1
|
Bogart AH, Brooks ER. Canonical Wnt pathway modulation is required to correctly execute multiple independent cellular dynamic programs during cranial neural tube closure. Dev Biol 2025; 523:115-131. [PMID: 40280384 DOI: 10.1016/j.ydbio.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Defects in cranial neural tube closure are among the most common and deleterious human structural birth defects. Correct cranial closure requires the coordination of multiple cell dynamic programs including cell proliferation and cell shape change. Mutations that impact Wnt signaling, including loss of the pathway co-receptor LRP6, lead to defects in cranial neural tube closure, but the cellular dynamics under control of the Wnt pathway during this critical morphogenetic process remain unclear. Here, we use mice mutant for LRP6 to examine the consequences of conditional and global reduction in Wnt signaling and mutants with conditional inactivation of APC to examine the consequences of pathway hyperactivation. Strikingly, we find that regulated Wnt signaling is required for two independent events during cranial neural tube closure. First, global reduction of Wnt leads to a surprising hyperplasia of the cranial neural folds driven by excessive cell proliferation at early pre-elevation stages, with the increased tissue volume creating a mechanical blockade to efficient closure despite normal apical constriction and cell polarization at later stages. Conversely, conditional hyperactivation of the pathway at later elevation stages prevents correct actin organization, blocking apical constriction and neural fold elevation without impacting tissue scaling. Together these data reveal that Wnt signaling levels must be modulated to restrict proliferation at early stages and promote apical constriction at later elevation stages to drive efficient closure of the cranial neural tube.
Collapse
Affiliation(s)
- Amber Huffine Bogart
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, United States
| | - Eric R Brooks
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, United States.
| |
Collapse
|
2
|
Bhasker A, Veleri S. Fundamental origins of neural tube defects with a basis in genetics and nutrition. Exp Brain Res 2025; 243:79. [PMID: 40025180 DOI: 10.1007/s00221-025-07016-9] [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: 10/07/2024] [Accepted: 01/30/2025] [Indexed: 03/04/2025]
Abstract
Neural tube defects (NTDs) are leading congenital malformations. Its global prevalence is one in 1000 pregnancies and it has high morbidity and mortality. It has multiple risk factors like genetic errors and environmental stressors like maternal malnutrition and in utero exposure to pollutants like chemicals. The genetic program determines neural tube development based on timely expression of many genes involved in developmental signaling pathways like BMP, PCP and SHH. BMP expression defines ectoderm. SOX represses BMP in ectoderm and convertes to the neuroectoderm. Subsequently, PCP molecules define the tissue patterning for convergent-extension, a critical step in neural tube genesis. Further, SHH sets spatial patterning of the neural tube. Nutrients are the essential major environmental input for embryogenesis. But it may also carry risk factors. Malnutrition, especially folate deficiency, during embryogenesis is a major cause for NTDs. Folate is integral in the One Carbon metabolic pathway. Its deficiency and error in the pathway are implicated in NTDs. Folate supplementation alone is insufficient to prevent NTDs. Thus, a comprehensive understanding of the various risk factors is necessary to strategize reduction of NTDs. We review the current knowledge of various risk factors, like genetic, metabolic, nutritional, and drugs causing NTDs and discuss the steps required to identify them in the early embryogenesis to avoid NTDs.
Collapse
Affiliation(s)
- Anjusha Bhasker
- Drug Safety Division, ICMR-National Institute of Nutrition, Department of Health Research, Ministry of Health & Family Welfare, Govt. of India, Hyderabad, 500007, India
| | - Shobi Veleri
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
3
|
Palicharla VR, Badgandi HB, Hwang SH, Legué E, Liem KF, Mukhopadhyay S. A defined tubby domain β-barrel surface region of TULP3 mediates ciliary trafficking of diverse cargoes. Mol Biol Cell 2025; 36:ar1. [PMID: 39565681 PMCID: PMC11742108 DOI: 10.1091/mbc.e24-09-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
The primary cilium is a paradigmatic subcellular compartment at the nexus of numerous cellular and morphogenetic pathways. The tubby family protein TULP3 acts as an adapter of the intraflagellar transport complex A in transporting integral membrane and membrane-associated lipidated proteins into cilia. However, the mechanisms by which TULP3 coordinates ciliary transport of diverse cargoes is not well understood. Here, we provide molecular insights into TULP3-mediated ciliary cargo recognition. We screened for critical TULP3 residues by proximity biotinylation-mass spectrometry, structural analysis, and testing TULP3 variants in human patients with hepatorenal fibrocystic disease and spina bifida. The TULP3 residues we identified 1) were located on one side of the β-barrel of the tubby domain away from the phosphoinositide binding site, 2) mediated ciliary trafficking of lipidated and transmembrane cargoes, and 3) determined proximity with these cargoes in vivo without affecting ciliary localization, phosphoinositide binding or hydrodynamic properties of TULP3. Overall, these findings implicate a specific region of one of the surfaces of the TULP3 β-barrel in ciliary trafficking of diverse cargoes. This region overlooks the β-strands 8-12 of the β-barrel and is away from the membrane anchoring phosphoinositide binding site. Targeting the TULP3-cargo interactions could provide therapeutics in ciliary trafficking diseases.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Hemant B. Badgandi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Karel F. Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
4
|
Jiang Z, Chen X, Ruan L, Xu Y, Li K. Molecular analyses of the tubby-like protein gene family and their response to salt and high temperature in the foxtail millet (Setaria italica). Funct Integr Genomics 2024; 24:170. [PMID: 39317784 DOI: 10.1007/s10142-024-01458-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Tubby-like proteins (TLPs) are a group of proteins found in both eukaryotes and prokaryotes. They are significant in various physiological and biochemical processes, especially in plants' response to abiotic stress. However, the role of TLP in foxtail millet (Setaria italica) remains unclear. The millet genome has 16 members of the TLP family with typical Tub domains, which can be sorted into five subgroups based on gene structure, motif, and protein domain distribution. SiTLPs were discovered to be predominantly located in the nucleus and also had extracellular distribution. The interspecific evolutionary analysis indicated that SiTLPs had a closer evolutionary relationship with monocots and were consistent with the morphological classification of foxtail millet. When subjected to salt stress, the abundance of SiTLP was affected, and qRT-PCR results showed that the expression levels of certain SiTLP members were induced by salt stress while others remained unresponsive. Except for SiTLP14, all other SiTLP genes were up-regulated in response to high-temperature stress, implying a potentially crucial role for SiTLP in mitigating high-temperature-induced damage. This study provides valuable insights into understanding the functional significance of the TLP gene family in foxtail millet.
Collapse
Affiliation(s)
- Zhuanzhuan Jiang
- Provincial Key Laboratory of the Biodiversity Study and Ecology Conservation in Southwest Anhui, Anqing, 246133, China.
- College of Life Sciences, Anqing Normal University, Anqing, 246133, China.
| | - Xiaoqi Chen
- Provincial Key Laboratory of the Biodiversity Study and Ecology Conservation in Southwest Anhui, Anqing, 246133, China
- College of Life Sciences, Anqing Normal University, Anqing, 246133, China
| | - Lingling Ruan
- Provincial Key Laboratory of the Biodiversity Study and Ecology Conservation in Southwest Anhui, Anqing, 246133, China
- College of Life Sciences, Anqing Normal University, Anqing, 246133, China
| | - Yan Xu
- Provincial Key Laboratory of the Biodiversity Study and Ecology Conservation in Southwest Anhui, Anqing, 246133, China
- College of Life Sciences, Anqing Normal University, Anqing, 246133, China
| | - Ke Li
- Provincial Key Laboratory of the Biodiversity Study and Ecology Conservation in Southwest Anhui, Anqing, 246133, China
- College of Life Sciences, Anqing Normal University, Anqing, 246133, China
| |
Collapse
|
5
|
Reddy Palicharla V, Mukhopadhyay S. Molecular and structural perspectives on protein trafficking to the primary cilium membrane. Biochem Soc Trans 2024; 52:1473-1487. [PMID: 38864436 PMCID: PMC11346432 DOI: 10.1042/bst20231403] [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: 03/20/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/13/2024]
Abstract
The primary cilium is a dynamic subcellular compartment templated from the mother centriole or basal body. Cilia are solitary and tiny, but remarkably consequential in cellular pathways regulating proliferation, differentiation, and maintenance. Multiple transmembrane proteins such as G-protein-coupled receptors, channels, enzymes, and membrane-associated lipidated proteins are enriched in the ciliary membrane. The precise regulation of ciliary membrane content is essential for effective signal transduction and maintenance of tissue homeostasis. Surprisingly, a few conserved molecular factors, intraflagellar transport complex A and the tubby family adapter protein TULP3, mediate the transport of most membrane cargoes into cilia. Recent advances in cryogenic electron microscopy provide fundamental insights into these molecular players. Here, we review the molecular players mediating cargo delivery into the ciliary membrane through the lens of structural biology. These mechanistic insights into ciliary transport provide a framework for understanding of disease variants in ciliopathies, enable precise manipulation of cilia-mediated pathways, and provide a platform for the development of targeted therapeutics.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| |
Collapse
|
6
|
Jäger R, Geyer SH, Kavirayani A, Kiss MG, Waltenberger E, Rülicke T, Binder CJ, Weninger WJ, Kralovics R. Effects of Tulp4 deficiency on murine embryonic development and adult phenotype. Microsc Res Tech 2024; 87:854-866. [PMID: 38115643 DOI: 10.1002/jemt.24476] [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: 09/26/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Genetically engineered mouse models have the potential to unravel fundamental biological processes and provide mechanistic insights into the pathogenesis of human diseases. We have previously observed that germline genetic variation at the TULP4 locus influences clinical characteristics in patients with myeloproliferative neoplasms. To elucidate the role of TULP4 in pathological and physiological processes in vivo, we generated a Tulp4 knockout mouse model. Systemic Tulp4 deficiency exerted a strong impact on embryonic development in both Tulp4 homozygous null (Tulp4-/-) and heterozygous (Tulp4+/-) knockout mice, the former exhibiting perinatal lethality. High-resolution episcopic microscopy (HREM) of day 14.5 embryos allowed for the identification of multiple developmental defects in Tulp4-/- mice, including severe heart defects. Moreover, in Tulp4+/- embryos HREM revealed abnormalities of several organ systems, which per se do not affect prenatal or postnatal survival. In adult Tulp4+/- mice, extensive examinations of hematopoietic and cardiovascular features, involving histopathological surveys of multiple tissues as well as blood counts and immunophenotyping, did not provide evidence for anomalies as observed in corresponding embryos. Finally, evaluating a potential obesity-related phenotype as reported for other TULP family members revealed a trend for increased body weight of Tulp4+/- mice. RESEARCH HIGHLIGHTS: To study the role of the TULP4 gene in vivo, we generated a Tulp4 knockout mouse model. Correlative analyses involving HREM revealed a strong impact of Tulp4 deficiency on murine embryonic development.
Collapse
Affiliation(s)
- Roland Jäger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Stefan H Geyer
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical Imaging Cluster, Medical University of Vienna, Vienna, Austria
| | - Anoop Kavirayani
- Vienna BioCenter Core Facilities GmbH, Austrian BioImaging/CMI, Vienna, Austria
| | - Máté G Kiss
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Waltenberger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Rülicke
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang J Weninger
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical Imaging Cluster, Medical University of Vienna, Vienna, Austria
| | - Robert Kralovics
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Maniou E, Farah F, Marshall AR, Crane-Smith Z, Krstevski A, Stathopoulou A, Greene NDE, Copp AJ, Galea GL. Caudal Fgfr1 disruption produces localised spinal mis-patterning and a terminal myelocystocele-like phenotype in mice. Development 2023; 150:dev202139. [PMID: 37756583 PMCID: PMC10617625 DOI: 10.1242/dev.202139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
Closed spinal dysraphisms are poorly understood malformations classified as neural tube (NT) defects. Several, including terminal myelocystocele, affect the distal spine. We have previously identified a NT closure-initiating point, Closure 5, in the distal spine of mice. Here, we document equivalent morphology of the caudal-most closing posterior neuropore (PNP) in mice and humans. Closure 5 forms in a region of active FGF signalling, and pharmacological FGF receptor blockade impairs its formation in cultured mouse embryos. Conditional genetic deletion of Fgfr1 in caudal embryonic tissues with Cdx2Cre diminishes neuroepithelial proliferation, impairs Closure 5 formation and delays PNP closure. After closure, the distal NT of Fgfr1-disrupted embryos dilates to form a fluid-filled sac overlying ventrally flattened spinal cord. This phenotype resembles terminal myelocystocele. Histological analysis reveals regional and progressive loss of SHH- and FOXA2-positive ventral NT domains, resulting in OLIG2 labelling of the ventral-most NT. The OLIG2 domain is also subsequently lost, eventually producing a NT that is entirely positive for the dorsal marker PAX3. Thus, a terminal myelocystocele-like phenotype can arise after completion of NT closure with localised spinal mis-patterning caused by disruption of FGFR1 signalling.
Collapse
Affiliation(s)
- Eirini Maniou
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Faduma Farah
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Abigail R. Marshall
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Zoe Crane-Smith
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Andrea Krstevski
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Athanasia Stathopoulou
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Nicholas D. E. Greene
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Andrew J. Copp
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Gabriel L. Galea
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| |
Collapse
|
8
|
Bi Y, Ren D, Yuan F, Zhang Z, Zhou D, Yi X, Ji L, Li K, Yang F, Wu X, Li X, Xu Y, Liu Y, Wang P, Cai C, Liu C, Ma Q, He L, Shi Y, He G. TULP4, a novel E3 ligase gene, participates in neuronal migration as a candidate in schizophrenia. CNS Neurosci Ther 2023. [PMID: 37650344 DOI: 10.1111/cns.14423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/26/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND TUB-like protein 4 (TULP4) is one of the distant members of tubby family proteins, whose function remains largely unknown. In the present study, we intend to identify the role of TULP4 in schizophrenia from human samples and animal models. METHODS Whole-exome sequencing was used to detect the four schizophrenia families collected. In different cell lines, the effects of identified variants in TULP4 gene on its expression and localization were analyzed. Knockdown models in utero and adult mice were employed to investigate the role of Tulp4 on neuronal migration and schizophrenia-related behavior. Subsequently, co-IP assays were used to search for proteins that interact with TULP4 and the effects of mutants on the molecular function of TULP4. RESULTS For the first time, we identified five rare variants in TULP4 from schizophrenia families, of which three significantly reduced TULP4 protein expression. Knockdown the expression of Tulp4 delayed neuronal migration during embryological development and consequently triggered abnormal behaviors in adult mice, including impaired sensorimotor gating and cognitive dysfunction. Furthermore, we confirmed that TULP4 is involved in the formation of a novel E3 ligase through interaction with CUL5-ELOB/C-RNF7 and the three deleterious variants affected the binding amount of TULP4 and CUL5 to a certain extent. CONCLUSIONS Together, we believe TULP4 plays an important role in neurodevelopment and subsequent schizophrenic-related phenotypes through its E3 ubiquitin ligase function.
Collapse
Affiliation(s)
- Yan Bi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Decheng Ren
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Yuan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Burning Rock Biotech, Guangzhou, China
| | - Daizhan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Yi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Ji
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keyi Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengping Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifeng Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Wang
- Wuhu Fourth People's Hospital, Wuhu, China
| | | | - Chuanxin Liu
- School of Mental Health, Jining Medical University, Jining, China
| | - Qian Ma
- Laboratory Animal Centre, Shanghai Jiao Tong University, Shanghai, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai, China
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Rai S, Leydier L, Sharma S, Katwala J, Sahu A. A quest for genetic causes underlying signaling pathways associated with neural tube defects. Front Pediatr 2023; 11:1126209. [PMID: 37284286 PMCID: PMC10241075 DOI: 10.3389/fped.2023.1126209] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/28/2023] [Indexed: 06/08/2023] Open
Abstract
Neural tube defects (NTDs) are serious congenital deformities of the nervous system that occur owing to the failure of normal neural tube closures. Genetic and non-genetic factors contribute to the etiology of neural tube defects in humans, indicating the role of gene-gene and gene-environment interaction in the occurrence and recurrence risk of neural tube defects. Several lines of genetic studies on humans and animals demonstrated the role of aberrant genes in the developmental risk of neural tube defects and also provided an understanding of the cellular and morphological programs that occur during embryonic development. Other studies observed the effects of folate and supplementation of folic acid on neural tube defects. Hence, here we review what is known to date regarding altered genes associated with specific signaling pathways resulting in NTDs, as well as highlight the role of various genetic, and non-genetic factors and their interactions that contribute to NTDs. Additionally, we also shine a light on the role of folate and cell adhesion molecules (CAMs) in neural tube defects.
Collapse
Affiliation(s)
- Sunil Rai
- Department of Molecular Biology, Medical University of the Americas, Charlestown, Saint Kitts and Nevis
| | - Larissa Leydier
- Department of Molecular Biology, Medical University of the Americas, Charlestown, Saint Kitts and Nevis
| | - Shivani Sharma
- Department of Molecular Biology, Medical University of the Americas, Charlestown, Saint Kitts and Nevis
| | - Jigar Katwala
- Department of Molecular Biology, Medical University of the Americas, Charlestown, Saint Kitts and Nevis
| | - Anurag Sahu
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
10
|
Yan L, Yin H, Mi Y, Wu Y, Zheng Y. Deficiency of Wdr60 and Wdr34 cause distinct neural tube malformation phenotypes in early embryos. Front Cell Dev Biol 2023; 11:1084245. [PMID: 37228654 PMCID: PMC10203710 DOI: 10.3389/fcell.2023.1084245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Cilia are specialized organelles that extend from plasma membrane, functioning as antennas for signal transduction and are involved in embryonic morphogenesis. Dysfunction of cilia lead to many developmental defects, including neural tube defects (NTDs). Heterodimer WDR60-WDR34 (WD repeat domain 60 and 34) are intermediate chains of motor protein dynein-2, which play important roles in ciliary retrograde transport. It has been reported that disruption of Wdr34 in mouse model results in NTDs and defects of Sonic Hedgehog (SHH) signaling. However, no Wdr60 deficiency mouse model has been reported yet. In this study, piggyBac (PB) transposon is used to interfere Wdr60 and Wdr34 expression respectively to establish Wdr60 PB/PB and Wdr34 PB/PB mouse models. We found that the expression of Wdr60 or Wdr34 is significantly decreased in the homozygote mice. Wdr60 homozygote mice die around E13.5 to E14.5, while Wdr34 homozygote mice die around E10.5 to E11.5. WDR60 is highly expressed in the head region at E10.5 and Wdr60 PB/PB embryos have head malformation. RNAseq and qRT-PCR experiments revealed that Sonic Hedgehog signaling is also downregulated in Wdr60 PB/PB head tissue, demonstrating that WDR60 is also required for promoting SHH signaling. Further experiments on mouse embryos also revealed that the expression levels of planar cell polarity (PCP) components such as CELSR1 and downstream signal molecule c-Jun were downregulated in WDR34 homozygotes compared to wildtype littermates. Coincidently, we observed much higher ratio of open cranial and caudal neural tube in Wdr34 PB/PB mice. CO-IP experiment showed that WDR60 and WDR34 both interact with IFT88, but only WDR34 interacts with IFT140. Taken together, WDR60 and WDR34 play overlapped and distinct functions in modulating neural tube development.
Collapse
Affiliation(s)
- Lu Yan
- Obstetrics and Gynecology Hospital, The Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
- Department of Cellular and Developmental Biology, School of Life Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Hailing Yin
- Obstetrics and Gynecology Hospital, The Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
- Obstetrics Department of the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yiwei Mi
- Department of Cellular and Developmental Biology, School of Life Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yu Wu
- Department of Cellular and Developmental Biology, School of Life Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yufang Zheng
- Obstetrics and Gynecology Hospital, The Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China
- Department of Cellular and Developmental Biology, School of Life Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Palicharla VR, Hwang SH, Somatilaka BN, Legué E, Shimada IS, Familiari NE, Tran VM, Woodruff JB, Liem KF, Mukhopadhyay S. Interactions between TULP3 tubby domain and ARL13B amphipathic helix promote lipidated protein transport to cilia. Mol Biol Cell 2023; 34:ar18. [PMID: 36652335 PMCID: PMC10011728 DOI: 10.1091/mbc.e22-10-0473] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The primary cilium is a nexus for cell signaling and relies on specific protein trafficking for function. The tubby family protein TULP3 transports integral membrane proteins into cilia through interactions with the intraflagellar transport complex-A (IFT-A) and phosphoinositides. It was previously shown that short motifs called ciliary localization sequences (CLSs) are necessary and sufficient for TULP3-dependent ciliary trafficking of transmembrane cargoes. However, the mechanisms by which TULP3 regulates ciliary compartmentalization of nonintegral, membrane-associated proteins and whether such trafficking requires TULP3-dependent CLSs is unknown. Here we show that TULP3 is required for ciliary transport of the Joubert syndrome-linked palmitoylated GTPase ARL13B through a CLS. An N-terminal amphipathic helix, preceding the GTPase domain of ARL13B, couples with the TULP3 tubby domain for ciliary trafficking, irrespective of palmitoylation. ARL13B transport requires TULP3 binding to IFT-A but not to phosphoinositides, indicating strong membrane-proximate interactions, unlike transmembrane cargo transport requiring both properties of TULP3. TULP3-mediated trafficking of ARL13B also regulates ciliary enrichment of farnesylated and myristoylated downstream effectors of ARL13B. The lipidated cargoes show distinctive depletion kinetics from kidney epithelial cilia with relation to Tulp3 deletion-induced renal cystogenesis. Overall, these findings indicate an expanded role of the tubby domain in capturing analogous helical secondary structural motifs from diverse cargoes.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Issei S Shimada
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nicole E Familiari
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Vanna M Tran
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jeffrey B Woodruff
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Karel F Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
12
|
Jafari Khamirani H, Palicharla VR, Dastgheib SA, Dianatpour M, Imanieh MH, Tabei SS, Besse W, Mukhopadhyay S, Liem KF. A pathogenic variant of TULP3 causes renal and hepatic fibrocystic disease. Front Genet 2022; 13:1021037. [PMID: 36276950 PMCID: PMC9585244 DOI: 10.3389/fgene.2022.1021037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022] Open
Abstract
Patient variants in Tubby Like Protein-3 (TULP3) have recently been associated with progressive fibrocystic disease in tissues and organs. TULP3 is a ciliary trafficking protein that links membrane-associated proteins to the intraflagellar transport complex A. In mice, mutations in Tulp3 drive phenotypes consistent with ciliary dysfunction which include renal cystic disease, as part of a ciliopathic spectrum. Here we report two sisters from consanguineous parents with fibrocystic renal and hepatic disease harboring a homozygous missense mutation in TULP3 (NM_003324.5: c.1144C>T, p.Arg382Trp). The R382W patient mutation resides within the C-terminal Tubby domain, a conserved domain required for TULP3 to associate with phosphoinositides. We show that inner medullary collecting duct-3 cells expressing the TULP3 R382W patient variant have a severely reduced ability to localize the membrane-associated proteins ARL13b, INPP5E, and GPR161 to the cilium, consistent with a loss of TULP3 function. These studies establish Arginine 382 as a critical residue in the Tubby domain, which is essential for TULP3-mediated protein trafficking within the cilium, and expand the phenotypic spectrum known to result from recessive deleterious mutations in TULP3.
Collapse
Affiliation(s)
| | - Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | | | - Mehdi Dianatpour
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Imanieh
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Sajjad Tabei
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Whitney Besse
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT, United States
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Karel F. Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
13
|
Fan L, Jin P, Qian Y, Shen G, Shen X, Dong M. Case Report: Prenatal Diagnosis of Postaxial Polydactyly With Bi-Allelic Variants in Smoothened (SMO). Front Genet 2022; 13:887082. [PMID: 35812756 PMCID: PMC9257524 DOI: 10.3389/fgene.2022.887082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/13/2022] [Indexed: 12/02/2022] Open
Abstract
Postaxial polydactyly is a common congenital malformation which involves complex genetic factors. This retrospective study analyzed the cytogenetic and molecular results of a Chinese fetus diagnosed with postaxial polydactyly of all four limbs. Fetal karyotyping and chromosomal microarray analysis (CMA) did not find any abnormality while trio whole-exome sequencing (trio-WES) identified bi-allelic variants in smoothened (SMO) and (NM_005631.5: c.1219C > G, NP_005622.1: p. Pro407Ala, and NM_005631.5: c.1619C > T, NP_005622.1: p. Ala540Val). Sanger sequencing validated these variants. The mutations are highly conserved across multiple species. In-depth bioinformatics analysis and familial co-segregation implied the compound heterozygous variants as the likely cause of postaxial polydactyly in this fetus. Our findings provided the basis for genetic counseling and will contribute to a better understanding of the complex genetic mechanism that underlies postaxial polydactyly.
Collapse
Affiliation(s)
- Lihong Fan
- Center of Prenatal Diagnosis, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Pengzhen Jin
- Women’s Hospital, School of Medicine Zhejiang University, Hangzhou, China
| | - Yeqing Qian
- Women’s Hospital, School of Medicine Zhejiang University, Hangzhou, China
| | - Guosong Shen
- Center of Prenatal Diagnosis, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Xueping Shen
- Center of Prenatal Diagnosis, Huzhou Maternity & Child Health Care Hospital, Huzhou, China
| | - Minyue Dong
- Women’s Hospital, School of Medicine Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
- *Correspondence: Minyue Dong,
| |
Collapse
|
14
|
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
|
15
|
Song J, Fu Q, Liu G, Zhang C, Wang Y, Tao S, Liu R, Li Z. TULP3 silencing suppresses cell proliferation, migration and invasion in gastric cancer via the PTEN/Akt/Snail pathway. Cancer Treat Res Commun 2022; 31:100551. [PMID: 35344762 DOI: 10.1016/j.ctarc.2022.100551] [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: 01/15/2022] [Revised: 03/11/2022] [Accepted: 03/20/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Tubby-like protein 3 (TULP3) is a member of the tubby family, has been related to the development of nervous system by gene knockout researches. Nevertheless, the role of TULP3 in the gastric cancer is not clear. METHODS Western blotting and real-time polymerase chain reaction (PCR) were employed for the quantitative detection of TULP3 expression in the gastric cancer and consecutive non-cancerous tissues, and gastric cancer cells. The roles of TULP3 in invasion, migration as well as proliferation of the gastric cancer cell in vivo and in vitro through utilizing colony formation, MTT, wound-healing, transwell and mouse xenograft model. Western blotting assay was implemented in order to clarify the potential molecular mechanisms. Furthermore, electron microscopy and western blot were evaluated TULP3 expression in gastric cancer patient extracted serum exosomes. RESULTS TULP3 expression levels were remarkably upregulated in the gastric cancer tissues and cells. Subsequent functional assays demonstrated that TULP3 downregulation suppressed invasion, migration as well as the proliferation of the gastric cancer cell. Mechanism assays depicted that the PTEN/Akt/Snail signaling pathway can inhibit invasion, migration as well as the proliferation of the gastric cancer cell via TULP3 silencing. Finally, we found that the expression of TULP3 could be determined in the extracted serum exosomes. The expression of TULP3 in gastric cancer group was higher in comparison with normal group. CONCLUSIONS Our results reveal that TULP3 might serve as a potential prognostic biomarker and therapeutic target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Jun Song
- Department of Laboratory Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Qingsheng Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Gang Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Chengxiong Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Yingying Wang
- Department of Nuclear medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Shaoneng Tao
- Department of Nuclear medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China.
| | - Rui Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China.
| | - Zhi Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China.
| |
Collapse
|
16
|
Liu Y, Yuan Q, Wang Z, Ding L, Kong N, Liu J, Hu Y, Zhang Y, Li C, Yan G, Jiang Y, Sun H. A high level of KLF12 causes folic acid-resistant neural tube defects by activating the Shh signaling pathway in mice†. Biol Reprod 2021; 105:837-845. [PMID: 34104947 DOI: 10.1093/biolre/ioab111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/26/2021] [Accepted: 06/02/2021] [Indexed: 11/13/2022] Open
Abstract
Although adequate periconceptional folic acid (FA) supplementation has reduced the occurrence of pregnancies affected by neural tube defects (NTDs), the mechanisms underlying FA-resistant NTDs are poorly understood, and thus NTDs still remain a global public health concern. A high level of Krüppel-like factor 12 (KLF12) exerts deleterious effects on heath in most cases, but evidence for its roles in development has not been published. We observed KLF12-overexpressing mice showed disturbed neural tube development. KLF12-overexpressing fetuses died in utero at approximately 10.5 days post-coitus, with 100% presenting cranial NTDs. Neither FA nor formate promoted normal neural tube closure in mutant fetuses. The RNA-seq results showed that a high level of KLF12 caused NTDs in mice via overactivating the sonic hedgehog (Shh) signaling pathway, leading to the upregulation of patched 1, GLI-Krüppel family member GLI1, hedgehog-interacting protein, etc., whereas FA metabolism-related enzymes did not express differently. PF-5274857, an antagonist of the Shh signaling pathway, significantly promoted dorsolateral hinge point formation and partially rescued the NTDs. The regulatory hierarchy between a high level of KLF12 and FA-resistant NTDs might provide new insights into the diagnosis and treatment of unexplained NTDs in the future.
Collapse
Affiliation(s)
- Yang Liu
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Qiong Yuan
- Department of Obstetrics and Gynecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Zhilong Wang
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Lijun Ding
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Na Kong
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Jingyu Liu
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Yali Hu
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Yang Zhang
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Chaojun Li
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Guijun Yan
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yue Jiang
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Haixiang Sun
- Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, Jiangsu, People's Republic of China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Reproductive Medicine Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
17
|
Ma N, Pan J, Wen Y, Wu Q, Yu B, Chen X, Wan J, Zhang W. RETRACTED: circTulp4 functions in Alzheimer’s disease pathogenesis by regulating its parental gene, Tulp4. Mol Ther 2021; 29:2167-2181. [PMID: 33578037 PMCID: PMC8178447 DOI: 10.1016/j.ymthe.2021.02.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 12/20/2020] [Accepted: 02/04/2021] [Indexed: 01/04/2023] Open
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern has been raised by a reader that research reported in the article duplicates figures and reuses data from prior publications without attribution. After investigation of the concerns raised, the editors found the following reuse of data without attribution: Figure 1B in this article is identical to Ma et al., 2020, FASEB J. 34, 10342–10356, https://doi.org/10.1096/fj.201903157R, Figure 2B. Figure 1D in this article is identical to Ma et al., 2019, Mol. Ther. Nucleic Acids 18, 1049–1062, https://doi.org/10.1016/j.omtn.2019.10.030, Figure 2A. Figure 1F in this article is identical to Ma et al., 2019, Mol. Ther. Nucleic Acids 18, 1049–1062, https://doi.org/10.1016/j.omtn.2019.10.030, Figure 2B. Figure 1A in this article reuses data from Ma et al., 2020, FASEB J. 34, 10342–10356, https://doi.org/10.1096/fj.201903157R, Figure 2E. Figure S1 in this article is identical to Ma et al., 2019, Mol. Ther. Nucleic Acids 18, 1049–1062, https://doi.org/10.1016/j.omtn.2019.10.030, Figure 3 One of the conditions of submission of a paper for publication is that authors declare explicitly that the paper has not been previously published and is not under consideration for publication elsewhere. As such this article represents a misuse of the scientific publishing system. The corresponding authors have agreed to the retraction.
Collapse
Affiliation(s)
- Nana Ma
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China; Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Beijing, China
| | - Jie Pan
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China; Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Yi Wen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Qi Wu
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Bo Yu
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China; Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Xi Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jun Wan
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China; Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China; Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, China.
| | - Wei Zhang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China; Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China.
| |
Collapse
|
18
|
Hall ET, Dillard ME, Stewart DP, Zhang Y, Wagner B, Levine RM, Pruett-Miller SM, Sykes A, Temirov J, Cheney RE, Mori M, Robinson CG, Ogden SK. Cytoneme delivery of Sonic Hedgehog from ligand-producing cells requires Myosin 10 and a Dispatched-BOC/CDON co-receptor complex. eLife 2021; 10:61432. [PMID: 33570491 PMCID: PMC7968926 DOI: 10.7554/elife.61432] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Morphogens function in concentration-dependent manners to instruct cell fate during tissue patterning. The cytoneme morphogen transport model posits that specialized filopodia extend between morphogen-sending and responding cells to ensure that appropriate signaling thresholds are achieved. How morphogens are transported along and deployed from cytonemes, how quickly a cytoneme-delivered, receptor-dependent signal is initiated, and whether these processes are conserved across phyla are not known. Herein, we reveal that the actin motor Myosin 10 promotes vesicular transport of Sonic Hedgehog (SHH) morphogen in mouse cell cytonemes, and that SHH morphogen gradient organization is altered in neural tubes of Myo10-/- mice. We demonstrate that cytoneme-mediated deposition of SHH onto receiving cells induces a rapid, receptor-dependent signal response that occurs within seconds of ligand delivery. This activity is dependent upon a novel Dispatched (DISP)-BOC/CDON co-receptor complex that functions in ligand-producing cells to promote cytoneme occurrence and facilitate ligand delivery for signal activation.
Collapse
Affiliation(s)
- Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Miriam E Dillard
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Daniel P Stewart
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Yan Zhang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Ben Wagner
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, United States
| | - Rachel M Levine
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - April Sykes
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, United States
| | - Jamshid Temirov
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Richard E Cheney
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, United States
| | - Motomi Mori
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, United States
| | - Camenzind G Robinson
- Cell and Tissue Imaging Center, St. Jude Children's Research Hospital, Memphis, United States
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| |
Collapse
|
19
|
Kopinke D, Norris AM, Mukhopadhyay S. Developmental and regenerative paradigms of cilia regulated hedgehog signaling. Semin Cell Dev Biol 2021; 110:89-103. [PMID: 32540122 PMCID: PMC7736055 DOI: 10.1016/j.semcdb.2020.05.029] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 01/08/2023]
Abstract
Primary cilia are immotile appendages that have evolved to receive and interpret a variety of different extracellular cues. Cilia play crucial roles in intercellular communication during development and defects in cilia affect multiple tissues accounting for a heterogeneous group of human diseases called ciliopathies. The Hedgehog (Hh) signaling pathway is one of these cues and displays a unique and symbiotic relationship with cilia. Not only does Hh signaling require cilia for its function but the majority of the Hh signaling machinery is physically located within the cilium-centrosome complex. More specifically, cilia are required for both repressing and activating Hh signaling by modifying bifunctional Gli transcription factors into repressors or activators. Defects in balancing, interpreting or establishing these repressor/activator gradients in Hh signaling either require cilia or phenocopy disruption of cilia. Here, we will summarize the current knowledge on how spatiotemporal control of the molecular machinery of the cilium allows for a tight control of basal repression and activation states of the Hh pathway. We will then discuss several paradigms on how cilia influence Hh pathway activity in tissue morphogenesis during development. Last, we will touch on how cilia and Hh signaling are being reactivated and repurposed during adult tissue regeneration. More specifically, we will focus on mesenchymal stem cells within the connective tissue and discuss the similarities and differences of how cilia and ciliary Hh signaling control the formation of fibrotic scar and adipose tissue during fatty fibrosis of several tissues.
Collapse
Affiliation(s)
- Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA.
| | - Alessandra M Norris
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
20
|
Ishida Y, Kobayashi T, Chiba S, Katoh Y, Nakayama K. Molecular basis of ciliary defects caused by compound heterozygous IFT144/WDR19 mutations found in cranioectodermal dysplasia. Hum Mol Genet 2021; 30:213-225. [PMID: 33517396 DOI: 10.1093/hmg/ddab034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/28/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Primary cilia contain specific proteins to achieve their functions as cellular antennae. Ciliary protein trafficking is mediated by the intraflagellar transport (IFT) machinery containing the IFT-A and IFT-B complexes. Mutations in genes encoding the IFT-A subunits (IFT43, IFT121/WDR35, IFT122, IFT139/TTC21B, IFT140 and IFT144/WDR19) often result in skeletal ciliopathies, including cranioectodermal dysplasia (CED). We here characterized the molecular and cellular defects of CED caused by compound heterozygous mutations in IFT144 [the missense variant IFT144(L710S) and the nonsense variant IFT144(R1103*)]. These two variants were distinct with regard to their interactions with other IFT-A subunits and with the IFT-B complex. When exogenously expressed in IFT144-knockout (KO) cells, IFT144(L710S) as well as IFT144(WT) rescued both moderately compromised ciliogenesis and the abnormal localization of ciliary proteins. As the homozygous IFT144(L710S) mutation was found to cause autosomal recessive retinitis pigmentosa, IFT144(L710S) is likely to be hypomorphic at the cellular level. In striking contrast, the exogenous expression of IFT144(R1103*) in IFT144-KO cells exacerbated the ciliogenesis defects. The expression of IFT144(R1103*) together with IFT144(WT) restored the abnormal phenotypes of IFT144-KO cells. However, the coexpression of IFT144(R1103*) with the hypomorphic IFT144(L710S) variant in IFT144-KO cells, which mimics the genotype of compound heterozygous CED patients, resulted in severe ciliogenesis defects. Taken together, these observations demonstrate that compound heterozygous mutations in IFT144 cause severe ciliary defects via a complicated mechanism, where one allele can cause severe ciliary defects when combined with a hypomorphic allele.
Collapse
Affiliation(s)
- Yamato Ishida
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takuya Kobayashi
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shuhei Chiba
- Department of Genetic Disease Research, Graduate School of Medicine, Osaka City University, Abeno-ku, Osaka 545-8585, Japan
| | - Yohei Katoh
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuhisa Nakayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
21
|
Cousminer DL, Wagley Y, Pippin JA, Elhakeem A, Way GP, Pahl MC, McCormack SE, Chesi A, Mitchell JA, Kindler JM, Baird D, Hartley A, Howe L, Kalkwarf HJ, Lappe JM, Lu S, Leonard ME, Johnson ME, Hakonarson H, Gilsanz V, Shepherd JA, Oberfield SE, Greene CS, Kelly A, Lawlor DA, Voight BF, Wells AD, Zemel BS, Hankenson KD, Grant SFA. Genome-wide association study implicates novel loci and reveals candidate effector genes for longitudinal pediatric bone accrual. Genome Biol 2021; 22:1. [PMID: 33397451 PMCID: PMC7780623 DOI: 10.1186/s13059-020-02207-9] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Bone accrual impacts lifelong skeletal health, but genetic discovery has been primarily limited to cross-sectional study designs and hampered by uncertainty about target effector genes. Here, we capture this dynamic phenotype by modeling longitudinal bone accrual across 11,000 bone scans in a cohort of healthy children and adolescents, followed by genome-wide association studies (GWAS) and variant-to-gene mapping with functional follow-up. RESULTS We identify 40 loci, 35 not previously reported, with various degrees of supportive evidence, half residing in topological associated domains harboring known bone genes. Of several loci potentially associated with later-life fracture risk, a candidate SNP lookup provides the most compelling evidence for rs11195210 (SMC3). Variant-to-gene mapping combining ATAC-seq to assay open chromatin with high-resolution promoter-focused Capture C identifies contacts between GWAS loci and nearby gene promoters. siRNA knockdown of gene expression supports the putative effector gene at three specific loci in two osteoblast cell models. Finally, using CRISPR-Cas9 genome editing, we confirm that the immediate genomic region harboring the putative causal SNP influences PRPF38A expression, a location which is predicted to coincide with a set of binding sites for relevant transcription factors. CONCLUSIONS Using a new longitudinal approach, we expand the number of genetic loci putatively associated with pediatric bone gain. Functional follow-up in appropriate cell models finds novel candidate genes impacting bone accrual. Our data also raise the possibility that the cell fate decision between osteogenic and adipogenic lineages is important in normal bone accrual.
Collapse
Affiliation(s)
- Diana L Cousminer
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Yadav Wagley
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James A Pippin
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ahmed Elhakeem
- MRC Integrative Epidemiology Unit, Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gregory P Way
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, 02140, USA
| | - Matthew C Pahl
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shana E McCormack
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan A Mitchell
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joseph M Kindler
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Denis Baird
- MRC Integrative Epidemiology Unit, Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - April Hartley
- MRC Integrative Epidemiology Unit, Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Laura Howe
- MRC Integrative Epidemiology Unit, Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Heidi J Kalkwarf
- Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - Joan M Lappe
- Department of Medicine and College of Nursing, Creighton University School of Medicine, Omaha, NB, USA
| | - Sumei Lu
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michelle E Leonard
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew E Johnson
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Vicente Gilsanz
- Center for Endocrinology, Diabetes & Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - John A Shepherd
- Department of Epidemiology and Population Science, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Sharon E Oberfield
- Division of Pediatric Endocrinology, Columbia University Medical Center, New York, NY, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Childhood Cancer Data Lab, Alex's Lemonade Stand Foundation, Philadelphia, PA, USA
| | - Andrea Kelly
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Babette S Zemel
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kurt D Hankenson
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Struan F A Grant
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Brooks ER, Islam MT, Anderson KV, Zallen JA. Sonic hedgehog signaling directs patterned cell remodeling during cranial neural tube closure. eLife 2020; 9:60234. [PMID: 33103996 PMCID: PMC7655103 DOI: 10.7554/elife.60234] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022] Open
Abstract
Neural tube closure defects are a major cause of infant mortality, with exencephaly accounting for nearly one-third of cases. However, the mechanisms of cranial neural tube closure are not well understood. Here, we show that this process involves a tissue-wide pattern of apical constriction controlled by Sonic hedgehog (Shh) signaling. Midline cells in the mouse midbrain neuroepithelium are flat with large apical surfaces, whereas lateral cells are taller and undergo synchronous apical constriction, driving neural fold elevation. Embryos lacking the Shh effector Gli2 fail to produce appropriate midline cell architecture, whereas embryos with expanded Shh signaling, including the IFT-A complex mutants Ift122 and Ttc21b and embryos expressing activated Smoothened, display apical constriction defects in lateral cells. Disruption of lateral, but not midline, cell remodeling results in exencephaly. These results reveal a morphogenetic program of patterned apical constriction governed by Shh signaling that generates structural changes in the developing mammalian brain.
Collapse
Affiliation(s)
- Eric R Brooks
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Mohammed Tarek Islam
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, United States
| |
Collapse
|
23
|
Legué E, Liem KF. Mutations in Ciliary Trafficking Genes affect Sonic Hedgehog-dependent Neural Tube Patterning Differentially along the Anterior-Posterior Axis. Neuroscience 2020; 450:3-14. [PMID: 32682825 DOI: 10.1016/j.neuroscience.2020.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/30/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022]
Abstract
Cell specification in the ventral spinal cord is a well-studied model system to understand how tissue pattern develops in response to a morphogen gradient. Ventral cell types including motor neurons (MNs) are induced in the neural tube in response to graded Sonic Hedgehog (Shh) signaling. We performed a forward genetic screen in the mouse that incorporated a GFP-expressing transgene to visualize MNs to identify genes regulating ventral patterning. Here we contrast the neural patterning phenotypes of two mouse lines carrying induced mutations in ciliary trafficking genes. We show that a hypomorphic mutation in the gene Tubby-like protein 3 (Tulp3) resulted in a dorsal expansion of MNs consistent with an up-regulation of Shh signaling. Interestingly, patterning defects in Tulp3 mutants were restricted to posterior regions of the spinal cord as patterning was similar to WT in the anterior spinal cord. In contrast, a mutation in the ciliary trafficking gene cytoplasmic dynein 2 heavy chain 1 (Dync2h1), led to a complete loss of MNs in anterior regions of the spinal cord, indicating a strong down-regulation of Shh signaling. However, this severe phenotype was restricted to the cervical region as MNs developed posteriorly. Mutations in cilia trafficking genes affect Shh-dependent signaling in the neural tube differentially along the anterior-posterior (A-P) axis in a process that is not understood.
Collapse
Affiliation(s)
- Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, United States
| | - Karel F Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, United States.
| |
Collapse
|
24
|
Constable S, Long AB, Floyd KA, Schurmans S, Caspary T. The ciliary phosphatidylinositol phosphatase Inpp5e plays positive and negative regulatory roles in Shh signaling. Development 2020; 147:dev.183301. [PMID: 31964774 DOI: 10.1242/dev.183301] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/02/2020] [Indexed: 01/04/2023]
Abstract
Sonic hedgehog (Shh) signal transduction specifies ventral cell fates in the neural tube and is mediated by the Gli transcription factors that play both activator (GliA) and repressor (GliR) roles. Cilia are essential for Shh signal transduction and the ciliary phosphatidylinositol phosphatase Inpp5e is linked to Shh regulation. In the course of a forward genetic screen for recessive mouse mutants, we identified a functional null allele of inositol polyphosphate-5-phosphatase E (Inpp5e), ridge top (rdg), with expanded ventral neural cell fates at E10.5. By E12.5, Inpp5erdg/rdg embryos displayed normal neural patterning and this correction over time required Gli3, the predominant repressor in neural patterning. Inpp5erdg function largely depended on the presence of cilia and on smoothened, the obligate transducer of Shh signaling, indicating that Inpp5e functions within the cilium to regulate the pathway. These data indicate that Inpp5e plays a more complicated role in Shh signaling than previously appreciated. We propose that Inpp5e attenuates Shh signaling in the neural tube through regulation of the relative timing of GliA and GliR production, which is important in understanding how the duration of Shh signaling regulates neural tube patterning.
Collapse
Affiliation(s)
- Sandii Constable
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Alyssa B Long
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Katharine A Floyd
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA-R Centre, Université de Liège, Liège 4000, Belgium
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
25
|
Abstract
Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical Hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this Review, we highlight central mechanisms by which primary cilia coordinate HH, G protein-coupled receptor, WNT, receptor tyrosine kinase and transforming growth factor-β (TGFβ)/bone morphogenetic protein (BMP) signalling and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.
Collapse
|
26
|
Ma M, Legué E, Tian X, Somlo S, Liem KF. Cell-Autonomous Hedgehog Signaling Is Not Required for Cyst Formation in Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2019; 30:2103-2111. [PMID: 31451534 DOI: 10.1681/asn.2018121274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/15/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND PKD1 or PKD2, the two main causal genes for autosomal dominant polycystic kidney disease (ADPKD), encode the multipass transmembrane proteins polycystin-1 (PC1) and polycystin-2 (PC2), respectively. Polycystins localize to the primary cilium, an organelle essential for cell signaling, including signal transduction of the Hedgehog pathway. Mutations in ciliary genes that build and maintain the cilium also cause renal cystic disease through unknown pathways. Although recent studies have found alterations in Hedgehog signaling in ADPKD-related models and tissues, the relationship between Hedgehog and polycystic kidney disease is not known. METHODS To examine the potential role of cell-autonomous Hedgehog signaling in regulating kidney cyst formation in vivo in both early- and adult-onset mouse models of ADPKD, we used conditional inactivation of Pkd1 combined with conditional modulation of Hedgehog signaling components in renal epithelial cells, where mutations in Pkd1 initiate cyst formation. After increasing or decreasing levels of Hedgehog signaling in cells that underwent inactivation of Pkd1, we evaluated the effects of these genetic manipulations on quantitative parameters of polycystic kidney disease severity. RESULTS We found that in Pkd1 conditional mutant mouse kidneys, neither downregulation nor activation of the Hedgehog pathway in epithelial cells along the nephron significantly influenced the severity of the polycystic kidney phenotype in mouse models of developmental or adult-onset of ADPKD. CONCLUSIONS These data suggest that loss of Pkd1 function results in kidney cysts through pathways that are not affected by the activity of the Hedgehog pathway.
Collapse
Affiliation(s)
- Ming Ma
- Departments of Internal Medicine
| | - Emilie Legué
- Pediatrics, and.,Vertebrate Developmental Biology Program, Yale University, New Haven, Connecticut
| | - Xin Tian
- Departments of Internal Medicine
| | | | - Karel F Liem
- Pediatrics, and .,Vertebrate Developmental Biology Program, Yale University, New Haven, Connecticut
| |
Collapse
|
27
|
Legué E, Liem KF. Tulp3 Is a Ciliary Trafficking Gene that Regulates Polycystic Kidney Disease. Curr Biol 2019; 29:803-812.e5. [PMID: 30799240 DOI: 10.1016/j.cub.2019.01.054] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/12/2018] [Accepted: 01/21/2019] [Indexed: 12/17/2022]
Abstract
The primary cilium is an organelle essential for cell signaling pathways. One of the most common human genetic diseases is autosomal dominant polycystic kidney disease (ADPKD), which is caused by mutations in the PKD1 or PKD2 genes that encode Polycystin 1 and 2 (PC1/2), transmembrane proteins that translocate to the cilium. Mutations in genes that disrupt ciliogenesis also cause kidney cysts as part of a "ciliopathic" disease spectrum. The molecular mechanisms that link cilia function with renal cystic diseases are not well understood, and the mechanistic relationship between ADPKD and ciliopathic PKD is not known. Here we identify the gene Tubby-like protein-3 (Tulp3) as a key regulator of renal cystic disease from a forward genetic screen in the mouse. Mice homozygous for a hypomorphic missense mutation within the conserved Tubby domain of Tulp3 develop cysts at late embryonic stages, leading to severe postnatal loss of kidney function. In contrast to other ciliopathic disease models, Tulp3 mutations do not affect ciliogenesis. Instead, we demonstrate that Tulp3 is essential for the trafficking of the Joubert syndrome-associated small GTPase Arl13b into kidney cilia. We show that reduction of Pkd1 dosage promotes cystogenesis in the Tulp3 conditional ciliopathic PKD model. However, in an adult model of ADPKD utilizing inducible conditional Pkd1 deletion, concomitant removal of Tulp3 surprisingly ameliorates cystic disease. Therefore, Tulp3 controls distinct ciliary pathways that positively or negatively regulate cystogenesis depending on the cellular context.
Collapse
Affiliation(s)
- Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Karel F Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
28
|
Kim SE, Lei Y, Hwang SH, Wlodarczyk BJ, Mukhopadhyay S, Shaw GM, Ross ME, Finnell RH. Dominant negative GPR161 rare variants are risk factors of human spina bifida. Hum Mol Genet 2019; 28:200-208. [PMID: 30256984 PMCID: PMC6321953 DOI: 10.1093/hmg/ddy339] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/18/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022] Open
Abstract
Spina bifida (SB) is a complex disorder of failed neural tube closure during the first month of human gestation, with a suspected etiology involving multiple gene and environmental interactions. GPR161 is a ciliary G-protein coupled receptor that regulates Sonic Hedgehog (Shh) signaling. Gpr161 null and hypomorphic mutations cause neural tube defects (NTDs) in mouse models. Herein we show that several genes involved in Shh and Wnt signaling were differentially expressed in the Gpr161 null embryos using RNA-seq analysis. To determine whether there exists an association between GPR161 and SB in humans, we performed direct Sanger sequencing on the GPR161 gene in a cohort of 384 SB patients and 190 healthy controls. We identified six rare variants of GPR161 in six SB cases, of which two of the variants were novel and did not exist in any databases. Both of these variants were predicted to be damaging by SIFT and/or PolyPhen analysis. The novel GPR161 rare variants mislocalized to the primary cilia, dysregulated Shh and Wnt signaling and inhibited cell proliferation in vitro. Our results demonstrate that GPR161 mutations cause NTDs via dysregulation of Shh and Wnt signaling in mice, and novel rare variants of GPR161 can be risk factors for SB in humans.
Collapse
Affiliation(s)
- Sung-Eun Kim
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, USA
| | - Yunping Lei
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, USA
- Departments of Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sun-Hee Hwang
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bogdan J Wlodarczyk
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, USA
- Departments of Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Richard H Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, USA
- Departments of Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
29
|
Insights into the Etiology of Mammalian Neural Tube Closure Defects from Developmental, Genetic and Evolutionary Studies. J Dev Biol 2018; 6:jdb6030022. [PMID: 30134561 PMCID: PMC6162505 DOI: 10.3390/jdb6030022] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023] Open
Abstract
The human neural tube defects (NTD), anencephaly, spina bifida and craniorachischisis, originate from a failure of the embryonic neural tube to close. Human NTD are relatively common and both complex and heterogeneous in genetic origin, but the genetic variants and developmental mechanisms are largely unknown. Here we review the numerous studies, mainly in mice, of normal neural tube closure, the mechanisms of failure caused by specific gene mutations, and the evolution of the vertebrate cranial neural tube and its genetic processes, seeking insights into the etiology of human NTD. We find evidence of many regions along the anterior–posterior axis each differing in some aspect of neural tube closure—morphology, cell behavior, specific genes required—and conclude that the etiology of NTD is likely to be partly specific to the anterior–posterior location of the defect and also genetically heterogeneous. We revisit the hypotheses explaining the excess of females among cranial NTD cases in mice and humans and new developments in understanding the role of the folate pathway in NTD. Finally, we demonstrate that evidence from mouse mutants strongly supports the search for digenic or oligogenic etiology in human NTD of all types.
Collapse
|
30
|
Shimada IS, Mukhopadhyay S. G-protein-coupled receptor signaling and neural tube closure defects. Birth Defects Res 2018; 109:129-139. [PMID: 27731925 DOI: 10.1002/bdra.23567] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Disruption of the normal mechanisms that mediate neural tube closure can result in neural tube defects (NTDs) with devastating consequences in affected patients. With the advent of next-generation sequencing, we are increasingly detecting mutations in multiple genes in NTD cases. However, our ability to determine which of these genes contribute to the malformation is limited by our understanding of the pathways controlling neural tube closure. G-protein-coupled receptors (GPCRs) comprise the largest family of transmembrane receptors in humans and have been historically favored as drug targets. Recent studies implicate several GPCRs and downstream signaling pathways in neural tube development and closure. In this review, we will discuss our current understanding of GPCR signaling pathways in pathogenesis of NTDs. Notable examples include the orphan primary cilia-localized GPCR, Gpr161 that regulates the basal suppression machinery of sonic hedgehog pathway by means of activation of cAMP-protein kinase A signaling in the neural tube, and protease-activated receptors that are activated by a local network of membrane-tethered proteases during neural tube closure involving the surface ectoderm. Understanding the role of these GPCR-regulated pathways in neural tube development and closure is essential toward identification of underlying genetic causes to prevent NTDs. Birth Defects Research 109:129-139, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Issei S Shimada
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
31
|
Garcia G, Raleigh DR, Reiter JF. How the Ciliary Membrane Is Organized Inside-Out to Communicate Outside-In. Curr Biol 2018; 28:R421-R434. [PMID: 29689227 PMCID: PMC6434934 DOI: 10.1016/j.cub.2018.03.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cilia, organelles that move to execute functions like fertilization and signal to execute functions like photoreception and embryonic patterning, are composed of a core of nine-fold doublet microtubules overlain by a membrane. Distinct types of cilia display distinct membrane morphologies, ranging from simple domed cylinders to the highly ornate invaginations and membrane disks of photoreceptor outer segments. Critical for the ability of cilia to signal, both the protein and the lipid compositions of ciliary membranes are different from those of other cellular membranes. This specialization presents a unique challenge for the cell as, unlike membrane-bounded organelles, the ciliary membrane is contiguous with the surrounding plasma membrane. This distinct ciliary membrane is generated in concert with multiple membrane remodeling events that comprise the process of ciliogenesis. Once the cilium is formed, control of ciliary membrane composition relies on discrete molecular machines, including a barrier to membrane proteins entering the cilium at a specialized region of the base of the cilium called the transition zone and a trafficking adaptor that controls G protein-coupled receptor (GPCR) localization to the cilium called the BBSome. The ciliary membrane can be further remodeled by the removal of membrane proteins by the release of ciliary extracellular vesicles that may function in intercellular communication, removal of unneeded proteins or ciliary disassembly. Here, we review the structures and transport mechanisms that control ciliary membrane composition, and discuss how membrane specialization enables the cilium to function as the antenna of the cell.
Collapse
Affiliation(s)
- Galo Garcia
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - David R Raleigh
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA; Department of Radiation Oncology, University of California, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
32
|
Sahakyan V, Duelen R, Tam WL, Roberts SJ, Grosemans H, Berckmans P, Ceccarelli G, Pelizzo G, Broccoli V, Deprest J, Luyten FP, Verfaillie CM, Sampaolesi M. Folic Acid Exposure Rescues Spina Bifida Aperta Phenotypes in Human Induced Pluripotent Stem Cell Model. Sci Rep 2018; 8:2942. [PMID: 29440666 PMCID: PMC5811493 DOI: 10.1038/s41598-018-21103-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/30/2018] [Indexed: 12/30/2022] Open
Abstract
Neural tube defects (NTDs) are severe congenital abnormalities, caused by failed closure of neural tube during early embryonic development. Periconceptional folic acid (FA) supplementation greatly reduces the risk of NTDs. However, the molecular mechanisms behind NTDs and the preventive role of FA remain unclear. Here, we use human induced pluripotent stem cells (iPSCs) derived from fetuses with spina bifida aperta (SBA) to study the pathophysiology of NTDs and explore the effects of FA exposure. We report that FA exposure in SBA model is necessary for the proper formation and maturation of neural tube structures and robust differentiation of mesodermal derivatives. Additionally, we show that the folate antagonist methotrexate dramatically affects the formation of neural tube structures and FA partially reverts this aberrant phenotype. In conclusion, we present a novel model for human NTDs and provide evidence that it is a powerful tool to investigate the molecular mechanisms underlying NTDs, test drugs for therapeutic approaches.
Collapse
Affiliation(s)
- Vardine Sahakyan
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology Unit, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Robin Duelen
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology Unit, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Wai Long Tam
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, and Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Scott J Roberts
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, and Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London, The Royal National Orthopaedic Hospital, London, UK
| | - Hanne Grosemans
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology Unit, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Pieter Berckmans
- Stem Cell Institute and Stem Cell Biology and Embryology Unit, Department Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Gabriele Ceccarelli
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Gloria Pelizzo
- Pediatric Surgery Department, Istituto Mediterraneo di Eccellenza Pediatrica (ISMEP), Children's Hospital "G di Cristina", Palermo, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
- CNR-Institute of Neuroscience, Milan, Italy
| | - Jan Deprest
- Department of Obstetrics and Gynecology, Division Woman and Child, Fetal Medicine Unit, University Hospitals KU Leuven, Leuven, Belgium
- Institute for Women's Health (IWH), University College London, London, United Kingdom
| | - Frank P Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, and Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Catherine M Verfaillie
- Stem Cell Institute and Stem Cell Biology and Embryology Unit, Department Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology Unit, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
33
|
Wang M, Xu Z, Kong Y. The tubby-like proteins kingdom in animals and plants. Gene 2018; 642:16-25. [PMID: 29109004 DOI: 10.1016/j.gene.2017.10.077] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/15/2017] [Accepted: 10/27/2017] [Indexed: 11/28/2022]
|
34
|
Hwang SH, White KA, Somatilaka BN, Shelton JM, Richardson JA, Mukhopadhyay S. The G protein-coupled receptor Gpr161 regulates forelimb formation, limb patterning and skeletal morphogenesis in a primary cilium-dependent manner. Development 2018; 145:dev.154054. [PMID: 29222391 DOI: 10.1242/dev.154054] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022]
Abstract
The role of basal suppression of the sonic hedgehog (Shh) pathway and its interaction with Indian hedgehog (Ihh) signaling during limb/skeletal morphogenesis is not well understood. The orphan G protein-coupled receptor Gpr161 localizes to primary cilia and functions as a negative regulator of Shh signaling by promoting Gli transcriptional repressor versus activator formation. Here, we show that forelimb buds are not formed in Gpr161 knockout mouse embryos despite establishment of prospective limb fields. Limb-specific deletion of Gpr161 resulted in prematurely expanded Shh signaling and ectopic Shh-dependent patterning defects resulting in polysyndactyly. In addition, endochondral bone formation in forearms, including formation of both trabecular bone and bone collar was prevented. Endochondral bone formation defects resulted from accumulation of proliferating round/periarticular-like chondrocytes, lack of differentiation into columnar chondrocytes, and corresponding absence of Ihh signaling. Gpr161 deficiency in craniofacial mesenchyme also prevented intramembranous bone formation in calvarium. Defects in limb patterning, endochondral and intramembranous skeletal morphogenesis were suppressed in the absence of cilia. Overall, Gpr161 promotes forelimb formation, regulates limb patterning, prevents periarticular chondrocyte proliferation and drives osteoblastogenesis in intramembranous bones in a cilium-dependent manner.
Collapse
Affiliation(s)
- Sun-Hee Hwang
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kevin A White
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | - John M Shelton
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | - Saikat Mukhopadhyay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
35
|
Abstract
Signaling pathways direct organogenesis, often through concentration-dependent effects on cells. The hedgehog pathway enables cells to sense and respond to hedgehog ligands, of which the best studied is sonic hedgehog. Hedgehog signaling is essential for development, proliferation, and stem cell maintenance, and it is a driver of certain cancers. Lipid metabolism has a profound influence on both hedgehog signal transduction and the properties of the ligands themselves, leading to changes in the strength of hedgehog signaling and cellular functions. Here we review the evolving understanding of the relationship between lipids and hedgehog signaling.
Collapse
Affiliation(s)
- Robert Blassberg
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - John Jacob
- Nuffield Department of Clinical Neurosciences (NDCN), Level 6, West Wing, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK. .,Department of Neurology, West Wing, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK. .,Milton Keynes University Hospital, Standing Way, Eaglestone, Milton Keynes, MK6 5LD, UK.
| |
Collapse
|
36
|
Zhao L, Wang L, Chi C, Lan W, Su Y. The emerging roles of phosphatases in Hedgehog pathway. Cell Commun Signal 2017; 15:35. [PMID: 28931407 PMCID: PMC5607574 DOI: 10.1186/s12964-017-0191-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/14/2017] [Indexed: 01/12/2023] Open
Abstract
Hedgehog signaling is evolutionarily conserved and plays a pivotal role in cell fate determination, embryonic development, and tissue renewal. As aberrant Hedgehog signaling is tightly associated with a broad range of human diseases, its activities must be precisely controlled. It has been known that several core components of Hedgehog pathway undergo reversible phosphorylations mediated by protein kinases and phosphatases, which acts as an effective regulatory mechanism to modulate Hedgehog signal activities. In contrast to kinases that have been extensively studied in these phosphorylation events, phosphatases were thought to function in an unspecific manner, thus obtained much less emphasis in the past. However, in recent years, increasing evidence has implicated that phosphatases play crucial and specific roles in the context of developmental signaling, including Hedgehog signaling. In this review, we present a summary of current progress on phosphatase studies in Hedgehog pathway, emphasizing the multiple employments of protein serine/threonine phosphatases during the transduction of morphogenic Hedgehog signal in both Drosophila and vertebrate systems, all of which provide insights into the importance of phosphatases in the specific regulation of Hedgehog signaling.
Collapse
Affiliation(s)
- Long Zhao
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Liguo Wang
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Chunli Chi
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Wenwen Lan
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
37
|
Nakagawa N, Li J, Yabuno-Nakagawa K, Eom TY, Cowles M, Mapp T, Taylor R, Anton ES. APC sets the Wnt tone necessary for cerebral cortical progenitor development. Genes Dev 2017; 31:1679-1692. [PMID: 28916710 PMCID: PMC5647938 DOI: 10.1101/gad.302679.117] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/21/2017] [Indexed: 01/10/2023]
Abstract
Nakagawa et al. show that the maintenance of appropriate β-catenin-mediated Wnt tone is necessary for the orderly differentiation of cortical progenitors and the resultant formation of the cerebral cortex. Adenomatous polyposis coli (APC) regulates the activity of β-catenin, an integral component of Wnt signaling. However, the selective role of the APC–β-catenin pathway in cerebral cortical development is unknown. Here we genetically dissected the relative contributions of APC-regulated β-catenin signaling in cortical progenitor development, a necessary early step in cerebral cortical formation. Radial progenitor-specific inactivation of the APC–β-catenin pathway indicates that the maintenance of appropriate β-catenin-mediated Wnt tone is necessary for the orderly differentiation of cortical progenitors and the resultant formation of the cerebral cortex. APC deletion deregulates β-catenin, leads to high Wnt tone, and disrupts Notch1 signaling and primary cilium maintenance necessary for radial progenitor functions. β-Catenin deregulation directly disrupts cilium maintenance and signaling via Tulp3, essential for intraflagellar transport of ciliary signaling receptors. Surprisingly, deletion of β-catenin or inhibition of β-catenin activity in APC-null progenitors rescues the APC-null phenotype. These results reveal that APC-regulated β-catenin activity in cortical progenitors sets the appropriate Wnt tone necessary for normal cerebral cortical development.
Collapse
Affiliation(s)
- Naoki Nakagawa
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Jingjun Li
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Keiko Yabuno-Nakagawa
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Tae-Yeon Eom
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Martis Cowles
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Tavien Mapp
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Robin Taylor
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - E S Anton
- University of North Carolina Neuroscience Center, Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
38
|
Mitchison HM, Valente EM. Motile and non-motile cilia in human pathology: from function to phenotypes. J Pathol 2017; 241:294-309. [PMID: 27859258 DOI: 10.1002/path.4843] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 12/13/2022]
Abstract
Ciliopathies are inherited human disorders caused by both motile and non-motile cilia dysfunction that form an important and rapidly expanding disease category. Ciliopathies are complex conditions to diagnose, being multisystem disorders characterized by extensive genetic heterogeneity and clinical variability with high levels of lethality. There is marked phenotypic overlap among distinct ciliopathy syndromes that presents a major challenge for their recognition, diagnosis, and clinical management, in addition to posing an on-going task to develop the most appropriate family counselling. The impact of next-generation sequencing and high-throughput technologies in the last decade has significantly improved our understanding of the biological basis of ciliopathy disorders, enhancing our ability to determine the possible reasons for the extensive overlap in their symptoms and genetic aetiologies. Here, we review the diverse functions of cilia in human health and disease and discuss a growing shift away from the classical clinical definitions of ciliopathy syndromes to a more functional categorization. This approach arises from our improved understanding of this unique organelle, revealed through new genetic and cell biological insights into the discrete functioning of subcompartments of the cilium (basal body, transition zone, intraflagellar transport, motility). Mutations affecting these distinct ciliary protein modules can confer different genetic diseases and new clinical classifications are possible to define, according to the nature and extent of organ involvement. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hannah M Mitchison
- Genetics and Genomic Medicine Programme, University College London, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Enza Maria Valente
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy.,Neurogenetics Unit, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 00143, Rome, Italy
| |
Collapse
|
39
|
Bangs F, Anderson KV. Primary Cilia and Mammalian Hedgehog Signaling. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028175. [PMID: 27881449 DOI: 10.1101/cshperspect.a028175] [Citation(s) in RCA: 430] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
It has been a decade since it was discovered that primary cilia have an essential role in Hedgehog (Hh) signaling in mammals. This discovery came from screens in the mouse that identified a set of genes that are required for both normal Hh signaling and for the formation of primary cilia. Since then, dozens of mouse mutations have been identified that disrupt cilia in a variety of ways and have complex effects on Hedgehog signaling. Here, we summarize the genetic and developmental studies used to deduce how Hedgehog signal transduction is linked to cilia and the complex effects that perturbation of cilia structure can have on Hh signaling. We conclude by describing the current status of our understanding of the cell-type-specific regulation of ciliogenesis and how that determines the ability of cells to respond to Hedgehog ligands.
Collapse
Affiliation(s)
- Fiona Bangs
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
40
|
Kim S, Sung HJ, Lee JW, Kim YH, Oh YS, Yoon KA, Heo K, Suh PG. C-terminally mutated tubby protein accumulates in aggresomes. BMB Rep 2017; 50:37-42. [PMID: 27697107 PMCID: PMC5319663 DOI: 10.5483/bmbrep.2017.50.1.140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Indexed: 11/24/2022] Open
Abstract
The tubby protein (Tub), a putative transcription factor, plays important roles in the maintenance and function of neuronal cells. A splicing defect-causing mutation in the 3′-end of the tubby gene, which is predicted to disrupt the carboxy-terminal region of the Tub protein, causes maturity-onset obesity, blindness, and deafness in mice. Although this pathological Tub mutation leads to a loss of function, the precise mechanism has not yet been investigated. Here, we found that the mutant Tub proteins were mostly localized to puncta found in the perinuclear region and that the C-terminus was important for its solubility. Immunocytochemical analysis revealed that puncta of mutant Tub co-localized with the aggresome. Moreover, whereas wild-type Tub was translocated to the nucleus by extracellular signaling, the mutant forms failed to undergo such translocation. Taken together, our results suggest that the malfunctions of the Tub mutant are caused by its misfolding and subsequent localization to aggresomes.
Collapse
Affiliation(s)
- Sunshin Kim
- Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Ho Jin Sung
- Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Ji Won Lee
- Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Yun Hee Kim
- Research Institute and Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea
| | - Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Kyong-Ah Yoon
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Kyun Heo
- Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Pann-Ghill Suh
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Korea
| |
Collapse
|
41
|
Lange A, Müller GB. Polydactyly in Development, Inheritance, and Evolution. QUARTERLY REVIEW OF BIOLOGY 2017; 92:1-38. [DOI: 10.1086/690841] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
42
|
Genetic Research of Hand Congenital Deformities and Advancement in Plastic and Reconstructive Treatment. Plast Reconstr Surg 2017. [DOI: 10.1007/978-981-10-5101-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
43
|
Hong CJ, Hamilton BA. Zfp423 Regulates Sonic Hedgehog Signaling via Primary Cilium Function. PLoS Genet 2016; 12:e1006357. [PMID: 27727273 PMCID: PMC5065120 DOI: 10.1371/journal.pgen.1006357] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 09/09/2016] [Indexed: 11/17/2022] Open
Abstract
Zfp423 encodes a 30-zinc finger transcription factor that intersects several canonical signaling pathways. Zfp423 mutations result in ciliopathy-related phenotypes, including agenesis of the cerebellar vermis in mice and Joubert syndrome (JBTS19) and nephronophthisis (NPHP14) in humans. Unlike most ciliopathy genes, Zfp423 encodes a nuclear protein and its developmental expression is complex, leading to alternative proposals for cellular mechanisms. Here we show that Zfp423 is expressed by cerebellar granule cell precursors, that loss of Zfp423 in these precursors leads to cell-intrinsic reduction in proliferation, loss of response to Shh, and primary cilia abnormalities that include diminished frequency of both Smoothened and IFT88 localization. Loss of Zfp423 alters expression of several genes encoding key cilium components, including increased expression of Tulp3. Tulp3 is a direct binding target of Zfp423 and reducing the overexpression of Tulp3 in Zfp423-deficient cells suppresses Smoothened translocation defects. These results define Zfp423 deficiency as a bona fide ciliopathy, acting upstream of Shh signaling, and indicate a mechanism intrinsic to granule cell precursors for the resulting cerebellar hypoplasia.
Collapse
Affiliation(s)
- Chen-Jei Hong
- Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Department of Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Moores UCSD Cancer Center, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Institute for Genomic Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America
| | - Bruce A Hamilton
- Department of Cellular & Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Department of Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Moores UCSD Cancer Center, University of California, San Diego School of Medicine, La Jolla, California, United States of America.,Institute for Genomic Medicine, University of California, San Diego School of Medicine, La Jolla, California, United States of America
| |
Collapse
|
44
|
Hayashi S, Akiyama R, Wong J, Tahara N, Kawakami H, Kawakami Y. Gata6-Dependent GLI3 Repressor Function is Essential in Anterior Limb Progenitor Cells for Proper Limb Development. PLoS Genet 2016; 12:e1006138. [PMID: 27352137 PMCID: PMC4924869 DOI: 10.1371/journal.pgen.1006138] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/31/2016] [Indexed: 01/20/2023] Open
Abstract
Gli3 is a major regulator of Hedgehog signaling during limb development. In the anterior mesenchyme, GLI3 is proteolytically processed into GLI3R, a truncated repressor form that inhibits Hedgehog signaling. Although numerous studies have identified mechanisms that regulate Gli3 function in vitro, it is not completely understood how Gli3 function is regulated in vivo. In this study, we show a novel mechanism of regulation of GLI3R activities in limb buds by Gata6, a member of the GATA transcription factor family. We show that conditional inactivation of Gata6 prior to limb outgrowth by the Tcre deleter causes preaxial polydactyly, the formation of an anterior extra digit, in hindlimbs. A recent study suggested that Gata6 represses Shh transcription in hindlimb buds. However, we found that ectopic Hedgehog signaling precedes ectopic Shh expression. In conjunction, we observed Gata6 and Gli3 genetically interact, and compound heterozygous mutants develop preaxial polydactyly without ectopic Shh expression, indicating an additional prior mechanism to prevent polydactyly. These results support the idea that Gata6 possesses dual roles during limb development: enhancement of Gli3 repressor function to repress Hedgehog signaling in the anterior limb bud, and negative regulation of Shh expression. Our in vitro and in vivo studies identified that GATA6 physically interacts with GLI3R to facilitate nuclear localization of GLI3R and repressor activities of GLI3R. Both the genetic and biochemical data elucidates a novel mechanism by Gata6 to regulate GLI3R activities in the anterior limb progenitor cells to prevent polydactyly and attain proper development of the mammalian autopod. Gli3 is a major regulator of Hedgehog signaling in the limb, where Gli3 counteracts Sonic hedgehog (Shh) for patterning and proliferative expansion of limb progenitor cells. In the anterior limb mesenchyme, GLI3 is proteolytically processed into GLI3R, a truncated repressor form that inhibits Hedgehog signaling. In this study, we show a novel mechanism of regulation of GLI3R activities in limb buds by Gata6, a member of GATA transcription factor family. Conditional inactivation of Gata6 in mice caused formation of an extra digit in the anterior hindlimbs, a common congenital limb malformation. This phenotype was associated with ectopic Hedgehog signaling activation, and later ectopic Shh expression, in the anterior of hindlimb buds. We show that Gata6; Gli3 compound heterozygous mutants developed anterior extradigit without ectopic Shh expression, indicating there to be an additional and prior mechanism before ectopic Shh activation that induces extradigit formation. We identified that GATA6 physically interacts with GLI3R and that the interaction facilitates nuclear localization of GLI3R and repressor activities of GLI3R. Therefore, our study identified a novel mechanism by Gata6 to regulate GLI3R activities in the anterior limb mesenchyme to prevent extra digit formation and proper development of the mammalian autopod.
Collapse
Affiliation(s)
- Shinichi Hayashi
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ryutaro Akiyama
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Julia Wong
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Naoyuki Tahara
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Hiroko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
45
|
Francesca LC, Claudia R, Molinario C, Annamaria M, Chiara F, Natalia C, Emanuele A, Valentina P, Giovanni N, Costantino R, Eugenio S, Fiorella G. Variants in TNIP1, a regulator of the NF-kB pathway, found in two patients with neural tube defects. Childs Nerv Syst 2016; 32:1061-7. [PMID: 27125519 DOI: 10.1007/s00381-016-3087-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/11/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Neural tube defects (NTDs) occur in 1:1000 births. The etiology is complex, with the influence of environmental and genetic factors. Environmental factors, such as folate deficiency, diabetes, or hypoxia strongly contribute to the occurrence of NTD. Also, there is a strong genetic contribution to NTD, as highlighted by the number of genes so far identified in several different developmental pathways usually altered in NTD. Each gene identified so far accounts for a small percentage of all NTD cases, indicating a very high heterogeneity. METHODS Exome sequencing was performed in seven sporadic patients with severe mielomeningocele. Novel coding variants shared by two or more patients were selected for further analysis. RESULTS We identified in two unrelated patients two different variants in TNIP1, a gene not previously involved in NTD whose main role is downregulation of the NF-kB pathway. One variant, c.1089T>G (p.Phe363Leu), is de novo, whereas the c.1781C>T (p.Pro594Leu) is absent in the mother, but could not be tested in the father, as he was unavailable. The latter variant is a very rare variant in the ExAC database. CONCLUSIONS These findings suggest that TNIP1 is a new potential predisposing gene to spina bifida (SB) and its pathway needs to be investigated in human NTD in order to confirm its role and to plan appropriate counseling to families.
Collapse
Affiliation(s)
- La Carpia Francesca
- Department of Pathology and Cell Biology, Columbia University Medical Center, Columbia, NY, USA
| | - Rendeli Claudia
- Istituto di Pediatria, Università Cattolica del Sacro Cuore "A. Gemelli", Rome, Italy
| | - Clelia Molinario
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore "A. Gemelli", L.go Francesco Vito 1, 00168, Rome, Italy
| | - Milillo Annamaria
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore "A. Gemelli", L.go Francesco Vito 1, 00168, Rome, Italy
| | - Farroni Chiara
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore "A. Gemelli", L.go Francesco Vito 1, 00168, Rome, Italy
| | - Cannelli Natalia
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore "A. Gemelli", L.go Francesco Vito 1, 00168, Rome, Italy
| | - Ausili Emanuele
- Istituto di Pediatria, Università Cattolica del Sacro Cuore "A. Gemelli", Rome, Italy
| | - Paolucci Valentina
- Istituto di Pediatria, Università Cattolica del Sacro Cuore "A. Gemelli", Rome, Italy
| | - Neri Giovanni
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore "A. Gemelli", L.go Francesco Vito 1, 00168, Rome, Italy
| | - Romagnoli Costantino
- Istituto di Pediatria, Università Cattolica del Sacro Cuore "A. Gemelli", Rome, Italy
| | - Sangiorgi Eugenio
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore "A. Gemelli", L.go Francesco Vito 1, 00168, Rome, Italy
| | - Gurrieri Fiorella
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore "A. Gemelli", L.go Francesco Vito 1, 00168, Rome, Italy.
| |
Collapse
|
46
|
Hilgendorf KI, Johnson CT, Jackson PK. The primary cilium as a cellular receiver: organizing ciliary GPCR signaling. Curr Opin Cell Biol 2016; 39:84-92. [PMID: 26926036 DOI: 10.1016/j.ceb.2016.02.008] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/12/2022]
Abstract
The primary cilium is an antenna-like cellular protrusion mediating sensory and neuroendocrine signaling. Its localization within tissue architecture and a growing list of cilia-localized receptors, in particular G-protein-coupled receptors, determine a host of crucial physiologies, which are disrupted in human ciliopathies. Here, we discuss recent advances in the identification and characterization of ciliary signaling components and pathways. Recent studies have highlighted the unique signaling environment of the primary cilium and we are just beginning to understand how this design allows for highly amplified and regulated signaling.
Collapse
Affiliation(s)
- Keren I Hilgendorf
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carl T Johnson
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stem Cell and Regenerative Medicine PhD Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter K Jackson
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
47
|
Garcia-Gonzalo FR, Phua SC, Roberson EC, Garcia G, Abedin M, Schurmans S, Inoue T, Reiter JF. Phosphoinositides Regulate Ciliary Protein Trafficking to Modulate Hedgehog Signaling. Dev Cell 2015; 34:400-409. [PMID: 26305592 DOI: 10.1016/j.devcel.2015.08.001] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/01/2015] [Accepted: 08/05/2015] [Indexed: 12/17/2022]
Abstract
Primary cilia interpret vertebrate Hedgehog (Hh) signals. Why cilia are essential for signaling is unclear. One possibility is that some forms of signaling require a distinct membrane lipid composition, found at cilia. We found that the ciliary membrane contains a particular phosphoinositide, PI(4)P, whereas a different phosphoinositide, PI(4,5)P2, is restricted to the membrane of the ciliary base. This distribution is created by Inpp5e, a ciliary phosphoinositide 5-phosphatase. Without Inpp5e, ciliary PI(4,5)P2 levels are elevated and Hh signaling is disrupted. Inpp5e limits the ciliary levels of inhibitors of Hh signaling, including Gpr161 and the PI(4,5)P2-binding protein Tulp3. Increasing ciliary PI(4,5)P2 levels or conferring the ability to bind PI(4)P on Tulp3 increases the ciliary localization of Tulp3. Lowering Tulp3 in cells lacking Inpp5e reduces ciliary Gpr161 levels and restores Hh signaling. Therefore, Inpp5e regulates ciliary membrane phosphoinositide composition, and Tulp3 reads out ciliary phosphoinositides to control ciliary protein localization, enabling Hh signaling.
Collapse
Affiliation(s)
- Francesc R Garcia-Gonzalo
- Department of Biochemistry and Biophysics and Cardiovascular Research Institute, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Siew C Phua
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Elle C Roberson
- Department of Biochemistry and Biophysics and Cardiovascular Research Institute, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Galo Garcia
- Department of Biochemistry and Biophysics and Cardiovascular Research Institute, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Monika Abedin
- Department of Biochemistry and Biophysics and Cardiovascular Research Institute, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA-Research Centre, Université de Liège, 4000-Liège, Belgium
| | - Takanari Inoue
- Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics and Cardiovascular Research Institute, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| |
Collapse
|
48
|
Tsurubuchi T, Allender EV, Siddiqui MR, Shim KW, Ichi S, Boshnjaku V, Mania-Farnell B, Xi G, Finnell RH, McLone DG, Tomita T, Mayanil CS. A critical role of noggin in developing folate-nonresponsive NTD in Fkbp8 -/- embryos. Childs Nerv Syst 2014; 30:1343-53. [PMID: 24817375 DOI: 10.1007/s00381-014-2428-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 04/16/2014] [Indexed: 01/07/2023]
Abstract
PURPOSE Maternal folate intake has reduced the incidence of human neural tube defects by 60-70 %. However, 30-40 % of cases remain nonresponsive to folate intake. The main purpose of this study was to understand the molecular mechanism of folate nonresponsiveness in a mouse model of neural tube defect. METHODS We used a folate-nonresponsive Fkbp8 knockout mouse model to elucidate the molecular mechanism(s) of folate nonresponsiveness. Neurospheres were grown from neural stem cells isolated from the lumbar neural tube of E9.5 Fkbp8 (-/-) and wild-type embryos. Immunostaining was used to determine the protein levels of oligodendrocyte transcription factor 2 (Olig2), Nkx6.1, class III beta-tubulin (TuJ1), O4, glial fibrillary acidic protein (GFAP), histone H3 Lys27 trimethylation (H3K27me3), ubiquitously transcribed tetratricopeptide repeat (UTX), and Msx2, and quantitative real-time (RT)-PCR was used to determine the message levels of Olig2, Nkx6.1, Msx2, and noggin in neural stem cells differentiated in the presence and absence of folic acid. RESULTS Fkbp8 (-/-)-derived neural stem cells showed (i) increased noggin expression; (ii) decreased Msx2 expression; (iii) premature differentiation--neurogenesis, oligodendrogenesis (Olig2 expression), and gliogenesis (GFAP expression); and (iv) increased UTX expression and decreased H3K27me3 polycomb modification. Exogenous folic acid did not reverse these markers. CONCLUSIONS Folate nonresponsiveness could be attributed in part to increased noggin expression in Fkbp8 (-/-) embryos, resulting in decreased Msx2 expression. Folate treatment further increases Olig2 and noggin expression, thereby exacerbating ventralization.
Collapse
Affiliation(s)
- Takao Tsurubuchi
- Developmental Neurobiology Laboratory, Department of Pediatric Neurosurgery, Developmental Biology Program, Lurie Children's Research Center, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, 60614, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Korzh V. Neurulation continues: The parade commander is… apical constriction. Russ J Dev Biol 2014. [DOI: 10.1134/s1062360414040055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
A novel mouse Fgfr2 mutant, hobbyhorse (hob), exhibits complete XY gonadal sex reversal. PLoS One 2014; 9:e100447. [PMID: 24956260 PMCID: PMC4067367 DOI: 10.1371/journal.pone.0100447] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 05/23/2014] [Indexed: 12/13/2022] Open
Abstract
The secreted molecule fibroblast growth factor 9 (FGF9) plays a critical role in testis determination in the mouse. In embryonic gonadal somatic cells it is required for maintenance of SOX9 expression, a key determinant of Sertoli cell fate. Conditional gene targeting studies have identified FGFR2 as the main gonadal receptor for FGF9 during sex determination. However, such studies can be complicated by inefficient and variable deletion of floxed alleles, depending on the choice of Cre deleter strain. Here, we report a novel, constitutive allele of Fgfr2, hobbyhorse (hob), which was identified in an ENU-based forward genetic screen for novel testis-determining loci. Fgr2hob is caused by a C to T mutation in the invariant exon 7, resulting in a polypeptide with a mis-sense mutation at position 263 (Pro263Ser) in the third extracellular immunoglobulin-like domain of FGFR2. Mutant homozygous embryos show severe limb and lung defects and, when on the sensitised C57BL/6J (B6) genetic background, undergo complete XY gonadal sex reversal associated with failure to maintain expression of Sox9. Genetic crosses employing a null mutant of Fgfr2 suggest that Fgr2hob is a hypomorphic allele, affecting both the FGFR2b and FGFR2c splice isoforms of the receptor. We exploited the consistent phenotype of this constitutive mutant by analysing MAPK signalling at the sex-determining stage of gonad development, but no significant abnormalities in mutant embryos were detected.
Collapse
|