1
|
Wang K, Tang Z, Yang Y, Guo Y, Liu Z, Su Z, Li X, Xiao G. Zebrafish as a Model Organism for Congenital Hydrocephalus: Characteristics and Insights. Zebrafish 2024; 21:361-384. [PMID: 39510565 DOI: 10.1089/zeb.2024.0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Hydrocephalus is a cerebrospinal fluid-related disease that usually manifests as abnormal dilation of the ventricles, with a triad of clinical findings including walking difficulty, reduced attention span, and urinary frequency or incontinence. The onset of congenital hydrocephalus is closely related to mutations in genes that regulate brain development. Currently, our understanding of the mechanisms of congenital hydrocephalus remains limited, and the prognosis of existing treatments is unsatisfactory. Additionally, there are no suitable or dedicated model organisms for congenital hydrocephalus. Therefore, it is significant to determine the mechanism and develop special animal models of congenital hydrocephalus. Recently, zebrafish have emerged as a popular model organism in many fields, including developmental biology, genetics, and toxicology. Its genome shares high similarity with that of humans, and it has fast and low-cost reproduction. These advantages make it suitable for studying the pathogenesis and therapeutic approaches for various diseases, specifically congenital diseases. This study explored the possibility of using zebrafish as a model organism for congenital hydrocephalus. This review describes the characteristics of zebrafish and discusses specific congenital hydrocephalus models. The advantages and limitations of using zebrafish for hydrocephalus research are highlighted, and insights for further model development are provided.
Collapse
Affiliation(s)
- Kaiyue Wang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhi Tang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yating Guo
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, United Kingdom
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| |
Collapse
|
2
|
Nguyen TK, Baker S, Rodriguez JM, Arceri L, Wingert RA. Using Zebrafish to Study Multiciliated Cell Development and Disease States. Cells 2024; 13:1749. [PMID: 39513856 PMCID: PMC11545745 DOI: 10.3390/cells13211749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Multiciliated cells (MCCs) serve many important functions, including fluid propulsion and chemo- and mechanosensing. Diseases ranging from rare conditions to the recent COVID-19 global health pandemic have been linked to MCC defects. In recent years, the zebrafish has emerged as a model to investigate the biology of MCCs. Here, we review the major events in MCC formation including centriole biogenesis and basal body docking. Then, we discuss studies on the role of MCCs in diseases of the brain, respiratory, kidney and reproductive systems, as well as recent findings about the link between MCCs and SARS-CoV-2. Next, we explore why the zebrafish is a useful model to study MCCs and provide a comprehensive overview of previous studies of genetic components essential for MCC development and motility across three major tissues in the zebrafish: the pronephros, brain ependymal cells and nasal placode. Taken together, here we provide a cohesive summary of MCC research using the zebrafish and its future potential for expanding our understanding of MCC-related disease states.
Collapse
Affiliation(s)
- Thanh Khoa Nguyen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (S.B.); (J.-M.R.); (L.A.)
| | | | | | | | - Rebecca A. Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (S.B.); (J.-M.R.); (L.A.)
| |
Collapse
|
3
|
Liu XY, Song X, Czosnyka M, Robba C, Czosnyka Z, Summers JL, Yu HJ, Gao GY, Smielewski P, Guo F, Pang MJ, Ming D. Congenital hydrocephalus: a review of recent advances in genetic etiology and molecular mechanisms. Mil Med Res 2024; 11:54. [PMID: 39135208 PMCID: PMC11318184 DOI: 10.1186/s40779-024-00560-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/28/2024] [Indexed: 08/15/2024] Open
Abstract
The global prevalence rate for congenital hydrocephalus (CH) is approximately one out of every five hundred births with multifaceted predisposing factors at play. Genetic influences stand as a major contributor to CH pathogenesis, and epidemiological evidence suggests their involvement in up to 40% of all cases observed globally. Knowledge about an individual's genetic susceptibility can significantly improve prognostic precision while aiding clinical decision-making processes. However, the precise genetic etiology has only been pinpointed in fewer than 5% of human instances. More occurrences of CH cases are required for comprehensive gene sequencing aimed at uncovering additional potential genetic loci. A deeper comprehension of its underlying genetics may offer invaluable insights into the molecular and cellular basis of this brain disorder. This review provides a summary of pertinent genes identified through gene sequencing technologies in humans, in addition to the 4 genes currently associated with CH (two X-linked genes L1CAM and AP1S2, two autosomal recessive MPDZ and CCDC88C). Others predominantly participate in aqueduct abnormalities, ciliary movement, and nervous system development. The prospective CH-related genes revealed through animal model gene-editing techniques are further outlined, focusing mainly on 4 pathways, namely cilia synthesis and movement, ion channels and transportation, Reissner's fiber (RF) synthesis, cell apoptosis, and neurogenesis. Notably, the proper functioning of motile cilia provides significant impulsion for cerebrospinal fluid (CSF) circulation within the brain ventricles while mutations in cilia-related genes constitute a primary cause underlying this condition. So far, only a limited number of CH-associated genes have been identified in humans. The integration of genotype and phenotype for disease diagnosis represents a new trend in the medical field. Animal models provide insights into the pathogenesis of CH and contribute to our understanding of its association with related complications, such as renal cysts, scoliosis, and cardiomyopathy, as these genes may also play a role in the development of these diseases. Genes discovered in animals present potential targets for new treatments but require further validation through future human studies.
Collapse
Affiliation(s)
- Xiu-Yun Liu
- Medical School, Tianjin University, Tianjin, 300072, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, 300072, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300380, China
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Xin Song
- Medical School, Tianjin University, Tianjin, 300072, China
| | - Marek Czosnyka
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Chiara Robba
- San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, 16132, Genoa, Italy
| | - Zofia Czosnyka
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Jennifer Lee Summers
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Hui-Jie Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Guo-Yi Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Peter Smielewski
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Fang Guo
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Mei-Jun Pang
- Medical School, Tianjin University, Tianjin, 300072, China.
| | - Dong Ming
- Medical School, Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, 300072, China.
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300380, China.
| |
Collapse
|
4
|
Penny GM, Dutcher SK. Gene dosage of independent dynein arm motor preassembly factors influences cilia assembly in Chlamydomonas reinhardtii. PLoS Genet 2024; 20:e1011038. [PMID: 38498551 PMCID: PMC11020789 DOI: 10.1371/journal.pgen.1011038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/16/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Motile cilia assembly utilizes over 800 structural and cytoplasmic proteins. Variants in approximately 58 genes cause primary ciliary dyskinesia (PCD) in humans, including the dynein arm (pre)assembly factor (DNAAF) gene DNAAF4. In humans, outer dynein arms (ODAs) and inner dynein arms (IDAs) fail to assemble motile cilia when DNAAF4 function is disrupted. In Chlamydomonas reinhardtii, a ciliated unicellular alga, the DNAAF4 ortholog is called PF23. The pf23-1 mutant assembles short cilia and lacks IDAs, but partially retains ODAs. The cilia of a new null allele (pf23-4) completely lack ODAs and IDAs and are even shorter than cilia from pf23-1. In addition, PF23 plays a role in the cytoplasmic modification of IC138, a protein of the two-headed IDA (I1/f). As most PCD variants in humans are recessive, we sought to test if heterozygosity at two genes affects ciliary function using a second-site non-complementation (SSNC) screening approach. We asked if phenotypes were observed in diploids with pairwise heterozygous combinations of 21 well-characterized ciliary mutant Chlamydomonas strains. Vegetative cultures of single and double heterozygous diploid cells did not show SSNC for motility phenotypes. When protein synthesis is inhibited, wild-type Chlamydomonas cells utilize the pool of cytoplasmic proteins to assemble half-length cilia. In this sensitized assay, 8 double heterozygous diploids with pf23 and other DNAAF mutations show SSNC; they assemble shorter cilia than wild-type. In contrast, double heterozygosity of the other 203 strains showed no effect on ciliary assembly. Immunoblots of diploids heterozygous for pf23 and wdr92 or oda8 show that PF23 is reduced by half in these strains, and that PF23 dosage affects phenotype severity. Reductions in PF23 and another DNAAF in diploids affect the ability to assemble ODAs and IDAs and impedes ciliary assembly. Thus, dosage of multiple DNAAFs is an important factor in cilia assembly and regeneration.
Collapse
Affiliation(s)
- Gervette M. Penny
- Department of Genetics, Washington University in Saint Louis, Saint Louis,Missouri, United States of America
| | - Susan K. Dutcher
- Department of Genetics, Washington University in Saint Louis, Saint Louis,Missouri, United States of America
| |
Collapse
|
5
|
Calì F, Di Blasi FD, Avola E, Vinci M, Musumeci A, Gloria A, Greco D, Raciti DR, Zagami A, Rizzo B, Città S, Federico C, Vetri L, Saccone S, Buono S. Specific Learning Disorders: Variation Analysis of 15 Candidate Genes in 9 Multiplex Families. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1503. [PMID: 37629793 PMCID: PMC10456226 DOI: 10.3390/medicina59081503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/13/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
Background and Objectives: Specific Learning Disorder (SLD) is a complex neurobiological disorder characterized by a persistent difficult in reading (dyslexia), written expression (dysgraphia), and mathematics (dyscalculia). The hereditary and genetic component is one of the underlying causes of SLD, but the relationship between genes and the environment should be considered. Several genetic studies were performed in different populations to identify causative genes. Materials and Methods: Here, we show the analysis of 9 multiplex families with at least 2 individuals diagnosed with SLD per family, with a total of 37 persons, 21 of whom are young subjects with SLD, by means of Next-Generation Sequencing (NGS) to identify possible causative mutations in a panel of 15 candidate genes: CCPG1, CYP19A1, DCDC2, DGKI, DIP2A, DYM, GCFC2, KIAA0319, MC5R, MRPL19, NEDD4L, PCNT, PRMT2, ROBO1, and S100B. Results: We detected, in eight families out nine, SNP variants in the DGKI, DIP2A, KIAA0319, and PCNT genes, even if in silico analysis did not show any causative effect on this behavioral condition. In all cases, the mutation was transmitted by one of the two parents, thus excluding the case of de novo mutation. Moreover, the parent carrying the allelic variant transmitted to the children, in six out of seven families, reports language difficulties. Conclusions: Although the present results cannot be considered conclusive due to the limited sample size, the identification of genetic variants in the above genes can provide input for further research on the same, as well as on other genes/mutations, to better understand the genetic basis of this disorder, and from this perspective, to better understand also the neuropsychological and social aspects connected to this disorder, which affects an increasing number of young people.
Collapse
Affiliation(s)
- Francesco Calì
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | | | - Emanuela Avola
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Mirella Vinci
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Antonino Musumeci
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Angelo Gloria
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Donatella Greco
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Daniela Rita Raciti
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Alessandro Zagami
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Biagio Rizzo
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Santina Città
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Concetta Federico
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Luigi Vetri
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| | - Salvatore Saccone
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Serafino Buono
- Oasi Research Institute—IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy; (F.C.); (F.D.D.B.); (S.B.)
| |
Collapse
|
6
|
Bieder A, Chandrasekar G, Wason A, Erkelenz S, Gopalakrishnan J, Kere J, Tapia-Páez I. Genetic and protein interaction studies between the ciliary dyslexia candidate genes DYX1C1 and DCDC2. BMC Mol Cell Biol 2023; 24:20. [PMID: 37237337 DOI: 10.1186/s12860-023-00483-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND DYX1C1 (DNAAF4) and DCDC2 are two of the most replicated dyslexia candidate genes in genetic studies. They both have demonstrated roles in neuronal migration, in cilia growth and function and they both are cytoskeletal interactors. In addition, they both have been characterized as ciliopathy genes. However, their exact molecular functions are still incompletely described. Based on these known roles, we asked whether DYX1C1 and DCDC2 interact on the genetic and the protein level. RESULTS Here, we report the physical protein-protein interaction of DYX1C1 and DCDC2 as well as their respective interactions with the centrosomal protein CPAP (CENPJ) on exogenous and endogenous levels in different cell models including brain organoids. In addition, we show a synergistic genetic interaction between dyx1c1 and dcdc2b in zebrafish exacerbating the ciliary phenotype. Finally, we show a mutual effect on transcriptional regulation among DYX1C1 and DCDC2 in a cellular model. CONCLUSIONS In summary, we describe the physical and functional interaction between the two genes DYX1C1 and DCDC2. These results contribute to the growing understanding of the molecular roles of DYX1C1 and DCDC2 and set the stage for future functional studies.
Collapse
Affiliation(s)
- Andrea Bieder
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | - Arpit Wason
- Center for Molecular Medicine, Institute for Biochemistry I of the University of Cologne, Cologne, Germany
| | - Steffen Erkelenz
- Institute of Human Genetics, Universitätsklinikum, Heinrich Heine University, Düsseldorf, Germany
| | - Jay Gopalakrishnan
- Institute of Human Genetics, Universitätsklinikum, Heinrich Heine University, Düsseldorf, Germany
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Molecular Neurology Research Program, University of Helsinki, Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Isabel Tapia-Páez
- Department of Medicine, Solna, Karolinska Institutet, Solnavägen 30, SE-171 76, Solna, Sweden.
| |
Collapse
|
7
|
Galaburda AM. Animal models of developmental dyslexia. Front Neurosci 2022; 16:981801. [PMID: 36452335 PMCID: PMC9702821 DOI: 10.3389/fnins.2022.981801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/28/2022] [Indexed: 09/10/2024] Open
Abstract
As some critics have stated, the term "developmental dyslexia" refers to a strictly human disorder, relating to a strictly human capacity - reading - so it cannot be modeled in experimental animals, much less so in lowly rodents. However, two endophenotypes associated with developmental dyslexia are eminently suitable for animal modeling: Cerebral Lateralization, as illustrated by the association between dyslexia and non-righthandedness, and Cerebrocortical Dysfunction, as illustrated by the described abnormal structural anatomy and/or physiology and functional imaging of the dyslexic cerebral cortex. This paper will provide a brief review of these two endophenotypes in human beings with developmental dyslexia and will describe the animal work done in my laboratory and that of others to try to shed light on the etiology of and neural mechanisms underlying developmental dyslexia. Some thought will also be given to future directions of the research.
Collapse
Affiliation(s)
- Albert M. Galaburda
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
Ji W, Tang Z, Chen Y, Wang C, Tan C, Liao J, Tong L, Xiao G. Ependymal Cilia: Physiology and Role in Hydrocephalus. Front Mol Neurosci 2022; 15:927479. [PMID: 35903173 PMCID: PMC9315228 DOI: 10.3389/fnmol.2022.927479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/20/2022] [Indexed: 01/10/2023] Open
Abstract
Cerebrospinal fluid (CSF), a colorless liquid that generally circulates from the lateral ventricles to the third and fourth ventricles, provides essential nutrients for brain homeostasis and growth factors during development. As evidenced by an increasing corpus of research, CSF serves a range of important functions. While it is considered that decreased CSF flow is associated to the development of hydrocephalus, it has recently been postulated that motile cilia, which line the apical surfaces of ependymal cells (ECs), play a role in stimulating CSF circulation by cilia beating. Ependymal cilia protrude from ECs, and their synchronous pulsing transports CSF from the lateral ventricle to the third and fourth ventricles, and then to the subarachnoid cavity for absorption. As a result, we postulated that malfunctioning ependymal cilia could disrupt normal CSF flow, raising the risk of hydrocephalus. This review aims to demonstrate the physiological functions of ependymal cilia, as well as how cilia immobility or disorientation causes problems. We also conclude conceivable ways of treatment of hydrocephalus currently for clinical application and provide theoretical support for regimen improvements by investigating the relationship between ependymal cilia and hydrocephalus development.
Collapse
Affiliation(s)
- Weiye Ji
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Tang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yibing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chuansen Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Changwu Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Tong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Gelei Xiao,
| |
Collapse
|
9
|
KIAA0319 influences cilia length, cell migration and mechanical cell-substrate interaction. Sci Rep 2022; 12:722. [PMID: 35031635 PMCID: PMC8760330 DOI: 10.1038/s41598-021-04539-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 12/17/2021] [Indexed: 01/11/2023] Open
Abstract
Following its association with dyslexia in multiple genetic studies, the KIAA0319 gene has been extensively investigated in different animal models but its function in neurodevelopment remains poorly understood. We developed the first human cellular knockout model for KIAA0319 in RPE1 retinal pigment epithelia cells via CRISPR-Cas9n to investigate its role in processes suggested but not confirmed in previous studies, including cilia formation and cell migration. We observed in the KIAA0319 knockout increased cilia length and accelerated cell migration. Using Elastic Resonator Interference Stress Microscopy (ERISM), we detected an increase in cellular force for the knockout cells that was restored by a rescue experiment. Combining ERISM and immunostaining we show that RPE1 cells exert highly dynamic, piconewton vertical pushing forces through actin-rich protrusions that are surrounded by vinculin-rich pulling sites. This protein arrangement and force pattern has previously been associated to podosomes in other cells. KIAA0319 depletion reduces the fraction of cells forming these actin-rich protrusions. Our results suggest an involvement of KIAA0319 in cilia biology and cell-substrate force regulation.
Collapse
|
10
|
Johnson JL. Mutations in Hsp90 Cochaperones Result in a Wide Variety of Human Disorders. Front Mol Biosci 2021; 8:787260. [PMID: 34957217 PMCID: PMC8694271 DOI: 10.3389/fmolb.2021.787260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 12/19/2022] Open
Abstract
The Hsp90 molecular chaperone, along with a set of approximately 50 cochaperones, mediates the folding and activation of hundreds of cellular proteins in an ATP-dependent cycle. Cochaperones differ in how they interact with Hsp90 and their ability to modulate ATPase activity of Hsp90. Cochaperones often compete for the same binding site on Hsp90, and changes in levels of cochaperone expression that occur during neurodegeneration, cancer, or aging may result in altered Hsp90-cochaperone complexes and client activity. This review summarizes information about loss-of-function mutations of individual cochaperones and discusses the overall association of cochaperone alterations with a broad range of diseases. Cochaperone mutations result in ciliary or muscle defects, neurological development or degeneration disorders, and other disorders. In many cases, diseases were linked to defects in established cochaperone-client interactions. A better understanding of the functional consequences of defective cochaperones will provide new insights into their functions and may lead to specialized approaches to modulate Hsp90 functions and treat some of these human disorders.
Collapse
Affiliation(s)
- Jill L Johnson
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, ID, United States
| |
Collapse
|
11
|
Animal models of developmental dyslexia: Where we are and what we are missing. Neurosci Biobehav Rev 2021; 131:1180-1197. [PMID: 34699847 DOI: 10.1016/j.neubiorev.2021.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022]
Abstract
Developmental dyslexia (DD) is a complex neurodevelopmental disorder and the most common learning disability among both school-aged children and across languages. Recently, sensory and cognitive mechanisms have been reported to be potential endophenotypes (EPs) for DD, and nine DD-candidate genes have been identified. Animal models have been used to investigate the etiopathological pathways that underlie the development of complex traits, as they enable the effects of genetic and/or environmental manipulations to be evaluated. Animal research designs have also been linked to cutting-edge clinical research questions by capitalizing on the use of EPs. For the present scoping review, we reviewed previous studies of murine models investigating the effects of DD-candidate genes. Moreover, we highlighted the use of animal models as an innovative way to unravel new insights behind the pathophysiology of reading (dis)ability and to assess cutting-edge preclinical models.
Collapse
|
12
|
Kumar V, Umair Z, Kumar S, Goutam RS, Park S, Kim J. The regulatory roles of motile cilia in CSF circulation and hydrocephalus. Fluids Barriers CNS 2021; 18:31. [PMID: 34233705 PMCID: PMC8261947 DOI: 10.1186/s12987-021-00265-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background Cerebrospinal fluid (CSF) is an ultra-filtrated colorless brain fluid that circulates within brain spaces like the ventricular cavities, subarachnoid space, and the spine. Its continuous flow serves many primary functions, including nourishment, brain protection, and waste removal. Main body The abnormal accumulation of CSF in brain cavities triggers severe hydrocephalus. Accumulating evidence had indicated that synchronized beats of motile cilia (cilia from multiciliated cells or the ependymal lining in brain ventricles) provide forceful pressure to generate and restrain CSF flow and maintain overall CSF circulation within brain spaces. In humans, the disorders caused by defective primary and/or motile cilia are generally referred to as ciliopathies. The key role of CSF circulation in brain development and its functioning has not been fully elucidated. Conclusions In this review, we briefly discuss the underlying role of motile cilia in CSF circulation and hydrocephalus. We have reviewed cilia and ciliated cells in the brain and the existing evidence for the regulatory role of functional cilia in CSF circulation in the brain. We further discuss the findings obtained for defective cilia and their potential involvement in hydrocephalus. Furthermore, this review will reinforce the idea of motile cilia as master regulators of CSF movements, brain development, and neuronal diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Shiv Kumar
- School of Psychology and Neuroscience, University of St. Andrews, St. Mary's Quad, South Street. St. Andrews, Fife, KY16 9JP, UK
| | - Ravi Shankar Goutam
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Soochul Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
13
|
The Polygenic Nature and Complex Genetic Architecture of Specific Learning Disorder. Brain Sci 2021; 11:brainsci11050631. [PMID: 34068951 PMCID: PMC8156942 DOI: 10.3390/brainsci11050631] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/16/2022] Open
Abstract
Specific Learning Disorder (SLD) is a multifactorial, neurodevelopmental disorder which may involve persistent difficulties in reading (dyslexia), written expression and/or mathematics. Dyslexia is characterized by difficulties with speed and accuracy of word reading, deficient decoding abilities, and poor spelling. Several studies from different, but complementary, scientific disciplines have investigated possible causal/risk factors for SLD. Biological, neurological, hereditary, cognitive, linguistic-phonological, developmental and environmental factors have been incriminated. Despite worldwide agreement that SLD is highly heritable, its exact biological basis remains elusive. We herein present: (a) an update of studies that have shaped our current knowledge on the disorder’s genetic architecture; (b) a discussion on whether this genetic architecture is ‘unique’ to SLD or, alternatively, whether there is an underlying common genetic background with other neurodevelopmental disorders; and, (c) a brief discussion on whether we are at a position of generating meaningful correlations between genetic findings and anatomical data from neuroimaging studies or specific molecular/cellular pathways. We conclude with open research questions that could drive future research directions.
Collapse
|
14
|
Falkenberg LG, Beckman SA, Ravisankar P, Dohn TE, Waxman JS. Ccdc103 promotes myeloid cell proliferation and migration independent of motile cilia. Dis Model Mech 2021; 14:dmm048439. [PMID: 34028558 PMCID: PMC8214733 DOI: 10.1242/dmm.048439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/31/2021] [Indexed: 01/01/2023] Open
Abstract
The pathology of primary ciliary dyskinesia (PCD) is predominantly attributed to impairment of motile cilia. However, PCD patients also have perplexing functional defects in myeloid cells, which lack motile cilia. Here, we show that coiled-coil domain-containing protein 103 (CCDC103), one of the genes that, when mutated, is known to cause PCD, is required for the proliferation and directed migration of myeloid cells. CCDC103 is expressed in human myeloid cells, where it colocalizes with cytoplasmic microtubules. Zebrafish ccdc103/schmalhans (smh) mutants have macrophages and neutrophils with reduced proliferation, abnormally rounded cell morphology and an inability to migrate efficiently to the site of sterile wounds, all of which are consistent with a loss of cytoplasmic microtubule stability. Furthermore, we demonstrate that direct interactions between CCDC103 and sperm associated antigen 6 (SPAG6), which also promotes microtubule stability, are abrogated by CCDC103 mutations from PCD patients, and that spag6 zebrafish mutants recapitulate the myeloid defects observed in smh mutants. In summary, we have illuminated a mechanism, independent of motile cilia, to explain functional defects in myeloid cells from PCD patients. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lauren G. Falkenberg
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati OH 45267, USA
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sarah A. Beckman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Padmapriyadarshini Ravisankar
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tracy E. Dohn
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati OH 45267, USA
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
15
|
McKnight I, Hart C, Park IH, Shim JW. Genes causing congenital hydrocephalus: Their chromosomal characteristics of telomere proximity and DNA compositions. Exp Neurol 2021; 335:113523. [PMID: 33157092 PMCID: PMC7750280 DOI: 10.1016/j.expneurol.2020.113523] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/10/2020] [Accepted: 10/30/2020] [Indexed: 01/06/2023]
Abstract
Congenital hydrocephalus (CH) is caused by genetic mutations, but whether factors impacting human genetic mutations are disease-specific remains elusive. Given two factors associated with high mutation rates, we reviewed how many disease-susceptible genes match with (i) proximity to telomeres or (ii) high adenine and thymine (A + T) content in human CH as compared to other disorders of the central nervous system (CNS). We extracted genomic information using a genome data viewer. Importantly, 98 of 108 genes causing CH satisfied (i) or (ii), resulting in >90% matching rate. However, such a high accordance no longer sustained as we checked two factors in Alzheimer's disease (AD) and/or familial Parkinson's disease (fPD), resulting in 84% and 59% matching, respectively. A disease-specific matching of telomere proximity or high A + T content predicts causative genes of CH much better than neurodegenerative diseases and other CNS conditions, likely due to sufficient number of known causative genes (n = 108) and precise determination and classification of the genotype and phenotype. Our analysis suggests a need for identifying genetic basis of both factors before human clinical studies, to prioritize putative genes found in preclinical models into the likely (meeting at least one) and more likely candidate (meeting both), which predisposes human genes to mutations.
Collapse
Affiliation(s)
- Ian McKnight
- Department of Biomedical Engineering, Marshall University, Huntington, WV 25755, USA
| | - Christoph Hart
- Department of Biomedical Engineering, Marshall University, Huntington, WV 25755, USA
| | - In-Hyun Park
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Joon W Shim
- Department of Biomedical Engineering, Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
16
|
Bieder A, Yoshihara M, Katayama S, Krjutškov K, Falk A, Kere J, Tapia-Páez I. Dyslexia Candidate Gene and Ciliary Gene Expression Dynamics During Human Neuronal Differentiation. Mol Neurobiol 2020; 57:2944-2958. [PMID: 32445086 PMCID: PMC7320047 DOI: 10.1007/s12035-020-01905-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/19/2020] [Indexed: 11/30/2022]
Abstract
Developmental dyslexia (DD) is a neurodevelopmental condition with complex genetic mechanisms. A number of candidate genes have been identified, some of which are linked to neuronal development and migration and to ciliary functions. However, expression and regulation of these genes in human brain development and neuronal differentiation remain uncharted. Here, we used human long-term self-renewing neuroepithelial stem (lt-NES, here termed NES) cells derived from human induced pluripotent stem cells to study neuronal differentiation in vitro. We characterized gene expression changes during differentiation by using RNA sequencing and validated dynamics for selected genes by qRT-PCR. Interestingly, we found that genes related to cilia were significantly enriched among upregulated genes during differentiation, including genes linked to ciliopathies with neurodevelopmental phenotypes. We confirmed the presence of primary cilia throughout neuronal differentiation. Focusing on dyslexia candidate genes, 33 out of 50 DD candidate genes were detected in NES cells by RNA sequencing, and seven candidate genes were upregulated during differentiation to neurons, including DYX1C1 (DNAAF4), a highly replicated DD candidate gene. Our results suggest a role of ciliary genes in differentiating neuronal cells and show that NES cells provide a relevant human neuronal model to study ciliary and DD candidate genes.
Collapse
Affiliation(s)
- Andrea Bieder
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 9, 141 57, Huddinge, Sweden.
| | - Masahito Yoshihara
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 9, 141 57, Huddinge, Sweden
| | - Shintaro Katayama
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 9, 141 57, Huddinge, Sweden
| | - Kaarel Krjutškov
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 9, 141 57, Huddinge, Sweden.,Competence Centre on Health Technologies, Tartu, Estonia.,Research Program of Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 9, 141 57, Huddinge, Sweden. .,Research Program of Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland. .,Folkhälsan Institute of Genetics, Helsinki, Finland. .,School of Basic and Medical Biosciences, King's College London, London, UK.
| | | |
Collapse
|
17
|
Bieder A, Einarsdottir E, Matsson H, Nilsson HE, Eisfeldt J, Dragomir A, Paucar M, Granberg T, Li TQ, Lindstrand A, Kere J, Tapia-Páez I. Rare variants in dynein heavy chain genes in two individuals with situs inversus and developmental dyslexia: a case report. BMC MEDICAL GENETICS 2020; 21:87. [PMID: 32357925 PMCID: PMC7193346 DOI: 10.1186/s12881-020-01020-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/05/2020] [Indexed: 02/08/2023]
Abstract
Background Developmental dyslexia (DD) is a neurodevelopmental learning disorder with high heritability. A number of candidate susceptibility genes have been identified, some of which are linked to the function of the cilium, an organelle regulating left-right asymmetry development in the embryo. Furthermore, it has been suggested that disrupted left-right asymmetry of the brain may play a role in neurodevelopmental disorders such as DD. However, it is unknown whether there is a common genetic cause to DD and laterality defects or ciliopathies. Case presentation Here, we studied two individuals with co-occurring situs inversus (SI) and DD using whole genome sequencing to identify genetic variants of importance for DD and SI. Individual 1 had primary ciliary dyskinesia (PCD), a rare, autosomal recessive disorder with oto-sino-pulmonary phenotype and SI. We identified two rare nonsynonymous variants in the dynein axonemal heavy chain 5 gene (DNAH5): a previously reported variant c.7502G > C; p.(R2501P), and a novel variant c.12043 T > G; p.(Y4015D). Both variants are predicted to be damaging. Ultrastructural analysis of the cilia revealed a lack of outer dynein arms and normal inner dynein arms. MRI of the brain revealed no significant abnormalities. Individual 2 had non-syndromic SI and DD. In individual 2, one rare variant (c.9110A > G;p.(H3037R)) in the dynein axonemal heavy chain 11 gene (DNAH11), coding for another component of the outer dynein arm, was identified. Conclusions We identified the likely genetic cause of SI and PCD in one individual, and a possibly significant heterozygosity in the other, both involving dynein genes. Given the present evidence, it is unclear if the identified variants also predispose to DD and further studies into the association between laterality, ciliopathies and DD are needed.
Collapse
Affiliation(s)
- Andrea Bieder
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 141 83, Huddinge, Sweden.
| | - Elisabet Einarsdottir
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 141 83, Huddinge, Sweden.,Stem Cells and Metabolism Research Program (STEMM), University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of Technology, Solna, Sweden
| | - Hans Matsson
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Harriet E Nilsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 141 83, Huddinge, Sweden.,Department of Biomedical Engineering and Health Systems, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Huddinge, Sweden
| | - Jesper Eisfeldt
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Karolinska Institutet Science Park, Solna, Sweden
| | - Anca Dragomir
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Martin Paucar
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Tie-Qiang Li
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Lindstrand
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 141 83, Huddinge, Sweden.,Stem Cells and Metabolism Research Program (STEMM), University of Helsinki, Helsinki, Finland.,School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Isabel Tapia-Páez
- Department of Medicine, Solna, Karolinska Institutet, Solnavägen 30, 171 76 Solna, Stockholm, Sweden.
| |
Collapse
|
18
|
Osinka A, Poprzeczko M, Zielinska MM, Fabczak H, Joachimiak E, Wloga D. Ciliary Proteins: Filling the Gaps. Recent Advances in Deciphering the Protein Composition of Motile Ciliary Complexes. Cells 2019; 8:cells8070730. [PMID: 31319499 PMCID: PMC6678824 DOI: 10.3390/cells8070730] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/15/2022] Open
Abstract
Cilia are highly evolutionarily conserved, microtubule-based cell protrusions present in eukaryotic organisms from protists to humans, with the exception of fungi and higher plants. Cilia can be broadly divided into non-motile sensory cilia, called primary cilia, and motile cilia, which are locomotory organelles. The skeleton (axoneme) of primary cilia is formed by nine outer doublet microtubules distributed on the cilium circumference. In contrast, the skeleton of motile cilia is more complex: in addition to outer doublets, it is composed of two central microtubules and several diverse multi-protein complexes that are distributed periodically along both types of microtubules. For many years, researchers have endeavored to fully characterize the protein composition of ciliary macro-complexes and the molecular basis of signal transduction between these complexes. Genetic and biochemical analyses have suggested that several hundreds of proteins could be involved in the assembly and function of motile cilia. Within the last several years, the combined efforts of researchers using cryo-electron tomography, genetic and biochemical approaches, and diverse model organisms have significantly advanced our knowledge of the ciliary structure and protein composition. Here, we summarize the recent progress in the identification of the subunits of ciliary complexes, their precise intraciliary localization determined by cryo-electron tomography data, and the role of newly identified proteins in cilia.
Collapse
Affiliation(s)
- Anna Osinka
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Martyna Poprzeczko
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Magdalena M Zielinska
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| | - Dorota Wloga
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|
19
|
Full-Length Transcriptome Sequencing and the Discovery of New Transcripts in the Unfertilized Eggs of Zebrafish ( Danio rerio). G3-GENES GENOMES GENETICS 2019; 9:1831-1838. [PMID: 30872328 PMCID: PMC6553537 DOI: 10.1534/g3.119.200997] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Understanding early gene expression in zebrafish embryos is a prerequisite for developmental biology research. In this study, 1,629,447 polymerase reads were obtained from the unfertilized eggs of zebrafish via full-length transcriptome sequencing using the PacBio RS II platform first. Then, 102,920 unique isoforms were obtained by correction, clustering and comparison with the zebrafish genome. 12,782 genes in the genome were captured, accounting for 39.71% of the all annotated genes. Approximately 62.27% of the 12,782 genes have been alternatively spliced. GO and KEGG annotations revealed that the unfertilized eggs primarily stored genes that participate in RNA processing and nuclear protein complex composition. According to this PacBio data that aligned with the genome, 3,970 fusion genes, 819 ncRNAs, and 84 new transcripts were predicted. Illumina RNA-seq and RT-qPCR detection found that the expression of two new transcripts, PB.5289.1 and PB.10209.1, were significantly up-regulated at the 2-cell stage and down-regulated rapidly thereafter, suggesting their involvement in minor ZGA during early embryonic development. This study indicated that the unfertilized eggs of zebrafish may have retained genes directly related to cell division and development to initiate the subsequent development in a limited space and time. On the other hand, NTRs or new transcriptome regions in the genome were discovered, which provided new clues regarding ZGA of MZT during early embryonic development in fish.
Collapse
|
20
|
Gostic M, Martinelli A, Tucker C, Yang Z, Gasparoli F, Ewart JY, Dholakia K, Sillar KT, Tello JA, Paracchini S. The dyslexia susceptibility KIAA0319 gene shows a specific expression pattern during zebrafish development supporting a role beyond neuronal migration. J Comp Neurol 2019; 527:2634-2643. [PMID: 30950042 PMCID: PMC6767054 DOI: 10.1002/cne.24696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/20/2019] [Accepted: 03/29/2019] [Indexed: 01/17/2023]
Abstract
Dyslexia is a common neurodevelopmental disorder caused by a significant genetic component. The KIAA0319 gene is one of the most robust dyslexia susceptibility factors but its function remains poorly understood. Initial RNA-interference studies in rats suggested a role in neuronal migration whereas subsequent work with double knock-out mouse models for both Kiaa0319 and its paralogue Kiaa0319-like reported effects in the auditory system but not in neuronal migration. To further understand the role of KIAA0319 during neurodevelopment, we carried out an expression study of its zebrafish orthologue at different embryonic stages. We used different approaches including RNAscope in situ hybridization combined with light-sheet microscopy. The results show particularly high expression during the first few hours of development. Later, expression becomes localized in well-defined structures. In addition to high expression in the brain, we report for the first time expression in the eyes and the notochord. Surprisingly, kiaa0319-like, which generally shows a similar expression pattern to kiaa0319, was not expressed in the notochord suggesting a distinct role for kiaa0319 in this structure. This observation was supported by the identification of notochord enhancers enriched upstream of the KIAA0319 transcription start site, in both zebrafish and humans. This study supports a developmental role for KIAA0319 in the brain as well as in other developing structures, particularly in the notochord which, is key for establishing body patterning in vertebrates.
Collapse
Affiliation(s)
- Monika Gostic
- School of Medicine, University of St Andrews, St Andrews, UK.,Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Angela Martinelli
- School of Medicine, University of St Andrews, St Andrews, UK.,Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Carl Tucker
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Zhengyi Yang
- SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, UK.,School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | | | - Jade-Yi Ewart
- School of Medicine, University of St Andrews, St Andrews, UK.,School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | - Kishan Dholakia
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK.,SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, UK
| | - Keith T Sillar
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | - Javier A Tello
- School of Medicine, University of St Andrews, St Andrews, UK.,Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Silvia Paracchini
- School of Medicine, University of St Andrews, St Andrews, UK.,Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| |
Collapse
|
21
|
Xie S, Jin J, Xu Z, Huang Y, Zhang W, Zhao L, Lo LJ, Peng J, Liu W, Wang F, Shu Q, Zhou T. Centrosomal protein FOR20 is essential for cilia-dependent development in zebrafish embryos. FASEB J 2019; 33:3613-3622. [PMID: 30475641 DOI: 10.1096/fj.201801235rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Centrosomal proteins play critical roles in ciliogenesis. Mutations in many centrosomal proteins have been documented to contribute to developmental defects and cilium-related diseases. Centrosomal protein fibroblast growth factor receptor 1 oncogene partner-related protein of 20 kDa (FOR20) is crucial for ciliogenesis in mammalian cells and the unicellular eukaryote Paramecium; however, the biologic significance of FOR20 in vertebrate development remains unclear. We cloned the zebrafish homolog of the for20 gene and found that for20 mRNA is enriched in ciliated tissues during early zebrafish development. Knockdown of for20 by morpholino oligonucleotides in zebrafish results in multiple ciliary phenotypes, including curved body, hydrocephaly, pericardial edema, kidney cysts, and left-right asymmetry defects. for20 morphants show reduced number and length of cilia in Kupffer's vesicle and pronephric ducts. High-speed video microscopy reveals that cilia in most for20 morphants are consistently paralyzed or beat arrhythmically. To confirm the ciliary phenotypes of for20 morphants, we used the CRISPR/Cas9 system to disrupt for20 gene in zebrafish. for20 mutants exhibit multiple ciliary phenotypes resembling the defects in for20 morphants. All of these phenotypes in for20 morphants and mutants are significantly reversed by exogenous expression of for20 mRNA. Taken together, these data suggest that FOR20 is required for cilium-mediated processes during zebrafish embryogenesis.-Xie, S., Jin, J., Xu, Z., Huang, Y., Zhang, W., Zhao, L., Lo, L. J., Peng, J., Liu, W., Wang, F., Shu, Q., Zhou, T. Centrosomal protein FOR20 is essential for cilia-dependent development in zebrafish embryos.
Collapse
Affiliation(s)
- Shanshan Xie
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Jin
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhangqi Xu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuliang Huang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Zhang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lu Zhao
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Jan Lo
- Ministry of Education (MOE) Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, China; and
| | - Jinrong Peng
- Ministry of Education (MOE) Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, China; and
| | - Wei Liu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Shu
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Joint Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Hangzhou, China
| |
Collapse
|
22
|
Guidi LG, Velayos‐Baeza A, Martinez‐Garay I, Monaco AP, Paracchini S, Bishop DVM, Molnár Z. The neuronal migration hypothesis of dyslexia: A critical evaluation 30 years on. Eur J Neurosci 2018; 48:3212-3233. [PMID: 30218584 PMCID: PMC6282621 DOI: 10.1111/ejn.14149] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 12/29/2022]
Abstract
The capacity for language is one of the key features underlying the complexity of human cognition and its evolution. However, little is known about the neurobiological mechanisms that mediate normal or impaired linguistic ability. For developmental dyslexia, early postmortem studies conducted in the 1980s linked the disorder to subtle defects in the migration of neurons in the developing neocortex. These early studies were reinforced by human genetic analyses that identified dyslexia susceptibility genes and subsequent evidence of their involvement in neuronal migration. In this review, we examine recent experimental evidence that does not support the link between dyslexia and neuronal migration. We critically evaluate gene function studies conducted in rodent models and draw attention to the lack of robust evidence from histopathological and imaging studies in humans. Our review suggests that the neuronal migration hypothesis of dyslexia should be reconsidered, and the neurobiological basis of dyslexia should be approached with a fresh start.
Collapse
Affiliation(s)
- Luiz G. Guidi
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Antonio Velayos‐Baeza
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Isabel Martinez‐Garay
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Division of NeuroscienceSchool of BiosciencesCardiff UniversityCardiffUK
| | | | | | | | - Zoltán Molnár
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
23
|
Costa-Martins AG, Lima L, Alves JMP, Serrano MG, Buck GA, Camargo EP, Teixeira MMG. Genome-wide identification of evolutionarily conserved Small Heat-Shock and eight other proteins bearing α-crystallin domain-like in kinetoplastid protists. PLoS One 2018; 13:e0206012. [PMID: 30346990 PMCID: PMC6197667 DOI: 10.1371/journal.pone.0206012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/04/2018] [Indexed: 11/18/2022] Open
Abstract
Small Heat-Shock Proteins (sHSPs) and other proteins bearing alpha-crystallin domains (ACD) participate in defense against heat and oxidative stress and play important roles in cell cycle, cytoskeleton dynamics, and immunological and pathological mechanisms in eukaryotes. However, little is known about these proteins in early-diverging lineages of protists such as the kinetoplastids. Here, ACD-like proteins (ACDp) were investigated in genomes of 61 species of 12 kinetoplastid genera, including Trypanosoma spp. (23 species of mammals, reptiles and frogs), Leishmania spp. (mammals and lizards), trypanosomatids of insects, Phytomonas spp. of plants, and bodonids. Comparison of ACDps based on domain architecture, predicted tertiary structure, phylogeny and genome organization reveals a kinetoplastid evolutionarily conserved repertoire, which diversified prior to trypanosomatid adaptation to parasitic life. We identified 9 ACDp orthologs classified in 8 families of TryACD: four previously recognized (HSP20, Tryp23A, Tryp23B and ATOM69), and four characterized for the first time in kinetoplastids (TryACDP, TrySGT1, TryDYX1C1 and TryNudC). A single copy of each ortholog was identified in each genome alongside TryNudC1/TrypNudC2 homologs and, overall, ACDPs were under strong selection pressures at main phylogenetic lineages. Transcripts of all ACDPs were identified across the life stages of T. cruzi, T. brucei and Leishmania spp., but proteomic profiles suggested that most ACDPs may be species- and stage-regulated. Our findings establish the basis for functional studies, and provided evolutionary and structural support for an underestimated repertoire of ACDps in the kinetoplastids.
Collapse
Affiliation(s)
- André G Costa-Martins
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luciana Lima
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,INCT-EpiAmO-Instituto Nacional de Epidemiologia na Amazônia Ocidental, Porto Velho, RO, Brazil
| | - João Marcelo P Alves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Myrna G Serrano
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Gregory A Buck
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Erney P Camargo
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,INCT-EpiAmO-Instituto Nacional de Epidemiologia na Amazônia Ocidental, Porto Velho, RO, Brazil
| | - Marta M G Teixeira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,INCT-EpiAmO-Instituto Nacional de Epidemiologia na Amazônia Ocidental, Porto Velho, RO, Brazil
| |
Collapse
|
24
|
C11orf70 Mutations Disrupting the Intraflagellar Transport-Dependent Assembly of Multiple Axonemal Dyneins Cause Primary Ciliary Dyskinesia. Am J Hum Genet 2018; 102:956-972. [PMID: 29727692 DOI: 10.1016/j.ajhg.2018.03.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 03/23/2018] [Indexed: 01/05/2023] Open
Abstract
Primary ciliary dyskinesia (PCD) is a genetically and phenotypically heterogeneous disorder characterized by destructive respiratory disease and laterality abnormalities due to randomized left-right body asymmetry. PCD is mostly caused by mutations affecting the core axoneme structure of motile cilia that is essential for movement. Genes that cause PCD when mutated include a group that encode proteins essential for the assembly of the ciliary dynein motors and the active transport process that delivers them from their cytoplasmic assembly site into the axoneme. We screened a cohort of affected individuals for disease-causing mutations using a targeted next generation sequencing panel and identified two unrelated families (three affected children) with mutations in the uncharacterized C11orf70 gene (official gene name CFAP300). The affected children share a consistent PCD phenotype from early life with laterality defects and immotile respiratory cilia displaying combined loss of inner and outer dynein arms (IDA+ODA). Phylogenetic analysis shows C11orf70 is highly conserved, distributed across species similarly to proteins involved in the intraflagellar transport (IFT)-dependant assembly of axonemal dyneins. Paramecium C11orf70 RNAi knockdown led to combined loss of ciliary IDA+ODA with reduced cilia beating and swim velocity. Tagged C11orf70 in Paramecium and Chlamydomonas localizes mainly in the cytoplasm with a small amount in the ciliary component. IFT139/TTC21B (IFT-A protein) and FLA10 (IFT kinesin) depletion experiments show that its transport within cilia is IFT dependent. During ciliogenesis, C11orf70 accumulates at the ciliary tips in a similar distribution to the IFT-B protein IFT46. In summary, C11orf70 is essential for assembly of dynein arms and C11orf70 mutations cause defective cilia motility and PCD.
Collapse
|
25
|
Kumar D, Thomason RT, Yankova M, Gitlin JD, Mains RE, Eipper BA, King SM. Microvillar and ciliary defects in zebrafish lacking an actin-binding bioactive peptide amidating enzyme. Sci Rep 2018; 8:4547. [PMID: 29540787 PMCID: PMC5852006 DOI: 10.1038/s41598-018-22732-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 02/28/2018] [Indexed: 11/09/2022] Open
Abstract
The assembly of membranous extensions such as microvilli and cilia in polarized cells is a tightly regulated, yet poorly understood, process. Peptidylglycine α-amidating monooxygenase (PAM), a membrane enzyme essential for the synthesis of amidated bioactive peptides, was recently identified in motile and non-motile (primary) cilia and has an essential role in ciliogenesis in Chlamydomonas, Schmidtea and mouse. In mammalian cells, changes in PAM levels alter secretion and organization of the actin cytoskeleton. Here we show that lack of Pam in zebrafish recapitulates the lethal edematous phenotype observed in Pam -/- mice and reveals additional defects. The pam -/- zebrafish embryos display an initial striking loss of microvilli and subsequently impaired ciliogenesis in the pronephros. In multiciliated mouse tracheal epithelial cells, vesicular PAM staining colocalizes with apical actin, below the microvilli. In PAM-deficient Chlamydomonas, the actin cytoskeleton is dramatically reorganized, and expression of an actin paralogue is upregulated. Biochemical assays reveal that the cytosolic PAM C-terminal domain interacts directly with filamentous actin but does not alter the rate of actin polymerization or disassembly. Our results point to a critical role for PAM in organizing the actin cytoskeleton during development, which could in turn impact both microvillus formation and ciliogenesis.
Collapse
Affiliation(s)
- Dhivya Kumar
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Rebecca T Thomason
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- University of Virginia, Charlottesville, VA, 22904, USA
| | - Maya Yankova
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Electron Microscopy Facility, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Jonathan D Gitlin
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Betty A Eipper
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| | - Stephen M King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
- Electron Microscopy Facility, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
26
|
Yamamoto R, Obbineni JM, Alford LM, Ide T, Owa M, Hwang J, Kon T, Inaba K, James N, King SM, Ishikawa T, Sale WS, Dutcher SK. Chlamydomonas DYX1C1/PF23 is essential for axonemal assembly and proper morphology of inner dynein arms. PLoS Genet 2017; 13:e1006996. [PMID: 28892495 PMCID: PMC5608425 DOI: 10.1371/journal.pgen.1006996] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/21/2017] [Accepted: 08/22/2017] [Indexed: 12/26/2022] Open
Abstract
Cytoplasmic assembly of ciliary dyneins, a process known as preassembly, requires numerous non-dynein proteins, but the identities and functions of these proteins are not fully elucidated. Here, we show that the classical Chlamydomonas motility mutant pf23 is defective in the Chlamydomonas homolog of DYX1C1. The pf23 mutant has a 494 bp deletion in the DYX1C1 gene and expresses a shorter DYX1C1 protein in the cytoplasm. Structural analyses, using cryo-ET, reveal that pf23 axonemes lack most of the inner dynein arms. Spectral counting confirms that DYX1C1 is essential for the assembly of the majority of ciliary inner dynein arms (IDA) as well as a fraction of the outer dynein arms (ODA). A C-terminal truncation of DYX1C1 shows a reduction in a subset of these ciliary IDAs. Sucrose gradients of cytoplasmic extracts show that preassembled ciliary dyneins are reduced compared to wild-type, which suggests an important role in dynein complex stability. The role of PF23/DYX1C1 remains unknown, but we suggest that DYX1C1 could provide a scaffold for macromolecular assembly. Most animal cells have antenna-like organelles called “cilia”. These organelles have various important functions both in motility and sensing the environment. Motile cilia are essential for moving cells as well as moving fluids across a surface. The waveform of motile cilia requires large macromolecular motors; these are the ciliary dyneins. These dynein complexes are assembled in the cytoplasm in a pathway called preassembly, and then transported into cilia. Defects in this process cause a heterogeneous human disease called primary ciliary dyskinesia that results, for example, in the disruption of the motility of respiratory tract cilia, sperm and nodal cilia during development. The mechanisms of the preassembly pathway are not fully understood. In this study, we use a mutation in the well-conserved DYX1C1/PF23 gene of the green alga, Chlamydomonas reinhardtii. Loss of a conserved domain (DYX) reveals a failure to assemble most ciliary dyneins. Preassembly of inner arm dyneins is particularly affected. We find that if dynein arms are not assembled, dynein subunits in the cytoplasm are unstable. We suggest that DYX1C1 may play a role as a scaffold for other preassembly factors and the dynein subunits.
Collapse
Affiliation(s)
- Ryosuke Yamamoto
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Jagan M. Obbineni
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen PSI, Switzerland
| | - Lea M. Alford
- Department of Biology, Oglethorpe University, Atlanta, Georgia, United States of America
| | - Takahiro Ide
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Mikito Owa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Juyeon Hwang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Takahide Kon
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Kazuo Inaba
- Shimoda Marine Research Center, University of Tsukuba, Shizuoka, Japan
| | - Noliyanda James
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Stephen M. King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Takashi Ishikawa
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen PSI, Switzerland
- * E-mail: (TI); (WSS); (SKD)
| | - Winfield S. Sale
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (TI); (WSS); (SKD)
| | - Susan K. Dutcher
- Shimoda Marine Research Center, University of Tsukuba, Shizuoka, Japan
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (TI); (WSS); (SKD)
| |
Collapse
|
27
|
Trulioff A, Ermakov A, Malashichev Y. Primary Cilia as a Possible Link between Left-Right Asymmetry and Neurodevelopmental Diseases. Genes (Basel) 2017; 8:genes8020048. [PMID: 28125008 PMCID: PMC5333037 DOI: 10.3390/genes8020048] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/21/2016] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
Abstract
Cilia have multiple functions in the development of the entire organism, and participate in the development and functioning of the central nervous system. In the last decade, studies have shown that they are implicated in the development of the visceral left-right asymmetry in different vertebrates. At the same time, some neuropsychiatric disorders, such as schizophrenia, autism, bipolar disorder, and dyslexia, are known to be associated with lateralization failure. In this review, we consider possible links in the mechanisms of determination of visceral asymmetry and brain lateralization, through cilia. We review the functions of seven genes associated with both cilia, and with neurodevelopmental diseases, keeping in mind their possible role in the establishment of the left-right brain asymmetry.
Collapse
Affiliation(s)
- Andrey Trulioff
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
| | - Alexander Ermakov
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| | - Yegor Malashichev
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| |
Collapse
|
28
|
Identification of Novel Transcribed Regions in Zebrafish (Danio rerio) Using RNA-Sequencing. PLoS One 2016; 11:e0160197. [PMID: 27462902 PMCID: PMC4962977 DOI: 10.1371/journal.pone.0160197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 07/17/2016] [Indexed: 11/28/2022] Open
Abstract
Zebrafish (Danio rerio) has emerged as a model organism to investigate vertebrate development and human genetic diseases. However, the zebrafish genome annotation is still ongoing and incomplete, and there are still new gene transcripts to be found. With the introduction of massive parallel sequencing, whole transcriptome studies became possible. In the present study, we aimed to discover novel transcribed regions (NTRs) using developmental transcriptome data from RNA sequencing. In order to achieve this, we developed an in-house bioinformatics pipeline for NTR discovery. Using the pipeline, we detected 152 putative NTRs that at the time of discovery were not annotated in Ensembl and NCBI gene database. Four randomly selected NTRs were successfully validated using RT-PCR, and expression profiles of 10 randomly selected NTRs were evaluated using qRT-PCR. The identification of these 152 NTRs provide new information for zebrafish genome annotation as well as new candidates for studies of zebrafish gene function.
Collapse
|
29
|
Tammimies K, Bieder A, Lauter G, Sugiaman-Trapman D, Torchet R, Hokkanen ME, Burghoorn J, Castrén E, Kere J, Tapia-Páez I, Swoboda P. Ciliary dyslexia candidate genes DYX1C1 and DCDC2 are regulated by Regulatory Factor X (RFX) transcription factors through X-box promoter motifs. FASEB J 2016; 30:3578-3587. [PMID: 27451412 PMCID: PMC5024701 DOI: 10.1096/fj.201500124rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 07/05/2016] [Indexed: 11/11/2022]
Abstract
DYX1C1, DCDC2, and KIAA0319 are
three of the most replicated dyslexia candidate genes (DCGs). Recently, these DCGs
were implicated in functions at the cilium. Here, we investigate the regulation of
these DCGs by Regulatory Factor X transcription factors (RFX TFs), a gene family
known for transcriptionally regulating ciliary genes. We identify conserved X-box
motifs in the promoter regions of DYX1C1, DCDC2, and
KIAA0319 and demonstrate their functionality, as well as the
ability to recruit RFX TFs using reporter gene and electrophoretic mobility shift
assays. Furthermore, we uncover a complex regulation pattern between
RFX1, RFX2, and RFX3 and their
significant effect on modifying the endogenous expression of DYX1C1
and DCDC2 in a human retinal pigmented epithelial cell line
immortalized with hTERT (hTERT-RPE1). In addition, induction of ciliogenesis
increases the expression of RFX TFs and DCGs. At the protein level, we show that
endogenous DYX1C1 localizes to the base of the cilium, whereas DCDC2 localizes along
the entire axoneme of the cilium, thereby validating earlier localization studies
using overexpression models. Our results corroborate the emerging role of DCGs in
ciliary function and characterize functional noncoding elements, X-box promoter
motifs, in DCG promoter regions, which thus can be targeted for mutation screening in
dyslexia and ciliopathies associated with these genes.—Tammimies, K., Bieder,
A., Lauter, G., Sugiaman-Trapman, D., Torchet, R., Hokkanen, M.-E., Burghoorn, J.,
Castrén, E., Kere, J., Tapia-Páez, I., Swoboda, P. Ciliary dyslexia
candidate genes DYX1C1 and DCDC2 are regulated by
Regulatory Factor (RF) X transcription factors through X-box promoter motifs.
Collapse
Affiliation(s)
- Kristiina Tammimies
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden; Center of Neurodevelopmental Disorders (KIND), Pediatric Neuropsychiatry Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Bieder
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Gilbert Lauter
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | - Rachel Torchet
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | - Jan Burghoorn
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Eero Castrén
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden; Molecular Neurology Research Program, University of Helsinki, Helsinki, Finland; and Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Isabel Tapia-Páez
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden;
| | - Peter Swoboda
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden;
| |
Collapse
|
30
|
Gialluisi A, Visconti A, Willcutt EG, Smith SD, Pennington BF, Falchi M, DeFries JC, Olson RK, Francks C, Fisher SE. Investigating the effects of copy number variants on reading and language performance. J Neurodev Disord 2016; 8:17. [PMID: 27186239 PMCID: PMC4868026 DOI: 10.1186/s11689-016-9147-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/31/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Reading and language skills have overlapping genetic bases, most of which are still unknown. Part of the missing heritability may be caused by copy number variants (CNVs). METHODS In a dataset of children recruited for a history of reading disability (RD, also known as dyslexia) or attention deficit hyperactivity disorder (ADHD) and their siblings, we investigated the effects of CNVs on reading and language performance. First, we called CNVs with PennCNV using signal intensity data from Illumina OmniExpress arrays (~723,000 probes). Then, we computed the correlation between measures of CNV genomic burden and the first principal component (PC) score derived from several continuous reading and language traits, both before and after adjustment for performance IQ. Finally, we screened the genome, probe-by-probe, for association with the PC scores, through two complementary analyses: we tested a binary CNV state assigned for the location of each probe (i.e., CNV+ or CNV-), and we analyzed continuous probe intensity data using FamCNV. RESULTS No significant correlation was found between measures of CNV burden and PC scores, and no genome-wide significant associations were detected in probe-by-probe screening. Nominally significant associations were detected (p~10(-2)-10(-3)) within CNTN4 (contactin 4) and CTNNA3 (catenin alpha 3). These genes encode cell adhesion molecules with a likely role in neuronal development, and they have been previously implicated in autism and other neurodevelopmental disorders. A further, targeted assessment of candidate CNV regions revealed associations with the PC score (p~0.026-0.045) within CHRNA7 (cholinergic nicotinic receptor alpha 7), which encodes a ligand-gated ion channel and has also been implicated in neurodevelopmental conditions and language impairment. FamCNV analysis detected a region of association (p~10(-2)-10(-4)) within a frequent deletion ~6 kb downstream of ZNF737 (zinc finger protein 737, uncharacterized protein), which was also observed in the association analysis using CNV calls. CONCLUSIONS These data suggest that CNVs do not underlie a substantial proportion of variance in reading and language skills. Analysis of additional, larger datasets is warranted to further assess the potential effects that we found and to increase the power to detect CNV effects on reading and language.
Collapse
Affiliation(s)
- Alessandro Gialluisi
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands ; Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Alessia Visconti
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Erik G Willcutt
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO USA ; Department of Psychology and Neuroscience, University of Colorado, Boulder, CO USA
| | - Shelley D Smith
- Munroe Meyer Institute, University of Nebraska Medical Center, Omaha, NE USA
| | | | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - John C DeFries
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO USA ; Department of Psychology and Neuroscience, University of Colorado, Boulder, CO USA
| | - Richard K Olson
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO USA ; Department of Psychology and Neuroscience, University of Colorado, Boulder, CO USA
| | - Clyde Francks
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands ; Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands ; Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| |
Collapse
|
31
|
Paracchini S, Diaz R, Stein J. Advances in Dyslexia Genetics—New Insights Into the Role of Brain Asymmetries. ADVANCES IN GENETICS 2016; 96:53-97. [DOI: 10.1016/bs.adgen.2016.08.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
32
|
Kageyama H, Miyajima M, Ogino I, Nakajima M, Shimoji K, Fukai R, Miyake N, Nishiyama K, Matsumoto N, Arai H. Panventriculomegaly with a wide foramen of Magendie and large cisterna magna. J Neurosurg 2015; 124:1858-66. [PMID: 26636390 DOI: 10.3171/2015.6.jns15162] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The authors' goal in this paper is to provide the first clinical, radiological, and genetic studies of panventriculomegaly (PaVM) defined by a wide foramen of Magendie and large cisterna magna. METHODS Clinical and brain imaging data from 28 PaVM patients (including 10 patients from 5 families) were retrospectively studied. Five children were included. In adult patients, the age at onset was 56.0 ± 16.7 years. Tetraventricular dilation, aqueductal opening with flow void on T2-weighted images, and a wide foramen of Magendie and large cisterna magna (wide cerebrospinal fluid space at the fourth ventricle outlet) were essential MRI findings for PaVM diagnosis. 3D fast asymmetrical spin echo sequences were used for visualization of cistern membranes. Time-spatial labeling inversion pulse examination was performed to analyze cerebrospinal fluid movement. Copy number variations were determined using high-resolution microarray and were validated by quantitative polymerase chain reaction with breakpoint sequencing. RESULTS Adult patients showed gait disturbance, urinary dysfunction, and cognitive dysfunction. Five infant patients exhibited macrocranium. Patients were divided into 2 subcategories, those with or without downward bulging third ventricular floors and membranous structures in the prepontine cistern. Patients with bulging floors were successfully treated with endoscopic third ventriculostomy. Genetic analysis revealed a deletion in DNAH14 that encodes a dynein heavy chain protein associated with motile cilia function, and which co-segregated with patients in a family without a downward bulging third ventricular floor. CONCLUSIONS Panventriculomegaly with a wide foramen of Magendie and a large cisterna magna may belong to a subtype of congenital hydrocephalus with familial accumulation, younger age at onset, and symptoms of normal pressure hydrocephalus. In addition, a family with PaVM has a gene mutation associated with dysfunction of motile cilia.
Collapse
Affiliation(s)
- Hiroshi Kageyama
- Department of Neurosurgery, Graduate School of Medicine, Juntendo University, Tokyo;,Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama;,Department of Neurosurgery, Kuki General Hospital, Kuki, Saitama; and
| | - Masakazu Miyajima
- Department of Neurosurgery, Graduate School of Medicine, Juntendo University, Tokyo
| | - Ikuko Ogino
- Department of Neurosurgery, Graduate School of Medicine, Juntendo University, Tokyo
| | - Madoka Nakajima
- Department of Neurosurgery, Graduate School of Medicine, Juntendo University, Tokyo
| | - Kazuaki Shimoji
- Department of Neurosurgery, Graduate School of Medicine, Juntendo University, Tokyo
| | - Ryoko Fukai
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama
| | | | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama
| | - Hajime Arai
- Department of Neurosurgery, Graduate School of Medicine, Juntendo University, Tokyo
| |
Collapse
|
33
|
Abstract
Language is a defining characteristic of the human species, but its foundations remain mysterious. Heritable disorders offer a gateway into biological underpinnings, as illustrated by the discovery that FOXP2 disruptions cause a rare form of speech and language impairment. The genetic architecture underlying language-related disorders is complex, and although some progress has been made, it has proved challenging to pinpoint additional relevant genes with confidence. Next-generation sequencing and genome-wide association studies are revolutionizing understanding of the genetic bases of other neurodevelopmental disorders, like autism and schizophrenia, and providing fundamental insights into the molecular networks crucial for typical brain development. We discuss how a similar genomic perspective, brought to the investigation of language-related phenotypes, promises to yield equally informative discoveries. Moreover, we outline how follow-up studies of genetic findings using cellular systems and animal models can help to elucidate the biological mechanisms involved in the development of brain circuits supporting language.
Collapse
Affiliation(s)
- Sarah A Graham
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands;
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; .,Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 EN Nijmegen, The Netherlands;
| |
Collapse
|
34
|
Einarsdottir E, Svensson I, Darki F, Peyrard-Janvid M, Lindvall JM, Ameur A, Jacobsson C, Klingberg T, Kere J, Matsson H. Mutation in CEP63 co-segregating with developmental dyslexia in a Swedish family. Hum Genet 2015; 134:1239-48. [PMID: 26400686 PMCID: PMC4628622 DOI: 10.1007/s00439-015-1602-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/15/2015] [Indexed: 01/17/2023]
Abstract
Developmental dyslexia is the most common learning disorder in children. Problems in reading and writing are likely due to a complex interaction of genetic and environmental factors, resulting in reduced power of studies of the genetic factors underlying developmental dyslexia. Our approach in the current study was to perform exome sequencing of affected and unaffected individuals within an extended pedigree with a familial form of developmental dyslexia. We identified a two-base mutation, causing a p.R229L amino acid substitution in the centrosomal protein 63 kDa (CEP63), co-segregating with developmental dyslexia in this pedigree. This mutation is novel, and predicted to be highly damaging for the function of the protein. 3D modelling suggested a distinct conformational change caused by the mutation. CEP63 is localised to the centrosome in eukaryotic cells and is required for maintaining normal centriole duplication and control of cell cycle progression. We found that a common polymorphism in the CEP63 gene had a significant association with brain white matter volume. The brain regions were partly overlapping with the previously reported region influenced by polymorphisms in the dyslexia susceptibility genes DYX1C1 and KIAA0319. We hypothesise that CEP63 is particularly important for brain development and might control the proliferation and migration of cells when those two events need to be highly coordinated.
Collapse
Affiliation(s)
- Elisabet Einarsdottir
- Department of Biosciences and Nutrition, and Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden.
| | - Idor Svensson
- Department of Psychology, Linneaus University, Växjö, Sweden
| | - Fahimeh Darki
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Myriam Peyrard-Janvid
- Department of Biosciences and Nutrition, and Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Jessica M Lindvall
- Department of Biosciences and Nutrition, and Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden.,Bioinformatics Infrastructure for Life Sciences (BILS), Stockholm University, Stockholm, Sweden.,Science for Life Laboratory, Stockholm University, Stockholm, Sweden.,Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Adam Ameur
- Uppsala Genome Center, Uppsala University, Uppsala, Sweden
| | | | - Torkel Klingberg
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, and Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden.,Science for Life Laboratory, Stockholm University, Stockholm, Sweden.,Molecular Neurology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Hans Matsson
- Department of Biosciences and Nutrition, and Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
35
|
Lin H, Dutcher SK. Genetic and genomic approaches to identify genes involved in flagellar assembly in Chlamydomonas reinhardtii. Methods Cell Biol 2015; 127:349-86. [PMID: 25837400 DOI: 10.1016/bs.mcb.2014.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Flagellar assembly requires intraflagellar transport of components from the cell body to the flagellar tip for assembly. The understanding of flagellar assembly has been aided by the ease of biochemistry and the availability of mutants in the unicellular green alga, Chlamydomonas reinhardtii. In this chapter, we discuss means to identify genes involved in these processes using forward and reverse genetics. In particular, the ease and low cost of whole genome sequencing (WGS) will help to make gene identification easier and promote the understanding of this important process.
Collapse
Affiliation(s)
- Huawen Lin
- Department of Genetics, Washington University, St. Louis, MO, USA
| | - Susan K Dutcher
- Department of Genetics, Washington University, St. Louis, MO, USA.
| |
Collapse
|
36
|
DCDC2 polymorphism is associated with left temporoparietal gray and white matter structures during development. J Neurosci 2015; 34:14455-62. [PMID: 25339756 DOI: 10.1523/jneurosci.1216-14.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Three genes, DYX1C1, DCDC2, and KIAA0319, have been previously associated with dyslexia, neuronal migration, and ciliary function. Three polymorphisms within these genes, rs3743204 (DYX1C1), rs793842 (DCDC2), and rs6935076 (KIAA0319) have also been linked to normal variability of left temporoparietal white matter volume connecting the middle temporal cortex to the angular and supramarginal gyri. Here, we assessed whether these polymorphisms are also related to the cortical thickness of the associated regions during childhood development using a longitudinal dataset of 76 randomly selected children and young adults who were scanned up to three times each, 2 years apart. rs793842 in DCDC2 was significantly associated with the thickness of left angular and supramarginal gyri as well as the left lateral occipital cortex. The cortex was significantly thicker for T-allele carriers, who also had lower white matter volume and lower reading comprehension scores. There was a negative correlation between white matter volume and cortical thickness, but only white matter volume predicted reading comprehension 2 years after scanning. These results show how normal variability in reading comprehension is related to gene, white matter volume, and cortical thickness in the inferior parietal lobe. Possibly, the variability of gray and white matter structures could both be related to the role of DCDC2 in ciliary function, which affects both neuronal migration and axonal outgrowth.
Collapse
|
37
|
Schueler M, Braun DA, Chandrasekar G, Gee HY, Klasson TD, Halbritter J, Bieder A, Porath JD, Airik R, Zhou W, LoTurco JJ, Che A, Otto EA, Böckenhauer D, Sebire NJ, Honzik T, Harris PC, Koon SJ, Gunay-Aygun M, Saunier S, Zerres K, Bruechle NO, Drenth JPH, Pelletier L, Tapia-Páez I, Lifton RP, Giles RH, Kere J, Hildebrandt F. DCDC2 mutations cause a renal-hepatic ciliopathy by disrupting Wnt signaling. Am J Hum Genet 2015; 96:81-92. [PMID: 25557784 DOI: 10.1016/j.ajhg.2014.12.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022] Open
Abstract
Nephronophthisis-related ciliopathies (NPHP-RC) are recessive diseases characterized by renal dysplasia or degeneration. We here identify mutations of DCDC2 as causing a renal-hepatic ciliopathy. DCDC2 localizes to the ciliary axoneme and to mitotic spindle fibers in a cell-cycle-dependent manner. Knockdown of Dcdc2 in IMCD3 cells disrupts ciliogenesis, which is rescued by wild-type (WT) human DCDC2, but not by constructs that reflect human mutations. We show that DCDC2 interacts with DVL and DCDC2 overexpression inhibits β-catenin-dependent Wnt signaling in an effect additive to Wnt inhibitors. Mutations detected in human NPHP-RC lack these effects. A Wnt inhibitor likewise restores ciliogenesis in 3D IMCD3 cultures, emphasizing the importance of Wnt signaling for renal tubulogenesis. Knockdown of dcdc2 in zebrafish recapitulates NPHP-RC phenotypes, including renal cysts and hydrocephalus, which is rescued by a Wnt inhibitor and by WT, but not by mutant, DCDC2. We thus demonstrate a central role of Wnt signaling in the pathogenesis of NPHP-RC, suggesting an avenue for potential treatment of NPHP-RC.
Collapse
Affiliation(s)
- Markus Schueler
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gayathri Chandrasekar
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Heon Yung Gee
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy D Klasson
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Jan Halbritter
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrea Bieder
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Jonathan D Porath
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rannar Airik
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Weibin Zhou
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joseph J LoTurco
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Alicia Che
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Edgar A Otto
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109, USA
| | - Detlef Böckenhauer
- University College London, Institute of Child Health and Pediatric Nephrology, Great Ormond Street Hospital, London WC1N3JH, UK
| | - Neil J Sebire
- Department of Histopathology, Great Ormond Street Hospital, London WC1N3JH, UK
| | - Tomas Honzik
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 2, Prague 2, 128 08 Czech Republic
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarah J Koon
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Meral Gunay-Aygun
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sophie Saunier
- Inserm U574 and Department of Genetics, Paris 5 University, Necker Hospital, 75015 Paris, France
| | - Klaus Zerres
- Institute of Human Genetics, University Hospital, RWTH Aachen, 52074 Aachen, Germany
| | - Nadina Ortiz Bruechle
- Institute of Human Genetics, University Hospital, RWTH Aachen, 52074 Aachen, Germany
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud UMC, P.O. Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Laurence Pelletier
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Isabel Tapia-Páez
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Rachel H Giles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden; Molecular Neurology Research Program, University of Helsinki, and Folkhälsan Institute of Genetics, 00014 Helsinki, Finland; Science for Life Laboratory, Karolinska Institutet, 171 21 Solna, Sweden.
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
38
|
Ito-Amano M, Nakamura Y, Morisaki M, He X, Hayashi M, Watanapokasin R, Kato H. Temporal and spatial expression patterns of bone morphogenetic protein 3 in developing zebrafish. Open Rheumatol J 2014; 8:69-72. [PMID: 25328556 PMCID: PMC4196250 DOI: 10.2174/1874312901408010069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 07/18/2014] [Accepted: 07/19/2014] [Indexed: 11/22/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are important elements in bone biology. We herein report the expression profiles of zebrafish bmp3 (zbmp3) as demonstrated by real-time PCR and in situ hybridization. The expression of zbmp3 was highly detectable by real-time PCR from 1 day post-fertilization (1 dpf) to 2 weeks post-fertilization (2 wpf) and peaked at 1 wpf. For in situ hybridization experiments, zbmp3 was expressed in the otic vesicle at 1 dpf, 2 dpf, 3 dpf, and 5 dpf. It was also expressed in the pharyngeal arches, including the opercle, branchiostegal ray, and pectoral fins, at 2 dpf. Our results suggest that zbmp3 may play an important role in the skeletal biology of developing zebrafish.
Collapse
Affiliation(s)
- Midori Ito-Amano
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto 390-8621, Japan
| | - Yukio Nakamura
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto 390-8621, Japan
| | - Mika Morisaki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto 390-8621, Japan
| | - Xinjun He
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Masanori Hayashi
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto 390-8621, Japan
| | - Ramida Watanapokasin
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Hiroyuki Kato
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto 390-8621, Japan
| |
Collapse
|
39
|
Kere J. The molecular genetics and neurobiology of developmental dyslexia as model of a complex phenotype. Biochem Biophys Res Commun 2014; 452:236-43. [PMID: 25078623 DOI: 10.1016/j.bbrc.2014.07.102] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 07/22/2014] [Indexed: 10/25/2022]
Abstract
Among complex disorders, those concerning neuropsychiatric phenotypes involve particular challenges compared to disorders with more easily distinguished clinical signs and measures. One such common and unusually challenging phenotype to disentangle genetically is developmental dyslexia (DD), or reading disability, defined as the inability to learn to read and write for an otherwise normally intelligent child with normal senses and educational opportunity. There is presently ample evidence for the strongly biological etiology for DD, and a dozen susceptibility genes have been suggested. Many of these genes point to common but previously unsuspected biological mechanisms, such as neuronal migration and cilia functions. I discuss here the state-of-the-art in genomic and neurobiological aspects of DD research, starting with short general background to its history.
Collapse
Affiliation(s)
- Juha Kere
- Department of Biosciences and Nutrition, Centre for Innovative Medicine, Karolinska Institutet, Stockholm, Sweden; Molecular Neurology Research Program, University of Helsinki, Folkhälsan Institute of Genetics, Helsinki, Finland.
| |
Collapse
|
40
|
Reading and language disorders: the importance of both quantity and quality. Genes (Basel) 2014; 5:285-309. [PMID: 24705331 PMCID: PMC4094934 DOI: 10.3390/genes5020285] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 01/25/2023] Open
Abstract
Reading and language disorders are common childhood conditions that often co-occur with each other and with other neurodevelopmental impairments. There is strong evidence that disorders, such as dyslexia and Specific Language Impairment (SLI), have a genetic basis, but we expect the contributing genetic factors to be complex in nature. To date, only a few genes have been implicated in these traits. Their functional characterization has provided novel insight into the biology of neurodevelopmental disorders. However, the lack of biological markers and clear diagnostic criteria have prevented the collection of the large sample sizes required for well-powered genome-wide screens. One of the main challenges of the field will be to combine careful clinical assessment with high throughput genetic technologies within multidisciplinary collaborations.
Collapse
|
41
|
Brandler WM, Paracchini S. The genetic relationship between handedness and neurodevelopmental disorders. Trends Mol Med 2013; 20:83-90. [PMID: 24275328 PMCID: PMC3969300 DOI: 10.1016/j.molmed.2013.10.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/29/2013] [Accepted: 10/31/2013] [Indexed: 02/06/2023]
Abstract
Handedness and brain asymmetry have been linked to neurodevelopmental disorders such as dyslexia and schizophrenia. The genetic nature of this correlation is not understood. Recent discoveries have shown handedness is determined in part by the biological pathways that establish left/right (LR) body asymmetry during development. Cilia play a key role in this process, and candidate genes for dyslexia have also been recently shown to be involved in cilia formation. Defective cilia result not only in LR body asymmetry phenotypes but also brain midline phenotypes such as an absent corpus callosum. These findings suggest that the mechanisms for establishing LR asymmetry in the body are reused for brain midline development, which in turn influences traits such as handedness and reading ability.
Collapse
Affiliation(s)
- William M Brandler
- MRC Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3PT, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Silvia Paracchini
- School of Medicine, University of St Andrews, St Andrews, KY16 9TF, UK.
| |
Collapse
|
42
|
Carrion-Castillo A, Franke B, Fisher SE. Molecular genetics of dyslexia: an overview. DYSLEXIA (CHICHESTER, ENGLAND) 2013; 19:214-240. [PMID: 24133036 DOI: 10.1002/dys.1464] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 09/02/2013] [Indexed: 05/28/2023]
Abstract
Dyslexia is a highly heritable learning disorder with a complex underlying genetic architecture. Over the past decade, researchers have pinpointed a number of candidate genes that may contribute to dyslexia susceptibility. Here, we provide an overview of the state of the art, describing how studies have moved from mapping potential risk loci, through identification of associated gene variants, to characterization of gene function in cellular and animal model systems. Work thus far has highlighted some intriguing mechanistic pathways, such as neuronal migration, axon guidance, and ciliary biology, but it is clear that we still have much to learn about the molecular networks that are involved. We end the review by highlighting the past, present, and future contributions of the Dutch Dyslexia Programme to studies of genetic factors. In particular, we emphasize the importance of relating genetic information to intermediate neurobiological measures, as well as the value of incorporating longitudinal and developmental data into molecular designs.
Collapse
Affiliation(s)
- Amaia Carrion-Castillo
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands
| | | | | |
Collapse
|
43
|
Brandler WM, Morris AP, Evans DM, Scerri TS, Kemp JP, Timpson NJ, St Pourcain B, Smith GD, Ring SM, Stein J, Monaco AP, Talcott JB, Fisher SE, Webber C, Paracchini S. Common variants in left/right asymmetry genes and pathways are associated with relative hand skill. PLoS Genet 2013; 9:e1003751. [PMID: 24068947 PMCID: PMC3772043 DOI: 10.1371/journal.pgen.1003751] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 07/10/2013] [Indexed: 12/31/2022] Open
Abstract
Humans display structural and functional asymmetries in brain organization, strikingly with respect to language and handedness. The molecular basis of these asymmetries is unknown. We report a genome-wide association study meta-analysis for a quantitative measure of relative hand skill in individuals with dyslexia [reading disability (RD)] (n = 728). The most strongly associated variant, rs7182874 (P = 8.68 × 10(-9)), is located in PCSK6, further supporting an association we previously reported. We also confirmed the specificity of this association in individuals with RD; the same locus was not associated with relative hand skill in a general population cohort (n = 2,666). As PCSK6 is known to regulate NODAL in the development of left/right (LR) asymmetry in mice, we developed a novel approach to GWAS pathway analysis, using gene-set enrichment to test for an over-representation of highly associated variants within the orthologs of genes whose disruption in mice yields LR asymmetry phenotypes. Four out of 15 LR asymmetry phenotypes showed an over-representation (FDR ≤ 5%). We replicated three of these phenotypes; situs inversus, heterotaxia, and double outlet right ventricle, in the general population cohort (FDR ≤ 5%). Our findings lead us to propose that handedness is a polygenic trait controlled in part by the molecular mechanisms that establish LR body asymmetry early in development.
Collapse
Affiliation(s)
- William M. Brandler
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- MRC Functional Genomics Unit, Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - David M. Evans
- MRC Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Thomas S. Scerri
- The Walter and Eliza Hall Research Institute of Medical Research, Melbourne, Australia
| | - John P. Kemp
- MRC Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Nicholas J. Timpson
- MRC Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Beate St Pourcain
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Susan M. Ring
- MRC Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
| | - John Stein
- Department of Physiology, University of Oxford, Oxford, United Kingdom
| | - Anthony P. Monaco
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Joel B. Talcott
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Simon E. Fisher
- Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Caleb Webber
- MRC Functional Genomics Unit, Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | - Silvia Paracchini
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
44
|
Lyons PJ, Sapio MR, Fricker LD. Zebrafish cytosolic carboxypeptidases 1 and 5 are essential for embryonic development. J Biol Chem 2013; 288:30454-30462. [PMID: 24022483 DOI: 10.1074/jbc.m113.497933] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The cytosolic carboxypeptidases (CCPs) are a subfamily of metalloenzymes within the larger M14 family of carboxypeptidases that have been implicated in the post-translational modification of tubulin. It has been suggested that at least four of the six mammalian CCPs function as tubulin deglutamylases. However, it is not yet clear whether these enzymes play redundant or unique roles within the cell. To address this question, genes encoding CCPs were identified in the zebrafish genome. Analysis by quantitative polymerase chain reaction indicated that CCP1, CCP2, CCP5, and CCP6 mRNAs were detectable between 2 h and 8 days postfertilization with highest levels 5-8 days postfertilization. CCP1, CCP2, and CCP5 mRNAs were predominantly expressed in tissues such as the brain, olfactory placodes, and pronephric ducts. Morpholino oligonucleotide-mediated knockdown of CCP1 and CCP5 mRNA resulted in a common phenotype including ventral body curvature and hydrocephalus. Confocal microscopy of morphant zebrafish revealed olfactory placodes with defective morphology as well as pronephric ducts with increased polyglutamylation. These data suggest that CCP1 and CCP5 play important roles in developmental processes, particularly the development and functioning of cilia. The robust and similar defects upon knockdown suggest that each CCP may have a function in microtubule modification and ciliary function and that other CCPs are not able to compensate for the loss of one.
Collapse
Affiliation(s)
| | - Matthew R Sapio
- Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | | |
Collapse
|
45
|
Tarkar A, Loges NT, Slagle CE, Francis R, Dougherty GW, Tamayo JV, Shook B, Cantino M, Schwartz D, Jahnke C, Olbrich H, Werner C, Raidt J, Pennekamp P, Abouhamed M, Hjeij R, Köhler G, Griese M, Li Y, Lemke K, Klena N, Liu X, Gabriel G, Tobita K, Jaspers M, Morgan LC, Shapiro AJ, Letteboer SJ, Mans DA, Carson JL, Leigh MW, Wolf WE, Chen S, Lucas JS, Onoufriadis A, Plagnol V, Schmidts M, Boldt K, UK10K, Roepman R, Zariwala M, Lo CW, Mitchison HM, Knowles MR, Burdine RD, LoTurco JJ, Omran H. DYX1C1 is required for axonemal dynein assembly and ciliary motility. Nat Genet 2013; 45:995-1003. [PMID: 23872636 PMCID: PMC4000444 DOI: 10.1038/ng.2707] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 06/24/2013] [Indexed: 11/08/2022]
Abstract
DYX1C1 has been associated with dyslexia and neuronal migration in the developing neocortex. Unexpectedly, we found that deleting exons 2-4 of Dyx1c1 in mice caused a phenotype resembling primary ciliary dyskinesia (PCD), a disorder characterized by chronic airway disease, laterality defects and male infertility. This phenotype was confirmed independently in mice with a Dyx1c1 c.T2A start-codon mutation recovered from an N-ethyl-N-nitrosourea (ENU) mutagenesis screen. Morpholinos targeting dyx1c1 in zebrafish also caused laterality and ciliary motility defects. In humans, we identified recessive loss-of-function DYX1C1 mutations in 12 individuals with PCD. Ultrastructural and immunofluorescence analyses of DYX1C1-mutant motile cilia in mice and humans showed disruptions of outer and inner dynein arms (ODAs and IDAs, respectively). DYX1C1 localizes to the cytoplasm of respiratory epithelial cells, its interactome is enriched for molecular chaperones, and it interacts with the cytoplasmic ODA and IDA assembly factor DNAAF2 (KTU). Thus, we propose that DYX1C1 is a newly identified dynein axonemal assembly factor (DNAAF4).
Collapse
Affiliation(s)
- Aarti Tarkar
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-3156, USA
| | - Niki T. Loges
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | | | - Richard Francis
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - Gerard W. Dougherty
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Joel V. Tamayo
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Brett Shook
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-3156, USA
| | - Marie Cantino
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-3156, USA
| | - Daniel Schwartz
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-3156, USA
| | - Charlotte Jahnke
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Heike Olbrich
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Claudius Werner
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Johanna Raidt
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Petra Pennekamp
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Marouan Abouhamed
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Rim Hjeij
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| | - Gabriele Köhler
- Department of Pathology, University Hospital Muenster, 48149 Muenster, Germany
| | - Matthias Griese
- Dr. von Haunersches Children‘s Hospital, Ludwig Maximilian University, 80337 Munich, Germany
| | - You Li
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - Kristi Lemke
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - Nikolas Klena
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - Xiaoqin Liu
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - George Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - Kimimasa Tobita
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - Martine Jaspers
- University Hospital Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Lucy C. Morgan
- Department of Respiratory Medicine, Concord Hospital, Concord 2139, Australia
| | - Adam J. Shapiro
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC 27599, USA
| | - Stef J.F. Letteboer
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
- Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dorus A. Mans
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
- Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Johnny L. Carson
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC 27599, USA
| | - Margaret W. Leigh
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC 27599, USA
| | - Whitney E. Wolf
- Department of Medicine, UNC School of Medicine, Chapel Hill, NC 27599, USA
| | - Serafine Chen
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jane S. Lucas
- Primary Ciliary Dyskinesia Centre, NIHR Southampton Respiratory Biomedical Research Unit, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, SO17 1BJ, UK
| | - Alexandros Onoufriadis
- Molecular Medicine Unit, Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Vincent Plagnol
- University College London, Genetics Institute, London, WC1E 6BT, UK
| | - Miriam Schmidts
- Molecular Medicine Unit, Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Karsten Boldt
- Institute for Ophthalmic Research, Division of Experimental Ophthalmology and Medical Proteome Center, University of Tuebingen, D-72076 Tuebingen, Germany
| | | | - Ronald Roepman
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
- Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
- Institute for Genetic and Metabolic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Maimoona Zariwala
- Department of Pathology & Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC 27599, USA
| | - Cecilia W. Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201
| | - Hannah M. Mitchison
- Molecular Medicine Unit, Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Michael R. Knowles
- Department of Medicine, UNC School of Medicine, Chapel Hill, NC 27599, USA
| | - Rebecca D. Burdine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Joseph J. LoTurco
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-3156, USA
| | - Heymut Omran
- Department of Pediatrics, University Hospital Muenster, 48149 Muenster; Germany
| |
Collapse
|
46
|
Wang S, Dong Z. Primary cilia and kidney injury: current research status and future perspectives. Am J Physiol Renal Physiol 2013; 305:F1085-98. [PMID: 23904226 DOI: 10.1152/ajprenal.00399.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cilia, membrane-enclosed organelles protruding from the apical side of cells, can be divided into two classes: motile and primary cilia. During the past decades, motile cilia have been intensively studied. However, it was not until the 1990s that people began to realize the importance of primary cilia as cellular-specific sensors, particularly in kidney tubular epithelial cells. Furthermore, accumulating evidence indicates that primary cilia may be involved in the regulation of cell proliferation, differentiation, apoptosis, and planar cell polarity. Many signaling pathways, such as Wnt, Notch, Hedgehog, and mammalian target of rapamycin, have been located to the primary cilia. Thus primary cilia have been regarded as a hub that integrates signals from the extracellular environment. More importantly, dysfunction of this organelle may contribute to the pathogenesis of a large spectrum of human genetic diseases, named ciliopathies. The significance of primary cilia in acquired human diseases such as hypertension and diabetes has gradually drawn attention. Interestingly, recent reports disclosed that cilia length varies during kidney injury, and shortening of cilia enhances the sensitivity of epithelial cells to injury cues. This review briefly summarizes the current status of cilia research and explores the potential mechanisms of cilia-length changes during kidney injury as well as provides some thoughts to allure more insightful ideas and promotes the further study of primary cilia in the context of kidney injury.
Collapse
Affiliation(s)
- Shixuan Wang
- Dept. of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA 30912.
| | | |
Collapse
|