1
|
Radhakrishnan P, Quadri N, Erger F, Fuhrmann N, Geist OM, Netzer C, Khyriem I, Muranjan M, Udani V, Yeole M, Mascarenhas S, Limaye S, Siddiqui S, Upadhyai P, Shukla A. Biallelic Variants in LRRC45 Impair Ciliogenesis and Cause a Severe Neurological Disorder. Clin Genet 2025; 107:311-322. [PMID: 39638757 PMCID: PMC11790379 DOI: 10.1111/cge.14663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Leucine - rich repeat containing 45 protein (LRRC45) protein localizes at the proximal end of centrioles and forms a component of the proteinaceous linker between them, with an important role in centrosome cohesion. In addition, a pool of it localizes at the distal appendages of the modified parent centriole that forms the primary cilium and it has essential functions in the establishment of the transition zone and axonemal extension during early ciliogenesis. Here, we describe three individuals from two unrelated families with severe central nervous system anomalies. Exome sequencing identified biallelic variants in LRRC45 in the affected individuals: P1: c.1402-2A>G; P2 and P3: c.1262G>C (p.Arg421Thr). Investigation of the variant c.1402-2A>G in patient-derived skin fibroblasts revealed that it triggers aberrant splicing, leading to an abnormal LRRC45 transcript that lacks exon 14. Consistent with this the mRNA and protein levels of LRRC45 were drastically reduced in P1-derived fibroblast cells compared to the controls. P1 fibroblasts showed a significant reduction of primary cilia frequency and length. In silico modeling of the missense variant in P2/P3 suggested a destabilizing effect on LRRC45. Given these findings, we propose that the pathogenic loss-of-function variants in LRRC45 are associated with a novel spectrum of neurological ciliopathy phenotypes.
Collapse
Affiliation(s)
- Periyasamy Radhakrishnan
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Neha Quadri
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Florian Erger
- Center for Rare Diseases Cologne, University Hospital Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Nico Fuhrmann
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Otilia-Maria Geist
- Department of Gynecology and Obstetrics, Klinikum Leverkusen, Leverkusen, Germany
| | - Christian Netzer
- Center for Rare Diseases Cologne, University Hospital Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Ibakordor Khyriem
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Mamta Muranjan
- Department of Paediatrics, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Vrajesh Udani
- Department of Child Neurology, PD Hinduja National Hospital, Mumbai, India
| | - Mayuri Yeole
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Selinda Mascarenhas
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Sanket Limaye
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Shahyan Siddiqui
- Department of Neuro and Vascular Interventional Radiology, Yashoda Hospitals, Hyderabad, India
| | - Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
2
|
Wang Y, Kraemer N, Schneider J, Ninnemann O, Weng K, Hildebrand M, Reid J, Li N, Hu H, Mani S, Kaindl AM. Togaram1 is expressed in the neural tube and its absence causes neural tube closure defects. HGG ADVANCES 2025; 6:100363. [PMID: 39385469 PMCID: PMC11541697 DOI: 10.1016/j.xhgg.2024.100363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Neural tube closure defect pathomechanisms in human embryonic development are poorly understood. Here we identified spina bifida patients expressing novel variants of the TOGARAM gene family. TOGARAM1 has been associated with the ciliopathy Joubert syndrome, but its connection to spina bifida and role in neural development is unknown. We show that Togaram1 is expressed in the neural tube and Togaram1 knockout mice have abnormal cilia, reduced sonic hedgehog (Shh) signaling, abnormal neural tube patterning, and display neural tube closure defects. Neural stem cells from Togaram1 knockout embryos showed reduced cilia and defects in Shh signaling. Overexpression in IMCD3 and HEK293 cells of TOGARAM1 carrying the variant found in the spina bifida patient resulted in cilia defect along with reduced pericentriolar material one (PCM1), a critical constituent of centriolar satellites involved in transporting proteins toward the centrosome and primary cilia. Our results demonstrate the role of TOGARAM1 in regulating Shh signaling during early neural development that is critical for neural tube closure and elucidates potential mechanisms whereby the ciliopathy-associated gene TOGARAM1 gives rise to spina bifida aperta in humans.
Collapse
Affiliation(s)
- Yanyan Wang
- Institute of Cell Biology and Neurobiology, Charite - Universitatsmedizin Berlin, Berlin, Germany; Department of Pediatric Neurology, Charité - Universitatsmedizin Berlin, Berlin, Germany
| | - Nadine Kraemer
- Institute of Cell Biology and Neurobiology, Charite - Universitatsmedizin Berlin, Berlin, Germany; Department of Pediatric Neurology, Charité - Universitatsmedizin Berlin, Berlin, Germany
| | - Joanna Schneider
- Department of Pediatric Neurology, Charité - Universitatsmedizin Berlin, Berlin, Germany; Center for Chronically Sick Children, Charité - Universitatsmedizin Berlin, Berlin, Germany
| | - Olaf Ninnemann
- Institute of Cell Biology and Neurobiology, Charite - Universitatsmedizin Berlin, Berlin, Germany
| | - Kai Weng
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Michael Hildebrand
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Melbourne, VIC, Australia; Neuroscience Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Joshua Reid
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Melbourne, VIC, Australia
| | - Na Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hao Hu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Shyamala Mani
- Institute of Cell Biology and Neurobiology, Charite - Universitatsmedizin Berlin, Berlin, Germany; Department of Pediatric Neurology, Charité - Universitatsmedizin Berlin, Berlin, Germany
| | - Angela M Kaindl
- Institute of Cell Biology and Neurobiology, Charite - Universitatsmedizin Berlin, Berlin, Germany; Department of Pediatric Neurology, Charité - Universitatsmedizin Berlin, Berlin, Germany; Center for Chronically Sick Children, Charité - Universitatsmedizin Berlin, Berlin, Germany; German Epilepsy Center for Children and Adolescents, Charité - Universitatsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Kim SE, Kim HY, Wlodarczyk BJ, Finnell RH. Linkage between Fuz and Gpr161 genes regulates sonic hedgehog signaling during mouse neural tube development. Development 2024; 151:dev202705. [PMID: 39369306 PMCID: PMC11463954 DOI: 10.1242/dev.202705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 09/02/2024] [Indexed: 10/07/2024]
Abstract
Sonic hedgehog (Shh) signaling regulates embryonic morphogenesis utilizing the primary cilium, the cell's antenna, which acts as a signaling hub. Fuz, an effector of planar cell polarity signaling, regulates Shh signaling by facilitating cilia formation, and the G protein-coupled receptor 161 (Gpr161) is a negative regulator of Shh signaling. The range of phenotypic malformations observed in mice bearing mutations in either of the genes encoding these proteins is similar; however, their functional relationship has not been previously explored. This study identified the genetic and biochemical linkage between Fuz and Gpr161 in mouse neural tube development. Fuz was found to be genetically epistatic to Gpr161 with respect to regulation of Shh signaling in mouse neural tube development. The Fuz protein biochemically interacts with Gpr161, and Fuz regulates Gpr161-mediated ciliary localization, a process that might utilize β-arrestin 2. Our study characterizes a previously unappreciated Gpr161-Fuz axis that regulates Shh signaling during mouse neural tube development.
Collapse
Affiliation(s)
- Sung-Eun Kim
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin/Dell Medical School, Austin, TX 78723, USA
| | | | - Bogdan J. Wlodarczyk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard H. Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin/Dell Medical School, Austin, TX 78723, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Molecular and Cellular Biology, Molecular and Human Genetics, and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
4
|
Wang X, Yu J, Yue H, Li S, Yang A, Zhu Z, Guan Z, Wang J. Inpp5e Regulated the Cilium-Related Genes Contributing to the Neural Tube Defects Under 5-Fluorouracil Exposure. Mol Neurobiol 2024; 61:6189-6199. [PMID: 38285286 DOI: 10.1007/s12035-024-03946-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024]
Abstract
Primary cilia are crucial for neurogenesis, and cilium-related genes are involved in the closure of neural tubes. Inositol polyphosphate-5-phosphatase (Inpp5e) was enriched in primary cilia and closely related to the occurrence of neural tube defects (NTDs). However, the role of Inpp5e in the development of NTDs is not well-known. To investigate whether Inpp5e gene is associated with the neural tube closure, we established a mouse model of NTDs by 5-fluorouracil (5-FU) exposure at gestational day 7.5 (GD7.5). The Inpp5e knockdown (Inpp5e-/-) mouse embryonic stem cells (mESCs) were produced by CRISPR/Cas9 system. The expressions of Inpp5e and other cilium-related genes including intraflagellar transport 80 (Ift80), McKusick-Kaufman syndrome (Mkks), and Kirsten rat sarcoma viral oncogene homolog (Kras) were determined, utilizing quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR), western blot, PCR array, and immunofluorescence staining. The result showed that the incidence of NTDs was 37.10% (23 NTDs/62 total embryos) and significantly higher than that in the control group (P < 0.001). The neuroepithelial cells of neural tubes were obviously disarranged in NTD embryos. The mRNA and protein levels of Inpp5e, Ift80, Mkks, and Kras were significantly decreased in NTD embryonic brain tissues, compared to the control (P < 0.05). Knockdown of the Inpp5e (Inpp5e-/-) reduced the expressions of Ift80, Mkks, and Kras in mESCs. Furthermore, the levels of α-tubulin were significantly reduced in NTD embryonic neural tissue and Inpp5e-/- mESCs. These results suggested that maternal 5-FU exposure inhibited the expression of Inpp5e, which resulted in the downregulation of cilium-related genes (Ift80, Mkks, and Kras), leading to the impairment of primary cilium development, and ultimately disrupted the neural tube closure.
Collapse
Affiliation(s)
- Xiuwei Wang
- Laboratory of Translational Medicine, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Jialu Yu
- Laboratory of Translational Medicine, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Huixuan Yue
- Department of Pediatrics, Beijing Chaoyang Hospital of Capital Medical University, Beijing, 100020, China
| | - Shen Li
- Laboratory of Translational Medicine, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Aiyun Yang
- Laboratory of Translational Medicine, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zhiqiang Zhu
- Laboratory of Translational Medicine, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zhen Guan
- Laboratory of Translational Medicine, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.
| | - Jianhua Wang
- Laboratory of Translational Medicine, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
5
|
Kim SE, Kim HY, Wlodarczyk BJ, Finnell RH. The novel linkage between Fuz and Gpr161 genes regulates sonic hedgehog signaling during mouse embryonic development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575263. [PMID: 38260275 PMCID: PMC10802560 DOI: 10.1101/2024.01.11.575263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Sonic hedgehog (Shh) signaling regulates embryonic morphogenesis utilizing primary cilia, the cell antenna acting as a signaling hub. Fuz, an effector of planar cell polarity (PCP) signaling, involves Shh signaling via cilia formation, while the G protein-coupled receptor 161 (Gpr161) is a negative regulator of Shh signaling. The range of phenotypic malformations observed in mice bearing mutations in either of these two genes is similar; however, their functional relations have not been previously explored. This study identified the genetic and biochemical link between Fuz and Gpr161 in mouse embryonic development. Fuz was genetically epistatic to Gpr161 via Shh signaling during mouse embryonic development. The FUZ biochemically interacted with GPR161, and Fuz regulated Gpr161 ciliary trafficking via β-arrestin2. Our study suggested the novel Gpr161-Fuz axis that regulates Shh signaling during mouse embryonic development.
Collapse
Affiliation(s)
- Sung-Eun Kim
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin/Dell Medical School, Austin, TX, 78723, USA
| | | | - Bogdan J. Wlodarczyk
- Departments of Molecular and Cellular Biology, Molecular and Human Genetics, and Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Richard H. Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin/Dell Medical School, Austin, TX, 78723, USA
- Departments of Molecular and Cellular Biology, Molecular and Human Genetics, and Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
6
|
Kim SE, Chothani PJ, Shaik R, Pollard W, Finnell RH. Pax3 lineage-specific deletion of Gpr161 is associated with spinal neural tube and craniofacial malformations during embryonic development. Dis Model Mech 2023; 16:dmm050277. [PMID: 37885410 PMCID: PMC10694864 DOI: 10.1242/dmm.050277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Sonic hedgehog (Shh) signaling is the morphogen signaling that regulates embryonic craniofacial and neural tube development. G protein-coupled receptor 161 (Gpr161) is a negative regulator of Shh signaling, and its inactivation in mice results in embryo lethality associated with craniofacial defects and neural tube defects. However, the structural defects of later embryonic stages and cell lineages underlying abnormalities have not been well characterized due to the limited lifespan of Gpr161 null mice. We found that embryos with Pax3 lineage-specific deletion of Gpr161 presented with tectal hypertrophy (anterior dorsal neuroepithelium), cranial vault and facial bone hypoplasia (cranial neural crest), vertebral abnormalities (somite) and the closed form of spina bifida (posterior dorsal neuroepithelium). In particular, the closed form of spina bifida was partly due to reduced Pax3 and Cdx4 gene expression in the posterior dorsal neural tubes of Gpr161 mutant embryos with decreased Wnt signaling, whereas Shh signaling was increased. We describe a previously unreported role for Gpr161 in the development of posterior neural tubes and confirm its role in cranial neural crest- and somite-derived skeletogenesis and midbrain morphogenesis in mice.
Collapse
Affiliation(s)
- Sung-Eun Kim
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
| | - Pooja J. Chothani
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
| | - Rehana Shaik
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
| | - Westley Pollard
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
| | - Richard H. Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
Kim SE, Chothani PJ, Shaik R, Pollard W, Finnell RH. Pax3 lineage-specific deletion of Gpr161 is associated with spinal neural tube and craniofacial malformations during embryonic development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.548129. [PMID: 37461574 PMCID: PMC10350067 DOI: 10.1101/2023.07.07.548129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Shh signaling is the morphogen signaling that regulates embryonic craniofacial and neural tube development. G protein-coupled receptor 161 (Gpr161) is a negative regulator of Shh signaling, and its inactivation in mice results in embryo lethality with craniofacial and neural tube defects (NTDs). However, the structural defects of later embryonic stages in Gpr161 null mice and cell lineages underlying abnormalities were not well characterized due to their limited lifespan. We found the Pax3 lineage-specific deletion of Gpr161 in mice presented with tectal hypertrophy (anterior dorsal neuroepithelium), cranial vault and facial bone hypoplasia (cranial neural crest (CNC)), vertebral abnormalities (somite), and the closed form of spina bifida (posterior dorsal neuroepithelium). In particular, the closed form of spina bifida is partly due to the reduced Pax3 and Cdx4 gene expression of the posterior dorsal neural tubes of Gpr161 mutant embryos involving decreased Wnt signaling whereas Shh signaling was increased. This study provides the novel role of Gpr161 in the posterior neural tube development and confirms its role on CNC- and somite-derived skeletogenesis and midbrain morphogenesis in mice.
Collapse
|
8
|
Zheng NX, Miao YT, Zhang X, Huang MZ, Jahangir M, Luo S, Lang B. Primary cilia-associated protein IFT172 in ciliopathies. Front Cell Dev Biol 2023; 11:1074880. [PMID: 36733456 PMCID: PMC9887189 DOI: 10.3389/fcell.2023.1074880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Cilium is a highly conserved antenna-like structure protruding from the surface of the cell membrane, which is widely distributed on most mammalian cells. Two types of cilia have been described so far which include motile cilia and immotile cilia and the latter are also known as primary cilia. Dysfunctional primary cilia are commonly associated with a variety of congenital diseases called ciliopathies with multifaceted presentations such as retinopathy, congenital kidney disease, intellectual disability, cancer, polycystic kidney, obesity, Bardet Biedl syndrome (BBS), etc. Intraflagellar transport (IFT) is a bi-directional transportation process that helps maintain a balanced flow of proteins or signaling molecules essential for the communication between cilia and cytoplasm. Disrupted IFT contributes to the abnormal structure or function of cilia and frequently promotes the occurrence of ciliopathies. Intraflagellar transport 172 (IFT172) is a newly identified member of IFT proteins closely involved in some rare ciliopathies such as Mainzer-Saldino syndrome (MZSDS) and BBS, though the underpinning causal mechanisms remain largely elusive. In this review, we summarize the key findings on the genetic and protein characteristic of IFT172, as well as its function in intraflagellar transport, to provide comprehensive insights to understand IFT172-related ciliopathies.
Collapse
Affiliation(s)
- Nan-Xi Zheng
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ya-Ting Miao
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mu-Zhi Huang
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Jahangir
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shilin Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China,*Correspondence: Shilin Luo, ; Bing Lang,
| | - Bing Lang
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Shilin Luo, ; Bing Lang,
| |
Collapse
|
9
|
Whye D, Wood D, Kim K, Chen C, Makhortova N, Sahin M, Buttermore ED. Dynamic 3D Combinatorial Generation of hPSC-Derived Neuromesodermal Organoids With Diverse Regional and Cellular Identities. Curr Protoc 2022; 2:e568. [PMID: 36264199 PMCID: PMC9589923 DOI: 10.1002/cpz1.568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neuromesodermal progenitors represent a unique, bipotent population of progenitors residing in the tail bud of the developing embryo, which give rise to the caudal spinal cord cell types of neuroectodermal lineage as well as the adjacent paraxial somite cell types of mesodermal origin. With the advent of stem cell technologies, including induced pluripotent stem cells (iPSCs), the modeling of rare genetic disorders can be accomplished in vitro to interrogate cell-type specific pathological mechanisms in human patient conditions. Stem cell-derived models of neuromesodermal progenitors have been accomplished by several developmental biology groups; however, most employ a 2D monolayer format that does not fully reflect the complexity of cellular differentiation in the developing embryo. This article presents a dynamic 3D combinatorial method to generate robust populations of human pluripotent stem cell-derived neuromesodermal organoids with multi-cellular fates and regional identities. By utilizing a dynamic 3D suspension format for the differentiation process, the organoids differentiated by following this protocol display a hallmark of embryonic development that involves a morphological elongation known as axial extension. Furthermore, by employing a combinatorial screening assay, we dissect essential pathways for optimally directing the patterning of pluripotent stem cells into neuromesodermal organoids. This protocol highlights the influence of timing, duration, and concentration of WNT and fibroblast growth factor (FGF) signaling pathways on enhancing early neuromesodermal identity, and later, downstream cell fate specification through combined synergies of retinoid signaling and sonic hedgehog activation. Finally, through robust inhibition of the Notch signaling pathway, this protocol accelerates the acquisition of terminal cell identities. This enhanced organoid model can serve as a powerful tool for studying normal developmental processes as well as investigating complex neurodevelopmental disorders, such as neural tube defects. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Robust generation of 3D hPSC-derived spheroid populations in dynamic motion settings Support Protocol 1: Pluronic F-127 reagent preparation and coating to generate low-attachment suspension culture dishes Basic Protocol 2: Enhanced specification of hPSCs into NMP organoids Support Protocol 2: Combinatorial pathway assay for NMP organoid protocol optimization Basic Protocol 3: Differentiation of NMP organoids along diverse cellular trajectories and accelerated terminal fate specification into neurons, neural crest, and sclerotome derivatives.
Collapse
Affiliation(s)
- Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Delaney Wood
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Kristina Kim
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Cidi Chen
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Nina Makhortova
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Mustafa Sahin
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Elizabeth D. Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
10
|
Shiferaw MY, Akililu YB, Worku BY, T/Mariam TL, Aklilu AT. Multiple-site neural tube defects complicated by multiple-site split cord malformations and thickened filum terminale: experience at a pediatric neurosurgical teaching hospital in Ethiopia. Illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2022; 4:CASE22220. [PMID: 36593675 PMCID: PMC9514283 DOI: 10.3171/case22220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/17/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Multiple-site open neural tube defects (MNTDs) and multiple-site split cord malformations (MSCMs) are extremely rare congenital anomalies that are defined by the simultaneous noncontiguous occurrence of more than one neural tube defect (NTD) and split cord malformation (SCM), respectively, in a single case with normal neural tissue in between. This work shows the cooccurrence of MNTDs and MSCMs, which has never been reported in the literature. OBSERVATIONS A single-stage repair for a 13-day-old female neonate with a preoperative diagnosis of MNTDs (thoracic meningocele and thoracolumbar myelomeningocele) plus an additional intraoperative diagnosis of MSCMs (type 3c) of thoracic and thoracolumbar spine, and thickened filum terminale was done with a favorable smooth postoperative course. LESSONS The use of intraoperative meticulous surgical technique along with preoperative skin stigmata helped for anticipation, detection, and treatment of associated complex spinal MNTDs, especially in resource-limited settings, where preoperative magnetic resonance imaging is not routinely used. Whether to repair the MNTDs as a single- versus multiple-stage procedure is mainly a function of the patient's tolerance to the duration of anesthesia and the anticipated blood loss for the patient's age. The overall developmental biology and long-term clinical outcome of MNTDs compared to single NTD/SCM is poorly understood and needs further study.
Collapse
|
11
|
Cilia and their role in neural tube development and defects. REPRODUCTIVE AND DEVELOPMENTAL MEDICINE 2022. [DOI: 10.1097/rd9.0000000000000014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
12
|
Ondičová M, Irwin RE, Thursby SJ, Hilman L, Caffrey A, Cassidy T, McLaughlin M, Lees-Murdock DJ, Ward M, Murphy M, Lamers Y, Pentieva K, McNulty H, Walsh CP. Folic acid intervention during pregnancy alters DNA methylation, affecting neural target genes through two distinct mechanisms. Clin Epigenetics 2022; 14:63. [PMID: 35578268 PMCID: PMC9112484 DOI: 10.1186/s13148-022-01282-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/29/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We previously showed that continued folic acid (FA) supplementation beyond the first trimester of pregnancy appears to have beneficial effects on neurocognitive performance in children followed for up to 11 years, but the biological mechanism for this effect has remained unclear. Using samples from our randomized controlled trial of folic acid supplementation in second and third trimester (FASSTT), where significant improvements in cognitive and psychosocial performance were demonstrated in children from mothers supplemented in pregnancy with 400 µg/day FA compared with placebo, we examined methylation patterns from cord blood (CB) using the EPIC array which covers approximately 850,000 cytosine-guanine (CG) sites across the genome. Genes showing significant differences were verified using pyrosequencing and mechanistic approaches used in vitro to determine effects on transcription. RESULTS FA supplementation resulted in significant differences in methylation, particularly at brain-related genes. Further analysis showed these genes split into two groups. In one group, which included the CES1 gene, methylation changes at the promoters were important for regulating transcription. We also identified a second group which had a characteristic bimodal profile, with low promoter and high gene body (GB) methylation. In the latter, loss of methylation in the GB is linked to decreases in transcription: this group included the PRKAR1B/HEATR2 genes and the dopamine receptor regulator PDE4C. Overall, methylation in CB also showed good correlation with methylation profiles seen in a published data set of late gestation foetal brain samples. CONCLUSION We show here clear alterations in DNA methylation at specific classes of neurodevelopmental genes in the same cohort of children, born to FA-supplemented mothers, who previously showed improved cognitive and psychosocial performance. Our results show measurable differences at neural genes which are important for transcriptional regulation and add to the supporting evidence for continued FA supplementation throughout later gestation. This trial was registered on 15 May 2013 at www.isrctn.com as ISRCTN19917787.
Collapse
Affiliation(s)
- Miroslava Ondičová
- Genomic Medicine Research Group, Ulster University, Coleraine, Northern Ireland, UK
| | - Rachelle E Irwin
- Genomic Medicine Research Group, Ulster University, Coleraine, Northern Ireland, UK
| | - Sara-Jayne Thursby
- Genomic Medicine Research Group, Ulster University, Coleraine, Northern Ireland, UK
- The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Luke Hilman
- Genomic Medicine Research Group, Ulster University, Coleraine, Northern Ireland, UK
| | - Aoife Caffrey
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Tony Cassidy
- Psychology Institute, Ulster University, Coleraine, Northern Ireland, UK
| | - Marian McLaughlin
- Psychology Institute, Ulster University, Coleraine, Northern Ireland, UK
| | - Diane J Lees-Murdock
- Genomic Medicine Research Group, Ulster University, Coleraine, Northern Ireland, UK
| | - Mary Ward
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Michelle Murphy
- Unitat de Medicina Preventiva i Salut Pública, Facultat de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Yvonne Lamers
- Food, Nutrition, and Health Program, Faculty of Land and Food Systems, The University of British Columbia, and British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kristina Pentieva
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Helene McNulty
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Colum P Walsh
- Genomic Medicine Research Group, Ulster University, Coleraine, Northern Ireland, UK.
- Centre for Research and Development, Region Gävleborg/Uppsala University, Gävle, Sweden.
| |
Collapse
|
13
|
Bai Y, Wei C, Li P, Sun X, Cai G, Chen X, Hong Q. Primary cilium in kidney development, function and disease. Front Endocrinol (Lausanne) 2022; 13:952055. [PMID: 36072924 PMCID: PMC9441790 DOI: 10.3389/fendo.2022.952055] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
The primary cilium is a hair-like, microtubule-based organelle that is covered by the cell membrane and extends from the surface of most vertebrate cells. It detects and translates extracellular signals to direct various cellular signaling pathways to maintain homeostasis. It is mainly distributed in the proximal and distal tubules and collecting ducts in the kidney. Specific signaling transduction proteins localize to primary cilia. Defects in cilia structure and function lead to a class of diseases termed ciliopathies. The proper functioning of primary cilia is essential to kidney organogenesis and the maintenance of epithelial cell differentiation and proliferation. Persistent cilia dysfunction has a role in the early stages and progression of renal diseases, such as cystogenesis and acute tubular necrosis (ATN). In this review, we focus on the central role of cilia in kidney development and illustrate how defects in cilia are associated with renal disease progression.
Collapse
Affiliation(s)
- Yunfeng Bai
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Cuiting Wei
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xuefeng Sun
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Xiangmei Chen, ; Quan Hong,
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
- *Correspondence: Xiangmei Chen, ; Quan Hong,
| |
Collapse
|
14
|
Shiferaw MY, Awedew AF, T/Mariam TL, Aklilu AT, Akililu YB, Andualem AM. Multiple site neural tube defects at Zewuditu Memorial Hospital, Addis Ababa, Ethiopia: a case report. J Med Case Rep 2021; 15:429. [PMID: 34399841 PMCID: PMC8369642 DOI: 10.1186/s13256-021-02913-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/18/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Neural tube defects are common group of central nervous system anomalies of complex etiology and major public health importance worldwide. The occurrence of multiple neural tube defects, however, is an extremely rare event and has never been reported in Ethiopia so far. This study gives an insight into how the embryogenesis, management, and postoperative complications of multiple neural tube defects differ from the ordinary single neural tube defects on the basis of up-to-date existing literature. CASE PRESENTATION This paper highlights a case of an 8 days old female black race Ethiopian neonate who was brought by her mother with the chief complaint of lower back and lower neck swelling since birth. The findings were a 5 × 4 × 5 cm sized ulcerated placode at the mid-lumbosacral area and a 1.5 × 1.5 × 1 cm sized fluctuant, nontender, transilluminating mass with overlying unruptured defect dysplastic skin at the cervicothoracic junction. With a diagnosis of multiple neural tube defects secondary to unruptured cervicothoracic meningocele and ruptured lumbosacral myelomeningocele, single-stage repair of the defects was done with good outcome. CONCLUSION There is insufficient evidence as to the exact mechanism of development of multiple neural tube defects. Similarly, whether patients with multiple neural tube defects had increased risk of post repair hydrocephalus compared with patients who have single neural tube defect is unknown. Hence, more research on the embryogenesis, management, and long-term outcome of multiple neural tube defects in particular and single neural tube defects in general should be done to better help patients with this costly and crippling problem. Lastly, the practice of folic acid supplementation is very low in resource-limited countries such as Ethiopia and, hence, should be improved.
Collapse
Affiliation(s)
- Mestet Yibeltal Shiferaw
- Neurosurgery Division, Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Atalel Fentahun Awedew
- Neurosurgery Division, Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tsegazeab Laeke T/Mariam
- Neurosurgery Division, Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Abenezer Tirsit Aklilu
- Neurosurgery Division, Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Yemisirach Bizuneh Akililu
- Neurosurgery Division, Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Aemiro Mazengia Andualem
- Neurosurgery Division, Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
15
|
Au KS, Hebert L, Hillman P, Baker C, Brown MR, Kim DK, Soldano K, Garrett M, Ashley-Koch A, Lee S, Gleeson J, Hixson JE, Morrison AC, Northrup H. Human myelomeningocele risk and ultra-rare deleterious variants in genes associated with cilium, WNT-signaling, ECM, cytoskeleton and cell migration. Sci Rep 2021; 11:3639. [PMID: 33574475 PMCID: PMC7878900 DOI: 10.1038/s41598-021-83058-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/28/2021] [Indexed: 01/08/2023] Open
Abstract
Myelomeningocele (MMC) affects one in 1000 newborns annually worldwide and each surviving child faces tremendous lifetime medical and caregiving burdens. Both genetic and environmental factors contribute to disease risk but the mechanism is unclear. This study examined 506 MMC subjects for ultra-rare deleterious variants (URDVs, absent in gnomAD v2.1.1 controls that have Combined Annotation Dependent Depletion score ≥ 20) in candidate genes either known to cause abnormal neural tube closure in animals or previously associated with human MMC in the current study cohort. Approximately 70% of the study subjects carried one to nine URDVs among 302 candidate genes. Half of the study subjects carried heterozygous URDVs in multiple genes involved in the structure and/or function of cilium, cytoskeleton, extracellular matrix, WNT signaling, and/or cell migration. Another 20% of the study subjects carried heterozygous URDVs in candidate genes associated with gene transcription regulation, folate metabolism, or glucose metabolism. Presence of URDVs in the candidate genes involving these biological function groups may elevate the risk of developing myelomeningocele in the study cohort.
Collapse
Affiliation(s)
- K S Au
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - L Hebert
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - P Hillman
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - C Baker
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - M R Brown
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center At Houston, Houston, TX, 77030, USA
| | - D-K Kim
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center At Houston, Houston, TX, 77030, USA
| | - K Soldano
- Department of Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - M Garrett
- Department of Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - A Ashley-Koch
- Department of Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - S Lee
- Department of Neurosciences and Pediatrics, University of California-San Diego, La Jolla, CA, 92093, USA.,Rady Children's Institute for Genomic Medicine, San Diego, CA, 92025, USA
| | - J Gleeson
- Department of Neurosciences and Pediatrics, University of California-San Diego, La Jolla, CA, 92093, USA.,Rady Children's Institute for Genomic Medicine, San Diego, CA, 92025, USA
| | - J E Hixson
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center At Houston, Houston, TX, 77030, USA
| | - A C Morrison
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center At Houston, Houston, TX, 77030, USA
| | - H Northrup
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| |
Collapse
|
16
|
Zhang YC, Bai YF, Yuan JF, Shen XL, Xu YL, Jian XX, Li S, Song ZQ, Hu HB, Li PY, Tu HQ, Han QY, Wang N, Li AL, Zhang XM, Wu M, Zhou T, Li HY. CEP55 promotes cilia disassembly through stabilizing Aurora A kinase. J Cell Biol 2021; 220:211702. [PMID: 33475699 PMCID: PMC7829976 DOI: 10.1083/jcb.202003149] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Primary cilia protrude from the cell surface and have diverse roles during development and disease, which depends on the precise timing and control of cilia assembly and disassembly. Inactivation of assembly often causes cilia defects and underlies ciliopathy, while diseases caused by dysfunction in disassembly remain largely unknown. Here, we demonstrate that CEP55 functions as a cilia disassembly regulator to participate in ciliopathy. Cep55-/- mice display clinical manifestations of Meckel-Gruber syndrome, including perinatal death, polycystic kidneys, and abnormalities in the CNS. Interestingly, Cep55-/- mice exhibit an abnormal elongation of cilia on these tissues. Mechanistically, CEP55 promotes cilia disassembly by interacting with and stabilizing Aurora A kinase, which is achieved through facilitating the chaperonin CCT complex to Aurora A. In addition, CEP55 mutation in Meckel-Gruber syndrome causes the failure of cilia disassembly. Thus, our study establishes a cilia disassembly role for CEP55 in vivo, coupling defects in cilia disassembly to ciliopathy and further suggesting that proper cilia dynamics are critical for mammalian development.
Collapse
Affiliation(s)
- Yu-Cheng Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yun-Feng Bai
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Xiao Jian
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Huai-Bin Hu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China,School of Medicine, Tsinghua University, Beijing, China
| | - Hai-Qing Tu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Na Wang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xue-Min Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China,Min Wu:
| | - Tao Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China,Tao Zhou:
| | - Hui-Yan Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China,School of Basic Medical Sciences, Fudan University, Shanghai, China,Correspondence to Hui-Yan Li:
| |
Collapse
|
17
|
Ding X, Fragoza R, Singh P, Zhang S, Yu H, Schimenti JC. Variants in RABL2A causing male infertility and ciliopathy. Hum Mol Genet 2020; 29:3402-3411. [PMID: 33075816 PMCID: PMC7749704 DOI: 10.1093/hmg/ddaa230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
Approximately 7% of men worldwide suffer from infertility, with sperm abnormalities being the most common defect. Though genetic causes are thought to underlie a substantial fraction of idiopathic cases, the actual molecular bases are usually undetermined. Because the consequences of most genetic variants in populations are unknown, this complicates genetic diagnosis even after genome sequencing of patients. Some patients with ciliopathies, including primary ciliary dyskinesia and Bardet-Biedl syndrome, also suffer from infertility because cilia and sperm flagella share several characteristics. Here, we identified two deleterious alleles of RABL2A, a gene essential for normal function of cilia and flagella. Our in silico predictions and in vitro assays suggest that both alleles destabilize the protein. We constructed and analyzed mice homozygous for these two single-nucleotide polymorphisms, Rabl2L119F (rs80006029) and Rabl2V158F (rs200121688), and found that they exhibit ciliopathy-associated disorders including male infertility, early growth retardation, excessive weight gain in adulthood, heterotaxia, pre-axial polydactyly, neural tube defects and hydrocephalus. Our study provides a paradigm for triaging candidate infertility variants in the population for in vivo functional validation, using computational, in vitro and in vivo approaches.
Collapse
Affiliation(s)
- Xinbao Ding
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Robert Fragoza
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Priti Singh
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Shu Zhang
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Haiyuan Yu
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - John C Schimenti
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
18
|
Saternos H, Ley S, AbouAlaiwi W. Primary Cilia and Calcium Signaling Interactions. Int J Mol Sci 2020; 21:E7109. [PMID: 32993148 PMCID: PMC7583801 DOI: 10.3390/ijms21197109] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
The calcium ion (Ca2+) is a diverse secondary messenger with a near-ubiquitous role in a vast array of cellular processes. Cilia are present on nearly every cell type in either a motile or non-motile form; motile cilia generate fluid flow needed for a variety of biological processes, such as left-right body patterning during development, while non-motile cilia serve as the signaling powerhouses of the cell, with vital singling receptors localized to their ciliary membranes. Much of the research currently available on Ca2+-dependent cellular actions and primary cilia are tissue-specific processes. However, basic stimuli-sensing pathways, such as mechanosensation, chemosensation, and electrical sensation (electrosensation), are complex processes entangled in many intersecting pathways; an overview of proposed functions involving cilia and Ca2+ interplay will be briefly summarized here. Next, we will focus on summarizing the evidence for their interactions in basic cellular activities, including the cell cycle, cell polarity and migration, neuronal pattering, glucose-mediated insulin secretion, biliary regulation, and bone formation. Literature investigating the role of cilia and Ca2+-dependent processes at a single-cellular level appears to be scarce, though overlapping signaling pathways imply that cilia and Ca2+ interact with each other on this level in widespread and varied ways on a perpetual basis. Vastly different cellular functions across many different cell types depend on context-specific Ca2+ and cilia interactions to trigger the correct physiological responses, and abnormalities in these interactions, whether at the tissue or the single-cell level, can result in diseases known as ciliopathies; due to their clinical relevance, pathological alterations of cilia function and Ca2+ signaling will also be briefly touched upon throughout this review.
Collapse
Affiliation(s)
| | | | - Wissam AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (H.S.); (S.L.)
| |
Collapse
|
19
|
Ross MM, Piorczynski TB, Harvey J, Burnham TS, Francis M, Larsen MW, Roe K, Hansen JM, Stark MR. Ceramide: a novel inducer for neural tube defects. Dev Dyn 2019; 248:979-996. [PMID: 31390103 DOI: 10.1002/dvdy.93] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/02/2019] [Accepted: 07/21/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Circulating plasma ceramides, a class of bioactive sphingolipids, are elevated in metabolic disorders, including obesity. Infants of women with these disorders are at 2- to 3-fold greater risk for developing a neural tube defect (NTD). This study aimed to test the effects of embryonic exposure to C2-ceramides (C2) during neural tube closure. Preliminary data shows an increase in NTDs in chick embryos after C2 exposure, and addresses potential mechanisms. RESULTS Cell and embryo models were used to examine redox shifts after ceramide exposure. While undifferentiated P19 cells were resistant to ceramide exposure, neuronally differentiated P19 cells exhibited an oxidizing shift. Consistent with these observations, GSH E h curves revealed a shift to a more oxidized state in C2 treated embryos without increasing apoptosis or changing Pax3 expression, however cell proliferation was lower. Neural tube defects were observed in 45% of chick embryos exposed to C2, compared to 12% in control embryos. CONCLUSIONS C2 exposure during critical developmental stages increased the frequency of NTDs in the avian model. Increased ROS generation in cell culture, along with the more oxidative GSH E h profiles of C2 exposed cells and embryos, support a model wherein ceramide affects neural tube closure via altered tissue redox environments.
Collapse
Affiliation(s)
- Micah M Ross
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Ted B Piorczynski
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Jamison Harvey
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Tyson S Burnham
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Morgan Francis
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Madison W Larsen
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Kyle Roe
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Jason M Hansen
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| | - Michael R Stark
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, Utah
| |
Collapse
|
20
|
Park SM, Jang HJ, Lee JH. Roles of Primary Cilia in the Developing Brain. Front Cell Neurosci 2019; 13:218. [PMID: 31139054 PMCID: PMC6527876 DOI: 10.3389/fncel.2019.00218] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/30/2019] [Indexed: 01/07/2023] Open
Abstract
Essential to development, primary cilia are microtubule-based cellular organelles that protrude from the surface of cells. Acting as cellular antenna, primary cilia play central roles in transducing or regulating several signaling pathways, including Sonic hedgehog (Shh) and Wnt signaling. Defects in primary cilia contribute to a group of syndromic disorders known as “ciliopathies” and can adversely affect development of the brain and other essential organs, including the kidneys, eyes, and liver. The molecular mechanisms of how defective primary cilia contribute to neurological defects, however, remain poorly understood. In this mini review, we summarize recent advances in understanding of the interactions between primary cilia and signaling pathways essential to cellular homeostasis and brain development.
Collapse
Affiliation(s)
- Sang Min Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hee Jin Jang
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
21
|
Yue H, Zhu X, Li S, Wang F, Wang X, Guan Z, Zhu Z, Niu B, Zhang T, Guo J, Wang J. Relationship Between INPP5E Gene Expression and Embryonic Neural Development in a Mouse Model of Neural Tube Defect. Med Sci Monit 2018; 24:2053-2059. [PMID: 29626185 PMCID: PMC5903545 DOI: 10.12659/msm.906095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background The INPP5E gene encodes for the inositol polyphosphate-5-phosphatase (INPP5E) 72 kDa protein that regulates the phosphoinositide signaling pathway and other cellular activities, but the functional role of this gene in embryonic neurodevelopment and neural tube defect (NTD) remains unclear. The aim of this study was to use a mouse model of NTD to investigate the expression levels of the INPP5E gene during neural development and the occurrence of NTD. Material/Methods In an established NTD mouse model, stereoscopy was used to look for morphological defects. Transcription and expression levels of the INPP5E gene in neural tissues were detected using real-time fluorescence quantitative polymerase chain reaction (PCR) and Western blotting in the NTD mouse embryos and compared with control mouse embryos. Results The expression levels of the INPP5E gene decreased as embryonic development progressed in the neural tissue of control mice embryos, but showed no obvious trend in the neural tissues of the NTD mouse embryos. The expression levels of the INPP5E gene in NTD mouse embryos were significantly lower compared with control embryos, at the time of neural tube closure (gestational day 11.5). Conclusions The INPP5E gene regulates the process of embryonic neural development. Abnormal levels of expression of the INPP5E gene may contribute to NTDs. Increased knowledge of the expression pattern of the INPP5E gene may lead to an advanced understanding of the molecular mechanism of embryonic neurodevelopment and identify more specific directions to explore potential treatments for NTDs associated with abnormalities in INPP5E gene expression levels.
Collapse
Affiliation(s)
- Huixuan Yue
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Xiting Zhu
- Emory Rollins School of Public Health, Atlanta, GA, USA
| | - Shen Li
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Fang Wang
- Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Xiuwei Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Zhen Guan
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Zhiqiang Zhu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Bo Niu
- Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Ting Zhang
- Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Jin Guo
- Capital Institute of Pediatrics, Beijing, China (mainland)
| | - Jianhua Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China (mainland)
| |
Collapse
|
22
|
McAllister JP, Guerra MM, Ruiz LC, Jimenez AJ, Dominguez-Pinos D, Sival D, den Dunnen W, Morales DM, Schmidt RE, Rodriguez EM, Limbrick DD. Ventricular Zone Disruption in Human Neonates With Intraventricular Hemorrhage. J Neuropathol Exp Neurol 2017; 76:358-375. [PMID: 28521038 DOI: 10.1093/jnen/nlx017] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To determine if ventricular zone (VZ) and subventricular zone (SVZ) alterations are associated with intraventricular hemorrhage (IVH) and posthemorrhagic hydrocephalus, we compared postmortem frontal and subcortical brain samples from 12 infants with IVH and 3 nonneurological disease controls without hemorrhages or ventriculomegaly. Birth and expiration estimated gestational ages were 23.0-39.1 and 23.7-44.1 weeks, respectively; survival ranges were 0-42 days (median, 2.0 days). Routine histology and immunohistochemistry for neural stem cells (NSCs), neural progenitors (NPs), multiciliated ependymal cells (ECs), astrocytes (AS), and cell adhesion molecules were performed. Controls exhibited monociliated NSCs and multiciliated ECs lining the ventricles, abundant NPs in the SVZ, and medial vs. lateral wall differences with a complex mosaic organization in the latter. In IVH cases, normal VZ/SVZ areas were mixed with foci of NSC and EC loss, eruption of cells into the ventricle, cytoplasmic transposition of N-cadherin, subependymal rosettes, and periventricular heterotopia. Mature AS populated areas believed to be sites of VZ disruption. The cytopathology and extension of the VZ disruption correlated with developmental age but not with brain hemorrhage grade or location. These results corroborate similar findings in congenital hydrocephalus in animals and humans and indicate that VZ disruption occurs consistently in premature neonates with IVH.
Collapse
Affiliation(s)
- James P McAllister
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Maria Montserrat Guerra
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Leandro Castaneyra Ruiz
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Antonio J Jimenez
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Dolores Dominguez-Pinos
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Deborah Sival
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Wilfred den Dunnen
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Diego M Morales
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Robert E Schmidt
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - Esteban M Rodriguez
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| | - David D Limbrick
- From the Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri (JPM, LCR, DMM, DDL); Instituto de Antomía, Histologia y Patologia, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile (MMG, EMR); Instituto de Biología Celular, Genética y Fisiología Facultad de Ciencias, Universidad de Malaga, Malaga, Spain and Instituto de Investigación Biomédica (IBIMA), Malaga, Spain (AJJ, DDP); Departments of Pediatrics, Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (DS, WD); Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri (RES); and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri (DDL)
| |
Collapse
|
23
|
Manojlovic Z, Earwood R, Kato A, Perez D, Cabrera OA, Didier R, Megraw TL, Stefanovic B, Kato Y. La-related protein 6 controls ciliated cell differentiation. Cilia 2017; 6:4. [PMID: 28344782 PMCID: PMC5364628 DOI: 10.1186/s13630-017-0047-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 02/16/2017] [Indexed: 01/07/2023] Open
Abstract
Background La-related protein 6 (LARP6) is an evolutionally conserved RNA-binding protein. Vertebrate LARP6 binds the 5′ stem-loop found in mRNAs encoding type I collagen to regulate their translation, but other target mRNAs and additional functions for LARP6 are unknown. The aim of this study was to elucidate an additional function of LARP6 and to evaluate the importance of its function during development. Methods To uncover the role of LARP6 in development, we utilized Morpholino Oligos to deplete LARP6 protein in Xenopus embryos. Then, embryonic phenotypes and ciliary structures of LAPR6 morphants were examined. To identify the molecular mechanism underlying ciliogenesis regulated by LARP6, we tested the expression level of cilia-related genes, which play important roles in ciliogenesis, by RT-PCR or whole mount in situ hybridization (WISH). Results We knocked down LARP6 in Xenopus embryos and found neural tube closure defects. LARP6 mutant, which compromises the collagen synthesis, could rescue these defects. Neural tube closure defects are coincident with lack of cilia, antenna-like cellular organelles with motility- or sensory-related functions, in the neural tube. The absence of cilia at the epidermis was also observed in LARP6 morphants, and this defect was due to the absence of basal bodies which are formed from centrioles and required for ciliary assembly. In the process of multi-ciliated cell (MCC) differentiation, mcidas, which activates the transcription of genes required for centriole formation during ciliogenesis, could partially restore MCCs in LARP6 morphants. In addition, LARP6 likely controls the expression of mcidas in a Notch-independent manner. Conclusions La-related protein 6 is involved in ciliated cell differentiation during development by controlling the expression of cilia-related genes including mcidas. This LARP6 function involves a mechanism that is distinct from its established role in binding to collagen mRNAs and regulating their translation. Electronic supplementary material The online version of this article (doi:10.1186/s13630-017-0047-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zarko Manojlovic
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA.,Department of Translational Genomics, Keck School of Medicine of University of Southern California, Los Angeles, CA 90089-9601 USA
| | - Ryan Earwood
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| | - Akiko Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| | - Diana Perez
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| | - Oscar A Cabrera
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| | - Ruth Didier
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| | - Timothy L Megraw
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| | - Branko Stefanovic
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| | - Yoichi Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115W. Call Street, Tallahassee, FL 32306-4300 USA
| |
Collapse
|
24
|
|
25
|
Sawardekar KP. Meckel–Gruber syndrome: prevalence from a hospital-based study in Oman. J Matern Fetal Neonatal Med 2016; 29:3696-8. [DOI: 10.3109/14767058.2016.1141883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Regulation of primary cilia formation by the ubiquitin–proteasome system. Biochem Soc Trans 2016; 44:1265-1271. [DOI: 10.1042/bst20160174] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 11/17/2022]
Abstract
Primary cilia form at the surface of most vertebrate cell types, where they are essential signalling antennae for signal transduction pathways important for development and cancer, including Hedgehog. The importance of primary cilia in development is clearly demonstrated by numerous disorders (known as ciliopathies) associated with disrupted cilia formation (ciliogenesis). Recent advances describing functional regulators of the primary cilium highlight an emerging role for the ubiquitin–proteasome system (UPS) as a key regulator of ciliogenesis. Although there are well-documented examples of E3 ubiquitin ligases and deubiquitases in the regulation of cilia proteins, many putative components remain unvalidated. This review explores current understanding of how the UPS influences primary cilia formation, and also how recent screen data have identified more putative regulators of the UPS. Emerging research has identified many promising leads in the search for regulators of this important organelle and may identify potential novel therapeutic targets for intervention in cancer and other disease contexts.
Collapse
|
27
|
Bhattacharyya S, Rainey MA, Arya P, Mohapatra BC, Mushtaq I, Dutta S, George M, Storck MD, McComb RD, Muirhead D, Todd GL, Gould K, Datta K, Gelineau-van Waes J, Band V, Band H. Endocytic recycling protein EHD1 regulates primary cilia morphogenesis and SHH signaling during neural tube development. Sci Rep 2016; 6:20727. [PMID: 26884322 PMCID: PMC4756679 DOI: 10.1038/srep20727] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/11/2016] [Indexed: 12/20/2022] Open
Abstract
Members of the four-member C-terminal EPS15-Homology Domain-containing (EHD) protein family play crucial roles in endocytic recycling of cell surface receptors from endosomes to the plasma membrane. In this study, we show that Ehd1 gene knockout in mice on a predominantly B6 background is embryonic lethal. Ehd1-null embryos die at mid-gestation with a failure to complete key developmental processes including neural tube closure, axial turning and patterning of the neural tube. We found that Ehd1-null embryos display short and stubby cilia on the developing neuroepithelium at embryonic day 9.5 (E9.5). Loss of EHD1 also deregulates the ciliary SHH signaling with Ehd1-null embryos displaying features indicative of increased SHH signaling, including a significant downregulation in the formation of the GLI3 repressor and increase in the ventral neuronal markers specified by SHH. Using Ehd1-null MEFS we found that EHD1 protein co-localizes with the SHH receptor Smoothened in the primary cilia upon ligand stimulation. Under the same conditions, EHD1 was shown to co-traffic with Smoothened into the developing primary cilia and we identify EHD1 as a direct binding partner of Smoothened. Overall, our studies identify the endocytic recycling regulator EHD1 as a novel regulator of the primary cilium-associated trafficking of Smoothened and Hedgehog signaling.
Collapse
Affiliation(s)
- Sohinee Bhattacharyya
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark A Rainey
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Priyanka Arya
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | - Samikshan Dutta
- The Department of Biochemistry &Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Manju George
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney D McComb
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Muirhead
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gordon L Todd
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Karen Gould
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kaustubh Datta
- The Department of Biochemistry &Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Vimla Band
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hamid Band
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.,The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
28
|
Kosty J, Vogel TW. Insights into the development of molecular therapies for craniosynostosis. Neurosurg Focus 2016; 38:E2. [PMID: 25929964 DOI: 10.3171/2015.2.focus155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
For the past 2 decades, clinical and basic science researchers have gained significant insights into the molecular and genetic pathways associated with common forms of craniosynostosis. This has led to invaluable information for families and physicians in their attempts to understand the heterogeneity of craniosynostosis. Genetic mutations have been identified in the fibroblast growth factor receptors (FGFRs) as well as in other targets, including TWIST1, BMP, and RUNX2. Greater understanding of these and other pathways has led to the development of innovative approaches for applying medical therapies to the treatment of craniosynostosis, in particular by maintaining suture patency. In this article, the authors discuss the molecular pathophysiological mechanisms underlying various forms of craniosynostosis. They also highlight recent developments in the field of molecular craniosynostosis research with the hope of identifying targets for medical therapies that might augment the results of surgical intervention.
Collapse
Affiliation(s)
- Jennifer Kosty
- Department of Neurosurgery, University of Cincinnati; and
| | | |
Collapse
|
29
|
Yuan X, Serra RA, Yang S. Function and regulation of primary cilia and intraflagellar transport proteins in the skeleton. Ann N Y Acad Sci 2015; 1335:78-99. [PMID: 24961486 PMCID: PMC4334369 DOI: 10.1111/nyas.12463] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Primary cilia are microtubule-based organelles that project from the cell surface to enable transduction of various developmental signaling pathways. The process of intraflagellar transport (IFT) is crucial for the building and maintenance of primary cilia. Ciliary dysfunction has been found in a range of disorders called ciliopathies, some of which display severe skeletal dysplasias. In recent years, interest has grown in uncovering the function of primary cilia/IFT proteins in bone development, mechanotransduction, and cellular regulation. We summarize recent advances in understanding the function of cilia and IFT proteins in the regulation of cell differentiation in osteoblasts, osteocytes, chondrocytes, and mesenchymal stem cells (MSCs). We also discuss the mechanosensory function of cilia and IFT proteins in bone cells, cilia orientation, and other functions of cilia in chondrocytes.
Collapse
Affiliation(s)
- Xue Yuan
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, NY
| | - Rosa A. Serra
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shuying Yang
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, NY
- Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY
| |
Collapse
|
30
|
Developmental patterns of Ki-67, Oct-4 and α-tubulin proteins expression in the human spinal cord. Acta Histochem 2014; 116:619-26. [PMID: 24373696 DOI: 10.1016/j.acthis.2013.11.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 11/21/2022]
Abstract
The aim of this study was to analyze immunohistochemically the relationships between factors involved in processes of cell proliferation (Ki-67), differentiation (Oct-4) and primary cilia formation (α-tubulin) in the two parts of the developing human spinal cord (SC) of different origin in 11 human concepti (developmental weeks 5-10). Proliferation was highest in weeks 7-8 in the dorsal ventricular zones of the cranial (85.5%) and caudal (12.1%) SC. In the ventricular (VZ), intermediate (IZ) and marginal zones (MZ) of the cranial SC, α-tubulin and Oct-4 were moderately to strongly expressed. During weeks 5-6, moderate expression of α-tubulin and Oct-4 characterized the ventral part, with mild expression in the dorsal part of the caudal SC. In weeks 7-8, their expression increased in the VZ and IZ, and decreased in the MZ. In both parts of the SC Ki-67 and α-tubulin co-localized in the VZ. Oct-4 and Ki-67 co-localized only in the ependymal cells. In the cranial SC α-tubulin and Oct-4 co-localized (VZ and IZ), while the MZ expressed only α-tubulin. In the caudal SC, α-tubulin and Oct-4 co-localized in the VZ, while in the IZ some cells were only α-tubulin-positive. We suggest the importance of temporal-spatial expression of Ki-67 for the thickening of the cranial SC lateral wall. While in the cranial part of the SC, proliferation followed a ventral-dorsal direction, the caudal SC had a more irregular pattern. α-Tubulin was associated with cilia formation (ependymal cells) and axonic elongation of neuroblasts (MZ). Primary cilia signaling are important in control of SC proliferation and differentiation. Oct-4 expression in the SC coincided with presence of dividing neuroepithelial cells in the VZ and neuroblasts in the IZ, and could control the level of SC differentiation.
Collapse
|
31
|
Manojlovic Z, Earwood R, Kato A, Stefanovic B, Kato Y. RFX7 is required for the formation of cilia in the neural tube. Mech Dev 2014; 132:28-37. [PMID: 24530844 DOI: 10.1016/j.mod.2014.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 02/04/2014] [Indexed: 12/22/2022]
Abstract
Regulatory Factor X (RFX) transcription factors are important for development and are likely involved in the pathogenesis of serious human diseases including ciliopathies. While seven RFX genes have been identified in vertebrates and several RFX transcription factors have been reported to be regulators of ciliogenesis, the role of RFX7 in development including ciliogenesis is not known. Here we show that RFX7 in Xenopus laevis is expressed in the neural tube, eye, otic vesicles, and somites. Knockdown of RFX7 in Xenopus embryos resulted in a defect of ciliogenesis in the neural tube and failure of neural tube closure. RFX7 controlled the formation of cilia by regulating the expression of RFX4 gene, which has been reported to be required for ciliogenesis in the neural tube. Moreover, ectopic expression of Foxj1, which is a master regulator of motile cilia formation, suppressed the expression of RFX4 but not RFX7. Taken together, RFX7 plays an important role in the process of neural tube closure at the top of the molecular cascade which controls ciliogenesis in the neural tube.
Collapse
Affiliation(s)
- Zarko Manojlovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Ryan Earwood
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Akiko Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Branko Stefanovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| | - Yoichi Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
32
|
Barker AR, Thomas R, Dawe HR. Meckel-Gruber syndrome and the role of primary cilia in kidney, skeleton, and central nervous system development. Organogenesis 2013; 10:96-107. [PMID: 24322779 DOI: 10.4161/org.27375] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The ciliopathies are a group of related inherited diseases characterized by malformations in organ development. The diseases affect multiple organ systems, with kidney, skeleton, and brain malformations frequently observed. Research over the last decade has revealed that these diseases are due to defects in primary cilia, essential sensory organelles found on most cells in the human body. Here we discuss the genetic and cell biological basis of one of the most severe ciliopathies, Meckel-Gruber syndrome, and explain how primary cilia contribute to the development of the affected organ systems.
Collapse
Affiliation(s)
- Amy R Barker
- College of Life and Environmental Sciences; University of Exeter; Exeter, UK
| | - Rhys Thomas
- College of Life and Environmental Sciences; University of Exeter; Exeter, UK
| | - Helen R Dawe
- College of Life and Environmental Sciences; University of Exeter; Exeter, UK
| |
Collapse
|
33
|
Wallingford JB, Niswander LA, Shaw GM, Finnell RH. The continuing challenge of understanding, preventing, and treating neural tube defects. Science 2013; 339:1222002. [PMID: 23449594 DOI: 10.1126/science.1222002] [Citation(s) in RCA: 336] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human birth defects are a major public health burden: The Center for Disease Control estimates that 1 of every 33 United States newborns presents with a birth defect, and worldwide the estimate approaches 6% of all births. Among the most common and debilitating of human birth defects are those affecting the formation of the neural tube, the precursor to the central nervous system. Neural tube defects (NTDs) arise from a complex combination of genetic and environmental interactions. Although substantial advances have been made in the prevention and treatment of these malformations, NTDs remain a substantial public health problem, and we are only now beginning to understand their etiology. Here, we review the process of neural tube development and how defects in this process lead to NTDs, both in humans and in the animal models that serve to inform our understanding of these processes. The insights we are gaining will help generate new intervention strategies to tackle the clinical challenges and to alleviate the personal and societal burdens that accompany these defects.
Collapse
Affiliation(s)
- John B Wallingford
- Howard Hughes Medical Institute, The University of Texas at Austin, Austin, TX 78712, USA.
| | | | | | | |
Collapse
|
34
|
D'Angelo A, De Angelis A, Avallone B, Piscopo I, Tammaro R, Studer M, Franco B. Ofd1 controls dorso-ventral patterning and axoneme elongation during embryonic brain development. PLoS One 2012; 7:e52937. [PMID: 23300826 PMCID: PMC3531334 DOI: 10.1371/journal.pone.0052937] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 11/26/2012] [Indexed: 01/04/2023] Open
Abstract
Oral-facial-digital type I syndrome (OFDI) is a human X-linked dominant-male-lethal developmental disorder caused by mutations in the OFD1 gene. Similar to other inherited disorders associated to ciliary dysfunction OFD type I patients display neurological abnormalities. We characterized the neuronal phenotype that results from Ofd1 inactivation in early phases of mouse embryonic development and at post-natal stages. We determined that Ofd1 plays a crucial role in forebrain development, and in particular, in the control of dorso-ventral patterning and early corticogenesis. We observed abnormal activation of Sonic hedgehog (Shh), a major pathway modulating brain development. Ultrastructural studies demonstrated that early Ofd1 inactivation results in the absence of ciliary axonemes despite the presence of mature basal bodies that are correctly orientated and docked. Ofd1 inducible-mediated inactivation at birth does not affect ciliogenesis in the cortex, suggesting a developmental stage-dependent role for a basal body protein in ciliogenesis. Moreover, we showed defects in cytoskeletal organization and apical-basal polarity in Ofd1 mutant embryos, most likely due to lack of ciliary axonemes. Thus, the present study identifies Ofd1 as a developmental disease gene that is critical for forebrain development and ciliogenesis in embryonic life, and indicates that Ofd1 functions after docking and before elaboration of the axoneme in vivo.
Collapse
Affiliation(s)
- Anna D'Angelo
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, Naples, Italy
| | - Amalia De Angelis
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, Naples, Italy
| | - Bice Avallone
- Department of Biological Science, University of Naples “Federico II”, Naples, Italy
| | - Immacolata Piscopo
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, Naples, Italy
| | - Roberta Tammaro
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, Naples, Italy
| | - Michèle Studer
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, Naples, Italy
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, Naples, Italy
- Medical Genetics, Department of Pediatrics, Federico II University, Naples, Italy
- * E-mail:
| |
Collapse
|
35
|
Carter CS, Vogel TW, Zhang Q, Seo S, Swiderski RE, Moninger TO, Cassell MD, Thedens DR, Keppler-Noreuil KM, Nopoulos P, Nishimura DY, Searby CC, Bugge K, Sheffield VC. Abnormal development of NG2+PDGFR-α+ neural progenitor cells leads to neonatal hydrocephalus in a ciliopathy mouse model. Nat Med 2012; 18:1797-804. [PMID: 23160237 PMCID: PMC3684048 DOI: 10.1038/nm.2996] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 10/12/2012] [Indexed: 12/15/2022]
Abstract
Hydrocephalus is a common neurological disorder leading to expansion of the cerebral ventricles and is associated with significant morbidity and mortality. Most neonatal cases are of unknown etiology and are likely to display complex inheritance involving multiple genes and environmental factors. Identifying molecular mechanisms for neonatal hydrocephalus and developing non-invasive treatment modalities are high priorities. Here we employ a hydrocephalic mouse model of the human ciliopathy Bardet-Biedl Syndrome (BBS) and identify a role for neural progenitors in the pathogenesis of neonatal hydrocephalus. We found that hydrocephalus in this mouse model is caused by aberrant PDGFRα signaling, resulting in increased apoptosis and impaired proliferation of NG2+PDGFRα+ neural progenitors. Targeting this pathway with lithium treatment rescued NG2+PDGFRα+ progenitor cell proliferation in BBS mutant mice, reducing ventricular volume. Our findings demonstrate that neural progenitors are critical in the pathogenesis of neonatal hydrocephalus and we identify novel therapeutic targets for this common neurological disorder.
Collapse
Affiliation(s)
- Calvin S Carter
- Graduate Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|