1
|
Kou G, Zhou Y, Han H, Liu Z, Lai Y, Gao S. Comparative Analysis of Transcriptome Data of Wings from Different Developmental Stages of the Gynaephora qinghaiensis. Int J Mol Sci 2025; 26:3562. [PMID: 40332056 PMCID: PMC12026863 DOI: 10.3390/ijms26083562] [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: 02/03/2025] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 05/08/2025] Open
Abstract
Gynaephora qinghaiensis is a major pest in the alpine meadow regions of China. While the females are unable to fly, the males can fly and cause widespread damage. The aim of this study was to use transcriptome analysis to identify and verify genes expressed at different developmental stages of Gynaephora qinghaiensis, with particular emphasis on genes associated with wing development. High-throughput sequencing was performed on an Illumina HiSeqTM2000 platform to assess transcriptomic differences in the wings of male and female pupa and male and female adults of Gynaephora qinghaiensis, and the expression levels of the differentially expressed genes (DEGs) were verified by real-time fluorescence quantitative PCR (RT-qPCR). A total of 60,536 unigenes were identified from the transcriptome data, and 25,162 unigenes were obtained from a comparison with four major databases. Further analysis identified 18 DEGs associated with wing development in Gynaephora qinghaiensis. RT-qPCR verification of the expression levels showed consistency with the RNA sequencing results. Spatio-temporal expression profiling of the 18 genes indicated different levels of expression in the thoraces of male and female pupa, as well as between the wing buds of adult females and the wings of adult males. GO annotation analysis showed that the DEGs were associated with similar categories with no significant enrichment and were involved in cellular processes, cellular anatomical entities, and binding. KEGG analysis indicated that the DEGs were associated with endocytosis and metabolic pathways. The results of this study expand the information on genes associated with Gynaephora qinghaiensis wing development and provide support for further investigations of wing development at the molecular level.
Collapse
Affiliation(s)
- Guixiang Kou
- Grassland Research Institute, Chinese Academy of Agricultural Sciences, Hohhot 010010, China; (G.K.); (H.H.)
- Institute of Plant Protection, Qinghai Academy of Agriculture and Forestry, Xining 810016, China
| | - Yuantao Zhou
- College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China; (Y.Z.); (Z.L.)
| | - Haibing Han
- Grassland Research Institute, Chinese Academy of Agricultural Sciences, Hohhot 010010, China; (G.K.); (H.H.)
| | - Zhanling Liu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China; (Y.Z.); (Z.L.)
| | - Youpeng Lai
- Institute of Plant Protection, Qinghai Academy of Agriculture and Forestry, Xining 810016, China
| | - Shujing Gao
- Grassland Research Institute, Chinese Academy of Agricultural Sciences, Hohhot 010010, China; (G.K.); (H.H.)
| |
Collapse
|
2
|
LoPilato RK, Kroeger H, Mohan SK, Lauderdale JD, Grimsey N, Haltiwanger RS. Two NOTCH1 O-fucose sites have opposing functions in mouse retinal angiogenesis. Glycobiology 2023; 33:661-672. [PMID: 37329502 PMCID: PMC10560083 DOI: 10.1093/glycob/cwad048] [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: 03/19/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/19/2023] Open
Abstract
Previous in vitro studies demonstrated that Fringe glycosylation of the NOTCH1 extracellular domain at O-fucose residues in Epidermal Growth Factor-like Repeats (EGFs) 6 and 8 is a significant contributor to suppression of NOTCH1 activation by JAG1 or enhancement of NOTCH1 activation by DLL1, respectively. In this study, we sought to evaluate the significance of these glycosylation sites in a mammalian model by generating 2 C57BL/6J mouse lines carrying NOTCH1 point mutations, which eliminate O-fucosylation and Fringe activity at EGFs 6 (T232V) or 8 (T311V). We assessed changes to morphology during retinal angiogenesis, a process in which expression of Notch1, Jag1, Dll4, Lfng, Mfng, and Rfng genes coordinate cell-fate decisions to grow vessel networks. In the EGF6 O-fucose mutant (6f/6f) retinas, we observed reduced vessel density and branching, suggesting that this mutant is a Notch1 hypermorph. This finding agrees with prior cell-based studies showing that the 6f mutation increased JAG1 activation of NOTCH1 during co-expression with inhibitory Fringes. Although we predicted that the EGF8 O-fucose mutant (8f/8f) would not complete embryonic development due to the direct involvement of the O-fucose in engaging ligand, the 8f/8f mice were viable and fertile. In the 8f/8f retina, we measured increased vessel density consistent with established Notch1 hypomorphs. Overall, our data support the importance of NOTCH1 O-fucose residues for pathway function and confirms that single O-glycan sites are rich in signaling instructions for mammalian development.
Collapse
Affiliation(s)
- Rachel K LoPilato
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, United States
| | - Heike Kroeger
- Department of Cellular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602, United States
| | - Sneha K Mohan
- Neuroscience Division of Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA 30602, United States
| | - James D Lauderdale
- Department of Cellular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602, United States
- Neuroscience Division of Biomedical and Translational Sciences Institute, University of Georgia, Athens, GA 30602, United States
| | - Neil Grimsey
- Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, United States
| | - Robert S Haltiwanger
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, United States
| |
Collapse
|
3
|
Pandey A, Harvey BM, Lopez MF, Ito A, Haltiwanger RS, Jafar-Nejad H. Glycosylation of Specific Notch EGF Repeats by O-Fut1 and Fringe Regulates Notch Signaling in Drosophila. Cell Rep 2020; 29:2054-2066.e6. [PMID: 31722217 PMCID: PMC6866671 DOI: 10.1016/j.celrep.2019.10.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/13/2019] [Accepted: 10/07/2019] [Indexed: 12/27/2022] Open
Abstract
Fringe glycosyltransferases differentially modulate the binding of Notch receptors to Delta/DLL versus Serrate/Jagged ligands by adding GlcNAc to O-linked fucose on Notch epidermal growth factor-like (EGF) repeats. Although Notch has 22 O-fucosylation sites, the biologically relevant sites affecting Notch activity during animal development in vivo in the presence or absence of Fringe are not known. Using a variety of assays, we find important roles in Drosophila Notch signaling for GlcNAc-fucose-O glycans on three sites: EGF8, EGF9, and EGF12. O-Fucose monosaccharide on EGF12 (in the absence of Fringe) is essential for Delta-mediated lateral inhibition in embryos. However, wing vein development depends on the addition of GlcNAc to EGF8 and EGF12 by Fringe, with a minor contribution from EGF9. Fringe modifications of EGF8 and EGF12 together prevent Notch from cis-inhibiting Serrate, thereby promoting normal wing margin formation. Our work shows the combinatorial and context-dependent roles of GlcNAc-fucose-O glycans on these sites in Drosophila Notch-ligand interactions. POFUT1/O-Fut1 and Fringe glycosyltransferases regulate Notch signaling by adding fucose and GlcNAc, respectively, to Notch EGF repeats. Using in vitro and in vivo experiments, Pandey et al. define the critical target sites of these enzymes on Drosophila Notch and determine the distinct roles of each sugar in Notch-dependent processes.
Collapse
Affiliation(s)
- Ashutosh Pandey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Beth M Harvey
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mario F Lopez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Atsuko Ito
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Robert S Haltiwanger
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA; Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA.
| | - Hamed Jafar-Nejad
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Pandey A, Niknejad N, Jafar-Nejad H. Multifaceted regulation of Notch signaling by glycosylation. Glycobiology 2020; 31:8-28. [PMID: 32472127 DOI: 10.1093/glycob/cwaa049] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/18/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
To build a complex body composed of various cell types and tissues and to maintain tissue homeostasis in the postembryonic period, animals use a small number of highly conserved intercellular communication pathways. Among these is the Notch signaling pathway, which is mediated via the interaction of transmembrane Notch receptors and ligands usually expressed by neighboring cells. Maintaining optimal Notch pathway activity is essential for normal development, as evidenced by various human diseases caused by decreased and increased Notch signaling. It is therefore not surprising that multiple mechanisms are used to control the activation of this pathway in time and space. Over the last 20 years, protein glycosylation has been recognized as a major regulatory mechanism for Notch signaling. In this review, we will provide a summary of the various types of glycan that have been shown to modulate Notch signaling. Building on recent advances in the biochemistry, structural biology, cell biology and genetics of Notch receptors and the glycosyltransferases that modify them, we will provide a detailed discussion on how various steps during Notch activation are regulated by glycans. Our hope is that the current review article will stimulate additional research in the field of Notch glycobiology and will potentially be of benefit to investigators examining the contribution of glycosylation to other developmental processes.
Collapse
Affiliation(s)
| | | | - Hamed Jafar-Nejad
- Department of Molecular and Human Genetics.,Development, Disease Models & Therapeutics Graduate Program.,Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Wang Y, Wang J, Xia X, Wu G. Functional Identification of Px-fringe and Px-engrailed Genes under Heat Stress in Chlorpyrifos-Resistant and -Susceptible Plutela xylostella (Lepidoptera: Plutellidae). INSECTS 2020; 11:insects11050287. [PMID: 32392846 PMCID: PMC7290670 DOI: 10.3390/insects11050287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 11/16/2022]
Abstract
In our previous research, the fitness cost of resistance of the diamondback moth (DBM), Plutella xylostella found in insecticide-resistant DBM (Rc-DBM) under heat stress was based on heavier damage to wing veins when compared to insecticide-susceptible DBM (Sm-DBM). To investigate the molecular mechanism of the damage to the veins between Rc- and Sm-DBM, the full-length sequences of two related genes involved in the development of wing veins, fringe (Px-fng) and engrailed (Px-en) of DBM were cloned, and the mRNA expressions of both Px-fng and Px-en were studied. The Px-fng and Px-en cDNA contained 1038 bp and 1152 bp of open reading frames (ORFs), respectively, which encoded a putative protein comprising 345 and 383 amino acids with a calculated molecular weight of 39.59 kDa and 42.69 kDa. Significantly down regulated expressions of Px-fng and Px-en under heat stress were found in pupae and adults of Rc-DBM compared to Sm-DBM, and a result of higher damage to wing veins in Rc-DBM under heat stress. Based on RNAi experiments, significant inhibitions on expressions of Px-fng and Px-en in both Sm-DBM and Rc-DBM were found when the pupae were infected by dsFng or dsEn. Corresponding to these, infections of dsFng or dsEn resulted in significant decrease of eclosion rate and increase malformation rate of DBM. Our results suggest that the higher damage of wing veins in DBM might be related to the heavier inhibitions of Px-fng and Px-en expression, and the Px-fng and Px-en are involved in the development of wings and veins.
Collapse
Affiliation(s)
- Yu Wang
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jingnan Wang
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaofeng Xia
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Correspondence: (X.X.); (G.W.)
| | - Gang Wu
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Correspondence: (X.X.); (G.W.)
| |
Collapse
|
6
|
Moulton MJ, Humphreys GB, Kim A, Letsou A. O-GlcNAcylation Dampens Dpp/BMP Signaling to Ensure Proper Drosophila Embryonic Development. Dev Cell 2020; 53:330-343.e3. [DOI: 10.1016/j.devcel.2020.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/15/2020] [Accepted: 03/31/2020] [Indexed: 01/09/2023]
|
7
|
Li W, De Schutter K, Van Damme EJM, Smagghe G. Synthesis and biological roles of O-glycans in insects. Glycoconj J 2019; 37:47-56. [DOI: 10.1007/s10719-019-09867-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 03/15/2019] [Indexed: 11/24/2022]
|
8
|
Zhang L, Ten Hagen KG. O-Linked glycosylation in Drosophila melanogaster. Curr Opin Struct Biol 2019; 56:139-145. [PMID: 30852302 DOI: 10.1016/j.sbi.2019.01.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/20/2018] [Accepted: 01/24/2019] [Indexed: 12/14/2022]
Abstract
Glycosylation, or the addition of sugars to proteins, is a highly conserved protein modification defined by both the monosaccharide initially added as well as the amino acid to which it is attached. O-Linked glycosylation represents a diverse group of protein modifications occurring on the hydroxyl groups of serine and/or threonine residues. O-Glycosylation can have wide-ranging effects on protein stability and function, which translate into crucial consequences at the organismal level. This review will summarize structural and biological insights into the major O-glycans formed within the secretory apparatus (O-GalNAc, O-Man, O-Fuc, O-Glc and extracellular O-GlcNAc) from studies in the fruit fly Drosophila melanogaster. Drosophila has many advantages for investigating these complex modifications, boasting reduced functional redundancy within gene families, reduced length/complexity of glycan chains and sophisticated genetic tools. Gaining an understanding of the normal cellular and developmental roles of these conserved modifications in Drosophila will provide insight into how changes in O-glycans are involved in human disease and disease susceptibilities.
Collapse
Affiliation(s)
- Liping Zhang
- Developmental Glycobiology Section, NIDCR, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, United States
| | - Kelly G Ten Hagen
- Developmental Glycobiology Section, NIDCR, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, United States.
| |
Collapse
|
9
|
Otomo N, Mizumoto S, Lu HF, Takeda K, Campos-Xavier B, Mittaz-Crettol L, Guo L, Takikawa K, Nakamura M, Yamada S, Matsumoto M, Watanabe K, Ikegawa S. Identification of novel LFNG mutations in spondylocostal dysostosis. J Hum Genet 2018; 64:261-264. [DOI: 10.1038/s10038-018-0548-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/17/2018] [Accepted: 11/19/2018] [Indexed: 01/22/2023]
|
10
|
Takeda K, Kou I, Mizumoto S, Yamada S, Kawakami N, Nakajima M, Otomo N, Ogura Y, Miyake N, Matsumoto N, Kotani T, Sudo H, Yonezawa I, Uno K, Taneichi H, Watanabe K, Shigematsu H, Sugawara R, Taniguchi Y, Minami S, Nakamura M, Matsumoto M, Watanabe K, Ikegawa S. Screening of known disease genes in congenital scoliosis. Mol Genet Genomic Med 2018; 6:966-974. [PMID: 30196550 PMCID: PMC6305645 DOI: 10.1002/mgg3.466] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/18/2018] [Accepted: 08/09/2018] [Indexed: 01/29/2023] Open
Abstract
Background Congenital scoliosis (CS) is defined as a lateral curvature of the spine due to the vertebral malformations and has an incidence of 0.5–1/1,000 births. We previously examined TBX6 in Japanese CS patients and revealed that approximately 10% of CS was caused by TBX6 mutations. However, the genetic cause of remaining CS is unknown. Methods We recruited 78 CS patients without TBX6 mutations and major comorbidities, and investigated the genes previously reported to be associated with CS and congenital vertebral malformations by whole‐exome sequencing. Results We identified the compound heterozygous missense variants in LFNG in one patient. No likely disease‐causing variants were identified in other patients, however. LFNG encodes a GlcNAc‐transferase. The LFNG variants showed loss of their enzyme function. Conclusions A LFNG mutation is reported in a case of spondylocostal dysostosis (SCD), a skeletal dysplasia with severe malformations of vertebra and rib. The CS patient with LFNG mutations had multiple vertebral malformations including hemivertebrae, butterfly vertebrae, and block vertebrae, and rib malformations. LFNG mutations may cause a spectrum of phenotypes including CS and SCD. The current list of known disease genes could explain only a small fraction of genetic cause of CS.
Collapse
Affiliation(s)
- Kazuki Takeda
- Laboratory of Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ikuyo Kou
- Laboratory of Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Japan.,Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Noriaki Kawakami
- Department of Orthopaedic Surgery, Meijo Hospital, Nagoya, Japan
| | - Masahiro Nakajima
- Laboratory of Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| | - Nao Otomo
- Laboratory of Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoji Ogura
- Laboratory of Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toshiaki Kotani
- Department of Orthopaedic Surgery, Seirei Sakura Citizen Hospital, Sakura, Japan
| | - Hideki Sudo
- Department of Advanced Medicine for Spine and Spinal Cord Disorders, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ikuho Yonezawa
- Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Koki Uno
- Department of Orthopaedic Surgery, National Hospital Organization, Kobe Medical Center, Kobe, Japan
| | - Hiroshi Taneichi
- Department of Orthopaedic Surgery, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Kei Watanabe
- Department of Orthopaedic Surgery, Niigata University Hospital, Niigata, Japan
| | - Hideki Shigematsu
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Ryo Sugawara
- Department of Orthopedics, Jichi Medical University, Shimotsuke, Japan
| | - Yuki Taniguchi
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shohei Minami
- Department of Orthopaedic Surgery, Seirei Sakura Citizen Hospital, Sakura, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | - Kota Watanabe
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| |
Collapse
|
11
|
Lee TV, Pandey A, Jafar-Nejad H. Xylosylation of the Notch receptor preserves the balance between its activation by trans-Delta and inhibition by cis-ligands in Drosophila. PLoS Genet 2017; 13:e1006723. [PMID: 28394891 PMCID: PMC5402982 DOI: 10.1371/journal.pgen.1006723] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/24/2017] [Accepted: 03/29/2017] [Indexed: 01/02/2023] Open
Abstract
The Drosophila glucoside xylosyltransferase Shams xylosylates Notch and inhibits Notch signaling in specific contexts including wing vein development. However, the molecular mechanisms underlying context-specificity of the shams phenotype is not known. Considering the role of Delta-Notch signaling in wing vein formation, we hypothesized that Shams might affect Delta-mediated Notch signaling in Drosophila. Using genetic interaction studies, we find that altering the gene dosage of Delta affects the wing vein and head bristle phenotypes caused by loss of Shams or by mutations in the Notch xylosylation sites. Clonal analysis suggests that loss of shams promotes Delta-mediated Notch activation. Further, Notch trans-activation by ectopically overexpressed Delta shows a dramatic increase upon loss of shams. In agreement with the above in vivo observations, cell aggregation and ligand-receptor binding assays show that shams knock-down in Notch-expressing cells enhances the binding between Notch and trans-Delta without affecting the binding between Notch and trans-Serrate and cell surface levels of Notch. Loss of Shams does not impair the cis-inhibition of Notch by ectopic overexpression of ligands in vivo or the interaction of Notch and cis-ligands in S2 cells. Nevertheless, removing one copy of endogenous ligands mimics the effects of loss shams on Notch trans-activation by ectopic Delta. This favors the notion that trans-activation of Notch by Delta overcomes the cis-inhibition of Notch by endogenous ligands upon loss of shams. Taken together, our data suggest that xylosylation selectively impedes the binding of Notch with trans-Delta without affecting its binding with cis-ligands and thereby assists in determining the balance of Notch receptor's response to cis-ligands vs. trans-Delta during Drosophila development.
Collapse
Affiliation(s)
- Tom V. Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ashutosh Pandey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hamed Jafar-Nejad
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
12
|
Ling L, Ge X, Li Z, Zeng B, Xu J, Chen X, Shang P, James AA, Huang Y, Tan A. MiR-2 family targets awd and fng to regulate wing morphogenesis in Bombyx mori. RNA Biol 2016; 12:742-8. [PMID: 26037405 DOI: 10.1080/15476286.2015.1048957] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs) are post-transcriptional regulators that target specific mRNAs for repression and thus play key roles in many biological processes, including insect wing morphogenesis. miR-2 is an invertebrate-specific miRNA family that has been predicted in the fruit fly, Drosophila melanogaster, to be involved in regulating the Notch signaling pathway. We show here that miR-2 plays a critical role in wing morphogenesis in the silkworm, Bombyx mori, a lepidopteran model insect. Transgenic over-expression of a miR-2 cluster using a Gal4/UAS system results in deformed adult wings, supporting the conclusion that miR-2 regulates functions essential for normal wing morphogenesis. Two genes, abnormal wing disc (awd) and fringe (fng), which are positive regulators in Notch signaling, are identified as miR-2 targets and validated by a dual-luciferase reporter assay. The relative abundance of both awd and fng expression products was reduced significantly in transgenic animals, implicating them in the abnormal wing phenotype. Furthermore, somatic mutagenesis analysis of awd and fng using the CRISPR/Cas9 system and knock-out mutants also resulted in deformed wings similar to those observed in the miR-2 overexpression transgenic animals. The critical role of miR-2 in Bombyx wing morphogenesis may provide a potential target in future lepidopteran pest control.
Collapse
Affiliation(s)
- Lin Ling
- a Faculty of Life Sciences; Northwestern Polytechnical University ; Xi'an , China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kellokumpu S, Hassinen A, Glumoff T. Glycosyltransferase complexes in eukaryotes: long-known, prevalent but still unrecognized. Cell Mol Life Sci 2016; 73:305-25. [PMID: 26474840 PMCID: PMC7079781 DOI: 10.1007/s00018-015-2066-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/28/2015] [Accepted: 10/08/2015] [Indexed: 01/08/2023]
Abstract
Glycosylation is the most common and complex cellular modification of proteins and lipids. It is critical for multicellular life and its abrogation often leads to a devastating disease. Yet, the underlying mechanistic details of glycosylation in both health and disease remain unclear. Partly, this is due to the complexity and dynamicity of glycan modifications, and the fact that not all the players are taken into account. Since late 1960s, a vast number of studies have demonstrated that glycosyltransferases typically form homomeric and heteromeric complexes with each other in yeast, plant and animal cells. To propagate their acceptance, we will summarize here accumulated data for their prevalence and potential functional importance for glycosylation focusing mainly on their mutual interactions, the protein domains mediating these interactions, and enzymatic activity changes that occur upon complex formation. Finally, we will highlight the few existing 3D structures of these enzyme complexes to pinpoint their individual nature and to emphasize that their lack is the main obstacle for more detailed understanding of how these enzyme complexes interact and function in a eukaryotic cell.
Collapse
Affiliation(s)
- Sakari Kellokumpu
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland.
| | - Antti Hassinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland
| | - Tuomo Glumoff
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland
| |
Collapse
|
14
|
Tseng CY, Kao SH, Wan CL, Cho Y, Tung SY, Hsu HJ. Notch signaling mediates the age-associated decrease in adhesion of germline stem cells to the niche. PLoS Genet 2014; 10:e1004888. [PMID: 25521289 PMCID: PMC4270478 DOI: 10.1371/journal.pgen.1004888] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 11/12/2014] [Indexed: 11/18/2022] Open
Abstract
Stem cells have an innate ability to occupy their stem cell niche, which in turn, is optimized to house stem cells. Organ aging is associated with reduced stem cell occupancy in the niche, but the mechanisms involved are poorly understood. Here, we report that Notch signaling is increased with age in Drosophila female germline stem cells (GSCs), and this results in their removal from the niche. Clonal analysis revealed that GSCs with low levels of Notch signaling exhibit increased adhesiveness to the niche, thereby out-competing their neighbors with higher levels of Notch; adhesiveness is altered through regulation of E-cadherin expression. Experimental enhancement of Notch signaling in GSCs hastens their age-dependent loss from the niche, and such loss is at least partially mediated by Sex lethal. However, disruption of Notch signaling in GSCs does not delay GSC loss during aging, and nor does it affect BMP signaling, which promotes self-renewal of GSCs. Finally, we show that in contrast to GSCs, Notch activation in the niche (which maintains niche integrity, and thus mediates GSC retention) is reduced with age, indicating that Notch signaling regulates GSC niche occupancy both intrinsically and extrinsically. Our findings expose a novel role of Notch signaling in controlling GSC-niche adhesion in response to aging, and are also of relevance to metastatic cancer cells, in which Notch signaling suppresses cell adhesion. Aging is frequently associated with a decline in the size of stem cell pools, but little is known regarding the molecular mechanisms underlying this process. Here, we report that Notch signaling is increased in GSCs as they age, and this promotes their removal from the niche in an E-cadherin dependent manner. In contrast to GSCs, niche cells exhibit decreased Notch signaling with age; Notch signaling in these cells controls niche integrity, and consequently GSC retention. While Notch signaling in the niche is regulated by insulin signaling, Notch signaling in GSCs is controlled by Sex lethal, an RNA-binding protein. These results imply that Notch signaling is regulated in a cell-type-dependent manner, and coordination between GSCs and their niche facilitates the removal of cells from the niche during the aging process.
Collapse
Affiliation(s)
- Chen-Yuan Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Han Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chih-Ling Wan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yueh Cho
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Shu-Yun Tung
- Genomic Core Facility, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hwei-Jan Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
15
|
LeBon L, Lee TV, Sprinzak D, Jafar-Nejad H, Elowitz MB. Fringe proteins modulate Notch-ligand cis and trans interactions to specify signaling states. eLife 2014; 3:e02950. [PMID: 25255098 PMCID: PMC4174579 DOI: 10.7554/elife.02950] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 08/31/2014] [Indexed: 12/31/2022] Open
Abstract
The Notch signaling pathway consists of multiple types of receptors and ligands, whose interactions can be tuned by Fringe glycosyltransferases. A major challenge is to determine how these components control the specificity and directionality of Notch signaling in developmental contexts. Here, we analyzed same-cell (cis) Notch-ligand interactions for Notch1, Dll1, and Jag1, and their dependence on Fringe protein expression in mammalian cells. We found that Dll1 and Jag1 can cis-inhibit Notch1, and Fringe proteins modulate these interactions in a way that parallels their effects on trans interactions. Fringe similarly modulated Notch-ligand cis interactions during Drosophila development. Based on these and previously identified interactions, we show how the design of the Notch signaling pathway leads to a restricted repertoire of signaling states that promote heterotypic signaling between distinct cell types, providing insight into the design principles of the Notch signaling system, and the specific developmental process of Drosophila dorsal-ventral boundary formation. DOI:http://dx.doi.org/10.7554/eLife.02950.001 In animals, cells use a process called Notch signaling to communicate with neighboring cells. During this process, a protein known as a DSL ligand from one cell binds to a protein called a Notch receptor on a neighboring cell. This triggers a series of events in the neighboring cell that change how the genes in this cell are expressed. Notch signaling is involved in many processes including the early growth of embryos, the formation of organs and limbs, and the maintenance of stem cells throughout adult life. Enzymes called Fringe enzymes can control Notch signaling by blocking or promoting the formation of the DSL ligand-Notch receptor pairs. It is also possible for a DSL ligand and a Notch receptor from the same cell to interact. This is thought to be important because it prevents an individual cell from sending and receiving Notch signals at the same time. There are several different DSL ligands, Notch receptors and Fringe enzymes, so it is difficult to determine which configurations of receptors, ligands and Fringe enzymes can enable Notch signals to be sent or received. To address this problem, LeBon et al. investigated how Fringe enzymes acted on several different DSL-Notch receptor pairs in mammalian cells, and also in fruit flies. They focused in particular on the interactions that occurred within the same cell, as the role of Fringe enzymes in this type of interaction has not been examined previously. The experiments revealed that Fringe proteins modify specific same-cell interactions in a way that enables a cell to receive one type of Notch signal from a neighboring cell and send a different type of Notch signal to another cell at the same time. More generally, these results show how an unconventional, ‘bottom-up’ approach can reveal the design principles of cell signaling systems, and suggest that it should now be possible to use these principles to try to understand which cell types send signals to which other cell types in many different contexts. DOI:http://dx.doi.org/10.7554/eLife.02950.002
Collapse
Affiliation(s)
- Lauren LeBon
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, United States
| | - Tom V Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - David Sprinzak
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv, Israel
| | | | - Michael B Elowitz
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, United States
| |
Collapse
|
16
|
Humphreys GB, Jud MC, Monroe KM, Kimball SS, Higley M, Shipley D, Vrablik MC, Bates KL, Letsou A. Mummy, A UDP-N-acetylglucosamine pyrophosphorylase, modulates DPP signaling in the embryonic epidermis of Drosophila. Dev Biol 2013; 381:434-45. [PMID: 23796903 DOI: 10.1016/j.ydbio.2013.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 06/06/2013] [Indexed: 12/29/2022]
Abstract
The evolutionarily conserved JNK/AP-1 (Jun N-terminal kinase/activator protein 1) and BMP (Bone Morphogenetic Protein) signaling cascades are deployed hierarchically to regulate dorsal closure in the fruit fly Drosophila melanogaster. In this developmental context, the JNK/AP-1 signaling cascade transcriptionally activates BMP signaling in leading edge epidermal cells. Here we show that the mummy (mmy) gene product, which is required for dorsal closure, functions as a BMP signaling antagonist. Genetic and biochemical tests of Mmy's role as a BMP-antagonist indicate that its function is independent of AP-1, the transcriptional trigger of BMP signal transduction in leading edge cells. pMAD (phosphorylated Mothers Against Dpp) activity data show the mmy gene product to be a new type of epidermal BMP regulator - one which transforms a BMP ligand from a long- to a short-range signal. mmy codes for the single UDP-N-acetylglucosamine pyrophosphorylase in Drosophila, and its requirement for attenuating epidermal BMP signaling during dorsal closure points to a new role for glycosylation in defining a highly restricted BMP activity field in the fly. These findings add a new dimension to our understanding of mechanisms modulating the BMP signaling gradient.
Collapse
Affiliation(s)
- Gregory B Humphreys
- Department of Human Genetics, Eccles Institute of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Filipek-Górniok B, Holmborn K, Haitina T, Habicher J, Oliveira MB, Hellgren C, Eriksson I, Kjellén L, Kreuger J, Ledin J. Expression of chondroitin/dermatan sulfate glycosyltransferases during early zebrafish development. Dev Dyn 2013; 242:964-75. [PMID: 23703795 DOI: 10.1002/dvdy.23981] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 03/08/2013] [Accepted: 04/08/2013] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Chondroitin/dermatan sulfate (CS/DS) proteoglycans present in the extracellular matrix have important structural and regulatory functions. RESULTS Six human genes have previously been shown to catalyze CS/DS polymerization. Here we show that one of these genes, chpf, is represented by two copies in the zebrafish genome, chpfa and chpfb, while the other five human CS/DS glycosyltransferases csgalnact1, csgalnact2, chpf2, chsy1, and chsy3 all have single zebrafish orthologues. The putative zebrafish CS/DS glycosyltransferases are spatially and temporally expressed. Interestingly, overlapping expression of multiple glycosyltransferases coincides with high CS/DS deposition. Finally, whereas the relative levels of the related polysaccharide HS reach steady-state at around 2 days post fertilization, there is a continued relative increase of the CS amounts per larvae during the first 6 days of development, matching the increased cartilage formation. CONCLUSIONS There are 7 CS/DS glycosyltransferases in zebrafish, which, based on homology, can be divided into the CSGALNACT, CHSY, and CHPF families. The overlap between intense CS/DS production and the expression of multiple CS/DS glycosyltransferases suggests that efficient CS/DS biosynthesis requires a combination of several glycosyltransferases.
Collapse
Affiliation(s)
- Beata Filipek-Górniok
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Giampietro PF, Dunwoodie SL, Kusumi K, Pourquié O, Tassy O, Offiah AC, Cornier AS, Alman BA, Blank RD, Raggio CL, Glurich I, Turnpenny PD. Molecular diagnosis of vertebral segmentation disorders in humans. ACTA ACUST UNITED AC 2013; 2:1107-21. [PMID: 23496422 DOI: 10.1517/17530059.2.10.1107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Vertebral malformations contribute substantially to the pathophysiology of kyphosis and scoliosis, common health problems associated with back and neck pain, disability, cosmetic disfigurement and functional distress. OBJECTIVE To provide an overview of the current understanding of vertebral malformations, at both the clinical level and the molecular level, and factors that contribute to their occurrence. METHODS The literature related to the following was reviewed: recent advances in the understanding of the molecular embryology underlying vertebral development and relevance to elucidation of etiologies of several known human vertebral malformation syndromes; outcomes of molecular studies elucidating genetic contributions to congenital and sporadic vertebral malformations; and complex interrelationships between genetic and environmental factors that contribute to the pathogenesis of isolated syndromic and non-syndromic congenital vertebral malformations. RESULTS/CONCLUSION Expert opinions extend to discussion of the importance of establishing improved classification systems for vertebral malformation, future directions in molecular and genetic research approaches to vertebral malformation and translational value of research efforts to clinical management and genetic counseling of affected individuals and their families.
Collapse
Affiliation(s)
- Philip F Giampietro
- Marshfield Clinic, Department of Genetic Services, 1000 N. Oak Avenue, Marshfield, WI 54449, USA +1 715 221 7410 ; +1 715 389 4399 ;
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Vandenborre G, Smagghe G, Ghesquière B, Menschaert G, Nagender Rao R, Gevaert K, Van Damme EJM. Diversity in protein glycosylation among insect species. PLoS One 2011; 6:e16682. [PMID: 21373189 PMCID: PMC3044136 DOI: 10.1371/journal.pone.0016682] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 12/23/2010] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND A very common protein modification in multicellular organisms is protein glycosylation or the addition of carbohydrate structures to the peptide backbone. Although the Class of the Insecta is the largest animal taxon on Earth, almost all information concerning glycosylation in insects is derived from studies with only one species, namely the fruit fly Drosophila melanogaster. METHODOLOGY/PRINCIPAL FINDINGS In this report, the differences in glycoproteomes between insects belonging to several economically important insect orders were studied. Using GNA (Galanthus nivalis agglutinin) affinity chromatography, different sets of glycoproteins with mannosyl-containing glycan structures were purified from the flour beetle (Tribolium castaneum), the silkworm (Bombyx mori), the honeybee (Apis mellifera), the fruit fly (D. melanogaster) and the pea aphid (Acyrthosiphon pisum). To identify and characterize the purified glycoproteins, LC-MS/MS analysis was performed. For all insect species, it was demonstrated that glycoproteins were related to a broad range of biological processes and molecular functions. Moreover, the majority of glycoproteins retained on the GNA column were unique to one particular insect species and only a few glycoproteins were present in the five different glycoprotein sets. Furthermore, these data support the hypothesis that insect glycoproteins can be decorated with mannosylated O-glycans. CONCLUSIONS/SIGNIFICANCE The results presented here demonstrate that oligomannose N-glycosylation events are highly specific depending on the insect species. In addition, we also demonstrated that protein O-mannosylation in insect species may occur more frequently than currently believed.
Collapse
Affiliation(s)
- Gianni Vandenborre
- Laboratory of Agrozoology, Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Laboratory of Biochemistry and Glycobiology, Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Guy Smagghe
- Laboratory of Agrozoology, Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Bart Ghesquière
- Department of Medical Protein Research, VIB, Ghent, Belgium
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Gerben Menschaert
- Laboratory for Bioinformatics and Computational Genomics, Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Rameshwaram Nagender Rao
- Laboratory of Agrozoology, Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Laboratory of Biochemistry and Glycobiology, Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kris Gevaert
- Department of Medical Protein Research, VIB, Ghent, Belgium
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Els J. M. Van Damme
- Laboratory of Biochemistry and Glycobiology, Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
20
|
Drosophila glial glutamate transporter Eaat1 is regulated by fringe-mediated notch signaling and is essential for larval locomotion. J Neurosci 2010; 30:14446-57. [PMID: 20980602 DOI: 10.1523/jneurosci.1021-10.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In the mammalian CNS, glial cells expressing excitatory amino acid transporters (EAATs) tightly regulate extracellular glutamate levels to control neurotransmission and protect neurons from excitotoxic damage. Dysregulated EAAT expression is associated with several CNS pathologies in humans, yet mechanisms of EAAT regulation and the importance of glutamate transport for CNS development and function in vivo remain incompletely understood. Drosophila is an advanced genetic model with only a single high-affinity glutamate transporter termed Eaat1. We found that Eaat1 expression in CNS glia is regulated by the glycosyltransferase Fringe, which promotes neuron-to-glia signaling through the Delta-Notch ligand-receptor pair during embryogenesis. We made Eaat1 loss-of-function mutations and found that homozygous larvae could not perform the rhythmic peristaltic contractions required for crawling. We found no evidence for excitotoxic cell death or overt defects in the development of neurons and glia, and the crawling defect could be induced by postembryonic inactivation of Eaat1. Eaat1 fully rescued locomotor activity when expressed in only a limited subpopulation of glial cells situated near potential glutamatergic synapses within the CNS neuropil. Eaat1 mutants had deficits in the frequency, amplitude, and kinetics of synaptic currents in motor neurons whose rhythmic patterns of activity may be regulated by glutamatergic neurotransmission among premotor interneurons; similar results were seen with pharmacological manipulations of glutamate transport. Our findings indicate that Eaat1 expression is promoted by Fringe-mediated neuron-glial communication during development and suggest that Eaat1 plays an essential role in regulating CNS neural circuits that control locomotion in Drosophila.
Collapse
|
21
|
Giampietro PF, Dunwoodie SL, Kusumi K, Pourquié O, Tassy O, Offiah AC, Cornier AS, Alman BA, Blank RD, Raggio CL, Glurich I, Turnpenny PD. Progress in the understanding of the genetic etiology of vertebral segmentation disorders in humans. Ann N Y Acad Sci 2009; 1151:38-67. [PMID: 19154516 DOI: 10.1111/j.1749-6632.2008.03452.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vertebral malformations contribute substantially to the pathophysiology of kyphosis and scoliosis, common health problems associated with back and neck pain, disability, cosmetic disfigurement, and functional distress. This review explores (1) recent advances in the understanding of the molecular embryology underlying vertebral development and relevance to elucidation of etiologies of several known human vertebral malformation syndromes; (2) outcomes of molecular studies elucidating genetic contributions to congenital and sporadic vertebral malformation; and (3) complex interrelationships between genetic and environmental factors that contribute to the pathogenesis of isolated syndromic and nonsyndromic congenital vertebral malformation. Discussion includes exploration of the importance of establishing improved classification systems for vertebral malformation, future directions in molecular and genetic research approaches to vertebral malformation, and translational value of research efforts to clinical management and genetic counseling of affected individuals and their families.
Collapse
Affiliation(s)
- Philip F Giampietro
- Department of Medical Genetic Services, Marshfield Clinic, 1000 North Oak Avenue, Marshfield, WI 54449, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mutation of the fucose-specific β1,3 N-acetylglucosaminyltransferase LFNG results in abnormal formation of the spine. Biochim Biophys Acta Mol Basis Dis 2009; 1792:100-11. [DOI: 10.1016/j.bbadis.2008.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 10/31/2008] [Accepted: 11/04/2008] [Indexed: 01/24/2023]
|
23
|
Luther KB, Schindelin H, Haltiwanger RS. Structural and mechanistic insights into lunatic fringe from a kinetic analysis of enzyme mutants. J Biol Chem 2009; 284:3294-3305. [PMID: 19028689 PMCID: PMC2631955 DOI: 10.1074/jbc.m805502200] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 11/19/2008] [Indexed: 12/25/2022] Open
Abstract
The Notch receptor is critical for proper development where it orchestrates numerous cell fate decisions. The Fringe family of beta1,3-N-acetylglucosaminyltransferases are regulators of this pathway. Fringe enzymes add N-acetylglucosamine to O-linked fucose on the epidermal growth factor repeats of Notch. Here we have analyzed the reaction catalyzed by Lunatic Fringe (Lfng) in detail. A mutagenesis strategy for Lfng was guided by a multiple sequence alignment of Fringe proteins and solutions from docking an epidermal growth factor-like O-fucose acceptor substrate onto a homology model of Lfng. We targeted three main areas as follows: residues that could help resolve where the fucose binds, residues in two conserved loops not observed in the published structure of Manic Fringe, and residues predicted to be involved in UDP-N-acetylglucosamine (UDP-GlcNAc) donor specificity. We utilized a kinetic analysis of mutant enzyme activity toward the small molecule acceptor substrate 4-nitrophenyl-alpha-L-fucopyranoside to judge their effect on Lfng activity. Our results support the positioning of O-fucose in a specific orientation to the catalytic residue. We also found evidence that one loop closes off the active site coincident with, or subsequent to, substrate binding. We propose a mechanism whereby the ordering of this short loop may alter the conformation of the catalytic aspartate. Finally, we identify several residues near the UDP-GlcNAc-binding site, which are specifically permissive toward UDP-GlcNAc utilization.
Collapse
Affiliation(s)
- Kelvin B Luther
- Department of Biochemistry and Cell Biology, Institute for Cell and Developmental Biology, Stony Brook University, Stony Brook, New York 11794-5215
| | - Hermann Schindelin
- Department of Biochemistry and Cell Biology, Institute for Cell and Developmental Biology, Stony Brook University, Stony Brook, New York 11794-5215
| | - Robert S Haltiwanger
- Department of Biochemistry and Cell Biology, Institute for Cell and Developmental Biology, Stony Brook University, Stony Brook, New York 11794-5215.
| |
Collapse
|
24
|
Turnpenny PD. Defective somitogenesis and abnormal vertebral segmentation in man. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 638:164-89. [PMID: 21038776 DOI: 10.1007/978-0-387-09606-3_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
In recent years molecular genetics has revolutionized the study of somitogenesis in developmental biology and advances that have taken place in animal models have been applied successfully to human disease. Abnormal segmentation in man is a relatively common birth defect and advances in understanding have come through the study of cases clustered in families using DNA linkage analysis and candidate gene approaches, the latter stemming directly from knowledge gained through the study of animal models. Only a minority of abnormal segmentation phenotypes appear to follow Mendelian inheritance but three genes--DLL3, MESP2 and LNFG--have now been identified for spondylocostal dysostosis (SCD), a spinal malformation characterized by extensive hemivertebrae, trunkal shortening and abnormally aligned ribs with points of fusion. In affected families autosomal recessive inheritance is followed. These genes are all important components of the Notch signaling pathway. Other genes within the pathway cause diverse phenotypes such as Alagille syndrome (AGS) and CADASIL, conditions that may have their origin in defective vasculogenesis. This review deals mainly with SCD, with some consideration of AGS. Significant future challenges lie in identifying causes of the many abnormal segmentation phenotypes in man but it is hoped that combined approaches in collaboration with developmental biologists will reap rewards.
Collapse
Affiliation(s)
- Peter D Turnpenny
- Clinical Genetics Department, Royal Devon & Exeter Hospital, Gladstone Road, Exeter EX1 2ED, United Kingdom.
| |
Collapse
|
25
|
Lin YR, Reddy BVVG, Irvine KD. Requirement for a core 1 galactosyltransferase in the Drosophila nervous system. Dev Dyn 2008; 237:3703-14. [PMID: 18985719 PMCID: PMC2722375 DOI: 10.1002/dvdy.21775] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mucin type O-glycosylation is a widespread modification of eukaryotic proteins, but its functional requirements remain incompletely understood. It is initiated by the attachment of N-acetylgalactosamine (GalNAc) to Ser or Thr residues, and then elongated by additional sugars. We have examined requirements for mucin-type glycosylation in Drosophila by characterizing the expression and phenotypes of core 1 galactosyltransferases (core 1 GalTs), which elongate O-GalNAc by adding galactose in a beta1,3 linkage. Drosophila encode several putative core 1 GalTs, each expressed in distinct patterns. CG9520 (C1GalTA) is expressed in the amnioserosa and central nervous system. A null mutation in C1GalTA is lethal, and mutant animals exhibit a striking morphogenetic defect in which the ventral nerve cord is greatly elongated and the brain hemispheres are misshapen. Lectin staining and blotting experiments confirmed that C1GalTA contributes to the synthesis of Gal-beta1,3-GalNAc in vivo. Our results identify a role for mucin-type O-glycosylation during neural development in Drosophila.
Collapse
Affiliation(s)
- Yuh-Ru Lin
- Howard Hughes Medical Institute, Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
26
|
Aoki K, Porterfield M, Lee SS, Dong B, Nguyen K, McGlamry KH, Tiemeyer M. The diversity of O-linked glycans expressed during Drosophila melanogaster development reflects stage- and tissue-specific requirements for cell signaling. J Biol Chem 2008; 283:30385-400. [PMID: 18725413 DOI: 10.1074/jbc.m804925200] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Appropriate glycoprotein O-glycosylation is essential for normal development and tissue function in multicellular organisms. To comprehensively assess the developmental and functional impact of altered O-glycosylation, we have extensively analyzed the non-glycosaminoglycan, O-linked glycans expressed in Drosophila embryos. Through multidimensional mass spectrometric analysis of glycans released from glycoproteins by beta-elimination, we detected novel as well as previously reported O-glycans that exhibit developmentally modulated expression. The core 1 mucin-type disaccharide (Galbeta1-3GalNAc) is the predominant glycan in the total profile. HexNAcitol, hexitol, xylosylated hexitol, and branching extension of core 1 with HexNAc (to generate core 2 glycans) were also evident following release and reduction. After Galbeta1-3GalNAc, the next most prevalent glycans were a mixture of novel, isobaric, linear, and branched forms of a glucuronyl core 1 disaccharide. Other less prevalent structures were also extended with HexA, including an O-fucose glycan. Although the expected disaccharide product of the Fringe glycosyltransferase, (GlcNAcbeta1-3)fucitol, was not detectable in whole embryos, mass spectrometry fragmentation and exoglycosidase sensitivity defined a novel glucuronyl trisaccharide as GlcNAcbeta1-3(GlcAbeta1-4)fucitol. Consistent with the spatial distribution of the Fringe function, the GlcA-extended form of the Fringe product was enriched in the dorsal portion of the wing imaginal disc. Furthermore, loss of Fringe activity reduced the prevalence of the O-Fuc trisaccharide. Therefore, O-Fuc glycans necessary for the modulation of important signaling events in Drosophila are, as in vertebrates, substrates for extension beyond the addition of a single HexNAc.
Collapse
Affiliation(s)
- Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602-4712, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Positional cloning of a Bombyx wingless locus flugellos (fl) reveals a crucial role for fringe that is specific for wing morphogenesis. Genetics 2008; 179:875-85. [PMID: 18505883 DOI: 10.1534/genetics.107.082784] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mutations at the flügellos (fl) locus in Bombyx mori produce wingless pupae and moths because of the repressed response of wing discs to ecdysteroid. Four recessive fl alleles occurred spontaneously and were mapped at 13.0 of the silkworm genetic linkage group 10. By positional cloning, we confirmed that the gene responsible for fl is fringe (fng) encoding Fng glycosyltransferase, which is involved in regulating the Notch signaling pathway. In four different fl alleles, we detected a large deletion of the fng gene in fl(k) and nonsense mutations in fl, fl(o), and fl(n). In the wild-type (WT) silkworm, fng is expressed actively in the wing discs, brain, and reproductive organs from the fourth to final instars but barely in the other tissues tested. In situ hybridization showed that fng mRNA is expressed in the dorsal layer of the WT wing discs. The wingless (wg) mRNA, a downstream marker of Fng-mediated Notch signaling, is localized at the dorsoventral boundary in the WT wing discs but repressed markedly in the fl wing discs. Although null mutants of Drosophila fng result in postembryonic lethality, loss of fng function in Bombyx affects only wing morphogenesis, suggesting different essential roles for fng in tissue differentiation among insects.
Collapse
|
28
|
Xu A, Haines N, Dlugosz M, Rana NA, Takeuchi H, Haltiwanger RS, Irvine KD. In Vitro Reconstitution of the Modulation of Drosophila Notch-Ligand Binding by Fringe. J Biol Chem 2007; 282:35153-62. [DOI: 10.1074/jbc.m707040200] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
29
|
Turnpenny PD, Alman B, Cornier AS, Giampietro PF, Offiah A, Tassy O, Pourquié O, Kusumi K, Dunwoodie S. Abnormal vertebral segmentation and the notch signaling pathway in man. Dev Dyn 2007; 236:1456-74. [PMID: 17497699 DOI: 10.1002/dvdy.21182] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Abnormal vertebral segmentation (AVS) in man is a relatively common congenital malformation but cannot be subjected to the scientific analysis that is applied in animal models. Nevertheless, some spectacular advances in the cell biology and molecular genetics of somitogenesis in animal models have proved to be directly relevant to human disease. Some advances in our understanding have come through DNA linkage analysis in families demonstrating a clustering of AVS cases, as well as adopting a candidate gene approach. Only rarely do AVS phenotypes follow clear Mendelian inheritance, but three genes-DLL3, MESP2, and LNFG-have now been identified for spondylocostal dysostosis (SCD). SCD is characterized by extensive hemivertebrae, trunkal shortening, and abnormally aligned ribs with points of fusion. In familial cases clearly following a Mendelian pattern, autosomal recessive inheritance is more common than autosomal dominant and the genes identified are functional within the Notch signaling pathway. Other genes within the pathway cause diverse phenotypes such as Alagille syndrome (AGS) and CADASIL, conditions that may have their origin in defective vasculogenesis. Here, we deal mainly with SCD and AGS, and present a new classification system for AVS phenotypes, for which, hitherto, the terminology has been inconsistent and confusing.
Collapse
Affiliation(s)
- Peter D Turnpenny
- Clinical Genetics, Royal Devon & Exeter Hospital, and Peninsula Medical School, Exeter, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Thomas GB, van Meyel DJ. The glycosyltransferase Fringe promotes Delta-Notch signaling between neurons and glia, and is required for subtype-specific glial gene expression. Development 2007; 134:591-600. [PMID: 17215308 DOI: 10.1242/dev.02754] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The development, organization and function of central nervous systems depend on interactions between neurons and glial cells. However, the molecular signals that regulate neuron-glial communication remain elusive. In the ventral nerve cord of Drosophila, the close association of the longitudinal glia (LG) with the neuropil provides an excellent opportunity to identify and characterize neuron-glial signals in vivo. We have found that the activity and restricted expression of the glycosyltransferase Fringe (Fng)renders a subset of LG sensitive to activation of signaling through the Notch(N) receptor. This is the first report showing that modulation of N signaling by Fng is important for central nervous system development in any organism. In each hemisegment of the nerve cord the transcription factor Prospero (Pros) is selectively expressed in the six most anterior LG. Pros expression is specifically reduced in fng mutants, and is blocked by antagonism of the N pathway. The N ligand Delta (Dl), which is expressed by a subset of neurons, cooperates with Fng for N signaling in the anterior LG, leading to subtype-specific expression of Pros. Furthermore, ectopic Pros expression in posterior LG can be triggered by Fng, and by Dl derived from neurons but not glia. This effect can be mimicked by direct activation of the N pathway within glia. Our genetic studies suggest that Fng sensitizes N on glia to axon-derived Dl and that enhanced neuron-glial communication through this ligand-receptor pair is required for the proper molecular diversity of glial cell subtypes in the developing nervous system.
Collapse
Affiliation(s)
- Graham B Thomas
- Graduate Program in Neurological Sciences, Montreal, QC, Canada
| | | |
Collapse
|
31
|
Sasaki N, Yoshida H, Fuwa TJ, Kinoshita-Toyoda A, Toyoda H, Hirabayashi Y, Ishida H, Ueda R, Nishihara S. Drosophila beta 1,4-N-acetylgalactosaminyltransferase-A synthesizes the LacdiNAc structures on several glycoproteins and glycosphingolipids. Biochem Biophys Res Commun 2007; 354:522-7. [PMID: 17239818 DOI: 10.1016/j.bbrc.2007.01.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2006] [Accepted: 01/04/2007] [Indexed: 10/23/2022]
Abstract
The GalNAcbeta1,4GlcNAc (LacdiNAc or LDN) structure is a more common structural feature in invertebrate glycoconjugates when compared with the Galbeta1,4GlcNAc structure. Recently, beta1,4-N-acetylgalactosaminyltransferase (beta4GalNAcT) was identified in some invertebrates including Drosophila. However, the LDN structure has not been reported in Drosophila, and the biological function of LDN remains to be determined. In this study, we examined acceptor substrate specificity of Drosophila beta4GalNAcTA by using some N- and O-glycans on glycoproteins and neutral glycosphingolipids (GSLs). GalNAc was efficiently transferred toward N-glycans, O-glycans, and the arthro-series GSLs. Moreover, we showed that dbeta4GalNAcTA contributed to the synthesis of the LDN structure in vivo. The dbeta4GalNAcTA mRNA was highly expressed in the developmental and adult neuronal tissues. Thus, these results suggest that dbeta4GalNAcTA acts on the terminal GlcNAc residue of some glycans for the synthesis of LDN, and the LDN structure may play a role in the physiological or neuronal development of Drosophila.
Collapse
Affiliation(s)
- Norihiko Sasaki
- Laboratory of Cell Biology, Department of Bioinformatics, Faculty of Engineering, Soka University, 1-236 Hachioji, Tokyo 192-8577, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sparrow DB, Chapman G, Turnpenny PD, Dunwoodie SL. Disruption of the somitic molecular clock causes abnormal vertebral segmentation. ACTA ACUST UNITED AC 2007; 81:93-110. [PMID: 17600782 DOI: 10.1002/bdrc.20093] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Somites are the precursors of the vertebral column. They segment from the presomitic mesoderm (PSM) that is caudally located and newly generated from the tailbud. Somites form in synchrony on either side of the embryonic midline in a reiterative manner. A molecular clock that operates in the PSM drives this reiterative process. Genetic manipulation in mouse, chick and zebrafish has revealed that the molecular clock controls the activity of the Notch and WNT signaling pathways in the PSM. Disruption of the molecular clock impacts on somite formation causing abnormal vertebral segmentation (AVS). A number of dysmorphic syndromes manifest AVS defects. Interaction between developmental biologists and clinicians has lead to groundbreaking research in this area with the identification that spondylocostal dysostosis (SCD) is caused by mutation in Delta-like 3 (DLL3), Mesoderm posterior 2 (MESP2), and Lunatic fringe (LFNG); three genes that are components of the Notch signaling pathway. This review describes our current understanding of the somitic molecular clock and highlights how key findings in developmental biology can impact on clinical practice.
Collapse
Affiliation(s)
- Duncan B Sparrow
- Developmental Biology Program, Victor Chang Cardiac Research Institute, Sydney, Australia
| | | | | | | |
Collapse
|
33
|
Luo Y, Koles K, Vorndam W, Haltiwanger RS, Panin VM. Protein O-fucosyltransferase 2 adds O-fucose to thrombospondin type 1 repeats. J Biol Chem 2006; 281:9393-9. [PMID: 16464857 DOI: 10.1074/jbc.m511975200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
O-Fucose is an unusual form of glycosylation found on epidermal growth factor-like (EGF) repeats and thrombospondin type 1 repeats (TSRs) in many secreted and transmembrane proteins. Recently O-fucose on EGF repeats was shown to play important roles in Notch signaling. In contrast, physiological roles for O-fucose on TSRs are unknown. In the accompanying paper (Luo, Y., Nita-Lazar, A., and Haltiwanger, R. S. (2006) J. Biol. Chem. 281, 9385-9392), we demonstrated that an enzyme distinct from protein O-fucosyltransferase 1 adds O-fucose to TSRs. A known homologue of O-fucosyltransferase 1 is putative protein O-fucosyltransferase 2. The cDNA sequence encoding O-fucosyltransferase 2 was originally identified during a data base search for fucosyltransferases in Drosophila. Like O-fucosyltransferase 1, O-fucosyltransferase 2 is conserved from Caenorhabditis elegans to humans. Although O-fucosyltransferase 2 was assumed to be another protein O-fucosyltransferase, no biochemical characterization existed supporting this contention. Here we show that RNAi-mediated reduction of the O-fucosyltransferase 2 message significantly decreased TSR-specific O-fucosyltransferase activity in Drosophila S2 cells. We also found that O-fucosyltransferase 2 is predominantly localized in the endoplasmic reticulum compartment of these cells. Furthermore, we expressed recombinant Drosophila O-fucosyltransferase 2 and showed that it O-fucosylates TSRs but not EGF repeats in vitro. These results demonstrate that O-fucosyltransferase 2 is in fact a TSR-specific O-fucosyltransferase.
Collapse
Affiliation(s)
- Yi Luo
- Department of Biochemistry and Cell Biology, Institute for Cell and Developmental Biology, Stony Brook University, Stony Brook, New York 11794-5215, USA
| | | | | | | | | |
Collapse
|
34
|
Sparrow DB, Chapman G, Wouters MA, Whittock NV, Ellard S, Fatkin D, Turnpenny PD, Kusumi K, Sillence D, Dunwoodie SL. Mutation of the LUNATIC FRINGE gene in humans causes spondylocostal dysostosis with a severe vertebral phenotype. Am J Hum Genet 2006; 78:28-37. [PMID: 16385447 PMCID: PMC1380221 DOI: 10.1086/498879] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Accepted: 10/05/2005] [Indexed: 01/15/2023] Open
Abstract
The spondylocostal dysostoses (SCDs) are a heterogeneous group of vertebral malsegmentation disorders that arise during embryonic development by a disruption of somitogenesis. Previously, we had identified two genes that cause a subset of autosomal recessive forms of this disease: DLL3 (SCD1) and MESP2 (SCD2). These genes are important components of the Notch signaling pathway, which has multiple roles in development and disease. Here, we have used a candidate-gene approach to identify a mutation in a third Notch pathway gene, LUNATIC FRINGE (LFNG), in a family with autosomal recessive SCD. LFNG encodes a glycosyltransferase that modifies the Notch family of cell-surface receptors, a key step in the regulation of this signaling pathway. A missense mutation was identified in a highly conserved phenylalanine close to the active site of the enzyme. Functional analysis revealed that the mutant LFNG was not localized to the correct compartment of the cell, was unable to modulate Notch signaling in a cell-based assay, and was enzymatically inactive. This represents the first known mutation in the human LFNG gene and reinforces the hypothesis that proper regulation of the Notch signaling pathway is an absolute requirement for the correct patterning of the axial skeleton.
Collapse
Affiliation(s)
- D. B. Sparrow
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - G. Chapman
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - M. A. Wouters
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - N. V. Whittock
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - S. Ellard
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - D. Fatkin
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - P. D. Turnpenny
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - K. Kusumi
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - D. Sillence
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| | - S. L. Dunwoodie
- Developmental Biology Program, Computational Biology and Bioinformatics Program, and Sr. Bernice Research Program in Inherited Heart Diseases, Victor Chang Cardiac Research Institute, Cardiology Department, St. Vincent’s Hospital, Faculties of Medicine and Science, University of New South Wales, and Department of Medical Genetics, The Children’s Hospital at Westmead, Sydney; Institute of Biomedical and Clinical Science, Peninsular Medical School, United Kingdom; Clinical Genetics Department, Royal Devon & Exeter Hospital, Exeter, United Kingdom; and Divisions of Human Genetics and Orthopedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia
| |
Collapse
|
35
|
Arboleda-Velasquez JF, Rampal R, Fung E, Darland DC, Liu M, Martinez MC, Donahue CP, Navarro-Gonzalez MF, Libby P, D'Amore PA, Aikawa M, Haltiwanger RS, Kosik KS. CADASIL mutations impair Notch3 glycosylation by Fringe. Hum Mol Genet 2005; 14:1631-9. [PMID: 15857853 DOI: 10.1093/hmg/ddi171] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mutations in the NOTCH3 gene trigger adult-onset stroke and vascular dementia in patients with CADASIL (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy). All CADASIL mutations described to date affect the epidermal growth factor-like (EGF-like) repeats located in the extracellular domain of the Notch3 receptor. These domains are also the target of sequential complex O-linked glycosylation mediated by protein O-fucosyltransferase 1 and Fringe. We investigated whether O-fucosylation or Fringe-mediated elongation of O-fucose on Notch3 is impaired by CADASIL mutations. Biochemical studies of a Notch3 fragment containing the first five EGF-like repeats of Notch3, including the mutational hot spot, showed that CADASIL mutations do not affect the addition of O-fucose but do impair carbohydrate chain elongation by Fringe. CADASIL changes also induced aberrant homodimerization of mutant Notch3 fragments and heterodimerization of mutant Notch3 with Lunatic Fringe itself. Together, these data suggest that Fringe plays a role in CADASIL pathophysiology.
Collapse
|
36
|
Haines N, Irvine KD. Functional analysis of Drosophila beta1,4-N-acetlygalactosaminyltransferases. Glycobiology 2004; 15:335-46. [PMID: 15563714 DOI: 10.1093/glycob/cwi017] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Members of the mammalian beta1,4-galactosyltransferase family are among the best studied glycosyltransferases, but the requirements for all members of this family within an animal have not previously been determined. Here, we describe analysis of two Drosophila genes, beta4GalNAcTA (CG8536) and beta4GalNAcTB (CG14517), that are homologous to mammalian beta1,4-galactosyltransferases. Like their mammalian homologs, these glycosyltransferases use N-acetylglucosamine as an acceptor substrate. However, they transfer N-acetylgalactosamine rather than galactose. This activity, together with amino acid sequence similarity, places them among a group of recently identified invertebrate beta1,4-N-acetylgalactosaminyltransferases. To investigate the biological functions of these genes, null mutations were generated by imprecise excision of a transposable element (beta4GalNAcTA) or by gene-targeted homologous recombination (beta4GalNAcTB). Flies mutant for beta4GalNAcTA are viable and fertile but display behavioral phenotypes suggestive of essential roles for GalNAc-beta1,4-GlcNAc containing glycoconjugates in neuronal and/or muscular function. beta4GalNAcTB mutants are viable and display no evident morphological or behavioral phenotypes. Flies doubly mutant for both genes display only the behavioral phenotypes associated with mutation of beta4GalNAcTA. Thus Drosophila homologs of the mammalian beta4GalT family are essential for neuromuscular physiology or development but are not otherwise required for viability, fertility, or external morphology.
Collapse
Affiliation(s)
- Nicola Haines
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway NJ 08854, USA
| | | |
Collapse
|
37
|
Weber K, Johnson N, Champlin D, Patty A. Many P-element insertions affect wing shape in Drosophila melanogaster. Genetics 2004; 169:1461-75. [PMID: 15545659 PMCID: PMC1449561 DOI: 10.1534/genetics.104.027748] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A screen of random, autosomal, homozygous-viable P-element insertions in D. melanogaster found small effects on wing shape in 11 of 50 lines. The effects were due to single insertions and remained stable and significant for over 5 years, in repeated, high-resolution measurements. All 11 insertions were within or near protein-coding transcription units, none of which were previously known to affect wing shape. Many sites in the genome can affect wing shape.
Collapse
Affiliation(s)
- Kenneth Weber
- Department of Biological Sciences, University of Southern Maine, Portland, 04104-9300, USA.
| | | | | | | |
Collapse
|
38
|
Affiliation(s)
- W W Young
- Department of Molecular, Cellular, and Craniofacial Biology, School of Dentistry, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
39
|
Abstract
Intracellular post-translational modifications such as phosphorylation and ubiquitylation have been well studied for their roles in regulating diverse signalling pathways, but we are only just beginning to understand how differential glycosylation is used to regulate intercellular signalling. Recent studies make clear that extracellular post-translational modifications, in the form of glycosylation, are essential for the Notch signalling pathway, and that differences in the extent of glycosylation are a significant mechanism by which this pathway is regulated.
Collapse
Affiliation(s)
- Nicola Haines
- Howard Hughes Medical Institute, Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers: The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|