51
|
Yakar O, Tatar A. INTU-related oral-facial-digital syndrome XVII: Clinical spectrum of a rare disorder. Am J Med Genet A 2021; 188:590-594. [PMID: 34623732 DOI: 10.1002/ajmg.a.62527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/28/2021] [Accepted: 09/15/2021] [Indexed: 12/18/2022]
Abstract
Oral-facial-digital syndromes (OFDSs) as a subgroup of ciliopathies are rare genetic disorders characterized by the association of abnormalities of the face, oral cavity, and extremities. OFDS XVII is a recently described subtype of OFDS that presents with developmental delay, facial dysmorphism, high palate, tongue nodules, brain malformations, cardiac anomaly, polydactyly, renal malformation, and various other findings. OFDS XVII is caused by biallelic variants in INTU gene and is inherited autosomal recessively. Intu is part of the CPLANE protein module that has an essential role in the ciliary transport system and function. INTU pathogenic variants have been reported in two patients with OFDS XVII, in two patients with short-rib thoracic dysplasia-20 with polydactyly (SRTD20), and one with nephronophthisis so far. We report the third family in the literature with OFDS XVII, with urogenital malformations as an additional finding.
Collapse
Affiliation(s)
- Omer Yakar
- Department of Medical Genetics, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Abdulgani Tatar
- Department of Medical Genetics, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
52
|
Focșa IO, Budișteanu M, Bălgrădean M. Clinical and genetic heterogeneity of primary ciliopathies (Review). Int J Mol Med 2021; 48:176. [PMID: 34278440 PMCID: PMC8354309 DOI: 10.3892/ijmm.2021.5009] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/28/2021] [Indexed: 01/11/2023] Open
Abstract
Ciliopathies comprise a group of complex disorders, with involvement of the majority of organs and systems. In total, >180 causal genes have been identified and, in addition to Mendelian inheritance, oligogenicity, genetic modifications, epistatic interactions and retrotransposon insertions have all been described when defining the ciliopathic phenotype. It is remarkable how the structural and functional impairment of a single, minuscule organelle may lead to the pathogenesis of highly pleiotropic diseases. Thus, combined efforts have been made to identify the genetic substratum and to determine the pathophysiological mechanism underlying the clinical presentation, in order to diagnose and classify ciliopathies. Yet, predicting the phenotype, given the intricacy of the genetic cause and overlapping clinical characteristics, represents a major challenge. In the future, advances in proteomics, cell biology and model organisms may provide new insights that could remodel the field of ciliopathies.
Collapse
Affiliation(s)
- Ina Ofelia Focșa
- Department of Medical Genetics, University of Medicine and Pharmacy 'Carol Davila', 021901 Bucharest, Romania
| | - Magdalena Budișteanu
- Department of Pediatric Neurology, 'Prof. Dr. Alexandru Obregia' Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Mihaela Bălgrădean
- Department of Pediatrics and Pediatric Nephrology, Emergency Clinical Hospital for Children 'Maria Skłodowska Curie', 077120 Bucharest, Romania
| |
Collapse
|
53
|
Ningappa M, Adenuga M, Ngo KA, Mohamed N, Narayanan T, Prasadan K, Ashokkumar C, Das J, Schmitt L, Hartman H, Sehrawat A, Salgado CM, Reyes-Mugica M, Gittes GK, Lo CW, Subramaniam S, Sindhi R. Mechanisms of Impaired Lung Development and Ciliation in Mannosidase-1-Alpha-2 ( Man1a2) Mutants. Front Physiol 2021; 12:658518. [PMID: 34366878 PMCID: PMC8343402 DOI: 10.3389/fphys.2021.658518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/03/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Ciliary defects cause heterogenous phenotypes related to mutation burden which lead to impaired development. A previously reported homozygous deletion in the Man1a2 gene causes lethal respiratory failure in newborn pups and decreased lung ciliation compared with wild type (WT) pups. The effects of heterozygous mutation, and the potential for rescue are not known. PURPOSE We hypothesized that survival and lung ciliation, (a) would decrease progressively in Man1a2 +/- heterozygous and Man1a2 -/- null newborn pups compared with WT, and (b) could be enhanced by gestational treatment with N-Acetyl-cysteine (NAC), an antioxidant. METHODS Man1a2+/- adult mice were fed NAC or placebo from a week before breeding through gestation. Survival of newborn pups was monitored for 24 h. Lungs, liver and tails were harvested for morphology, genotyping, and transcriptional profiling. RESULTS Survival (p = 0.0001, Kaplan-Meier) and percent lung ciliation (p = 0.0001, ANOVA) measured by frequency of Arl13b+ respiratory epithelial cells decreased progressively, as hypothesized. Compared with placebo, gestational NAC treatment enhanced (a) lung ciliation in pups with each genotype, (b) survival in heterozygous pups (p = 0.017) but not in WT or null pups. Whole transcriptome of lung but not liver demonstrated patterns of up- and down-regulated genes that were identical in living heterozygous and WT pups, and completely opposite to those in dead heterozygous and null pups. Systems biology analysis enabled reconstruction of protein interaction networks that yielded functionally relevant modules and their interactions. In these networks, the mutant Man1a2 enzyme contributes to abnormal synthesis of proteins essential for lung development. The associated unfolded protein, hypoxic and oxidative stress responses can be mitigated with NAC. Comparisons with the developing human fetal lung transcriptome show that NAC likely restores normal vascular and epithelial tube morphogenesis in Man1a2 mutant mice. CONCLUSION Survival and lung ciliation in the Man1a2 mutant mouse, and its improvement with N-Acetyl cysteine is genotype-dependent. NAC-mediated rescue depends on the central role for oxidative and hypoxic stress in regulating ciliary function and organogenesis during development.
Collapse
Affiliation(s)
- Mylarappa Ningappa
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Morayooluwa Adenuga
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Kim A. Ngo
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
| | - Nada Mohamed
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Tejaswini Narayanan
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
| | - Krishna Prasadan
- Rangos Research Center Animal Imaging Core, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Chethan Ashokkumar
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Jishnu Das
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
- Departments of Immunology and Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lori Schmitt
- Histology Core Laboratory Manager, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Hannah Hartman
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Anuradha Sehrawat
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Claudia M. Salgado
- Division of Pediatric Pathology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Miguel Reyes-Mugica
- Division of Pediatric Pathology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - George K. Gittes
- Surgeon-in-Chief Emeritus, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Cecilia W. Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, La Jolla, CA, United States
- Department of Computer Science and Engineering, and Nanoengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
| | - Rakesh Sindhi
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| |
Collapse
|
54
|
Langhans MT, Gao J, Tang Y, Wang B, Alexander P, Tuan RS. Wdpcp regulates cellular proliferation and differentiation in the developing limb via hedgehog signaling. BMC DEVELOPMENTAL BIOLOGY 2021; 21:10. [PMID: 34225660 PMCID: PMC8258940 DOI: 10.1186/s12861-021-00241-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/07/2021] [Indexed: 12/27/2022]
Abstract
Background Mice with a loss of function mutation in Wdpcp were described previously to display severe birth defects in the developing heart, neural tube, and limb buds. Further characterization of the skeletal phenotype of Wdpcp null mice was limited by perinatal lethality. Results We utilized Prx1-Cre mice to generate limb bud mesenchyme specific deletion of Wdpcp. These mice recapitulated the appendicular skeletal phenotype of the Wdpcp null mice including polydactyl and limb bud signaling defects. Examination of late stages of limb development demonstrated decreased size of cartilage anlagen, delayed calcification, and abnormal growth plates. Utilizing in vitro assays, we demonstrated that loss of Wdpcp in skeletal progenitors lead to loss of hedgehog signaling responsiveness and associated proliferative response. In vitro chondrogenesis assays showed this loss of hedgehog and proliferative response was associated with decreased expression of early chondrogenic marker N-Cadherin. E14.5 forelimbs demonstrated delayed ossification and expression of osteoblast markers Runx2 and Sp7. P0 growth plates demonstrated loss of hedgehog signaling markers and expansion of the hypertrophic zones of the growth plate. In vitro osteogenesis assays demonstrated decreased osteogenic differentiation of Wdpcp null mesenchymal progenitors in response to hedgehog stimulation. Conclusions These findings demonstrate how Wdpcp and associated regulation of the hedgehog signaling pathway plays an important role at multiple stages of skeletal development. Wdpcp is necessary for positive regulation of hedgehog signaling and associated proliferation is key to the initiation of chondrogenesis. At later stages, Wdpcp facilitates the robust hedgehog response necessary for chondrocyte hypertrophy and osteogenic differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s12861-021-00241-9.
Collapse
Affiliation(s)
- Mark T Langhans
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219-3143, USA
| | - Jingtao Gao
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219-3143, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219-3143, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219-3143, USA
| | - Peter Alexander
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219-3143, USA
| | - Rocky S Tuan
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA, 15219-3143, USA. .,Present Address: Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
55
|
Zhang C, Sun Z, Xu L, Che F, Liu S. Novel compound heterozygous CPLANE1 variants identified in a Chinese family with Joubert syndrome. Int J Dev Neurosci 2021; 81:529-538. [PMID: 34091942 DOI: 10.1002/jdn.10135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/30/2021] [Indexed: 11/08/2022] Open
Abstract
Joubert syndrome (JS) and JS-related disorders (JSRD) are a group of neurodevelopmental diseases that share the "molar tooth sign" on axial brain magnetic resonance imaging (MRI), accompanied by cerebellar vermis hypoplasia, ataxia, hypotonia, and developmental delay. To identify variants responsible for the clinical symptoms of a Chinese family with JS and to explore the genotype-phenotype associations, we conducted a series of clinical examinations, including blood tests, brain MRI scans, ultrasound imaging, and ophthalmologic examination. Genomic DNA was extracted from the peripheral blood of the six-person family, and the pathogenic variants were detected by whole-exome sequencing (WES) and verified by Sanger sequencing. WES revealed two novel compound heterozygous variants in CPLANE1: c.1270C>T (p.Arg424*) in exon 10 and c.8901C>A (p.Tyr2967*) in exon 48 of one child, inherited from each parent. Both variants were absent in ethnically matched Chinese control individuals and were either absent or present at very low frequencies in public databases, suggesting that these variants could be the pathogenic triggers of the JS phenotype. Notably, these CPLANE1 sequence variants were related to the pathogenesis of autosomal recessive JS in this study. The newly discovered variants expand the mutation spectrum of CPLANE1, which assists in understanding the molecular mechanism underlying JS and improving the recognition of genetic counseling, particularly for families with a history of autosomal recessive JS.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, China.,Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhenchao Sun
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, China
| | - Lulu Xu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fengyuan Che
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, China
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
56
|
Torban E, Sokol SY. Planar cell polarity pathway in kidney development, function and disease. Nat Rev Nephrol 2021; 17:369-385. [PMID: 33547419 PMCID: PMC8967065 DOI: 10.1038/s41581-021-00395-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Planar cell polarity (PCP) refers to the coordinated orientation of cells in the tissue plane. Originally discovered and studied in Drosophila melanogaster, PCP is now widely recognized in vertebrates, where it is implicated in organogenesis. Specific sets of PCP genes have been identified. The proteins encoded by these genes become asymmetrically distributed to opposite sides of cells within a tissue plane and guide many processes that include changes in cell shape and polarity, collective cell movements or the uniform distribution of cell appendages. A unifying characteristic of these processes is that they often involve rearrangement of actomyosin. Mutations in PCP genes can cause malformations in organs of many animals, including humans. In the past decade, strong evidence has accumulated for a role of the PCP pathway in kidney development including outgrowth and branching morphogenesis of ureteric bud and podocyte development. Defective PCP signalling has been implicated in the pathogenesis of developmental kidney disorders of the congenital anomalies of the kidney and urinary tract spectrum. Understanding the origins, molecular constituents and cellular targets of PCP provides insights into the involvement of PCP molecules in normal kidney development and how dysfunction of PCP components may lead to kidney disease.
Collapse
Affiliation(s)
- Elena Torban
- McGill University and McGill University Health Center Research Institute, 1001 Boulevard Decarie, Block E, Montreal, Quebec, Canada, H4A3J1.,Corresponding authors: Elena Torban (); Sergei Sokol ()
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, USA,Corresponding authors: Elena Torban (); Sergei Sokol ()
| |
Collapse
|
57
|
Hammarsjö A, Pettersson M, Chitayat D, Handa A, Anderlid BM, Bartocci M, Basel D, Batkovskyte D, Beleza-Meireles A, Conner P, Eisfeldt J, Girisha KM, Chung BHY, Horemuzova E, Hyodo H, Korņejeva L, Lagerstedt-Robinson K, Lin AE, Magnusson M, Moosa S, Nayak SS, Nilsson D, Ohashi H, Ohashi-Fukuda N, Stranneheim H, Taylan F, Traberg R, Voss U, Wirta V, Nordgren A, Nishimura G, Lindstrand A, Grigelioniene G. High diagnostic yield in skeletal ciliopathies using massively parallel genome sequencing, structural variant screening and RNA analyses. J Hum Genet 2021; 66:995-1008. [PMID: 33875766 PMCID: PMC8472897 DOI: 10.1038/s10038-021-00925-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022]
Abstract
Skeletal ciliopathies are a heterogenous group of disorders with overlapping clinical and radiographic features including bone dysplasia and internal abnormalities. To date, pathogenic variants in at least 30 genes, coding for different structural cilia proteins, are reported to cause skeletal ciliopathies. Here, we summarize genetic and phenotypic features of 34 affected individuals from 29 families with skeletal ciliopathies. Molecular diagnostic testing was performed using massively parallel sequencing (MPS) in combination with copy number variant (CNV) analyses and in silico filtering for variants in known skeletal ciliopathy genes. We identified biallelic disease-causing variants in seven genes: DYNC2H1, KIAA0753, WDR19, C2CD3, TTC21B, EVC, and EVC2. Four variants located in non-canonical splice sites of DYNC2H1, EVC, and KIAA0753 led to aberrant splicing that was shown by sequencing of cDNA. Furthermore, CNV analyses showed an intragenic deletion of DYNC2H1 in one individual and a 6.7 Mb de novo deletion on chromosome 1q24q25 in another. In five unsolved cases, MPS was performed in family setting. In one proband we identified a de novo variant in PRKACA and in another we found a homozygous intragenic deletion of IFT74, removing the first coding exon and leading to expression of a shorter message predicted to result in loss of 40 amino acids at the N-terminus. These findings establish IFT74 as a new skeletal ciliopathy gene. In conclusion, combined single nucleotide variant, CNV and cDNA analyses lead to a high yield of genetic diagnoses (90%) in a cohort of patients with skeletal ciliopathies.
Collapse
Affiliation(s)
- Anna Hammarsjö
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden.
| | - Maria Pettersson
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - David Chitayat
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, and Mt. Sinai Hospital, Toronto, ON, Canada.,The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Atsuhiko Handa
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Britt-Marie Anderlid
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Bartocci
- Department of Women's and Children's Health, Neonatology, Karolinska Institutet, Stockholm, Sweden
| | - Donald Basel
- Division of Medical Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Dominyka Batkovskyte
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ana Beleza-Meireles
- Department of Clinical Genetics, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Peter Conner
- Department of Women's and Children's Health, Karolinska Institutet and Center for Fetal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jesper Eisfeldt
- Science for Life Laboratory, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Katta M Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, India
| | - Brian Hon-Yin Chung
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong and Shenzhen Hospital, Futian District, Shenzhen, China.,Department of Pediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Eva Horemuzova
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Women's and Children's Health, Karolinska Institutet and Paediatric Endocrinology Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Hironobu Hyodo
- Department of Obstetrics and Gynecology, Tokyo Metropolitan Bokutoh Hospital, Kotobashi, Sumida-ku, Tokyo, Japan
| | - Liene Korņejeva
- Department of Prenatal Diagnostics, Riga Maternity Hospital, Riga, Latvia
| | - Kristina Lagerstedt-Robinson
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Angela E Lin
- Medical Genetics, MassGeneral Hospital for Children, Boston, MA, USA
| | - Måns Magnusson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Centre for Inherited Metabolic Diseases, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden.,Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Shahida Moosa
- Medical Genetics, Tygerberg Hospital and Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Shalini S Nayak
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, India
| | - Daniel Nilsson
- Science for Life Laboratory, Department of Molecular Medicine and Surgery, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Hirofumi Ohashi
- Division of Medical Genetics, Saitama Children's Medical Center, Saitama, Japan
| | - Naoko Ohashi-Fukuda
- Department of Obstetrics and Gynecology, Tokyo Metropolitan Bokutoh Hospital, Kotobashi, Sumida-ku, Tokyo, Japan
| | - Henrik Stranneheim
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Centre for Inherited Metabolic Diseases, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Fulya Taylan
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Rasa Traberg
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ulrika Voss
- Department of Pediatric Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Valtteri Wirta
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Ann Nordgren
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Gen Nishimura
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Imaging, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Giedre Grigelioniene
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, and Department of Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
58
|
Hibbard JVK, Vazquez N, Satija R, Wallingford JB. Protein turnover dynamics suggest a diffusion-to-capture mechanism for peri-basal body recruitment and retention of intraflagellar transport proteins. Mol Biol Cell 2021; 32:1171-1180. [PMID: 33826363 PMCID: PMC8351562 DOI: 10.1091/mbc.e20-11-0717] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Intraflagellar transport (IFT) is essential for construction and maintenance of cilia. IFT proteins concentrate at the basal body where they are thought to assemble into trains and bind cargoes for transport. To study the mechanisms of IFT recruitment to this peri-basal body pool, we quantified protein dynamics of eight IFT proteins, as well as five other basal body localizing proteins using fluorescence recovery after photobleaching in vertebrate multiciliated cells. We found that members of the IFT-A and IFT-B protein complexes show distinct turnover kinetics from other basal body components. Additionally, known IFT subcomplexes displayed shared dynamics, suggesting shared basal body recruitment and/or retention mechanisms. Finally, we evaluated the mechanisms of basal body recruitment by depolymerizing cytosolic MTs, which suggested that IFT proteins are recruited to basal bodies through a diffusion-to-capture mechanism. Our survey of IFT protein dynamics provides new insights into IFT recruitment to basal bodies, a crucial step in ciliogenesis and ciliary signaling.
Collapse
Affiliation(s)
- Jaime V K Hibbard
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Neftali Vazquez
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Rohit Satija
- California Institute of Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| |
Collapse
|
59
|
Zhang X, Shen Y, Li P, Cai R, Lu C, Li Q, Chen C, Yu Y, Cheng T, Wang X, Luo M, Cao M, Cao Z, Ma X. Clinical heterogeneity and intrafamilial variability of Joubert syndrome in two siblings with CPLANE1 variants. Mol Genet Genomic Med 2021; 9:e1682. [PMID: 33822487 PMCID: PMC8222854 DOI: 10.1002/mgg3.1682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 11/18/2022] Open
Abstract
Background Joubert syndrome (JBTS) is a rare genetic disorder that is characterized by midbrain‐hindbrain malformations. Multiple variants in genes that affect ciliary function contribute to the genetic and clinical heterogeneity of JBTS and its subtypes. However, the correlation between genotype and phenotype has not been elucidated due to the limited number of patients available. Methods In this study, we observed different clinical features in two siblings from the same family. The older sibling was classified as a pure JBTS patient, whereas her younger sibling displayed oral‐facial‐digital defects and was therefore classified as an oral‐facial‐digital syndrome type VI (OFD VI) patient. Next, we performed human genetic tests to identify the potential pathogenic variants in the two siblings. Results Genetic sequencing indicated that both siblings harbored compound heterozygous variants of a missense variant (c.1067C>T, p.S356F) and a frameshift variant (c.8377_8378del, p.E2793Lfs*24) in CPLANE1 (NM_023073.3). Conclusion This study reports that two novel CPLANE1 variants are associated with the occurrence of JBTS and OFD VI. These results help elucidate the intrafamilial phenotypic variability associated with CPLANE1 variants.
Collapse
Affiliation(s)
- Xiujuan Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Shen
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Ping Li
- Department of Developmental Pediatrics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ruikun Cai
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Chao Lu
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Qian Li
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Cuixia Chen
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Yufei Yu
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Tingting Cheng
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Minna Luo
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zongfu Cao
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Xu Ma
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| |
Collapse
|
60
|
Drivas TG, Lucas A, Zhang X, Ritchie MD. Mendelian pathway analysis of laboratory traits reveals distinct roles for ciliary subcompartments in common disease pathogenesis. Am J Hum Genet 2021; 108:482-501. [PMID: 33636100 PMCID: PMC8008498 DOI: 10.1016/j.ajhg.2021.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/05/2021] [Indexed: 12/17/2022] Open
Abstract
Rare monogenic disorders of the primary cilium, termed ciliopathies, are characterized by extreme presentations of otherwise common diseases, such as diabetes, hepatic fibrosis, and kidney failure. However, despite a recent revolution in our understanding of the cilium's role in rare disease pathogenesis, the organelle's contribution to common disease remains largely unknown. Hypothesizing that common genetic variants within Mendelian ciliopathy genes might contribute to common complex diseases pathogenesis, we performed association studies of 16,874 common genetic variants across 122 ciliary genes with 12 quantitative laboratory traits characteristic of ciliopathy syndromes in 452,593 individuals in the UK Biobank. We incorporated tissue-specific gene expression analysis, expression quantitative trait loci, and Mendelian disease phenotype information into our analysis and replicated our findings in meta-analysis. 101 statistically significant associations were identified across 42 of the 122 examined ciliary genes (including eight novel replicating associations). These ciliary genes were widely expressed in tissues relevant to the phenotypes being studied, and eQTL analysis revealed strong evidence for correlation between ciliary gene expression levels and laboratory traits. Perhaps most interestingly, our analysis identified different ciliary subcompartments as being specifically associated with distinct sets of phenotypes. Taken together, our data demonstrate the utility of a Mendelian pathway-based approach to genomic association studies, challenge the widely held belief that the cilium is an organelle important mainly in development and in rare syndromic disease pathogenesis, and provide a framework for the continued integration of common and rare disease genetics to provide insight into the pathophysiology of human diseases of immense public health burden.
Collapse
Affiliation(s)
- Theodore George Drivas
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19194, USA; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Anastasia Lucas
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19194, USA
| | - Xinyuan Zhang
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19194, USA
| | - Marylyn DeRiggi Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19194, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19194, USA.
| |
Collapse
|
61
|
Abstract
Ciliogenesis describes the assembly of cilia in interphase cells. Several hundred proteins have been linked to ciliogenesis, which proceeds through a highly coordinated multistage process at the distal end of centrioles requiring membranes. In this short review, we focus on recently reported insights into the biogenesis of the primary cilium membrane and its association with other ciliogenic processes in the intracellular ciliogenesis pathway.
Collapse
Affiliation(s)
- Saurabh Shakya
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Laboratory of Cellular and Developmental Signaling, Frederick, MD 21702, USA
| | - Christopher J Westlake
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Laboratory of Cellular and Developmental Signaling, Frederick, MD 21702, USA
| |
Collapse
|
62
|
Rao VG, Kulkarni SS. Xenopus to the rescue: A model to validate and characterize candidate ciliopathy genes. Genesis 2021; 59:e23414. [PMID: 33576572 DOI: 10.1002/dvg.23414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022]
Abstract
Cilia are present on most vertebrate cells and play a central role in development, growth, and homeostasis. Thus, cilia dysfunction can manifest into an array of diseases, collectively termed ciliopathies, affecting millions of lives worldwide. Yet, our understanding of the gene regulatory networks that control cilia assembly and functions remain incomplete. With the advances in next-generation sequencing technologies, we can now rapidly predict pathogenic variants from hundreds of ciliopathy patients. While the pace of candidate gene discovery is exciting, most of these genes have never been previously implicated in cilia assembly or function. This makes assigning the disease causality difficult. This review discusses how Xenopus, a genetically tractable and high-throughput vertebrate model, has played a central role in identifying, validating, and characterizing candidate ciliopathy genes. The review is focused on multiciliated cells (MCCs) and diseases associated with MCC dysfunction. MCCs harbor multiple motile cilia on their apical surface to generate extracellular fluid flow inside the airway, the brain ventricles, and the oviduct. In Xenopus, these cells are external and present on the embryonic epidermal epithelia, facilitating candidate genes analysis in MCC development in vivo. The ability to introduce patient variants to study their effects on disease progression makes Xenopus a powerful model to improve our understanding of the underlying disease mechanisms and explain the patient phenotype.
Collapse
Affiliation(s)
- Venkatramanan G Rao
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Saurabh S Kulkarni
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA.,Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
63
|
Latour BL, Van De Weghe JC, Rusterholz TD, Letteboer SJ, Gomez A, Shaheen R, Gesemann M, Karamzade A, Asadollahi M, Barroso-Gil M, Chitre M, Grout ME, van Reeuwijk J, van Beersum SE, Miller CV, Dempsey JC, Morsy H, Bamshad MJ, Nickerson DA, Neuhauss SC, Boldt K, Ueffing M, Keramatipour M, Sayer JA, Alkuraya FS, Bachmann-Gagescu R, Roepman R, Doherty D. Dysfunction of the ciliary ARMC9/TOGARAM1 protein module causes Joubert syndrome. J Clin Invest 2021; 130:4423-4439. [PMID: 32453716 DOI: 10.1172/jci131656] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Joubert syndrome (JBTS) is a recessive neurodevelopmental ciliopathy characterized by a pathognomonic hindbrain malformation. All known JBTS genes encode proteins involved in the structure or function of primary cilia, ubiquitous antenna-like organelles essential for cellular signal transduction. Here, we used the recently identified JBTS-associated protein armadillo repeat motif-containing 9 (ARMC9) in tandem-affinity purification and yeast 2-hybrid screens to identify a ciliary module whose dysfunction underlies JBTS. In addition to the known JBTS-associated proteins CEP104 and CSPP1, we identified coiled-coil domain containing 66 (CCDC66) and TOG array regulator of axonemal microtubules 1 (TOGARAM1) as ARMC9 interaction partners. We found that TOGARAM1 variants cause JBTS and disrupt TOGARAM1 interaction with ARMC9. Using a combination of protein interaction analyses, characterization of patient-derived fibroblasts, and analysis of CRISPR/Cas9-engineered zebrafish and hTERT-RPE1 cells, we demonstrated that dysfunction of ARMC9 or TOGARAM1 resulted in short cilia with decreased axonemal acetylation and polyglutamylation, but relatively intact transition zone function. Aberrant serum-induced ciliary resorption and cold-induced depolymerization in ARMC9 and TOGARAM1 patient cell lines suggest a role for this new JBTS-associated protein module in ciliary stability.
Collapse
Affiliation(s)
- Brooke L Latour
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Tamara Ds Rusterholz
- Institute of Medical Genetics, and.,Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Stef Jf Letteboer
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Arianna Gomez
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Ranad Shaheen
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Matthias Gesemann
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Arezou Karamzade
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Asadollahi
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Miguel Barroso-Gil
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Manali Chitre
- Department of Paediatric Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Megan E Grout
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jeroen van Reeuwijk
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sylvia Ec van Beersum
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Caitlin V Miller
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jennifer C Dempsey
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Heba Morsy
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, Washington, USA.,The University of Washington Center for Mendelian Genomics is detailed in Supplemental Acknowledgments.,University of Washington Center for Mendelian Genomics, Seattle, Washington, USA.,Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Deborah A Nickerson
- The University of Washington Center for Mendelian Genomics is detailed in Supplemental Acknowledgments.,University of Washington Center for Mendelian Genomics, Seattle, Washington, USA
| | - Stephan Cf Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Karsten Boldt
- Medical Proteome Center, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Marius Ueffing
- Medical Proteome Center, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Mohammad Keramatipour
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ruxandra Bachmann-Gagescu
- Institute of Medical Genetics, and.,Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Ronald Roepman
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Dan Doherty
- Department of Pediatrics, University of Washington, Seattle, Washington, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
64
|
Andreu-Cervera A, Catala M, Schneider-Maunoury S. Cilia, ciliopathies and hedgehog-related forebrain developmental disorders. Neurobiol Dis 2020; 150:105236. [PMID: 33383187 DOI: 10.1016/j.nbd.2020.105236] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Development of the forebrain critically depends on the Sonic Hedgehog (Shh) signaling pathway, as illustrated in humans by the frequent perturbation of this pathway in holoprosencephaly, a condition defined as a defect in the formation of midline structures of the forebrain and face. The Shh pathway requires functional primary cilia, microtubule-based organelles present on virtually every cell and acting as cellular antennae to receive and transduce diverse chemical, mechanical or light signals. The dysfunction of cilia in humans leads to inherited diseases called ciliopathies, which often affect many organs and show diverse manifestations including forebrain malformations for the most severe forms. The purpose of this review is to provide the reader with a framework to understand the developmental origin of the forebrain defects observed in severe ciliopathies with respect to perturbations of the Shh pathway. We propose that many of these defects can be interpreted as an imbalance in the ratio of activator to repressor forms of the Gli transcription factors, which are effectors of the Shh pathway. We also discuss the complexity of ciliopathies and their relationships with forebrain disorders such as holoprosencephaly or malformations of cortical development, and emphasize the need for a closer examination of forebrain defects in ciliopathies, not only through the lens of animal models but also taking advantage of the increasing potential of the research on human tissues and organoids.
Collapse
Affiliation(s)
- Abraham Andreu-Cervera
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS) UMR7622, Institut national pour la Santé et la Recherche Médicale (Inserm) U1156, Institut de Biologie Paris Seine - Laboratoire de Biologie du Développement (IBPS-LBD), 9 Quai Saint-Bernard, 75005 Paris, France; Instituto de Neurociencias, Universidad Miguel Hernández - CSIC, Campus de San Juan; Avda. Ramón y Cajal s/n, 03550 Alicante, Spain
| | - Martin Catala
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS) UMR7622, Institut national pour la Santé et la Recherche Médicale (Inserm) U1156, Institut de Biologie Paris Seine - Laboratoire de Biologie du Développement (IBPS-LBD), 9 Quai Saint-Bernard, 75005 Paris, France.
| | - Sylvie Schneider-Maunoury
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS) UMR7622, Institut national pour la Santé et la Recherche Médicale (Inserm) U1156, Institut de Biologie Paris Seine - Laboratoire de Biologie du Développement (IBPS-LBD), 9 Quai Saint-Bernard, 75005 Paris, France.
| |
Collapse
|
65
|
Serrano M, Ramón M, Calvo JH, Jiménez MÁ, Freire F, Vázquez JM, Arranz JJ. Genome-wide association studies for sperm traits in Assaf sheep breed. Animal 2020; 15:100065. [PMID: 33573944 DOI: 10.1016/j.animal.2020.100065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Sperm quality traits routinely collected by artificial insemination (AI) center for rams progeny test are related with the capacity to produce sperm doses for AI and, in more or less grade, with males' fertility. Low-quality ejaculates are unuseful to perform AI sperm doses, which suppose high economic loses for the AI center. Moreover, sperm quality traits have low heritability values which make traditional genetic selection little efficient to its improvement. In this work, a genome-wide association study (GWAS) was conducted by using sperm quality traits data and 50 K Affymetrix custom chip genotypes of 429 rams of Assaf breed from OVIGEN AI centre. Furthermore, 47 of these rams were also genotyped with the Illumina HD Ovine BeadChip, and therefore HD genotypes were imputed for all rams with phenotype data. Previous to the GWAS, a linear regression model was fitted including sperm traits as dependent variables; the flock of origin, date of sperm collection, and jump number as fixed effects; rams age at collection in months as covariate; and ram permanent effect as random. Pseudo-phenotypes obtained from this model were used as input for GWAS. Associations at the chromosome-wise level (FDR 10%) of 76 single-nucleotide polymorphisms (SNPs) in 4 chromosomes for ejaculate concentration (CON), 20 SNPs in 3 chromosomes for ejaculate volume (VOL), 32 SNPs in 1 chromosome for ejaculate number of spermatozoa (SPZ), and 23 SNPs for spermatozoa mass motility (MOT) in 17 chromosomes were found. Only SNPs associated with MOT overcame the genome-wide significance level. Some candidate genes for sperm traits variability were SLC9C1 (OAR1), TSN (OAR2), and FUT10 (OAR26) for MOT;. DOCK2, CPLANE1, SPEF2, and RAI14 (OAR16) for CON; SCAPER and PSMA4 (OAR18) for VOL; and PARM1 and LOC101110593 (OAR6) for SPZ. SNPs associated with sperm traits were not found to be correlated with milk production genetic variation; however, the high frequencies of some SNPs with negative effect over sperm traits found in animals at the top milk yield estimated breeding values (EBVs) ranking would allow to exert some selective presure to improve rams sperm performances. Effects and frequencies of some of the SNPs detected over sperm quality traits make these variants good candidates to be used in marker-assisted selection to improve sperm characteristics of Assaf rams and AI center efficiency to produce sperm doses.
Collapse
Affiliation(s)
- M Serrano
- Departamento de Mejora Genética Animal, INIA, 28040 Madrid, Spain.
| | - M Ramón
- IRIAF-CERSYRA, Valdepeñas 13300, Ciudad Real, Spain
| | - J H Calvo
- Unidad de Tecnología en Producción Animal, CITA, 59059 Zaragoza, Spain; ARAID, 50004 Zaragoza, Spain
| | - M Á Jiménez
- Departamento de Mejora Genética Animal, INIA, 28040 Madrid, Spain
| | - F Freire
- OVIGEN, Granja Florencia s/n, Ctra. Villalazán-Peleagonzalo, 49800 Toro, Zamora, Spain
| | - J M Vázquez
- OVIGEN, Granja Florencia s/n, Ctra. Villalazán-Peleagonzalo, 49800 Toro, Zamora, Spain
| | - J J Arranz
- Departamento de Producción Animal, Universidad de León, 24007 León, Spain
| |
Collapse
|
66
|
Brücker L, Kretschmer V, May-Simera HL. The entangled relationship between cilia and actin. Int J Biochem Cell Biol 2020; 129:105877. [PMID: 33166678 DOI: 10.1016/j.biocel.2020.105877] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Primary cilia are microtubule-based sensory cell organelles that are vital for tissue and organ development. They act as an antenna, receiving and transducing signals, enabling communication between cells. Defects in ciliogenesis result in severe genetic disorders collectively termed ciliopathies. In recent years, the importance of the direct and indirect involvement of actin regulators in ciliogenesis came into focus as it was shown that F-actin polymerisation impacts ciliation. The ciliary basal body was further identified as both a microtubule and actin organising centre. In the current review, we summarize recent studies on F-actin in and around primary cilia, focusing on different actin regulators and their effect on ciliogenesis, from the initial steps of basal body positioning and regulation of ciliary assembly and disassembly. Since primary cilia are also involved in several intracellular signalling pathways such as planar cell polarity (PCP), subsequently affecting actin rearrangements, the multiple effectors of this pathway are highlighted in more detail with a focus on the feedback loops connecting actin networks and cilia proteins. Finally, we elucidate the role of actin regulators in the development of ciliopathy symptoms and cancer.
Collapse
Affiliation(s)
- Lena Brücker
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany
| | - Viola Kretschmer
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany
| | - Helen Louise May-Simera
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany.
| |
Collapse
|
67
|
Bosakova M, Abraham SP, Nita A, Hruba E, Buchtova M, Taylor SP, Duran I, Martin J, Svozilova K, Barta T, Varecha M, Balek L, Kohoutek J, Radaszkiewicz T, Pusapati GV, Bryja V, Rush ET, Thiffault I, Nickerson DA, Bamshad MJ, University of Washington Center for Mendelian Genomics, Rohatgi R, Cohn DH, Krakow D, Krejci P. Mutations in GRK2 cause Jeune syndrome by impairing Hedgehog and canonical Wnt signaling. EMBO Mol Med 2020; 12:e11739. [PMID: 33200460 PMCID: PMC7645380 DOI: 10.15252/emmm.201911739] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
Mutations in genes affecting primary cilia cause ciliopathies, a diverse group of disorders often affecting skeletal development. This includes Jeune syndrome or asphyxiating thoracic dystrophy (ATD), an autosomal recessive skeletal disorder. Unraveling the responsible molecular pathology helps illuminate mechanisms responsible for functional primary cilia. We identified two families with ATD caused by loss-of-function mutations in the gene encoding adrenergic receptor kinase 1 (ADRBK1 or GRK2). GRK2 cells from an affected individual homozygous for the p.R158* mutation resulted in loss of GRK2, and disrupted chondrocyte growth and differentiation in the cartilage growth plate. GRK2 null cells displayed normal cilia morphology, yet loss of GRK2 compromised cilia-based signaling of Hedgehog (Hh) pathway. Canonical Wnt signaling was also impaired, manifested as a failure to respond to Wnt ligand due to impaired phosphorylation of the Wnt co-receptor LRP6. We have identified GRK2 as an essential regulator of skeletogenesis and demonstrate how both Hh and Wnt signaling mechanistically contribute to skeletal ciliopathies.
Collapse
Affiliation(s)
- Michaela Bosakova
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Sara P Abraham
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Alexandru Nita
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Eva Hruba
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Marcela Buchtova
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - S Paige Taylor
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Ivan Duran
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Jorge Martin
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Katerina Svozilova
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Tomas Barta
- Department of Histology and EmbryologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Miroslav Varecha
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Lukas Balek
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | | | - Tomasz Radaszkiewicz
- Institute of Experimental BiologyFaculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Ganesh V Pusapati
- Department of BiochemistryStanford UniversityPalo AltoCAUSA
- Department of MedicineStanford UniversityPalo AltoCAUSA
| | - Vitezslav Bryja
- Institute of Experimental BiologyFaculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Eric T Rush
- Children's Mercy Kansas City, Center for Pediatric Genomic MedicineKansas CityMOUSA
- Department of PediatricsUniversity of MissouriKansas CityMOUSA
| | - Isabelle Thiffault
- Children's Mercy Kansas City, Center for Pediatric Genomic MedicineKansas CityMOUSA
- Department of PediatricsUniversity of MissouriKansas CityMOUSA
| | | | - Michael J Bamshad
- Department of Genome SciencesUniversity of WashingtonSeattleWAUSA
- Department of PediatricsUniversity of WashingtonSeattleWAUSA
- Division of Genetic MedicineSeattle Children's HospitalSeattleWAUSA
| | | | - Rajat Rohatgi
- Department of BiochemistryStanford UniversityPalo AltoCAUSA
- Department of MedicineStanford UniversityPalo AltoCAUSA
| | - Daniel H Cohn
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Molecular Cell and Developmental BiologyUniversity of California at Los AngelesLos AngelesCAUSA
| | - Deborah Krakow
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Human GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Obstetrics and GynecologyDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Pavel Krejci
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| |
Collapse
|
68
|
Karunakaran KB, Chaparala S, Lo CW, Ganapathiraju MK. Cilia interactome with predicted protein-protein interactions reveals connections to Alzheimer's disease, aging and other neuropsychiatric processes. Sci Rep 2020; 10:15629. [PMID: 32973177 PMCID: PMC7515907 DOI: 10.1038/s41598-020-72024-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cilia are dynamic microtubule-based organelles present on the surface of many eukaryotic cell types and can be motile or non-motile primary cilia. Cilia defects underlie a growing list of human disorders, collectively called ciliopathies, with overlapping phenotypes such as developmental delays and cognitive and memory deficits. Consistent with this, cilia play an important role in brain development, particularly in neurogenesis and neuronal migration. These findings suggest that a deeper systems-level understanding of how ciliary proteins function together may provide new mechanistic insights into the molecular etiologies of nervous system defects. Towards this end, we performed a protein-protein interaction (PPI) network analysis of known intraflagellar transport, BBSome, transition zone, ciliary membrane and motile cilia proteins. Known PPIs of ciliary proteins were assembled from online databases. Novel PPIs were predicted for each ciliary protein using a computational method we developed, called High-precision PPI Prediction (HiPPIP) model. The resulting cilia "interactome" consists of 165 ciliary proteins, 1,011 known PPIs, and 765 novel PPIs. The cilia interactome revealed interconnections between ciliary proteins, and their relation to several pathways related to neuropsychiatric processes, and to drug targets. Approximately 184 genes in the cilia interactome are targeted by 548 currently approved drugs, of which 103 are used to treat various diseases of nervous system origin. Taken together, the cilia interactome presented here provides novel insights into the relationship between ciliary protein dysfunction and neuropsychiatric disorders, for e.g. interconnections of Alzheimer's disease, aging and cilia genes. These results provide the framework for the rational design of new therapeutic agents for treatment of ciliopathies and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India
| | - Srilakshmi Chaparala
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
- Health Sciences Library System, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cecilia W Lo
- Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madhavi K Ganapathiraju
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA.
- Intelligent Systems Program, School of Computing and Information, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
69
|
Kim Y, Kim SH. WD40-Repeat Proteins in Ciliopathies and Congenital Disorders of Endocrine System. Endocrinol Metab (Seoul) 2020; 35:494-506. [PMID: 32894826 PMCID: PMC7520596 DOI: 10.3803/enm.2020.302] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
WD40-repeat (WDR)-containing proteins constitute an evolutionarily conserved large protein family with a broad range of biological functions. In human proteome, WDR makes up one of the most abundant protein-protein interaction domains. Members of the WDR protein family play important roles in nearly all major cellular signalling pathways. Mutations of WDR proteins have been associated with various human pathologies including neurological disorders, cancer, obesity, ciliopathies and endocrine disorders. This review provides an updated overview of the biological functions of WDR proteins and their mutations found in congenital disorders. We also highlight the significant role of WDR proteins in ciliopathies and endocrine disorders. The new insights may help develop therapeutic approaches targeting WDR motifs.
Collapse
Affiliation(s)
- Yeonjoo Kim
- Cell Biology Research Centre, Molecular and Clinical Sciences Research Institute, St. George’s, University of London, London, UK
| | - Soo-Hyun Kim
- Cell Biology Research Centre, Molecular and Clinical Sciences Research Institute, St. George’s, University of London, London, UK
- Corresponding author: Soo-Hyun Kim Cell Biology Research Centre, Molecular and Clinical Sciences Research Institute, St. George’s, University of London, Cranmer Terrace, London SW17 0RE, UK Tel: +44-208-266-6198, E-mail:
| |
Collapse
|
70
|
Panigrahi I, Ahuja C, Chaudhry C. Indian child with novel variant in OFD1 gene. Am J Med Genet A 2020; 182:2236-2238. [PMID: 32677760 DOI: 10.1002/ajmg.a.61768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/18/2020] [Accepted: 06/19/2020] [Indexed: 11/08/2022]
Abstract
Orofaciodigital syndrome (OFD) can have variable phenotype and presents with oral anomalies, facial dysmorphism, and digital malformations like syndactyly, and polydactyly. Other presentations also include renal and cardiac defects, and central nervous system anomalies like hydrocephalus and cerebellar abnormalities. OFD1 is a X-linked dominant form of the syndrome presenting in females with mutations in CXorf5 or OFD1 gene. We describe a young child with sparse hairs, milia over face and absence of corpus callosum. Next generation sequencing showed frameshift pathogenic variant in the exon 13 of the OFD1 gene, consistent with diagnosis of OFD1.
Collapse
Affiliation(s)
- Inusha Panigrahi
- Genetic Metabolic Unit, Department of Pediatrics, APC, PGIMER, Chandigarh, India
| | | | - Chakshu Chaudhry
- Genetic Metabolic Unit, Department of Pediatrics, APC, PGIMER, Chandigarh, India
| |
Collapse
|
71
|
Oboli G, Chukwuma D, Fagbule O, Abe E, Adisa A. MOLECULAR GENETICS OF CLEFT LIP AND PALATE: A REVIEW. Ann Ib Postgrad Med 2020; 18:S16-S21. [PMID: 33071691 PMCID: PMC7513377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- G.O. Oboli
- College of Medicine, University of Ibadan, Ibadan
| | - D.I. Chukwuma
- Department of Periodontology and Community Dentistry, University College Hospital, Ibadan
| | - O.F. Fagbule
- Department of Periodontology and Community Dentistry, University College Hospital, Ibadan
| | - E.O. Abe
- Department of Oral Pathology and Oral Medicine, University College Hospital, Ibadan
| | - A.O. Adisa
- College of Medicine, University of Ibadan, Ibadan, Department of Oral Pathology and Oral Medicine, University College Hospital, Ibadan
| |
Collapse
|
72
|
Gabriel GC, Young CB, Lo CW. Role of cilia in the pathogenesis of congenital heart disease. Semin Cell Dev Biol 2020; 110:2-10. [PMID: 32418658 DOI: 10.1016/j.semcdb.2020.04.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/13/2022]
Abstract
An essential role for cilia in the pathogenesis of congenital heart disease (CHD) has emerged from findings of a large-scale mouse forward genetic screen. High throughput screening with fetal ultrasound imaging followed by whole exome sequencing analysis recovered a preponderance of cilia related genes and cilia transduced cell signaling genes among mutations identified to cause CHD. The perturbation of left-right patterning in CHD pathogenesis is suggested by the association of CHD with heterotaxy, but also by the finding of the co-occurrence of laterality defects with CHD in birth defect registries. Many of the cilia and cilia cell signaling genes recovered were found to be related to Hedgehog signaling. Studies in mice showed cilia transduced hedgehog signaling coordinates left-right patterning with heart looping and differentiation of the heart tube. Cilia transduced Shh signaling also regulates later events in heart development, including outflow tract septation and formation of the atrioventricular septum. More recent work has shown mutations in cilia related genes may also contribute to valve disease that largely manifest in adult life. Overall, these and other findings show cilia play an important role in CHD and also in more common valve diseases.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States
| | - Cullen B Young
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States.
| |
Collapse
|
73
|
A novel variant in C5ORF42 gene is associated with Joubert syndrome. Mol Biol Rep 2020; 47:4099-4103. [PMID: 32367316 DOI: 10.1007/s11033-020-05465-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/25/2020] [Indexed: 10/24/2022]
Abstract
Joubert syndrome (JS) disease is a clinically and genetically heterogeneous disorder with mutations in more than 35 genes involved in its pathogenicity. Molecular genetic methods including next generation sequencing (NGS) and Sanger sequencing are effective techniques used for identifying rare genetic variants that have a strong effect on disease pathogenesis. In this study, we tested a large pedigree with a history of several affected members with JS. At first the proband was sequenced by NGS technique then, confirmed by sanger sequencing method. After this, all available members of the pedigree were subjected to molecular analysis by sanger sequencing technique. The results of this study showed a novel variant in the C5ORF42 gene c.3080A > T: p. D1027V leading to a substitution of a valine for aspartic acid (D1027V) and may be associated with JS. This variant was present in proband compatible with autosomal recessive pattern. Also this variant was present in all parents (both father and mother) of affected individuals in a heterozygous state. It seems that mutations in C5ORF42 gene are associated with JS. However, the substantial mechanism requires further investigation.
Collapse
|
74
|
Corral-Serrano JC, Lamers IJC, van Reeuwijk J, Duijkers L, Hoogendoorn ADM, Yildirim A, Argyrou N, Ruigrok RAA, Letteboer SJF, Butcher R, van Essen MD, Sakami S, van Beersum SEC, Palczewski K, Cheetham ME, Liu Q, Boldt K, Wolfrum U, Ueffing M, Garanto A, Roepman R, Collin RWJ. PCARE and WASF3 regulate ciliary F-actin assembly that is required for the initiation of photoreceptor outer segment disk formation. Proc Natl Acad Sci U S A 2020; 117:9922-9931. [PMID: 32312818 PMCID: PMC7211956 DOI: 10.1073/pnas.1903125117] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The outer segments (OS) of rod and cone photoreceptor cells are specialized sensory cilia that contain hundreds of opsin-loaded stacked membrane disks that enable phototransduction. The biogenesis of these disks is initiated at the OS base, but the driving force has been debated. Here, we studied the function of the protein encoded by the photoreceptor-specific gene C2orf71, which is mutated in inherited retinal dystrophy (RP54). We demonstrate that C2orf71/PCARE (photoreceptor cilium actin regulator) can interact with the Arp2/3 complex activator WASF3, and efficiently recruits it to the primary cilium. Ectopic coexpression of PCARE and WASF3 in ciliated cells results in the remarkable expansion of the ciliary tip. This process was disrupted by small interfering RNA (siRNA)-based down-regulation of an actin regulator, by pharmacological inhibition of actin polymerization, and by the expression of PCARE harboring a retinal dystrophy-associated missense mutation. Using human retinal organoids and mouse retina, we observed that a similar actin dynamics-driven process is operational at the base of the photoreceptor OS where the PCARE module and actin colocalize, but which is abrogated in Pcare-/- mice. The observation that several proteins involved in retinal ciliopathies are translocated to these expansions renders it a potential common denominator in the pathomechanisms of these hereditary disorders. Together, our work suggests that PCARE is an actin-associated protein that interacts with WASF3 to regulate the actin-driven expansion of the ciliary membrane at the initiation of new outer segment disk formation.
Collapse
Affiliation(s)
- Julio C Corral-Serrano
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Ideke J C Lamers
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Jeroen van Reeuwijk
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Lonneke Duijkers
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Anita D M Hoogendoorn
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Adem Yildirim
- Institute of Molecular Physiology, Johannes Gutenberg University of Mainz, 55099 Mainz, Germany
| | - Nikoleta Argyrou
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Renate A A Ruigrok
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Stef J F Letteboer
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Rossano Butcher
- Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114
| | - Max D van Essen
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Sanae Sakami
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Sylvia E C van Beersum
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Krzysztof Palczewski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106;
| | - Michael E Cheetham
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Qin Liu
- Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114
| | - Karsten Boldt
- Center of Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University of Mainz, 55099 Mainz, Germany
| | - Marius Ueffing
- Center of Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
| | - Alejandro Garanto
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
- Donders Institute for Cognitive Neuroscience, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Ronald Roepman
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands;
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Rob W J Collin
- Department of Human Genetics, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
- Donders Institute for Cognitive Neuroscience, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
75
|
Sanchez-Pulido L, Ponting CP. Hexa-Longin domain scaffolds for inter-Rab signalling. Bioinformatics 2020; 36:990-993. [PMID: 31562761 PMCID: PMC7703760 DOI: 10.1093/bioinformatics/btz739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/26/2019] [Accepted: 09/25/2019] [Indexed: 11/13/2022] Open
Abstract
Summary CPLANE is a protein complex required for assembly and maintenance of primary cilia. It contains several proteins, such as INTU, FUZ, WDPCP, JBTS17 and RSG1 (REM2- and RAB-like small GTPase 1), whose genes are mutated in ciliopathies. Using two contrasting evolutionary analyses, coevolution-based contact prediction and sequence conservation, we first identified the INTU/FUZ heterodimer as a novel member of homologous HerMon (Hermansky-Pudlak syndrome and MON1-CCZ1) complexes. Subsequently, we identified homologous Longin domains that are triplicated in each of these six proteins (MON1A, CCZ1, HPS1, HPS4, INTU and FUZ). HerMon complexes are known to be Rab effectors and Rab GEFs (Guanine nucleotide Exchange Factors) that regulate vesicular trafficking. Consequently, INTU/FUZ, their homologous complex, is likely to act as a GEF during activation of Rab GTPases involved in ciliogenesis. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Luis Sanchez-Pulido
- Medical Research Council Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Chris P Ponting
- Medical Research Council Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
76
|
Yang H, Huang K. Dissecting the Vesicular Trafficking Function of IFT Subunits. Front Cell Dev Biol 2020; 7:352. [PMID: 32010685 PMCID: PMC6974671 DOI: 10.3389/fcell.2019.00352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/05/2019] [Indexed: 12/22/2022] Open
Abstract
Intraflagellar transport (IFT) was initially identified as a transport machine with multiple protein subunits, and it is essential for the assembly, disassembly, and maintenance of cilium/flagellum, which serves as the nexus of extracellular-to-intracellular signal integration. To date, in addition to its well-established and indispensable roles in ciliated cells, most IFT subunits have presented more general functions of vesicular trafficking in the non-ciliated cells. Thus, this review aims to summarize the recent progress on the vesicular trafficking functions of the IFT subunits and to highlight the issues that may arise in future research.
Collapse
Affiliation(s)
- Huihui Yang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,Institute of Hydrobiology, University of Chinese Academy of Sciences, Beijing, China
| | - Kaiyao Huang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
77
|
Martínez-Hernández R, Serrano-Somavilla A, Ramos-Leví A, Sampedro-Nuñez M, Lens-Pardo A, Muñoz De Nova JL, Triviño JC, González MU, Torné L, Casares-Arias J, Martín-Cófreces NB, Sánchez-Madrid F, Marazuela M. Integrated miRNA and mRNA expression profiling identifies novel targets and pathological mechanisms in autoimmune thyroid diseases. EBioMedicine 2019; 50:329-342. [PMID: 31735554 PMCID: PMC6921241 DOI: 10.1016/j.ebiom.2019.10.061] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/31/2019] [Accepted: 10/31/2019] [Indexed: 12/18/2022] Open
Abstract
Background The mechanisms underlying autoimmune thyroid disease (AITD) remain elusive. Identification of such mechanisms would reveal novel and/or better therapeutic targets. Here, we use integrated analysis of miRNAs and mRNAs expression profiling to identify potential therapeutic targets involved in the mechanisms underlying AITD. Methods miRNA and mRNA from twenty fresh-frozen thyroid tissues (15 from AITD patients and 5 from healthy controls) were subjected to next-generation sequencing. An anti-correlated method revealed potential pathways and disease targets, including proteins involved in the formation of primary cilia. Thus, we examined the distribution and length of primary cilia in thyroid tissues from AITD and controls using immunofluorescence and scanning electron microscopy, and parsed cilia formation in thyroid cell lines in response to inflammatory stimuli in the presence of miRNA mimics. Findings We found that the expression of miR-21-5p, miR-146b-3p, miR-5571-3p and miR-6503-3p was anti-correlated with Enolase 4 (ENO4), in-turned planar cell polarity protein (INTU), kinesin family member 27 (KIF27), parkin co-regulated (PACRG) and serine/threonine kinase 36 (STK36) genes. Functional classification of these miRNA/mRNAs revealed that their differential expression was associated with cilia organization. We demonstrated that the number and length of primary cilia in thyroid tissues was significantly lower in AITD than in control (frequency of follicular ciliated cells in controls = 67.54% vs a mean of 22.74% and 21.61% in HT and GD respectively p = 0.0001, by one-way ANOVA test). In addition, pro-inflammatory cytokines (IFNγ and TNFα) and specific miRNA mimics for the newly identified target genes affected cilia appearance in thyroid cell lines. Interpretation Integrated miRNA/gene expression analysis has identified abnormal ciliogenesis as a novel susceptibility pathway that is involved in the pathogenesis of AITD. These results reflect that ciliogenesis plays a relevant role in AITD, and opens research pathways to design therapeutic targets in AITD. Funding Instituto de Salud Carlos III, Comunidad de Madrid, Grupo Español de Tumores Neuroendocrinos y Endocrinos, Ministerio de Economía y Empresa and FEDER.
Collapse
Affiliation(s)
- Rebeca Martínez-Hernández
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/ Diego de León 62, 28006 Madrid, Spain
| | - Ana Serrano-Somavilla
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/ Diego de León 62, 28006 Madrid, Spain
| | - Ana Ramos-Leví
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/ Diego de León 62, 28006 Madrid, Spain
| | - Miguel Sampedro-Nuñez
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/ Diego de León 62, 28006 Madrid, Spain
| | - Alberto Lens-Pardo
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/ Diego de León 62, 28006 Madrid, Spain
| | - José Luis Muñoz De Nova
- Department of Surgery, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/ Diego de León 62, 28006 Madrid, Spain
| | | | - María Ujue González
- Instituto de Micro y Nanotecnología, IMN-CNM, CSIC (CEI UAM+CSIC), Tres Cantos, Spain
| | - Lorena Torné
- Instituto de Micro y Nanotecnología, IMN-CNM, CSIC (CEI UAM+CSIC), Tres Cantos, Spain
| | - Javier Casares-Arias
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científcas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Noa B Martín-Cófreces
- Department of Immunology, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Spain
| | - Francisco Sánchez-Madrid
- Department of Immunology, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Spain
| | - Mónica Marazuela
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/ Diego de León 62, 28006 Madrid, Spain.
| |
Collapse
|
78
|
Bachmann-Gagescu R, Dempsey JC, Bulgheroni S, Chen ML, D'Arrigo S, Glass IA, Heller T, Héon E, Hildebrandt F, Joshi N, Knutzen D, Kroes HY, Mack SH, Nuovo S, Parisi MA, Snow J, Summers AC, Symons JM, Zein WM, Boltshauser E, Sayer JA, Gunay-Aygun M, Valente EM, Doherty D. Healthcare recommendations for Joubert syndrome. Am J Med Genet A 2019; 182:229-249. [PMID: 31710777 DOI: 10.1002/ajmg.a.61399] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 12/19/2022]
Abstract
Joubert syndrome (JS) is a recessive neurodevelopmental disorder defined by a characteristic cerebellar and brainstem malformation recognizable on axial brain magnetic resonance imaging as the "Molar Tooth Sign". Although defined by the neurological features, JS is associated with clinical features affecting many other organ systems, particularly progressive involvement of the retina, kidney, and liver. JS is a rare condition; therefore, many affected individuals may not have easy access to subspecialty providers familiar with JS (e.g., geneticists, neurologists, developmental pediatricians, ophthalmologists, nephrologists, hepatologists, psychiatrists, therapists, and educators). Expert recommendations can enable practitioners of all types to provide quality care to individuals with JS and know when to refer for subspecialty care. This need will only increase as precision treatments targeting specific genetic causes of JS emerge. The goal of these recommendations is to provide a resource for general practitioners, subspecialists, and families to maximize the health of individuals with JS throughout the lifespan.
Collapse
Affiliation(s)
- Ruxandra Bachmann-Gagescu
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Jennifer C Dempsey
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Sara Bulgheroni
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maida L Chen
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington.,Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, Seattle, Washington
| | - Stefano D'Arrigo
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ian A Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Theo Heller
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Elise Héon
- Department of Surgery, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Ophthalmology and Vision Science, University of Toronto, Toronto, Ontario, Canada
| | - Friedhelm Hildebrandt
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts.,Division of Nephrology, Boston Children's Hospital, Boston, Massachusetts
| | - Nirmal Joshi
- Department of Anesthesia, Deaconess Hospital, Evansville, Indiana.,Anesthesia Dynamics, LLC, Evansville, Indiana
| | - Dana Knutzen
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas.,The Children's Hospital of San Antonio, San Antonio, Texas
| | - Hester Y Kroes
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stephen H Mack
- Joubert Syndrome and Related Disorders Foundation, Petaluma, California
| | - Sara Nuovo
- Neurogenetics Lab, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Melissa A Parisi
- Intellectual and Developmental Disabilities Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Joseph Snow
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Angela C Summers
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.,Department of Psychology, Fordham University, Bronx, New York
| | - Jordan M Symons
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington.,Division of Nephrology, Seattle Children's Hospital, Seattle, Washington
| | - Wadih M Zein
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Eugen Boltshauser
- Department of Pediatric Neurology (emeritus), Children's University Hospital, Zürich, Switzerland
| | - John A Sayer
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK.,Renal Services, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne, UK
| | - Meral Gunay-Aygun
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland.,Department of Pediatrics and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Enza Maria Valente
- Neurogenetics Lab, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Dan Doherty
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
79
|
Sasai N, Toriyama M, Kondo T. Hedgehog Signal and Genetic Disorders. Front Genet 2019; 10:1103. [PMID: 31781166 PMCID: PMC6856222 DOI: 10.3389/fgene.2019.01103] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
The hedgehog (Hh) family comprises sonic hedgehog (Shh), Indian hedgehog (Ihh), and desert hedgehog (Dhh), which are versatile signaling molecules involved in a wide spectrum of biological events including cell differentiation, proliferation, and survival; establishment of the vertebrate body plan; and aging. These molecules play critical roles from embryogenesis to adult stages; therefore, alterations such as abnormal expression or mutations of the genes involved and their downstream factors cause a variety of genetic disorders at different stages. The Hh family involves many signaling mediators and functions through complex mechanisms, and achieving a comprehensive understanding of the entire signaling system is challenging. This review discusses the signaling mediators of the Hh pathway and their functions at the cellular and organismal levels. We first focus on the roles of Hh signaling mediators in signal transduction at the cellular level and the networks formed by these factors. Then, we analyze the spatiotemporal pattern of expression of Hh pathway molecules in tissues and organs, and describe the phenotypes of mutant mice. Finally, we discuss the genetic disorders caused by malfunction of Hh signaling-related molecules in humans.
Collapse
Affiliation(s)
- Noriaki Sasai
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Michinori Toriyama
- Systems Neurobiology and Medicine, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
80
|
Gerondopoulos A, Strutt H, Stevenson NL, Sobajima T, Levine TP, Stephens DJ, Strutt D, Barr FA. Planar Cell Polarity Effector Proteins Inturned and Fuzzy Form a Rab23 GEF Complex. Curr Biol 2019; 29:3323-3330.e8. [PMID: 31564489 PMCID: PMC6864590 DOI: 10.1016/j.cub.2019.07.090] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 01/12/2023]
Abstract
A subset of Rab GTPases have been implicated in cilium formation in cultured mammalian cells [1-6]. Rab11 and Rab8, together with their GDP-GTP exchange factors (GEFs), TRAPP-II and Rabin8, promote recruitment of the ciliary vesicle to the mother centriole and its subsequent maturation, docking, and fusion with the cell surface [2-5]. Rab23 has been linked to cilium formation and membrane trafficking at mature cilia [1, 7, 8]; however, the identity of the GEF pathway activating Rab23, a member of the Rab7 subfamily of Rabs, remains unclear. Longin-domain-containing complexes have been shown to act as GEFs for Rab7 subfamily GTPases [9-12]. Here, we show that Inturned and Fuzzy, proteins previously implicated as planar cell polarity (PCP) effectors and in developmentally regulated cilium formation [13, 14], contain multiple longin domains characteristic of the Mon1-Ccz1 family of Rab7 GEFs and form a specific Rab23 GEF complex. In flies, loss of Rab23 function gave rise to defects in planar-polarized trichome formation consistent with this biochemical relationship. In cultured human and mouse cells, Inturned and Fuzzy localized to the basal body and proximal region of cilia, and cilium formation was compromised by depletion of either Inturned or Fuzzy. Cilium formation arrested after docking of the ciliary vesicle to the mother centriole but prior to axoneme elongation and fusion of the ciliary vesicle and plasma membrane. These findings extend the family of longin domain GEFs and define a molecular activity linking Rab23-regulated membrane traffic to cilia and planar cell polarity.
Collapse
Affiliation(s)
- Andreas Gerondopoulos
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Helen Strutt
- Department of Biomedical Science, University of Sheffield, Firth Court, Sheffield S10 2TN, UK
| | - Nicola L Stevenson
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Tomoaki Sobajima
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Tim P Levine
- Institute of Ophthalmology, University College London, 11-43 Bath St., London EC1V 9EL, UK
| | - David J Stephens
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - David Strutt
- Department of Biomedical Science, University of Sheffield, Firth Court, Sheffield S10 2TN, UK
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
81
|
Gabriel GC, Lo CW. Novel insights into the genetic landscape of congenital heart disease with systems genetics. PROGRESS IN PEDIATRIC CARDIOLOGY 2019; 54. [PMID: 34404969 DOI: 10.1016/j.ppedcard.2019.101128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We recently conducted a large-scale mouse mutagenesis screen and uncovered a central role for cilia in the pathogenesis of congenital heart disease (CHD). Though our screen was phenotype based, most of the genes recovered were cilia-related, including genes encoding proteins important for ciliogenesis, cilia-transduced cell signaling, and vesicular trafficking. Also unexpected, many of the cilia related genes recovered are known direct protein-protein interactors, even though each gene was recovered independently in unrelated mouse lines. These findings suggest a cilia-based protein-protein interactome network may provide the context for congenital heart disease pathogenesis. This could explain the incomplete penetrance and variable expressivity of human CHD, and account for its complex non-Mendelian etiology. Supporting these findings in mice, a preponderance of cilia and cilia related cell signaling genes were observed among de novo pathogenic variants identified in a CHD patient cohort. Further clinical relevance unfolded with the observation of a high prevalence of respiratory cilia dysfunction in CHD patients. This was associated with increased postsurgical respiratory complications. Together these findings highlight the importance of cilia in CHD pathogenesis and suggest possible clinical translation with instituting pulmonary therapy to improve outcome for CHD patients undergoing congenital cardiac surgeries.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States of America
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States of America
| |
Collapse
|
82
|
Fujita A, Higashijima T, Shirozu H, Masuda H, Sonoda M, Tohyama J, Kato M, Nakashima M, Tsurusaki Y, Mitsuhashi S, Mizuguchi T, Takata A, Miyatake S, Miyake N, Fukuda M, Kameyama S, Saitsu H, Matsumoto N. Pathogenic variants of DYNC2H1, KIAA0556, and PTPN11 associated with hypothalamic hamartoma. Neurology 2019; 93:e237-e251. [PMID: 31197031 DOI: 10.1212/wnl.0000000000007774] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 03/04/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Intensive genetic analysis was performed to reveal comprehensive molecular insights into hypothalamic hamartoma (HH). METHODS Thirty-eight individuals with HH were investigated by whole exome sequencing, target capture-based deep sequencing, or single nucleotide polymorphism (SNP) array using DNA extracted from blood leukocytes or HH samples. RESULTS We identified a germline variant of KIAA0556, which encodes a ciliary protein, and 2 somatic variants of PTPN11, which forms part of the RAS/mitogen-activated protein kinase (MAPK) pathway, as well as variants in known genes associated with HH. An SNP array identified (among 3 patients) one germline copy-neutral loss of heterozygosity (cnLOH) at 6p22.3-p21.31 and 2 somatic cnLOH; one at 11q12.2-q25 that included DYNC2H1, which encodes a ciliary motor protein, and the other at 17p13.3-p11.2. A germline heterozygous variant and an identical somatic variant of DYNC2H1 arising from cnLOH at 11q12.2-q25 were confirmed in one patient (whose HH tissue, therefore, contains biallelic variants of DYNC2H1). Furthermore, a combination of a germline and a somatic DYNC2H1 variant was detected in another patient. CONCLUSIONS Overall, our cohort identified germline/somatic alterations in 34% (13/38) of patients with HH. Disruption of the Shh signaling pathway associated with cilia or the RAS/MAPK pathway may lead to the development of HH.
Collapse
Affiliation(s)
- Atsushi Fujita
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takefumi Higashijima
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroshi Shirozu
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroshi Masuda
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Masaki Sonoda
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Jun Tohyama
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Mitsuhiro Kato
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Mitsuko Nakashima
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yoshinori Tsurusaki
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Satomi Mitsuhashi
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takeshi Mizuguchi
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Atsushi Takata
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Satoko Miyatake
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Noriko Miyake
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Masafumi Fukuda
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Shigeki Kameyama
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hirotomo Saitsu
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Naomichi Matsumoto
- From the Departments of Human Genetics (A.F., S. Mitsuhashi, T.M., A.T., S. Miyatake, N. Miyake, N. Matsumoto) and Neurosurgery (M.S.), Yokohama City University Graduate School of Medicine; Departments of Functional Neurosurgery (T.H., H. Shirozu, H.M., M.F., S.K.) and Child Neurology (J.T.), Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital Niigata, Japan; Department of Pediatrics and Neurology (M.S.), Wayne State University, Children's Hospital of Michigan, Detroit Medical Center; Department of Pediatrics (M.K.), Showa University School of Medicine, Tokyo; Department of Biochemistry (M.N., H. Saitsu), Hamamatsu University School of Medicine; and Clinical Research Institute (Y.T.), Kanagawa Children's Medical Center, Yokohama, Japan.
| |
Collapse
|
83
|
Hong H, Joo K, Park SM, Seo J, Kim MH, Shin E, Cheong HI, Lee JH, Kim J. Extraciliary roles of the ciliopathy protein JBTS17 in mitosis and neurogenesis. Ann Neurol 2019; 86:99-115. [PMID: 31004438 DOI: 10.1002/ana.25491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE JBTS17 is a major gene mutated in ciliopathies such as Joubert syndrome and oral-facial-digital syndrome type VI. Most patients with loss of function mutations in JBTS17 exhibit cerebellar vermis hypoplasia and brainstem malformation. However, some patients with JBTS17 mutations show microcephaly and abnormal gyration. We examined potential roles of JBTS17 in neurogenesis to understand the pathological mechanism of JBTS17-related cortical abnormalities. METHODS We examined subcellular localization and cell-cycle-dependent expression of JBTS17 proteins using anti-JBTS17 antibodies and JBTS17 expression vectors. We also performed knockdown experiments to determined roles of JBTS17 in human cells, and demonstrated mitotic functions of JBTS17 using immunostaining and live imaging. We examined the involvement of JBTS17 in cortical neurogenesis using a mouse in utero electroporation technique. RESULTS We found that JBTS17 localizes to the kinetochore and the level of JBTS17 is regulated by cell-cycle-dependent proteolysis. Depletion of JBTS17 disrupts chromosome alignment and spindle pole orientation, resulting in mitotic delay. JBTS17 interacts with LIS1 and influences LIS1 localization. Depletion of Jbts17 in the developing mouse cortex interferes with the mitotic progression of neural progenitors and the migration of postmitotic neurons. INTERPRETATION LIS1 is implicated in lissencephaly, but altered dosage of LIS1 has been also associated with microcephaly syndromes. Our results suggest that JBTS17 contributes to mitotic progression by interacting with LIS1, and abnormal mitosis is an underlying mechanism of the microcephaly phenotype in JBTS17-related ciliopathies. We propose that understanding extraciliary roles of ciliopathy proteins is important to elucidate pathological mechanisms underlying diverse ciliopathy phenotypes. ANN NEUROL 2019.
Collapse
Affiliation(s)
- Hyowon Hong
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon
| | - Kwangsic Joo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam
| | - Sang Min Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon
| | - Jimyung Seo
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon
| | - Min Hwan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon
| | - EunBie Shin
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon
| | - Hae Il Cheong
- Department Pediatrics, Seoul National University Children's Hospital, Seoul; and 5Research Coordination Center for Rare Disease, Seoul National University Hospital, Seoul, Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon
| | - Joon Kim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon
| |
Collapse
|
84
|
Ryan R, Failler M, Reilly ML, Garfa-Traore M, Delous M, Filhol E, Reboul T, Bole-Feysot C, Nitschké P, Baudouin V, Amselem S, Escudier E, Legendre M, Benmerah A, Saunier S. Functional characterization of tektin-1 in motile cilia and evidence for TEKT1 as a new candidate gene for motile ciliopathies. Hum Mol Genet 2019; 27:266-282. [PMID: 29121203 DOI: 10.1093/hmg/ddx396] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/31/2017] [Indexed: 02/06/2023] Open
Abstract
A child presenting with Mainzer-Saldino syndrome (MZSDS), characterized by renal, retinal and skeletal involvements, was also diagnosed with lung infections and airway ciliary dyskinesia. These manifestations suggested dysfunction of both primary and motile cilia, respectively. Targeted exome sequencing identified biallelic mutations in WDR19, encoding an IFT-A subunit previously associated with MZSDS-related chondrodysplasia, Jeune asphyxiating thoracic dysplasia and cranioectodermal dysplasia, linked to primary cilia dysfunction, and in TEKT1 which encodes tektin-1 an uncharacterized member of the tektin family, mutations of which may cause ciliary dyskinesia. Tektin-1 localizes at the centrosome in cycling cells, at basal bodies of both primary and motile cilia and to the axoneme of motile cilia in airway cells. The identified mutations impaired these localizations. In addition, airway cells from the affected individual showed severe motility defects without major ultrastructural changes. Knockdown of tekt1 in zebrafish resulted in phenotypes consistent with a function for tektin-1 in ciliary motility, which was confirmed by live imaging. Finally, experiments in the zebrafish also revealed a synergistic effect of tekt1 and wdr19. Altogether, our data show genetic interactions between WDR19 and TEKT1 likely contributing to the overall clinical phenotype observed in the affected individual and provide strong evidence for TEKT1 as a new candidate gene for primary ciliary dyskinesia.
Collapse
Affiliation(s)
- Rebecca Ryan
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| | - Marion Failler
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| | - Madeline Louise Reilly
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France.,Paris Diderot University, Paris, France
| | - Meriem Garfa-Traore
- Cell Imaging Platform, INSERM US24 Structure Fédérative de Recherche Necker, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Marion Delous
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| | - Emilie Filhol
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| | - Thérèse Reboul
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| | - Christine Bole-Feysot
- Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France.,Bioinformatics Core Facility, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| | - Patrick Nitschké
- Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France.,INSERM UMR-1163, Genomic Core Facility, 75015 Paris, France
| | | | - Serge Amselem
- UMR-S 933, INSERM, Université Pierre et Marie Curie - Paris 6, Paris, France.,Service de Génétique et Embryologie Médicales, Assistance Publique - Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Estelle Escudier
- UMR-S 933, INSERM, Université Pierre et Marie Curie - Paris 6, Paris, France.,Service de Génétique et Embryologie Médicales, Assistance Publique - Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Marie Legendre
- UMR-S 933, INSERM, Université Pierre et Marie Curie - Paris 6, Paris, France.,Service de Génétique et Embryologie Médicales, Assistance Publique - Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Alexandre Benmerah
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| | - Sophie Saunier
- INSERM UMR 1163, Laboratory of Hereditary Kidney Diseases, 75015 Paris, France.,Imagine Institute, Paris Descartes - Sorbonne Paris Cité University, 75015 Paris, France
| |
Collapse
|
85
|
Konjikusic MJ, Yeetong P, Boswell CW, Lee C, Roberson EC, Ittiwut R, Suphapeetiporn K, Ciruna B, Gurnett CA, Wallingford JB, Shotelersuk V, Gray RS. Mutations in Kinesin family member 6 reveal specific role in ependymal cell ciliogenesis and human neurological development. PLoS Genet 2018; 14:e1007817. [PMID: 30475797 PMCID: PMC6307780 DOI: 10.1371/journal.pgen.1007817] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 12/27/2018] [Accepted: 11/07/2018] [Indexed: 01/06/2023] Open
Abstract
Cerebrospinal fluid flow is crucial for neurodevelopment and homeostasis of the ventricular system of the brain, with localized flow being established by the polarized beating of the ependymal cell (EC) cilia. Here, we report a homozygous one base-pair deletion, c.1193delT (p.Leu398Glnfs*2), in the Kinesin Family Member 6 (KIF6) gene in a child displaying neurodevelopmental defects and intellectual disability. To test the pathogenicity of this novel human KIF6 mutation we engineered an analogous C-terminal truncating mutation in mouse. These mutant mice display severe, postnatal-onset hydrocephalus. We generated a Kif6-LacZ transgenic mouse strain and report expression specifically and uniquely within the ependymal cells (ECs) of the brain, without labeling other multiciliated mouse tissues. Analysis of Kif6 mutant mice with scanning electron microscopy (SEM) and immunofluorescence (IF) revealed specific defects in the formation of EC cilia, without obvious effect of cilia of other multiciliated tissues. Dilation of the ventricular system and defects in the formation of EC cilia were also observed in adult kif6 mutant zebrafish. Finally, we report Kif6-GFP localization at the axoneme and basal bodies of multi-ciliated cells (MCCs) of the mucociliary Xenopus epidermis. Overall, this work describes the first clinically-defined KIF6 homozygous null mutation in human and defines KIF6 as a conserved mediator of neurological development with a specific role for EC ciliogenesis in vertebrates. Cerebrospinal fluid flow is crucial for neurodevelopment and homeostasis of the ventricular system of the brain. Localized flows of cerebrospinal fluid throughout the ventricular system of the brain are established from the polarized beating of the ependymal cell (EC) cilia. Here, we identified a homozygous truncating mutation in KIF6 in a child displaying neurodevelopmental defects and intellectual disability. To test the function of KIF6 in vivo, we engineered mutations of Kif6 in mouse. These Kif6 mutant mice display severe hydrocephalus, coupled with defects in the formation of EC cilia. Similarly, we observed hydrocephalus and a reduction in EC cilia in kif6 mutant zebrafish. Overall, this work describes the first clinically-defined KIF6 mutation in human, while our animal studies demonstrate the pathogenicity of mutations in KIF6 and establish KIF6 as a conserved mediator of ciliogenesis in ECs in vertebrates.
Collapse
Affiliation(s)
- Mia J. Konjikusic
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, Texas, United States of America
- Department of Molecular Biosciences, Patterson Labs, The University of Texas at Austin, Austin, Texas, United States of America
| | - Patra Yeetong
- Center of Excellence for Medical Genetics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Medical Genetics, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
- Division of Human Genetics, Department of Botany, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Curtis W. Boswell
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Molecular Genetics, The University of Toronto, Toronto, Ontario, Canada
| | - Chanjae Lee
- Department of Molecular Biosciences, Patterson Labs, The University of Texas at Austin, Austin, Texas, United States of America
| | - Elle C. Roberson
- Department of Molecular Biosciences, Patterson Labs, The University of Texas at Austin, Austin, Texas, United States of America
| | - Rungnapa Ittiwut
- Center of Excellence for Medical Genetics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Medical Genetics, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Kanya Suphapeetiporn
- Center of Excellence for Medical Genetics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Medical Genetics, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Brian Ciruna
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Molecular Genetics, The University of Toronto, Toronto, Ontario, Canada
| | - Christina A. Gurnett
- Department of Neurology, Division Pediatric Neurology, Washington University School of Medicine, St Louis, MO, United States of America
| | - John B. Wallingford
- Department of Molecular Biosciences, Patterson Labs, The University of Texas at Austin, Austin, Texas, United States of America
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genetics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Medical Genetics, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
- * E-mail: (VS); (RSG)
| | - Ryan S. Gray
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, Texas, United States of America
- * E-mail: (VS); (RSG)
| |
Collapse
|
86
|
Hua K, Ferland RJ. Primary Cilia Reconsidered in the Context of Ciliopathies: Extraciliary and Ciliary Functions of Cilia Proteins Converge on a Polarity theme? Bioessays 2018; 40:e1700132. [PMID: 29882973 PMCID: PMC6239423 DOI: 10.1002/bies.201700132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Once dismissed as vestigial organelles, primary cilia have garnered the interest of scientists, given their importance in development/signaling, and for their implication in a new disease category known as ciliopathies. However, many, if not all, "cilia" proteins also have locations/functions outside of the primary cilium. These extraciliary functions can complicate the interpretation of a particular ciliopathy phenotype: it may be a result of defects at the cilium and/or at extraciliary locations, and it could be broadly related to a unifying cellular process for these proteins, such as polarity. Assembly of a cilium has many similarities to the development of other polarized structures. This evolutionarily preserved process for the assembly of polarized cell structures offers a perspective on how the cilium may have evolved. We hypothesize that cilia proteins are critical for cell polarity, and that core polarity proteins may have been specialized to form various cellular protrusions, including primary cilia.
Collapse
Affiliation(s)
- Kiet Hua
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
- Department of Neurology, Albany Medical College, Albany, New York, USA, 12208
| |
Collapse
|
87
|
Abstract
Although tumours initiate from oncogenic changes in a cancer cell, subsequent tumour progression and therapeutic response depend on interactions between the cancer cells and the tumour microenvironment (TME). The primary monocilium, or cilium, provides a spatially localized platform for signalling by Hedgehog, Notch, WNT and some receptor tyrosine kinase pathways and mechanosensation. Changes in ciliation of cancer cells and/or cells of the TME during tumour development enforce asymmetric intercellular signalling in the TME. Growing evidence indicates that some oncogenic signalling pathways as well as some targeted anticancer therapies induce ciliation, while others repress it. The links between the genomic profile of cancer cells, drug treatment and ciliary signalling in the TME likely affect tumour growth and therapeutic response.
Collapse
Affiliation(s)
- Hanqing Liu
- School of Pharmacy, Jiangsu University, Jiangsu, China
| | - Anna A Kiseleva
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
- Kazan Federal University, Kazan, Russia
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
88
|
Bruel AL, Levy J, Elenga N, Defo A, Favre A, Lucron H, Capri Y, Perrin L, Passemard S, Vial Y, Tabet AC, Faivre L, Thauvin-Robinet C, Verloes A. INTU-related oral-facial-digital syndrome type VI: A confirmatory report. Clin Genet 2018; 93:1205-1209. [PMID: 29451301 DOI: 10.1111/cge.13238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 01/02/2023]
Abstract
Oral-facial-digital (OFD) syndromes are a subgroup of ciliopathies distinguished by the co-occurrence of hamartomas and/or multiple frenula of the oral region and digital anomalies. Several clinical forms of OFD syndromes are distinguished by their associated anomalies and/or inheritance patterns, and at least 20 genetic types of OFD syndromes have been delineated. We describe here a child with preaxial and postaxial polydactyly, lingual hamartoma, a congenital heart defect, delayed development and cerebellar peduncles displaying the molar tooth sign. Whole-exome sequencing and SNP array identified compound heterozygous variants in the INTU gene, which encodes a protein involved in the positioning of the ciliary basal body. INTU is a subunit of the CPLANE multiprotein complex essential for the assembly of IFT-A particles and intraflagellar transport. This report of a second patient with INTU-related OFD syndrome and the further delineation of its neuroimaging and skeletal phenotype now allow INTU-related OFD syndromes to be classified within the OFD syndrome type VI group. Patients display a phenotype similar to that of mice with a hypomorphic mutation of Intu, but with the addition of a heart defect.
Collapse
Affiliation(s)
- A-L Bruel
- UMR 1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne- et Franche-Comté, Dijon, France.,FHU TRANSLAD, Centre Hospitalier Universitaire de Bourgogne et Franche Comté, Dijon, France
| | - J Levy
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - N Elenga
- Department of Pediatrics Andrée Rosémon Regional Hospital, Cayenne, French Guiana, France.,Antilles-Guyane University, Cayenne, French Guiana, France
| | - A Defo
- Department of Pediatrics Andrée Rosémon Regional Hospital, Cayenne, French Guiana, France
| | - A Favre
- Neonatal intensive care Unit, Andrée Rosémon Regional Hospital, Cayenne, French Guiana, France
| | - H Lucron
- Antilles-Guyane M3C Pediatric Cardiology Center, Centre Hospitalier Universitaire de Martinique, Fort-de-France, France
| | - Y Capri
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - L Perrin
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - S Passemard
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France.,Denis Diderot Medical School, Sorbonne-Paris-Cité University and INSERM U1141, Paris, France
| | - Y Vial
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - A-C Tabet
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France
| | - L Faivre
- UMR 1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne- et Franche-Comté, Dijon, France.,FHU TRANSLAD, Centre Hospitalier Universitaire de Bourgogne et Franche Comté, Dijon, France.,Centre de Génétique, Centre de référence Labellisé Maladies rares Anomalies du Développement et syndromes malformatifs de l'est, Hôpital d'Enfants, CHU Dijon Bourgogne, Dijon, France
| | - C Thauvin-Robinet
- UMR 1231 GAD, Génétique des Anomalies du Développement, Université de Bourgogne- et Franche-Comté, Dijon, France.,FHU TRANSLAD, Centre Hospitalier Universitaire de Bourgogne et Franche Comté, Dijon, France.,Centre de Génétique, Centre de référence Labellisé Maladies rares Anomalies du Développement et syndromes malformatifs de l'est, Hôpital d'Enfants, CHU Dijon Bourgogne, Dijon, France
| | - A Verloes
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France.,Denis Diderot Medical School, Sorbonne-Paris-Cité University and INSERM U1141, Paris, France
| |
Collapse
|
89
|
Bonnard C, Shboul M, Tonekaboni SH, Ng AYJ, Tohari S, Ghosh K, Lai A, Lim JY, Tan EC, Devisme L, Stichelbout M, Alkindi A, Banu N, Yüksel Z, Ghoumid J, Elkhartoufi N, Boutaud L, Micalizzi A, Brett MS, Venkatesh B, Valente EM, Attié-Bitach T, Reversade B, Kariminejad A. Novel mutations in the ciliopathy-associated gene CPLANE1 (C5orf42) cause OFD syndrome type VI rather than Joubert syndrome. Eur J Med Genet 2018; 61:585-595. [PMID: 29605658 DOI: 10.1016/j.ejmg.2018.03.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 03/28/2018] [Accepted: 03/28/2018] [Indexed: 01/22/2023]
Abstract
Mutations in CPLANE1 (previously known as C5orf42) cause Oral-Facial-Digital Syndrome type VI (OFD6) as well as milder Joubert syndrome (JS) phenotypes. Seven new cases from five unrelated families diagnosed with pure OFD6 were systematically examined. Based on the clinical manifestations of these patients and those described in the literature, we revised the diagnostic features of OFD6 and include the seven most common characteristics: 1) molar tooth sign, 2) tongue hamartoma and/or lobulated tongue, 3) additional frenula, 4) mesoaxial polydactyly of hands, 5) preaxial polydactyly of feet, 6) syndactyly and/or bifid toe, and 7) hypothalamic hamartoma. By whole or targeted exome sequencing, we identified seven novel germline recessive mutations in CPLANE1, including missense, nonsense, frameshift and canonical splice site variants, all causing OFD6 in these patients. Since CPLANE1 is also mutated in JS patients, we examined whether a genotype-phenotype correlation could be established. We gathered and compared 46 biallelic CPLANE1 mutations reported in 32 JS and 26 OFD6 patients. Since no clear correlation between paired genotypes and clinical outcomes could be determined, we concluded that patient's genetic background and gene modifiers may modify the penetrance and expressivity of CPLANE1 causal alleles. To conclude, our study provides a comprehensive view of the phenotypic range, the genetic basis and genotype-phenotype association in OFD6 and JS. The updated phenotype scoring system together with the identification of new CPLANE1 mutations will help clinicians and geneticists reach a more accurate diagnosis for JS-related disorders.
Collapse
Affiliation(s)
- Carine Bonnard
- Institute of Medical Biology, A*STAR, Singapore, Singapore.
| | - Mohammad Shboul
- Institute of Medical Biology, A*STAR, Singapore, Singapore; Al-Balqa Applied University, Faculty of Science, Al-Salt, Jordan
| | | | - Alvin Yu Jin Ng
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Sumanty Tohari
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Kakaly Ghosh
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - Angeline Lai
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Jiin Ying Lim
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Ene Choo Tan
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | - Louise Devisme
- Institute of Pathology, Centre de Biologie Pathologie, CHRU Lille, France
| | | | - Adila Alkindi
- Genetics Department, Sultan Qaboos University Hospital, Oman
| | - Nazreen Banu
- Genetics Department, Sultan Qaboos University Hospital, Oman
| | - Zafer Yüksel
- Medical Genetics Department, School of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Jamal Ghoumid
- Service de Génétique Clinique et Université Lille 2, CHRU de Lille, Hôpital Jeanne de Flandre, Lille, France
| | - Nadia Elkhartoufi
- Département de Génétique, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Lucile Boutaud
- Département de Génétique, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France; INSERM U1163, Laboratoire d'Embryologie et Génétique des malformations congénitales, Université Paris Descartes, Sorbonne Paris Cite et Institute Imagine, Paris, France
| | | | | | - Byrappa Venkatesh
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Enza Maria Valente
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Tania Attié-Bitach
- Département de Génétique, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France; INSERM U1163, Laboratoire d'Embryologie et Génétique des malformations congénitales, Université Paris Descartes, Sorbonne Paris Cite et Institute Imagine, Paris, France
| | - Bruno Reversade
- Institute of Medical Biology, A*STAR, Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | | |
Collapse
|
90
|
Wang S, Liu A, Wu G, Ding HF, Huang S, Nahman S, Dong Z. The CPLANE protein Intu protects kidneys from ischemia-reperfusion injury by targeting STAT1 for degradation. Nat Commun 2018; 9:1234. [PMID: 29581513 PMCID: PMC5964315 DOI: 10.1038/s41467-018-03628-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 03/01/2018] [Indexed: 12/29/2022] Open
Abstract
Intu is known as a ciliogenesis and planar polarity effector (CPLANE) protein. Although roles for Intu have been reported during embryonic development and in the context of developmental disorders, its function and regulation in adult tissues remain poorly understood. Here we show that ablation of Intu specifically in kidney proximal tubules aggravates renal ischemia-reperfusion injury, and leads to defective post-injury ciliogenesis. We identify signal transducer and activator of transcription 1 (STAT1) as a novel interacting partner of Intu. In vitro, Intu and STAT1 colocalize at the centriole/basal body area, and Intu promotes proteasomal degradation of STAT1. During cell stress, Intu expression preserves cilia length and cell viability, and these actions are antagonized by STAT1 expression. Thus, we propose a role for Intu in protecting cells and tissues after injury by targeting STAT1 for degradation and maintaining primary cilia.
Collapse
Affiliation(s)
- Shixuan Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Aimin Liu
- Department of Biology, Eberly College of Sciences, Huck Institute of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Han-Fei Ding
- Cancer Center and Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Shuang Huang
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Stanley Nahman
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA. .,Department of Nephrology and Institute of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
91
|
Liu X, Wang Y, Liu F, Zhang M, Song H, Zhou B, Lo CW, Tong S, Hu Z, Zhang Z. Wdpcp promotes epicardial EMT and epicardium-derived cell migration to facilitate coronary artery remodeling. Sci Signal 2018; 11:11/519/eaah5770. [PMID: 29487191 DOI: 10.1126/scisignal.aah5770] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
During coronary vasculature development, endothelial cells enclose the embryonic heart to form the primitive coronary plexus. This structure is remodeled upon recruitment of epicardial cells that may undergo epithelial-mesenchymal transition (EMT) to enable migration and that give rise to smooth muscle cells. In mice expressing a loss-of-function mutant form of Wdpcp, a gene involved in ciliogenesis, the enclosure of the surface of the heart by the subepicardial coronary plexus was accelerated because of enhanced chemotactic responses to Shh. Coronary arteries, but not coronary veins in Wdpcp mutant mice, showed reduced smooth muscle cell coverage. In addition, Wdpcp mutant hearts had reduced expression of EMT and mesenchymal markers and had fewer epicardium-derived cells (EPDCs) that showed impaired migration. Epicardium-specific deletion of Wdpcp recapitulated the coronary artery defect of the Wdpcp mutant. Thus, Wdpcp promotes epithelial EMT and EPDC migration, processes that are required for remodeling of the coronary primitive plexus. The Wdpcp mutant mice will be a useful tool to dissect the molecular mechanisms that govern the remodeling of the primitive plexus during coronary development.
Collapse
Affiliation(s)
- Xiangyang Liu
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ye Wang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Feng Liu
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Min Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hejie Song
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Bin Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15201, USA
| | - Shilu Tong
- Department of Clinical Epidemiology and Biostatistics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhenlei Hu
- Department of Cardiovascular Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zhen Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
92
|
Tu F, Sedzinski J, Ma Y, Marcotte EM, Wallingford JB. Protein localization screening in vivo reveals novel regulators of multiciliated cell development and function. J Cell Sci 2018; 131:jcs.206565. [PMID: 29180514 DOI: 10.1242/jcs.206565] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/20/2017] [Indexed: 12/23/2022] Open
Abstract
Multiciliated cells (MCCs) drive fluid flow in diverse tubular organs and are essential for the development and homeostasis of the vertebrate central nervous system, airway and reproductive tracts. These cells are characterized by dozens or hundreds of motile cilia that beat in a coordinated and polarized manner. In recent years, genomic studies have not only elucidated the transcriptional hierarchy for MCC specification but also identified myriad new proteins that govern MCC ciliogenesis, cilia beating and cilia polarization. Interestingly, this burst of genomic data has also highlighted that proteins with no obvious role in cilia do, in fact, have important ciliary functions. Understanding the function of proteins with little prior history of study presents a special challenge, especially when faced with large numbers of such proteins. Here, we define the subcellular localization in MCCs of ∼200 proteins not previously implicated in cilia biology. Functional analyses arising from the screen provide novel links between actin cytoskeleton and MCC ciliogenesis.
Collapse
Affiliation(s)
- Fan Tu
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jakub Sedzinski
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.,The Danish Stem Cell Centre (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yun Ma
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.,The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen University, SunYat-sen University, Guangzhou, P.R. China
| | - Edward M Marcotte
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - John B Wallingford
- Dept. of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
93
|
Sigg MA, Menchen T, Lee C, Johnson J, Jungnickel MK, Choksi SP, Garcia G, Busengdal H, Dougherty GW, Pennekamp P, Werner C, Rentzsch F, Florman HM, Krogan N, Wallingford JB, Omran H, Reiter JF. Evolutionary Proteomics Uncovers Ancient Associations of Cilia with Signaling Pathways. Dev Cell 2018; 43:744-762.e11. [PMID: 29257953 DOI: 10.1016/j.devcel.2017.11.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 09/18/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022]
Abstract
Cilia are organelles specialized for movement and signaling. To infer when during evolution signaling pathways became associated with cilia, we characterized the proteomes of cilia from sea urchins, sea anemones, and choanoflagellates. We identified 437 high-confidence ciliary candidate proteins conserved in mammals and discovered that Hedgehog and G-protein-coupled receptor pathways were linked to cilia before the origin of bilateria and transient receptor potential (TRP) channels before the origin of animals. We demonstrated that candidates not previously implicated in ciliary biology localized to cilia and further investigated ENKUR, a TRP channel-interacting protein identified in the cilia of all three organisms. ENKUR localizes to motile cilia and is required for patterning the left-right axis in vertebrates. Moreover, mutation of ENKUR causes situs inversus in humans. Thus, proteomic profiling of cilia from diverse eukaryotes defines a conserved ciliary proteome, reveals ancient connections to signaling, and uncovers a ciliary protein that underlies development and human disease.
Collapse
Affiliation(s)
- Monika Abedin Sigg
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Tabea Menchen
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster 48149, Germany
| | - Chanjae Lee
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Jeffery Johnson
- Gladstone Institute of Cardiovascular Disease and Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA
| | - Melissa K Jungnickel
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Semil P Choksi
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Galo Garcia
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Henriette Busengdal
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen 5008, Norway
| | - Gerard W Dougherty
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster 48149, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster 48149, Germany
| | - Claudius Werner
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster 48149, Germany
| | - Fabian Rentzsch
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen 5008, Norway
| | - Harvey M Florman
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Nevan Krogan
- Gladstone Institute of Cardiovascular Disease and Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA
| | - John B Wallingford
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Heymut Omran
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster 48149, Germany
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
94
|
Zhang W, Taylor SP, Ennis HA, Forlenza KN, Duran I, Li B, Sanchez JAO, Nevarez L, Nickerson DA, Bamshad M, Lachman RS, Krakow D, Cohn DH. Expanding the genetic architecture and phenotypic spectrum in the skeletal ciliopathies. Hum Mutat 2018; 39:152-166. [PMID: 29068549 PMCID: PMC6198324 DOI: 10.1002/humu.23362] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/12/2017] [Accepted: 10/14/2017] [Indexed: 01/26/2023]
Abstract
Defects in the biosynthesis and/or function of primary cilia cause a spectrum of disorders collectively referred to as ciliopathies. A subset of these disorders is distinguished by profound abnormalities of the skeleton that include a long narrow chest with markedly short ribs, extremely short limbs, and polydactyly. These include the perinatal lethal short-rib polydactyly syndromes (SRPS) and the less severe asphyxiating thoracic dystrophy (ATD), Ellis-van Creveld (EVC) syndrome, and cranioectodermal dysplasia (CED) phenotypes. To identify new genes and define the spectrum of mutations in the skeletal ciliopathies, we analyzed 152 unrelated families with SRPS, ATD, and EVC. Causal variants were discovered in 14 genes in 120 families, including one newly associated gene and two genes previously associated with other ciliopathies. These three genes encode components of three different ciliary complexes; FUZ, which encodes a planar cell polarity complex molecule; TRAF3IP1, which encodes an anterograde ciliary transport protein; and LBR, which encodes a nuclear membrane protein with sterol reductase activity. The results established the molecular basis of SRPS type IV, in which mutations were identified in four different ciliary genes. The data provide systematic insight regarding the genotypes associated with a large cohort of these genetically heterogeneous phenotypes and identified new ciliary components required for normal skeletal development.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - S Paige Taylor
- Department of Human Genetics, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Hayley A Ennis
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Kimberly N Forlenza
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Ivan Duran
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), University of Malaga, Malaga, Spain
| | - Bing Li
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Jorge A Ortiz Sanchez
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Lisette Nevarez
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington
- University of Washington Center for Mendelian Genomics, University of Washington, Seattle, Washington
| | - Michael Bamshad
- Department of Genome Sciences, University of Washington, Seattle, Washington
- University of Washington Center for Mendelian Genomics, University of Washington, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, Washington
| | - Ralph S Lachman
- International Skeletal Dysplasia Registry at UCLA, Los Angeles, California
| | - Deborah Krakow
- Department of Human Genetics, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- International Skeletal Dysplasia Registry at UCLA, Los Angeles, California
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
| | - Daniel H Cohn
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California
- Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles, California
- International Skeletal Dysplasia Registry at UCLA, Los Angeles, California
| |
Collapse
|
95
|
Ma Y, Sun Y, Jiang L, Zuo K, Chen H, Guo J, Chen F, Lai Y, Shi J. WDPCP regulates the ciliogenesis of human sinonasal epithelial cells in chronic rhinosinusitis. Cytoskeleton (Hoboken) 2017; 74:82-90. [PMID: 28001338 DOI: 10.1002/cm.21351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 11/06/2022]
Abstract
Damage to the mucociliary clearance system is a typical change in the pathogenesis in chronic rhinosinusitis. However, the mechanisms underlying cilia loss remain unclear. WDPCP is a key protein essential for ciliogenesis, and is also an effector of the planar cell polarity signaling system. In this study, we sought to determine the role of WDPCP in cilia loss in patients with chronic rhinosinusitis. We demonstrated the expression of WDPCP in human sinonasal epithelium from patients with chronic rhinosinusitis and control subjects. We also used air-liquid interface to culture primary human sinonasal epithelial cells in-vitro model and to investigate WDPCP function. We then explored links between rhinosinusitis, WDPCP and inflammation. Accompanied with cilia loss, expression of WDPCP in human sinonasal epithelium from patients with chronic rhinosinusitis was decreased significantly compared with control subjects. In vitro study, we found that WDPCP level increased at first, and then decreased. Inhibiting WDPCP expression could lead to the poor quantity and length of cilia with reduced expression of Septin7. Also, Th1 type inflammatory mediators could decrease the expression of WDPCP. In conclusion, inflammatory cytokines cause reduced WDPCP expression, which contributes to impaired ciliogenesis in human rhinosinusitis.
Collapse
Affiliation(s)
- Yun Ma
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Yueqi Sun
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Lijie Jiang
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Kejun Zuo
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Hexin Chen
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Jiebo Guo
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Fenghong Chen
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Yinyan Lai
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| | - Jianbo Shi
- The Otorhinolaryngology Hospital, First Affiliated Hospital of Sun Yat-sen, University, SunYat-sen University, Guangzhou, P.R. China
| |
Collapse
|
96
|
Agbu SO, Liang Y, Liu A, Anderson KV. The small GTPase RSG1 controls a final step in primary cilia initiation. J Cell Biol 2017; 217:413-427. [PMID: 29038301 PMCID: PMC5748968 DOI: 10.1083/jcb.201604048] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/18/2016] [Accepted: 09/21/2017] [Indexed: 12/11/2022] Open
Abstract
Primary cilia are essential for normal development and tissue homeostasis, but the mechanisms that remodel the centriole to promote cilia initiation are not well understood. Agbu et al. report that mouse RSG1, a small GTPase, regulates a late step in cilia initiation, downstream of TTBK2 and the CPLANE protein INTU. Primary cilia, which are essential for normal development and tissue homeostasis, are extensions of the mother centriole, but the mechanisms that remodel the centriole to promote cilia initiation are poorly understood. Here we show that mouse embryos that lack the small guanosine triphosphatase RSG1 die at embryonic day 12.5, with developmental abnormalities characteristic of decreased cilia-dependent Hedgehog signaling. Rsg1 mutant embryos have fewer primary cilia than wild-type embryos, but the cilia that form are of normal length and traffic Hedgehog pathway proteins within the cilium correctly. Rsg1 mother centrioles recruit proteins required for cilia initiation and dock onto ciliary vesicles, but axonemal microtubules fail to elongate normally. RSG1 localizes to the mother centriole in a process that depends on tau tubulin kinase 2 (TTBK2), the CPLANE complex protein Inturned (INTU), and its own GTPase activity. The data suggest a specific role for RSG1 in the final maturation of the mother centriole and ciliary vesicle that allows extension of the ciliary axoneme.
Collapse
Affiliation(s)
- Stephanie O Agbu
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.,Biochemistry, Cell and Molecular Biology Program, Weill Graduate School of Medical Sciences of Cornell University, New York, NY
| | - Yinwen Liang
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Aimin Liu
- Department of Biology, Eberly College of Science, The Pennsylvania State University, University Park, PA
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
97
|
Single step genome-wide association studies based on genotyping by sequence data reveals novel loci for the litter traits of domestic pigs. Genomics 2017; 110:171-179. [PMID: 28943389 DOI: 10.1016/j.ygeno.2017.09.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 12/16/2022]
Abstract
In this study, data genotyping by sequence (GBS) was used to perform single step GWAS (ssGWAS) to identify SNPs associated with the litter traits in domestic pigs and search for candidate genes in the region of significant SNPs. After quality control, 167,355 high-quality SNPs from 532 pigs were obtained. Phenotypic traits on 2112 gilt litters from 532 pigs were recorded including total number born (TNB), number born alive (NBA), and litter weight born alive (LWB). A single-step genomic BLUP approach (ssGBLUP) was used to implement the genome-wide association analysis at a 5% genome-wide significance level. A total of 8, 23 and 20 significant SNPs were associated with TNB, NBA, and LWB, respectively, and these significant SNPs accounted for 62.78%, 79.75%, and 58.79% of genetic variance. Furthermore, 1 (SSC14: 16314857), 4 (SSC1: 81986236, SSC1: 66599775, SSC1: 161999013, and SSC1: 267883107), and 5 (SSC9: 29030061, SSC2: 32368561, SSC5: 110375350, SSC13: 45619882 and SSC13: 45647829) significant SNPs for TNB, NBA, and LWB were inferred to be novel loci. At SSC1, the AIM1 and FOXO3 genes were found to be associated with NBA; these genes increase ovarian reproductive capacity and follicle number and decrease gonadotropin levels. The genes SLC36A4 and INTU are involved in cell growth, cytogenesis and development were found to be associated with LWB. These significant SNPs can be used as an indication for regions in the Sus scrofa genome for variability in litter traits, but further studies are expected to confirm causative mutations.
Collapse
|
98
|
Wangler MF, Yamamoto S, Chao HT, Posey JE, Westerfield M, Postlethwait J, Hieter P, Boycott KM, Campeau PM, Bellen HJ. Model Organisms Facilitate Rare Disease Diagnosis and Therapeutic Research. Genetics 2017; 207:9-27. [PMID: 28874452 PMCID: PMC5586389 DOI: 10.1534/genetics.117.203067] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/06/2017] [Indexed: 12/29/2022] Open
Abstract
Efforts to identify the genetic underpinnings of rare undiagnosed diseases increasingly involve the use of next-generation sequencing and comparative genomic hybridization methods. These efforts are limited by a lack of knowledge regarding gene function, and an inability to predict the impact of genetic variation on the encoded protein function. Diagnostic challenges posed by undiagnosed diseases have solutions in model organism research, which provides a wealth of detailed biological information. Model organism geneticists are by necessity experts in particular genes, gene families, specific organs, and biological functions. Here, we review the current state of research into undiagnosed diseases, highlighting large efforts in North America and internationally, including the Undiagnosed Diseases Network (UDN) (Supplemental Material, File S1) and UDN International (UDNI), the Centers for Mendelian Genomics (CMG), and the Canadian Rare Diseases Models and Mechanisms Network (RDMM). We discuss how merging human genetics with model organism research guides experimental studies to solve these medical mysteries, gain new insights into disease pathogenesis, and uncover new therapeutic strategies.
Collapse
Affiliation(s)
- Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Hsiao-Tuan Chao
- Department of Pediatrics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Section of Child Neurology, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
| | - Monte Westerfield
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - John Postlethwait
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Philip Hieter
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4C, Canada
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ontario K1H 8L1, Canada
| | - Philippe M Campeau
- Department of Pediatrics, University of Montreal, Quebec H3T 1C5, Canada
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine (BCM), Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine (BCM), Houston, Texas 77030
- Howard Hughes Medical Institute, Baylor College of Medicine (BCM), Houston, Texas 77030
| |
Collapse
|
99
|
Prevo B, Scholey JM, Peterman EJG. Intraflagellar transport: mechanisms of motor action, cooperation, and cargo delivery. FEBS J 2017; 284:2905-2931. [PMID: 28342295 PMCID: PMC5603355 DOI: 10.1111/febs.14068] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/20/2017] [Accepted: 03/23/2017] [Indexed: 02/06/2023]
Abstract
Intraflagellar transport (IFT) is a form of motor-dependent cargo transport that is essential for the assembly, maintenance, and length control of cilia, which play critical roles in motility, sensory reception, and signal transduction in virtually all eukaryotic cells. During IFT, anterograde kinesin-2 and retrograde IFT dynein motors drive the bidirectional transport of IFT trains that deliver cargo, for example, axoneme precursors such as tubulins as well as molecules of the signal transduction machinery, to their site of assembly within the cilium. Following its discovery in Chlamydomonas, IFT has emerged as a powerful model system for studying general principles of motor-dependent cargo transport and we now appreciate the diversity that exists in the mechanism of IFT within cilia of different cell types. The absence of heterotrimeric kinesin-2 function, for example, causes a complete loss of both IFT and cilia in Chlamydomonas, but following its loss in Caenorhabditis elegans, where its primary function is loading the IFT machinery into cilia, homodimeric kinesin-2-driven IFT persists and assembles a full-length cilium. Generally, heterotrimeric kinesin-2 and IFT dynein motors are thought to play widespread roles as core IFT motors, whereas homodimeric kinesin-2 motors are accessory motors that mediate different functions in a broad range of cilia, in some cases contributing to axoneme assembly or the delivery of signaling molecules but in many other cases their ciliary functions, if any, remain unknown. In this review, we focus on mechanisms of motor action, motor cooperation, and motor-dependent cargo delivery during IFT.
Collapse
Affiliation(s)
- Bram Prevo
- Department of Cellular & Molecular Medicine, University of California San Diego, CA, USA
- Ludwig Institute for Cancer Research, San Diego, CA, USA
| | - Jonathan M Scholey
- Department of Molecular & Cell Biology, University of California Davis, CA, USA
| | - Erwin J G Peterman
- Department of Physics and Astronomy and LaserLaB Amsterdam, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
100
|
Abstract
Motile and non-motile (primary) cilia are nearly ubiquitous cellular organelles. The dysfunction of cilia causes diseases known as ciliopathies. The number of reported ciliopathies (currently 35) is increasing, as is the number of established (187) and candidate (241) ciliopathy-associated genes. The characterization of ciliopathy-associated proteins and phenotypes has improved our knowledge of ciliary functions. In particular, investigating ciliopathies has helped us to understand the molecular mechanisms by which the cilium-associated basal body functions in early ciliogenesis, as well as how the transition zone functions in ciliary gating, and how intraflagellar transport enables cargo trafficking and signalling. Both basic biological and clinical studies are uncovering novel ciliopathies and the ciliary proteins involved. The assignment of these proteins to different ciliary structures, processes and ciliopathy subclasses (first order and second order) provides insights into how this versatile organelle is built, compartmentalized and functions in diverse ways that are essential for human health.
Collapse
|