from wild type and nm1054 mice lacking CFAP221 on the B6 and 129 backgrounds. Expression differences were observed for a number of genes that cluster into distinct groups based on expression pattern and biological function, many of them implicated in cellular and biochemical processes essential for proper brain development. These include genes known to be functionally relevant to congenital hydrocephalus, as well as formation and function of both motile and sensory cilia. Identification of these genes provides important clues to mechanisms underlying congenital hydrocephalus severity.
Collapse
Affiliation(s)
- Casey W McKenzie
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA
| | - Claudia C Preston
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA
| | - Rozzy Finn
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA
| | - Kathleen M Eyster
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, 57069, USA
| | - Randolph S Faustino
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22nd Street, Sioux Falls, SD, 57105, USA
| | - Lance Lee
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th Street N., Sioux Falls, SD, 57104, USA. .,Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22nd Street, Sioux Falls, SD, 57105, USA.
| |
Collapse
|
147
|
Saternos HC, AbouAlaiwi WA. Signaling interplay between primary cilia and nitric oxide: A mini review. Nitric Oxide 2018; 80:108-112. [PMID: 30099097 DOI: 10.1016/j.niox.2018.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/01/2018] [Accepted: 08/06/2018] [Indexed: 01/12/2023]
Abstract
New discoveries into the functional role of primary cilia are on the rise. In little more than 20 years, research has shown the once vestigial organelle is a signaling powerhouse involved in a vast number of essential cellular processes. In the same decade that interest in primary cilia was burgeoning, nitric oxide won molecule of the year and a Nobel prize for its role as a near ubiquitous signaling molecule. Although primary cilia and nitric oxide are both involved in signaling, a direct relationship has not been investigated; however, after a quick review of the literature, parallels between their functions can be drawn. This review aims to suggest a possible interplay between primary cilia and nitric oxide signaling especially in the areas of vascular tissue homeostasis and cellular proliferation.
Collapse
Affiliation(s)
- Hannah C Saternos
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, USA
| | - Wissam A AbouAlaiwi
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology and Experimental Therapeutics, USA.
| |
Collapse
|
148
|
Park SM, Lim JS, Ramakrishina S, Kim SH, Kim WK, Lee J, Kang HC, Reiter JF, Kim DS, Kim HH, Lee JH. Brain Somatic Mutations in MTOR Disrupt Neuronal Ciliogenesis, Leading to Focal Cortical Dyslamination. Neuron 2018; 99:83-97.e7. [PMID: 29937275 DOI: 10.1016/j.neuron.2018.05.039] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/06/2018] [Accepted: 05/25/2018] [Indexed: 12/21/2022]
Abstract
Focal malformations of cortical development (FMCDs), including focal cortical dysplasia (FCD) and hemimegalencephaly (HME), are major etiologies of pediatric intractable epilepsies exhibiting cortical dyslamination. Brain somatic mutations in MTOR have recently been identified as a major genetic cause of FMCDs. However, the molecular mechanism by which these mutations lead to cortical dyslamination remains poorly understood. Here, using patient tissue, genome-edited cells, and mouse models with brain somatic mutations in MTOR, we discovered that disruption of neuronal ciliogenesis by the mutations underlies cortical dyslamination in FMCDs. We found that abnormal accumulation of OFD1 at centriolar satellites due to perturbed autophagy was responsible for the defective neuronal ciliogenesis. Additionally, we found that disrupted neuronal ciliogenesis accounted for cortical dyslamination in FMCDs by compromising Wnt signals essential for neuronal polarization. Altogether, this study describes a molecular mechanism by which brain somatic mutations in MTOR contribute to the pathogenesis of cortical dyslamination in FMCDs.
Collapse
Affiliation(s)
- Sang Min Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jae Seok Lim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Suresh Ramakrishina
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Woo Kyeong Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Junehawk Lee
- Biomedical HPC Technology Research Center, KISTI, Daejeon 34141, Republic of Korea
| | - Hoon-Chul Kang
- Division of Pediatric Neurology, Department of Pediatrics, Pediatric Epilepsy Clinics, Severance Children's Hospital, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dong Seok Kim
- Pediatric Neurosurgery, Severance Children's Hospital, Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology, Brain Korea 21 Plus Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon 34141, Republic of Korea.
| |
Collapse
|
149
|
Biallelic mutations in RTTN are associated with microcephaly, short stature and a wide range of brain malformations. Eur J Med Genet 2018; 61:733-737. [PMID: 29883675 DOI: 10.1016/j.ejmg.2018.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 05/07/2018] [Accepted: 06/02/2018] [Indexed: 12/12/2022]
Abstract
Biallelic mutations in the RTTN gene have been reported in association with microcephaly, short stature, developmental delay and malformations of cortical development. RTTN mutations have previously shown to link aberrant ciliary function with abnormal development and organization of the human cerebral cortex. We here report three individuals from two unrelated families with novel mutations in the RTTN gene. The phenotype consisted of microcephaly, short stature, pachygyria or polymicrogyria, colpocephaly, hypoplasia of the corpus callosum and superior vermis. These findings provide further confirmation of the phenotype related to pathogenic variants in RTTN.
Collapse
|
150
|
Ehrlich AT, Semache M, Bailly J, Wojcik S, Arefin TM, Colley C, Le Gouill C, Gross F, Lukasheva V, Hogue M, Darcq E, Harsan LA, Bouvier M, Kieffer BL. Mapping GPR88-Venus illuminates a novel role for GPR88 in sensory processing. Brain Struct Funct 2018; 223:1275-1296. [PMID: 29110094 PMCID: PMC5871604 DOI: 10.1007/s00429-017-1547-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/17/2017] [Indexed: 01/20/2023]
Abstract
GPR88 is an orphan G-protein coupled receptor originally characterized as a striatal-enriched transcript and is a potential target for neuropsychiatric disorders. At present, gene knockout studies in the mouse have essentially focused on striatal-related functions and a comprehensive knowledge of GPR88 protein distribution and function in the brain is still lacking. Here, we first created Gpr88-Venus knock-in mice expressing a functional fluorescent receptor to fine-map GPR88 localization in the brain. The receptor protein was detected in neuronal soma, fibers and primary cilia depending on the brain region, and remarkably, whole-brain mapping revealed a yet unreported layer-4 cortical lamination pattern specifically in sensory processing areas. The unique GPR88 barrel pattern in L4 of the somatosensory cortex appeared 3 days after birth and persisted into adulthood, suggesting a potential function for GPR88 in sensory integration. We next examined Gpr88 knockout mice for cortical structure and behavioral responses in sensory tasks. Magnetic resonance imaging of live mice revealed abnormally high fractional anisotropy, predominant in somatosensory cortex and caudate putamen, indicating significant microstructural alterations in these GPR88-enriched areas. Further, behavioral analysis showed delayed responses in somatosensory-, visual- and olfactory-dependent tasks, demonstrating a role for GPR88 in the integration rather than perception of sensory stimuli. In conclusion, our data show for the first time a prominent role for GPR88 in multisensory processing. Because sensory integration is disrupted in many psychiatric diseases, our study definitely positions GPR88 as a target to treat mental disorders perhaps via activity on cortical sensory networks.
Collapse
Affiliation(s)
- Aliza T Ehrlich
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Perry Pavilion Room E-3317.1, 6875 boulevard LaSalle, Montreal, QC, H4H 1R3, Canada
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, France
| | - Meriem Semache
- Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Julie Bailly
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Perry Pavilion Room E-3317.1, 6875 boulevard LaSalle, Montreal, QC, H4H 1R3, Canada
| | - Stefan Wojcik
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Perry Pavilion Room E-3317.1, 6875 boulevard LaSalle, Montreal, QC, H4H 1R3, Canada
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Tanzil M Arefin
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Bernard and Irene Schwartz Center for Biomedical Imaging, New York University School of Medicine, New York, USA
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, France
| | - Christine Colley
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Perry Pavilion Room E-3317.1, 6875 boulevard LaSalle, Montreal, QC, H4H 1R3, Canada
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Christian Le Gouill
- Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Florence Gross
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Perry Pavilion Room E-3317.1, 6875 boulevard LaSalle, Montreal, QC, H4H 1R3, Canada
- Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Viktoriya Lukasheva
- Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Mireille Hogue
- Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Emmanuel Darcq
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Perry Pavilion Room E-3317.1, 6875 boulevard LaSalle, Montreal, QC, H4H 1R3, Canada
| | - Laura-Adela Harsan
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Engineering Science, Computer Science and Imaging Laboratory (ICube), Integrative Multimodal Imaging in Healthcare, University of Strasbourg, CNRS, Strasbourg, France
- Department of Biophysics and Nuclear Medicine, Faculty of Medicine, University Hospital Strasbourg, Strasbourg, France
| | - Michel Bouvier
- Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Brigitte L Kieffer
- Department of Psychiatry, McGill University, Douglas Hospital Research Center, Perry Pavilion Room E-3317.1, 6875 boulevard LaSalle, Montreal, QC, H4H 1R3, Canada.
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, France.
| |
Collapse
|