1
|
Zhao Y, Geng J, Meng Z, Sun Y, Ou M, Xu L, Li M, Gan G, Rui M, Han J, Xie W. Neurexin facilitates glycosylation of Dystroglycan to sustain muscle architecture and function in Drosophila. Commun Biol 2024; 7:1481. [PMID: 39521920 PMCID: PMC11550397 DOI: 10.1038/s42003-024-07191-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Neurexin, a molecule associated with autism spectrum disorders, is thought to function mainly in neurons. Recently, it was reported that Neurexin is also present in muscle, but the role of Neurexin in muscle is still poorly understood. Here, we demonstrate that the overexpression of Neurexin in muscles effectively restored the locomotor function of Drosophila neurexin mutants, while rescuing effects are observed within the nervous. Notably, the defects in muscle structure and function caused by Neurexin deficiency were similar to those caused by mutations in dystroglycan, a gene associated with progressive muscular dystrophy. The absence of Neurexin leads to muscle attachment defects, emphasizing the essential role of Neurexin in muscle integrity. Furthermore, Neurexin deficiency reduces Dystroglycan glycosylation on the cell surface, which is crucial for maintaining proper muscle structure and function. Finally, Neurexin guides Dystroglycan to the glycosyltransferase complex through interactions with Rotated Abdomen, a homolog of mammalian POMT1. Our findings reveal that Neurexin mediates muscle development and function through Dystroglycan glycosylation, suggesting a potential association between autism spectrum disorders and muscular dystrophy.
Collapse
Affiliation(s)
- Yu Zhao
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Junhua Geng
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China.
| | - Zhu Meng
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Yichen Sun
- The Southern Modern Forestry Collaborative Innovation Center, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing, 210037, China
| | - Mengzhu Ou
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Lizhong Xu
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Moyi Li
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Guangming Gan
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
- Key Laboratory of Developmental Genes and Human Disease, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Menglong Rui
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Junhai Han
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Wei Xie
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
2
|
Mu M, Sun H, Geng S, Xu T, Sun C, Zhang Z, Meng S, Li M, Liu A, Yang Z, Xie W. Neurexin-3 in the paraventricular nucleus of the hypothalamus regulates body weight and glucose homeostasis independently of food intake. Mol Brain 2024; 17:49. [PMID: 39090731 PMCID: PMC11295692 DOI: 10.1186/s13041-024-01124-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024] Open
Abstract
Neurexin-3 (Nrxn3) has been genetically associated with obesity, but the underlying neural mechanisms remain poorly understood. This study aimed to investigate the role of Nrxn3 in the paraventricular nucleus of the hypothalamus (PVN) in regulating energy balance and glucose homeostasis. We found that Nrxn3 expression in the PVN was upregulated in response to metabolic stressors, including cold exposure and fasting. Using Cre-loxP technology, we selectively ablated Nrxn3 in CaMKIIα-expressing neurons of the PVN in male mice. This genetic manipulation resulted in marked weight gain attributable to increased adiposity and impaired glucose tolerance, without affecting food intake. Our findings identify PVN CaMKIIα-expressing neurons as a critical locus where Nrxn3 modulates energy balance by regulating adipogenesis and glucose metabolism, independently of appetite. These results reveal a novel neural mechanism potentially linking Nrxn3 dysfunction to obesity pathogenesis, suggesting that targeting PVN Nrxn3-dependent neural pathways may inform new therapeutic approaches for obesity prevention and treatment.
Collapse
Affiliation(s)
- Mingdao Mu
- School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, P. R. China.
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China.
| | - Haoyu Sun
- School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, P. R. China
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China
| | - Shuyan Geng
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China
| | - Tianxiang Xu
- School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, P. R. China
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China
| | - Chuanyao Sun
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China
| | - Zixu Zhang
- School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, P. R. China
| | - Sibie Meng
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China
| | - Moyi Li
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China
| | - An Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China
| | - Zhiyuan Yang
- School of Artificial Intelligence, Hangzhou Dianzi University, Hangzhou, P. R. China.
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, P. R. China.
| |
Collapse
|
3
|
Iwanaga R, Yahagi N, Hakeda‐Suzuki S, Suzuki T. Cell adhesion and actin dynamics factors promote axonal extension and synapse formation in transplanted Drosophila photoreceptor cells. Dev Growth Differ 2024; 66:205-218. [PMID: 38403285 PMCID: PMC11457513 DOI: 10.1111/dgd.12916] [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: 11/24/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/27/2024]
Abstract
Vision is formed by the transmission of light stimuli to the brain through axons extending from photoreceptor cells. Damage to these axons leads to loss of vision. Despite research on neural circuit regeneration through transplantation, achieving precise axon projection remains challenging. To achieve optic nerve regeneration by transplantation, we employed the Drosophila visual system. We previously established a transplantation method for Drosophila utilizing photoreceptor precursor cells extracted from the eye disc. However, little axonal elongation of transplanted cells into the brain, the lamina, was observed. We verified axonal elongation to the lamina by modifying the selection process for transplanted cells. Moreover, we focused on N-cadherin (Ncad), a cell adhesion factor, and Twinstar (Tsr), which has been shown to promote actin reorganization and induce axon elongation in damaged nerves. Overexpression of Ncad and tsr promoted axon elongation to the lamina, along with presynaptic structure formation in the elongating axons. Furthermore, overexpression of Neurexin-1 (Nrx-1), encoding a protein identified as a synaptic organizer, was found to not only promote presynapse formation but also enhance axon elongation. By introducing Ncad, tsr, and Nrx-1, we not only successfully achieved axonal projection of transplanted cells to the brain beyond the retina, but also confirmed the projection of transplanted cells into a deeper ganglion, the medulla. The present study offers valuable insights to realize regeneration through transplantation in a more complex nervous system.
Collapse
Affiliation(s)
- Riku Iwanaga
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| | - Nagisa Yahagi
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| | - Satoko Hakeda‐Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
- Research Initiatives and Promotion OrganizationYokohama National UniversityYokohamaJapan
| | - Takashi Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| |
Collapse
|
4
|
Lim-Kian-Siang G, Izawa-Ishiguro AR, Rao Y. Neurexin-1-dependent circuit activity is required for the maintenance of photoreceptor subtype identity in Drosophila. Mol Brain 2024; 17:2. [PMID: 38167109 PMCID: PMC10759516 DOI: 10.1186/s13041-023-01073-3] [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: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
In the human and Drosophila color vision system, each photoreceptor neuron (cone cell in humans and R7/R8 photoreceptor cell in Drosophila) makes a stochastic decision to express a single photopigment of the same family with the exclusion of the others. While recent studies have begun to reveal the mechanisms that specify the generation of cone subtypes during development in mammals, nothing is known about how the mosaic of mutually exclusive cone subtypes is maintained in the mammalian retina. In Drosophila, recent work has led to the identification of several intrinsic factors that maintain the identity of R8 photoreceptor subtypes in adults. Whether and how extrinsic mechanisms are involved, however, remain unknown. In this study, we present evidence that supports that the Drosophila transsynaptic adhesion molecule Neurexin 1 (Dnrx-1) is required non-cell autonomously in R8p subtypes for the maintenance of R8y subtype identity. Silencing the activity of R8p subtypes caused a phenotype identical to that in dnrx-1 mutants. These results support a novel role for Nrx-1-dependent circuit activity in mediating the communication between R8 photoreceptor subtypes for maintaining the subtype identity in the retina.
Collapse
Affiliation(s)
- Gabrielle Lim-Kian-Siang
- McGill Centre for Research in Neuroscience, Montreal, Canada
- Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Arianna R Izawa-Ishiguro
- McGill Centre for Research in Neuroscience, Montreal, Canada
- Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Yong Rao
- McGill Centre for Research in Neuroscience, Montreal, Canada.
- Department of Neurology and Neurosurgery, Montreal, Canada.
- Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
- Centre for Research in Neuroscience, McGill University Health Centre, Room L7-136, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
| |
Collapse
|
5
|
李 莉, 张 倩, 刘 欢, 吴 琼, 杨 亭, 陈 洁, 李 廷. Involvement of retinoic acid receptor α in the autistic-like behavior of rats with vitamin A deficiency by regulating neurexin 1 in the visual cortex: a mechanism study. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:928-935. [PMID: 36036133 PMCID: PMC9425865 DOI: 10.7499/j.issn.1008-8830.2204016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES To study the mechanism of retinoic acid receptor α (RARα) signal change to regulate neurexin 1 (NRXN1) in the visual cortex and participate in the autistic-like behavior in rats with vitamin A deficiency (VAD). METHODS The models of vitamin A normal (VAN) and VAD pregnant rats were established, and some VAD maternal and offspring rats were given vitamin A supplement (VAS) in the early postnatal period. Behavioral tests were performed on 20 offspring rats in each group at the age of 6 weeks. The three-chamber test and the open-field test were used to observe social behavior and repetitive stereotyped behavior. High-performance liquid chromatography was used to measure the serum level of retinol in the offspring rats in each group. Electrophysiological experiments were used to measure the long-term potentiation (LTP) level of the visual cortex in the offspring rats. Quantitative real-time PCR and Western blot were used to measure the expression levels of RARα, NRXN1, and N-methyl-D-aspartate receptor 1 (NMDAR1). Chromatin co-immunoprecipitation was used to measure the enrichment of RARα transcription factor in the promoter region of the NRXN1 gene. RESULTS The offspring rats in the VAD group had autistic-like behaviors such as impaired social interactions and repetitive stereotypical behaviors, and VAS started immediately after birth improved most of the behavioral deficits in offspring rats. The offspring rats in the VAD group had a significantly lower serum level of retinol than those in the VAN and VAS groups (P<0.05). Compared with the offspring rats in the VAN and VAS groups, the offspring rats in the VAD group had significant reductions in the mRNA and protein expression levels of NMDAR1, RARα, and NRXN1 and the LTP level of the visual cortex (P<0.05). The offspring rats in the VAD group had a significant reduction in the enrichment of RARα transcription factor in the promoter region of the NRXN1 gene in the visual cortex compared with those in the VAN and VAS groups (P<0.05). CONCLUSIONS RARα affects the synaptic plasticity of the visual cortex in VAD rats by regulating NRXN1, thereby participating in the formation of autistic-like behaviors in VAD rats.
Collapse
|
6
|
Liu Y, Shen L, Zhang Y, Zhao R, Liu C, Luo S, Chen J, Xia L, Li T, Peng Y, Xia K. Rare NRXN1 missense variants identified in autism interfered protein degradation and Drosophila sleeping. J Psychiatr Res 2021; 143:113-122. [PMID: 34487988 DOI: 10.1016/j.jpsychires.2021.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 11/29/2022]
Abstract
NRXN1 is involved in synaptogenesis and have been implicated in Autism spectrum disorders. However, many rare inherited missense variants of NRXN1 have not been thoroughly evaluated. Here, functional analyses in vitro and in Drosophila of three NRXN1 missense mutations, Y282H, L893V, and I1135V identified in ASD patients in our previous study were performed. Our results showed these three mutations interfered protein degradation compared with NRXN1-WT protein. Expressing human NRXN1 in Drosophila could lead to abnormal circadian rhythm and sleep behavior, and three mutated proteins caused milder phenotypes, indicating the mutations may change the function of NRXN1 slightly. These findings highlight the functional role of rare NRXN1 missense variants identified in autism patients, and provide clues for us to better understand the pathogenesis of abnormal circadian rhythm and sleep behavior of other organisms, including humans.
Collapse
Affiliation(s)
- Yalan Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, China; Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Otolaryngology Major Disease Research of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Shen
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Yaowen Zhang
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Rongjuan Zhao
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Cenying Liu
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Sanchuan Luo
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Jingjing Chen
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Lu Xia
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Taoxi Li
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, China; Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Otolaryngology Major Disease Research of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Peng
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; Key Laboratory of Animal Models for Human Diseases of Hunan Province, Central South University, Changsha, China
| | - Kun Xia
- Center for Medical Genetics and Hunan Provincial Key Laboratory for Medical Genetics, School of Life Sciences, Central South University, Changsha, China; CAS Center for Excellence in Brain Science and Intelligences Technology (CEBSIT), Shanghai, China; Key Laboratory of Medical Information Research, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Salim S, Banu A, Alwa A, Gowda SBM, Mohammad F. The gut-microbiota-brain axis in autism: what Drosophila models can offer? J Neurodev Disord 2021; 13:37. [PMID: 34525941 PMCID: PMC8442445 DOI: 10.1186/s11689-021-09378-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/06/2021] [Indexed: 12/28/2022] Open
Abstract
The idea that alterations in gut-microbiome-brain axis (GUMBA)-mediated communication play a crucial role in human brain disorders like autism remains a topic of intensive research in various labs. Gastrointestinal issues are a common comorbidity in patients with autism spectrum disorder (ASD). Although gut microbiome and microbial metabolites have been implicated in the etiology of ASD, the underlying molecular mechanism remains largely unknown. In this review, we have summarized recent findings in human and animal models highlighting the role of the gut-brain axis in ASD. We have discussed genetic and neurobehavioral characteristics of Drosophila as an animal model to study the role of GUMBA in ASD. The utility of Drosophila fruit flies as an amenable genetic tool, combined with axenic and gnotobiotic approaches, and availability of transgenic flies may reveal mechanistic insight into gut-microbiota-brain interactions and the impact of its alteration on behaviors relevant to neurological disorders like ASD.
Collapse
Affiliation(s)
- Safa Salim
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Ayesha Banu
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Amira Alwa
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Swetha B M Gowda
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Farhan Mohammad
- Division of Biological and Biomedical Sciences (BBS), College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar.
| |
Collapse
|
8
|
Mitra S, Ryoo HD. The role of Ire1 in Drosophila eye pigmentation revealed by an RNase dead allele. Dev Biol 2021; 478:205-211. [PMID: 34265355 DOI: 10.1016/j.ydbio.2021.07.008] [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: 04/17/2021] [Revised: 06/25/2021] [Accepted: 07/10/2021] [Indexed: 11/29/2022]
Abstract
Ire1 is an endoplasmic reticulum (ER) transmembrane RNase that cleaves substrate mRNAs to help cells adapt to ER stress. Because there are cell types with physiological ER stress, loss of Ire1 results in metabolic and developmental defects in diverse organisms. In Drosophila, Ire1 mutants show developmental defects at early larval stages and in pupal eye photoreceptor differentiation. These Drosophila studies relied on a single Ire1 loss of function allele with a Piggybac insertion in the coding sequence. Here, we report that an Ire1 allele with a specific impairment in the RNase domain, H890A, unmasks previously unrecognized Ire1 phenotypes in Drosophila eye pigmentation. Specifically, we found that the adult eye pigmentation is altered, and the pigment granules are compromised in Ire1H890A homozygous mosaic eyes. Furthermore, the Ire1H890A mutant eyes had dramatically reduced Rhodopsin-1 protein levels. Drosophila eye pigment granules are most notably associated with late endosome/lysosomal defects. Our results indicate that the loss of Ire1, which would impair ER homeostasis, also results in altered adult eye pigmentation.
Collapse
Affiliation(s)
- Sahana Mitra
- Department of Cell Biology, NYU Grossman School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Hyung Don Ryoo
- Department of Cell Biology, NYU Grossman School of Medicine, 550 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
9
|
Liu D, Wang X, Guo H, Zhang X, Zhang M, Tang W. Chromosome-level genome assembly of the endangered humphead wrasse Cheilinus undulatus: Insight into the expansion of opsin genes in fishes. Mol Ecol Resour 2021; 21:2388-2406. [PMID: 34003602 DOI: 10.1111/1755-0998.13429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
Wrasses are dominant components of major coral reef systems. Among wrasses, Cheilinus undulatus is an endangered species with high economic and ecological value that exhibits sex reversal of females to males, while sexual selection occurs in breeding aggregations. However, the molecular-associated mechanism underlying this remains unclear. Opsin gene diversification is regarded as a potent force in sexual selection. Here we present a genome assembly of C. undulatus, using Illumina, Nanopore and Hi-C sequencing. The 1.17 Gb genome was generated from 328 contigs with an N50 length of 16.5 Mb and anchored to 24 chromosomes. In total, 22,218 genes were functionally annotated, and 96.36% of BUSCO genes were fully represented. Transcriptomic analyses showed that 96.79% of the predicted genes were expressed. Transposons were most abundant, accounting for 39.88% of the genome, with low divergence, owing to their evolution with close species ~60.53 million years ago. In total, 567/1,826 gene families were expanded and contracted in the reconstructed phylogeny, respectively. Forty-six genes were under positive selection. Comparative genomic analyses with other fish revealed expansion of opsin SWS2B, LWS1 and Rh2. The elevated duplicates of SWS2B were generated by gene conversions via transposition of transposons followed by nonallelic homologous recombination. Amino acid substitutions of opsin paralogues occurred at key tuning sites, causing a spectral shift in maximal absorbance of visual pigment to capture functional changes. Among these opsin genes, SWS2B-3 and 4 and Rh1 are expressed in the retina. The genome sequence of C. undulatus provides valuable resources for future investigation of the conservation, evolution and behaviour of fishes.
Collapse
Affiliation(s)
- Dong Liu
- Shanghai Universities Key Laboratory of Marine Animal Taxonomy and Evolution, Shanghai Ocean University, Shanghai, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xinyang Wang
- Shanghai Universities Key Laboratory of Marine Animal Taxonomy and Evolution, Shanghai Ocean University, Shanghai, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Hongyi Guo
- Shanghai Universities Key Laboratory of Marine Animal Taxonomy and Evolution, Shanghai Ocean University, Shanghai, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xuguang Zhang
- Shanghai Universities Key Laboratory of Marine Animal Taxonomy and Evolution, Shanghai Ocean University, Shanghai, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, GA, USA
| | - Wenqiao Tang
- Shanghai Universities Key Laboratory of Marine Animal Taxonomy and Evolution, Shanghai Ocean University, Shanghai, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| |
Collapse
|
10
|
Parallel Synaptic Acetylcholine Signals Facilitate Large Monopolar Cell Repolarization and Modulate Visual Behavior in Drosophila. J Neurosci 2021; 41:2164-2176. [PMID: 33468565 DOI: 10.1523/jneurosci.2388-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/03/2020] [Accepted: 01/03/2021] [Indexed: 11/21/2022] Open
Abstract
Appropriate termination of the photoresponse in image-forming photoreceptors and downstream neurons is critical for an animal to achieve high temporal resolution. Although the cellular and molecular mechanisms of termination in image-forming photoreceptors have been extensively studied in Drosophila, the underlying mechanism of termination in their downstream large monopolar cells remains less explored. Here, we show that synaptic ACh signaling, from both amacrine cells (ACs) and L4 neurons, facilitates the rapid repolarization of L1 and L2 neurons. Intracellular recordings in female flies show that blocking synaptic ACh output from either ACs or L4 neurons leads to slow repolarization of L1 and L2 neurons. Genetic and electrophysiological studies in both male and female flies determine that L2 neurons express ACh receptors and directly receive ACh signaling. Moreover, our results demonstrate that synaptic ACh signaling from both ACs and L4 neurons simultaneously facilitates ERG termination. Finally, visual behavior studies in both male and female flies show that synaptic ACh signaling, from either ACs or L4 neurons to L2 neurons, is essential for the optomotor response of the flies in high-frequency light stimulation. Our study identifies parallel synaptic ACh signaling for repolarization of L1 and L2 neurons and demonstrates that synaptic ACh signaling facilitates L1 and L2 neuron repolarization to maintain the optomotor response of the fly on high-frequency light stimulation.SIGNIFICANCE STATEMENT The image-forming photoreceptor downstream neurons receive multiple synaptic inputs from image-forming photoreceptors and various types of interneurons. It remains largely unknown how these synaptic inputs modulate the neural activity and function of image-forming photoreceptor downstream neurons. We show that parallel synaptic ACh signaling from both amacrine cells and L4 neurons facilitates rapid repolarization of large monopolar cells in Drosophila and maintains the optomotor response of the fly on high-frequency light stimulation. This work is one of the first reports showing how parallel synaptic signaling modulates the activity of large monopolar cells and motion vision simultaneously.
Collapse
|
11
|
Yu Z, Du J, Hui H, Kan S, Huo T, Zhao K, Wu T, Guo Q, Lu N. LT-171-861, a novel FLT3 inhibitor, shows excellent preclinical efficacy for the treatment of FLT3 mutant acute myeloid leukemia. Am J Cancer Res 2021; 11:93-106. [PMID: 33391463 PMCID: PMC7681098 DOI: 10.7150/thno.46593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Acute myeloid leukemia (AML) is a common type of haematological malignancy. Several studies have shown that neoplasia in AML is enhanced by tyrosine kinase pathways. Recently, given that aberrant activation of Fms-like tyrosine receptor kinase 3 (FLT3) acts as a critical survival signal for cancer cells in 20‒30% patients with AML, inhibition of FLT3 may be a potential therapeutic strategy. Therefore, we identified LT-171-861, a novel kinase inhibitor with remarkable inhibitory activity against FLT3, in preclinical models of AML. Methods: We determined the inhibitory effects of LT-171-861 in vitro using AML cell lines and transformed BaF3 cells. Target engagement assays were used to verify the interaction between LT-171-861 and FLT3. Finally, a subcutaneous model and a bone marrow engrafted model were used to evaluate the antitumor effects of LT‑171‑861 in vivo. Results: Our data demonstrated that LT-171-861 had high affinity for FLT3 protein. We also showed that LT-171-861 had an inhibitory effect on FLT3 mutants in cellular assays. Moreover, LT-171-861 had a growth-inhibitory effect on human AML cell lines harboring FLT3 internal tandem duplications (FLT3-ITD) such as FLT3-D835Y, FLT3‑ITD-N676D, FLT3-ITD-D835Y, FLT3-ITD-F691L, FLT3-ITD-Y842C and AML blasts from patients with FLT3-ITD. Furthermore, LT-171-861 showed potent antileukemic efficacy against AML cells. We also show the efficacy of LT‑171-861 in a subcutaneous implantation model and a bone marrow engrafted model in vivo, where administration of LT-171-861 led to almost complete tumor regression and increased survival. Conclusions: Overall, this study not only identifies LT-171-861 as a potent FLT3 inhibitor, but also provides a rationale for the upcoming clinical trial of LT-171-861 in patients with AML and FLT3-ITD mutations.
Collapse
|
12
|
Gu Q, Wu J, Tian Y, Cheng S, Zhang ZC, Han J. Gαq splice variants mediate phototransduction, rhodopsin synthesis, and retinal integrity in Drosophila. J Biol Chem 2020; 295:5554-5563. [PMID: 32198182 PMCID: PMC7186184 DOI: 10.1074/jbc.ra120.012764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/19/2020] [Indexed: 11/06/2022] Open
Abstract
Heterotrimeric G proteins mediate a variety of signaling processes by coupling G protein-coupled receptors to intracellular effector molecules. In Drosophila, the Gαq gene encodes several Gαq splice variants, with the Gαq1 isoform protein playing a major role in fly phototransduction. However, Gαq1 null mutant flies still exhibit a residual light response, indicating that other Gαq splice variants or additional Gq α subunits are involved in phototransduction. Here, we isolated a mutant fly with no detectable light responses, decreased rhodopsin (Rh) levels, and rapid retinal degeneration. Using electrophysiological and genetic studies, biochemical assays, immunoblotting, real-time RT-PCR, and EM analysis, we found that mutations in the Gαq gene disrupt light responses and demonstrate that the Gαq3 isoform protein is responsible for the residual light response in Gαq1 null mutants. Moreover, we report that Gαq3 mediates rhodopsin synthesis. Depletion of all Gαq splice variants led to rapid light-dependent retinal degeneration, due to the formation stable Rh1-arrestin 2 (Arr2) complexes. Our findings clarify essential roles of several different Gαq splice variants in phototransduction and retinal integrity in Drosophila and reveal that Gαq3 functions in rhodopsin synthesis.
Collapse
Affiliation(s)
- Qiuxiang Gu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China
| | - Jinglin Wu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yao Tian
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China
| | - Shanshan Cheng
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China
| | - Zi Chao Zhang
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China.
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
13
|
Luo F, Sclip A, Jiang M, Südhof TC. Neurexins cluster Ca 2+ channels within the presynaptic active zone. EMBO J 2020; 39:e103208. [PMID: 32134527 PMCID: PMC7110102 DOI: 10.15252/embj.2019103208] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 02/05/2023] Open
Abstract
To achieve ultrafast neurotransmission, neurons assemble synapses with highly organized presynaptic and postsynaptic nanomachines that are aligned by synaptic adhesion molecules. How functional assembly of presynaptic active zones is controlled via trans-synaptic interactions remains unknown. Here, we conditionally deleted all three neurexin adhesion molecules from presynaptic neurons of the calyx of Held in the mouse auditory system, a model synapse that allows precise biophysical analyses of synaptic properties. The pan-neurexin deletion had no effect on synapse development or the basic release machinery, but dramatically impaired fast neurotransmitter release. The overall properties of presynaptic calcium ion channels appeared normal, as reflected by the similar characteristics of calcium currents recorded at the nerve terminals. However, the pan-neurexin deletion significantly impaired the tight coupling of calcium influx to exocytosis, thereby suppressing neurotransmitter release. Furthermore, the pan-neurexin deletion reduced the function of calcium-activated BK potassium channels, whose activation depends on their tight association with presynaptic calcium channels. Together, these results suggest that neurexins perform a major function at the calyx synapse in coupling presynaptic calcium channels to release sites.
Collapse
Affiliation(s)
- Fujun Luo
- Department of Molecular and Cellular PhysiologyHoward Hughes Medical InstituteStanford University Medical SchoolStanfordCAUSA
| | - Alessandra Sclip
- Department of Molecular and Cellular PhysiologyHoward Hughes Medical InstituteStanford University Medical SchoolStanfordCAUSA
| | - Man Jiang
- Department of Molecular and Cellular PhysiologyHoward Hughes Medical InstituteStanford University Medical SchoolStanfordCAUSA
| | - Thomas C Südhof
- Department of Molecular and Cellular PhysiologyHoward Hughes Medical InstituteStanford University Medical SchoolStanfordCAUSA
| |
Collapse
|
14
|
Katana R, Guan C, Zanini D, Larsen ME, Giraldo D, Geurten BRH, Schmidt CF, Britt SG, Göpfert MC. Chromophore-Independent Roles of Opsin Apoproteins in Drosophila Mechanoreceptors. Curr Biol 2019; 29:2961-2969.e4. [PMID: 31447373 DOI: 10.1016/j.cub.2019.07.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/24/2019] [Accepted: 07/11/2019] [Indexed: 12/23/2022]
Abstract
Rhodopsins, the major light-detecting molecules of animal visual systems [1], consist of opsin apoproteins that covalently bind a retinal chromophore with a conserved lysine residue [1, 2]. In addition to capturing photons, this chromophore contributes to rhodopsin maturation [3, 4], trafficking [3, 4], and stabilization [5], and defects in chromophore synthesis and recycling can cause dysfunction of the retina and dystrophy [6-9]. Indications that opsin apoproteins alone might have biological roles have come from archaebacteria and platyhelminths, which present opsin-like proteins that lack the chromophore binding site and are deemed to function independently of light [10, 11]. Light-independent sensory roles have been documented for Drosophila opsins [12-15], yet also these unconventional opsin functions are thought to require chromophore binding [12, 13, 15]. Unconjugated opsin apoproteins act as phospholipid scramblases in mammalian photoreceptor disks [16], yet chromophore-independent roles of opsin apoproteins outside of eyes have, to the best of our knowledge, hitherto not been described. Drosophila chordotonal mechanoreceptors require opsins [13, 15], and we find that their function remains uncompromised by nutrient carotenoid depletion. Disrupting carotenoid uptake and cleavage also left the mechanoreceptors unaffected, and manipulating the chromophore attachment site of the fly's major visual opsin Rh1 impaired photoreceptor, but not mechanoreceptor, function. Notwithstanding this chromophore independence, some proteins that process and recycle the chromophore in the retina are also required in mechanoreceptors, including visual cycle components that recycle the chromophore upon its photoisomerization. Our results thus establish biological function for unconjugated opsin apoproteins outside of eyes and, in addition, document chromophore-independent roles for chromophore pathway components.
Collapse
Affiliation(s)
- Radoslaw Katana
- Department of Cellular Neurobiology, University of Göttingen, 37077 Göttingen, Germany
| | - Chonglin Guan
- Faculty of Physics, Third Institute of Physics - Biophysics, University of Göttingen, 37077 Göttingen, Germany
| | - Damiano Zanini
- Department of Cellular Neurobiology, University of Göttingen, 37077 Göttingen, Germany
| | - Matthew E Larsen
- Departments of Neurology and Ophthalmology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - Diego Giraldo
- Department of Cellular Neurobiology, University of Göttingen, 37077 Göttingen, Germany
| | - Bart R H Geurten
- Department of Cellular Neurobiology, University of Göttingen, 37077 Göttingen, Germany
| | - Christoph F Schmidt
- Faculty of Physics, Third Institute of Physics - Biophysics, University of Göttingen, 37077 Göttingen, Germany; Department of Physics and Soft Matter Center, Duke University, Durham, NC 27708, USA
| | - Steven G Britt
- Departments of Neurology and Ophthalmology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - Martin C Göpfert
- Department of Cellular Neurobiology, University of Göttingen, 37077 Göttingen, Germany.
| |
Collapse
|
15
|
Proteomic insight into the pathogenesis of CAPN5-vitreoretinopathy. Sci Rep 2019; 9:7608. [PMID: 31110225 PMCID: PMC6527583 DOI: 10.1038/s41598-019-44031-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
CAPN5 Neovascular Inflammatory Vitreoretinopathy (CAPN5-NIV; OMIM 193235) is a poorly-understood rare, progressive inflammatory intraocular disease with limited therapeutic options. To profile disease effector proteins in CAPN5-NIV patient vitreous, liquid vitreous biopsies were collected from two groups: eyes from control subjects (n = 4) with idiopathic macular holes (IMH) and eyes from test subjects (n = 12) with different stages of CAPN5-NIV. Samples were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Protein expression changes were evaluated by principal component analysis, 1-way ANOVA (significant p-value < 0.05), hierarchical clustering, gene ontology, and pathway representation. There were 216 differentially-expressed proteins (between CAPN5-NIV and control vitreous), including those unique to and abundant in each clinical stage. Gene ontology analysis revealed decreased synaptic signaling proteins in CAPN5-NIV vitreous compared to controls. Pathway analysis revealed that inflammatory mediators of the acute phase response and the complement cascade were highly-represented. The CAPN5-NIV vitreous proteome displayed characteristic enrichment of proteins and pathways previously-associated with non-infectious posterior uveitis, rhegmatogenous retinal detachment (RRD), age-related macular degeneration (AMD), proliferative diabetic retinopathy (PDR), and proliferative vitreoretinopathy (PVR). This study expands our knowledge of affected molecular pathways in CAPN5-NIV using unbiased, shotgun proteomic analysis rather than targeted detection platforms. The high-levels and representation of acute phase response proteins suggests a functional role for the innate immune system in CAPN5-NIV pathogenesis.
Collapse
|
16
|
Cheng Y, Chen D. Fruit fly research in China. J Genet Genomics 2018; 45:583-592. [PMID: 30455037 DOI: 10.1016/j.jgg.2018.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/21/2018] [Accepted: 09/29/2018] [Indexed: 11/19/2022]
Abstract
Served as a model organism over a century, fruit fly has significantly pushed forward the development of global scientific research, including in China. The high similarity in genomic features between fruit fly and human enables this tiny insect to benefit the biomedical studies of human diseases. In the past decades, Chinese biologists have used fruit fly to make numerous achievements on understanding the fundamental questions in many diverse areas of biology. Here, we review some of the recent fruit fly studies in China, and mainly focus on those studies in the fields of stem cell biology, cancer therapy and regeneration medicine, neurological disorders and epigenetics.
Collapse
Affiliation(s)
- Ying Cheng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dahua Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
17
|
Xing G, Li M, Sun Y, Rui M, Zhuang Y, Lv H, Han J, Jia Z, Xie W. Neurexin-Neuroligin 1 regulates synaptic morphology and functions via the WAVE regulatory complex in Drosophila neuromuscular junction. eLife 2018. [PMID: 29537369 PMCID: PMC5873926 DOI: 10.7554/elife.30457] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuroligins are postsynaptic adhesion molecules that are essential for postsynaptic specialization and synaptic function. But the underlying molecular mechanisms of neuroligin functions remain unclear. We found that Drosophila Neuroligin 1 (DNlg1) regulates synaptic structure and function through WAVE regulatory complex (WRC)-mediated postsynaptic actin reorganization. The disruption of DNlg1, DNlg2, or their presynaptic partner neurexin (DNrx) led to a dramatic decrease in the amount of F-actin. Further study showed that DNlg1, but not DNlg2 or DNlg3, directly interacts with the WRC via its C-terminal interacting receptor sequence. That interaction is required to recruit WRC to the postsynaptic membrane to promote F-actin assembly. Furthermore, the interaction between DNlg1 and the WRC is essential for DNlg1 to rescue the morphological and electrophysiological defects in dnlg1 mutants. Our results reveal a novel mechanism by which the DNrx-DNlg1 trans-synaptic interaction coordinates structural and functional properties at the neuromuscular junction.
Collapse
Affiliation(s)
- Guanglin Xing
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Moyi Li
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yichen Sun
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Menglong Rui
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Yan Zhuang
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Huihui Lv
- The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Junhai Han
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhengping Jia
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,Neurosciences and Mental Health Program, The Hospital for Sick Children, University of Toronto, Ontario, Canada
| | - Wei Xie
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
18
|
A Single Residue Mutation in the Gα q Subunit of the G Protein Complex Causes Blindness in Drosophila. G3-GENES GENOMES GENETICS 2018; 8:363-371. [PMID: 29158337 PMCID: PMC5765363 DOI: 10.1534/g3.117.300340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Heterotrimeric G proteins play central roles in many signaling pathways, including the phototransduction cascade in animals. However, the degree of involvement of the G protein subunit Gαq is not clear since animals with previously reported strong loss-of-function mutations remain responsive to light stimuli. We recovered a new allele of Gαq in Drosophila that abolishes light response in a conventional electroretinogram assay, and reduces sensitivity in whole-cell recordings of dissociated cells by at least five orders of magnitude. In addition, mutant eyes demonstrate a rapid rate of degeneration in the presence of light. Our new allele is likely the strongest hypomorph described to date. Interestingly, the mutant protein is produced in the eyes but carries a single amino acid change of a conserved hydrophobic residue that has been assigned to the interface of interaction between Gαq and its downstream effector, PLC. Our study has thus uncovered possibly the first point mutation that specifically affects this interaction in vivo.
Collapse
|
19
|
Wu J, Tao N, Tian Y, Xing G, Lv H, Han J, Lin C, Xie W. Proteolytic maturation of Drosophila Neuroligin 3 by tumor necrosis factor α-converting enzyme in the nervous system. Biochim Biophys Acta Gen Subj 2017; 1862:440-450. [PMID: 29107812 DOI: 10.1016/j.bbagen.2017.10.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/18/2017] [Accepted: 10/27/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND The functions of autism-associated Neuroligins (Nlgs) are modulated by their post-translational modifications, such as proteolytic cleavage. A previous study has shown that there are different endogenous forms of DNlg3 in Drosophila, indicating it may undergo proteolytic processing. However, the molecular mechanism underlying DNlg3 proteolytic processing is unknown. Here, we report a novel proteolytic mechanism that is essential for DNlg3 maturation and function in the nervous system. METHODS Molecular cloning, cell culture, immunohistochemistry, western blotting and genetic studies were employed to map the DNlg3 cleavage region, identify the protease and characterize the cleavage manner. Behavior analysis, immunohistochemistry and genetic manipulations were employed to study the functions of different DNlg3 forms in the nervous system and neuromuscular junction (NMJs). RESULTS Tumor necrosis factor α-converting enzyme (TACE) cleaved DNlg3 exclusively at its extracellular acetylcholinesterase-like domain to generate the N-terminal fragment and the short membrane-anchored fragment (sDNlg3). DNlg3 was constitutively processed in an activity-independent manner. Interestingly, DNlg3 was cleaved intracellularly in the Golgi apparatus before it arrived at the cell surface, a unique cleavage mechanism that is distinct from 'conventional' ectodomain shedding of membrane proteins, including rodent Nlg1. Genetic studies showed that sDNlg3 was essential for maintaining proper locomotor activity in Drosophila. CONCLUSIONS Our results revealed a unique cleavage mechanism of DNlg3 and a neuron-specific role for DNlg3 maturation which is important in locomotor activity. GENERAL SIGNIFICANCE Our study provides a new insight into a cleavage mechanism of Nlgs maturation in the nervous system.
Collapse
Affiliation(s)
- Jun Wu
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China
| | - Nana Tao
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China
| | - Yao Tian
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Guanglin Xing
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Huihui Lv
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Junhai Han
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China; The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Chengqi Lin
- The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China
| | - Wei Xie
- Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, China; The Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing 210096, China.
| |
Collapse
|
20
|
Rui M, Qian J, Liu L, Cai Y, Lv H, Han J, Jia Z, Xie W. The neuronal protein Neurexin directly interacts with the Scribble-Pix complex to stimulate F-actin assembly for synaptic vesicle clustering. J Biol Chem 2017; 292:14334-14348. [PMID: 28710284 PMCID: PMC5582829 DOI: 10.1074/jbc.m117.794040] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/29/2017] [Indexed: 01/17/2023] Open
Abstract
Synaptic vesicles (SVs) form distinct pools at synaptic terminals, and this well-regulated separation is necessary for normal neurotransmission. However, how the SV cluster, in particular synaptic compartments, maintains normal neurotransmitter release remains a mystery. The presynaptic protein Neurexin (NRX) plays a significant role in synaptic architecture and function, and some evidence suggests that NRX is associated with neurological disorders, including autism spectrum disorders. However, the role of NRX in SV clustering is unclear. Here, using the neuromuscular junction at the 2-3 instar stages of Drosophila larvae as a model and biochemical imaging and electrophysiology techniques, we demonstrate that Drosophila NRX (DNRX) plays critical roles in regulating synaptic terminal clustering and release of SVs. We found that DNRX controls the terminal clustering and release of SVs by stimulating presynaptic F-actin. Furthermore, our results indicate that DNRX functions through the scaffold protein Scribble and the GEF protein DPix to activate the small GTPase Ras-related C3 Botulinum toxin substrate 1 (Rac1). We observed a direct interaction between the C-terminal PDZ-binding motif of DNRX and the PDZ domains of Scribble and that Scribble bridges DNRX to DPix, forming a DNRX-Scribble-DPix complex that activates Rac1 and subsequently stimulates presynaptic F-actin assembly and SV clustering. Taken together, our work provides important insights into the function of DNRX in regulating SV clustering, which could help inform further research into pathological neurexin-mediated mechanisms in neurological disorders such as autism.
Collapse
Affiliation(s)
- Menglong Rui
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Jinjun Qian
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Lijuan Liu
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Yihan Cai
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Huihui Lv
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Junhai Han
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| | - Zhengping Jia
- the Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada, and.,the Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Wei Xie
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China, .,the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| |
Collapse
|
21
|
Wang X, Mu Y, Sun M, Han J. Bidirectional regulation of fragile X mental retardation protein phosphorylation controls rhodopsin homoeostasis. J Mol Cell Biol 2017; 9:104-116. [PMID: 27702760 DOI: 10.1093/jmcb/mjw041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 09/29/2016] [Indexed: 01/03/2023] Open
Abstract
Homoeostatic regulation of the light sensor, rhodopsin, is critical for the maintenance of light sensitivity and survival of photoreceptors. The major fly rhodopsin, Rh1, undergoes light-induced endocytosis and degradation, but its protein and mRNA levels remain constant during light/dark cycles. It is not clear how translation of Rh1 is regulated. Here, we show that adult photoreceptors maintain a constant, abundant quantity of ninaE mRNA, which encodes Rh1. We demonstrate that the Fmr1 protein associates with ninaE mRNA and represses its translation. Further, light exposure triggers a calcium-dependent dephosphorylation of Fmr1, which relieves suppression of Rh1 translation. We demonstrate that Mts, the catalytic subunit of protein phosphatase 2A (PP2A), mediates light-induced Fmr1 dephosphorylation in a regulatory B subunit of PP2A (CKa)-dependent manner. Finally, we show that blocking light-induced Rh1 translation results in reduced light sensitivity. Our results reveal the molecular mechanism of Rh1 homoeostasis and physiological consequence of Rh1 dysregulation.
Collapse
Affiliation(s)
- Xiao Wang
- Institute of Life Sciences, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Yawen Mu
- Institute of Life Sciences, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Mengshi Sun
- Institute of Life Sciences, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Junhai Han
- Institute of Life Sciences, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| |
Collapse
|
22
|
Durand N, Chertemps T, Bozzolan F, Maïbèche M. Expression and modulation of neuroligin and neurexin in the olfactory organ of the cotton leaf worm Spodoptera littoralis. INSECT SCIENCE 2017; 24:210-221. [PMID: 26749290 DOI: 10.1111/1744-7917.12312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/27/2015] [Indexed: 06/05/2023]
Abstract
Carboxylesterases are enzymes widely distributed within living organisms. In insects, they have been mainly involved in dietary metabolism and detoxification function. Interestingly, several members of this family called carboxylesterase-like adhesion molecules (CLAMs) have lost their catalytic properties and are mainly involved in neuro/developmental functions. CLAMs include gliotactins, neurotactins, glutactins, and neuroligins. The latter have for binding partner the neurexin. In insects, the function of these proteins has been mainly studied in Drosophila central nervous system or neuromuscular junction. Some studies suggested a role of neuroligins and neurexin in sensory processing but CLAM expression within sensory systems has not been investigated. Here, we reported the identification of 5 putative CLAMs expressed in the olfactory system of the model pest insect Spodoptera littoralis. One neuroligin, Slnlg4-yll and its putative binding partner neurexin SlnrxI were the most expressed in the antennae and were surprisingly associated with olfactory sensilla. In addition, both transcripts were upregulated in male antennae after mating, known to modulate the sensitivity of the peripheral olfactory system in S. littoralis, suggesting that these molecules could be involved in sensory plasticity.
Collapse
Affiliation(s)
- Nicolas Durand
- Sorbonne Universités UPMC - Univ Paris 06, Institut d'Ecologie et des Sciences de 'Environnement de Paris, INRA, CNRS, IRD, UPEC, Département d'Ecologie Sensorielle, F-75252, Paris, France
| | - Thomas Chertemps
- Sorbonne Universités UPMC - Univ Paris 06, Institut d'Ecologie et des Sciences de 'Environnement de Paris, INRA, CNRS, IRD, UPEC, Département d'Ecologie Sensorielle, F-75252, Paris, France
| | - Françoise Bozzolan
- Sorbonne Universités UPMC - Univ Paris 06, Institut d'Ecologie et des Sciences de 'Environnement de Paris, INRA, CNRS, IRD, UPEC, Département d'Ecologie Sensorielle, F-75252, Paris, France
| | - Martine Maïbèche
- Sorbonne Universités UPMC - Univ Paris 06, Institut d'Ecologie et des Sciences de 'Environnement de Paris, INRA, CNRS, IRD, UPEC, Département d'Ecologie Sensorielle, F-75252, Paris, France
| |
Collapse
|
23
|
Liu L, Tian Y, Zhang XY, Zhang X, Li T, Xie W, Han J. Neurexin Restricts Axonal Branching in Columns by Promoting Ephrin Clustering. Dev Cell 2017; 41:94-106.e4. [PMID: 28366281 DOI: 10.1016/j.devcel.2017.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 02/03/2017] [Accepted: 03/03/2017] [Indexed: 11/18/2022]
Abstract
Columnar restriction of neurites is critical for forming nonoverlapping receptive fields and preserving spatial sensory information from the periphery in both vertebrate and invertebrate nervous systems, but the underlying molecular mechanisms remain largely unknown. Here, we demonstrate that Drosophila homolog of α-neurexin (DNrx) plays an essential role in columnar restriction during L4 axon branching. Depletion of DNrx from L4 neurons resulted in misprojection of L4 axonal branches into neighboring columns due to impaired ephrin clustering. The proper ephrin clustering requires its interaction with the intracellular region of DNrx. Furthermore, we find that Drosophila neuroligin 4 (DNlg4) in Tm2 neurons binds to DNrx and initiates DNrx clustering in L4 neurons, which subsequently induces ephrin clustering. Our study demonstrates that DNrx promotes ephrin clustering and reveals that ephrin/Eph signaling from adjacent L4 neurons restricts axonal branches of L4 neurons in columns.
Collapse
Affiliation(s)
- Lina Liu
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Yao Tian
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Xiao-Yan Zhang
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Xinwang Zhang
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Tao Li
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Wei Xie
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Junhai Han
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
24
|
Xiao X, Chen C, Yu TM, Ou J, Rui M, Zhai Y, He Y, Xue L, Ho MS. Molecular Chaperone Calnexin Regulates the Function of Drosophila Sodium Channel Paralytic. Front Mol Neurosci 2017; 10:57. [PMID: 28326013 PMCID: PMC5339336 DOI: 10.3389/fnmol.2017.00057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
Neuronal activity mediated by voltage-gated channels provides the basis for higher-order behavioral tasks that orchestrate life. Chaperone-mediated regulation, one of the major means to control protein quality and function, is an essential route for controlling channel activity. Here we present evidence that Drosophila ER chaperone Calnexin colocalizes and interacts with the α subunit of sodium channel Paralytic. Co-immunoprecipitation analysis indicates that Calnexin interacts with Paralytic protein variants that contain glycosylation sites Asn313, 325, 343, 1463, and 1482. Downregulation of Calnexin expression results in a decrease in Paralytic protein levels, whereas overexpression of the Calnexin C-terminal calcium-binding domain triggers an increase reversely. Genetic analysis using adult climbing, seizure-induced paralysis, and neuromuscular junction indicates that lack of Calnexin expression enhances Paralytic-mediated locomotor deficits, suppresses Paralytic-mediated ghost bouton formation, and regulates minature excitatory junction potentials (mEJP) frequency and latency time. Taken together, our findings demonstrate a need for chaperone-mediated regulation on channel activity during locomotor control, providing the molecular basis for channlopathies such as epilepsy.
Collapse
Affiliation(s)
- Xi Xiao
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Changyan Chen
- Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Institute of Intervention Vessel, Shanghai 10th People's Hospital, Tongji University Shanghai, China
| | - Tian-Ming Yu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Jiayao Ou
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Menglong Rui
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University Nanjing, China
| | - Yuanfen Zhai
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Yijing He
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Lei Xue
- Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Institute of Intervention Vessel, Shanghai 10th People's Hospital, Tongji University Shanghai, China
| | - Margaret S Ho
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of MedicineShanghai, China; Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| |
Collapse
|
25
|
Wang D, Pan J, Song G, Gao N, Zheng Y, Zhang Q, Li A. Abundance and Significance of Neuroligin-1 and Neurexin II in the Enteric Nervous System of Embryonic Rats. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1209360. [PMID: 28194405 PMCID: PMC5286486 DOI: 10.1155/2017/1209360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/19/2016] [Accepted: 01/04/2017] [Indexed: 12/30/2022]
Abstract
Aim. To investigate the abundance of neuroligin-1 and neurexin II in the enteric nervous system (ENS) of rats on different embryonic days and to explore their potential significance. Methods. The full-thickness colon specimens proximal to the ileocecal junction of rats on embryonic days 16, 18, and 20 and of newborns within 24 hours (E16, E18, E20, and Ep0) were studied, respectively. qRT-PCR was applied for detecting the expressions of neuroligin-1 and neurexin II on mRNA, and western blotting was employed for detecting their further expressions on the whole tissue. Finally, the histological appearance of neuroligin-1 and neurexin IIα was elucidated using immunohistochemical staining. Results. qRT-PCR showed that the neuroligin-1 and neurexin II mRNA expressions of groups E16, E18, E20, and Ep0 increased gradually with the growth of embryonic rats (P < 0.05). Western blotting confirmed the increasing tendency. In immunohistochemical staining, proteins neuroligin-1 and neurexin IIα positive cells concentrated mostly in the myenteric nerve plexus of the colon and their expressions depend on the embryonic time. Conclusion. Neuroligin-1 and neurexin II were both expressed in the ENS and have temporal correlation with the development of ENS, during which neuronal intestinal malformations (NIM) may occur due to their disruptions and consequent abnormal ENS development.
Collapse
Affiliation(s)
- Dongming Wang
- Department of Pediatric Surgery, Qilu Hospital, Shandong University, Shandong, China
| | - Jingnian Pan
- Internal Medicine, Qingzhou Clinical School, Weifang Medical College, Weifang, China
| | - Guoxin Song
- Department of Pediatric Surgery, Weihai Municipal Hospital, Shandong, China
| | - Ni Gao
- Department of Pediatric Surgery, Qilu Hospital, Shandong University, Shandong, China
| | - Yi Zheng
- Department of Pediatric Surgery, Qilu Hospital, Shandong University, Shandong, China
| | - Qiangye Zhang
- Department of Pediatric Surgery, Qilu Hospital, Shandong University, Shandong, China
| | - Aiwu Li
- Department of Pediatric Surgery, Qilu Hospital, Shandong University, Shandong, China
| |
Collapse
|
26
|
Neurexin regulates nighttime sleep by modulating synaptic transmission. Sci Rep 2016; 6:38246. [PMID: 27905548 PMCID: PMC5131284 DOI: 10.1038/srep38246] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/07/2016] [Indexed: 11/09/2022] Open
Abstract
Neurexins are cell adhesion molecules involved in synaptic formation and synaptic transmission. Mutations in neurexin genes are linked to autism spectrum disorders (ASDs), which are frequently associated with sleep problems. However, the role of neurexin-mediated synaptic transmission in sleep regulation is unclear. Here, we show that lack of the Drosophila α-neurexin homolog significantly reduces the quantity and quality of nighttime sleep and impairs sleep homeostasis. We report that neurexin expression in Drosophila mushroom body (MB) αβ neurons is essential for nighttime sleep. We demonstrate that reduced nighttime sleep in neurexin mutants is due to impaired αβ neuronal output, and show that neurexin functionally couples calcium channels (Cac) to regulate synaptic transmission. Finally, we determine that αβ surface (αβs) neurons release both acetylcholine and short neuropeptide F (sNPF), whereas αβ core (αβc) neurons release sNPF to promote nighttime sleep. Our findings reveal that neurexin regulates nighttime sleep by mediating the synaptic transmission of αβ neurons. This study elucidates the role of synaptic transmission in sleep regulation, and might offer insights into the mechanism of sleep disturbances in patients with autism disorders.
Collapse
|
27
|
Banerjee S, Venkatesan A, Bhat MA. Neurexin, Neuroligin and Wishful Thinking coordinate synaptic cytoarchitecture and growth at neuromuscular junctions. Mol Cell Neurosci 2016; 78:9-24. [PMID: 27838296 DOI: 10.1016/j.mcn.2016.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/19/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022] Open
Abstract
Trans-synaptic interactions involving Neurexins and Neuroligins are thought to promote adhesive interactions for precise alignment of the pre- and postsynaptic compartments and organize synaptic macromolecular complexes across species. In Drosophila, while Neurexin (Dnrx) and Neuroligins (Dnlg) are emerging as central organizing molecules at synapses, very little is known of the spectrum of proteins that might be recruited to the Dnrx/Dnlg trans-synaptic interface for organization and growth of the synapses. Using full length and truncated forms of Dnrx and Dnlg1 together with cell biological analyses and genetic interactions, we report novel functions of Dnrx and Dnlg1 in clustering of pre- and postsynaptic proteins, coordination of synaptic growth and ultrastructural organization. We show that Dnrx and Dnlg1 extracellular and intracellular regions are required for proper synaptic growth and localization of Dnlg1 and Dnrx, respectively. dnrx and dnlg1 single and double mutants display altered subcellular distribution of Discs large (Dlg), which is the homolog of mammalian post-synaptic density protein, PSD95. dnrx and dnlg1 mutants also display ultrastructural defects ranging from abnormal active zones, misformed pre- and post-synaptic areas with underdeveloped subsynaptic reticulum. Interestingly, dnrx and dnlg1 mutants have reduced levels of the Bone Morphogenetic Protein (BMP) receptor Wishful thinking (Wit), and Dnrx and Dnlg1 are required for proper localization and stability of Wit. In addition, the synaptic overgrowth phenotype resulting from the overexpression of Dnrx fails to manifest in wit mutants. Phenotypic analyses of dnrx/wit and dnlg1/wit mutants indicate that Dnrx/Dnlg1/Wit coordinate synaptic growth and architecture at the NMJ. Our findings also demonstrate that loss of Dnrx and Dnlg1 leads to decreased levels of the BMP co-receptor, Thickveins and the downstream effector phosphorylated Mad at the Neuromuscular Junction (NMJ) synapses indicating that Dnrx/Dnlg1 regulate components of the BMP signaling pathway. Together our findings reveal that Dnrx/Dnlg are at the core of a highly orchestrated process that combines adhesive and signaling mechanisms to ensure proper synaptic organization and growth during NMJ development.
Collapse
Affiliation(s)
- Swati Banerjee
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA.
| | - Anandakrishnan Venkatesan
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| |
Collapse
|
28
|
Temporal and spatial expression of Drosophila Neurexin during the life cycle visualized using a DNRX-Gal4/UAS-reporter. SCIENCE CHINA-LIFE SCIENCES 2015; 59:68-77. [PMID: 26501376 DOI: 10.1007/s11427-015-4946-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/22/2015] [Indexed: 10/22/2022]
Abstract
Drosophila neurexin (DNRX) plays a critical role in proper architecture development and synaptic function in vivo. However, the temporal and spatial expression pattern of DNRX still remains unclear. For this study, we generated a novel Drosophila transgenic strain termed the DNRX-Gal4 transgenic line, with characteristic features in agreement with the endogenous DNRX expression pattern. DNRX expression was examined by driving the expression of a GFP reporter (nuclear-localized and membrane- localized GFP) using the DNRX-Gal4 promoter. We found that DNRX was expressed preferentially in central and motor neurons in embryos, larvae and adults, but not in glial cells. DNRX was expressed in pre- and post-synaptic areas in third instar larvae neuromuscular junctions (NMJs). Reporter expression was also observed in the salivary glands, guts, wings and legs of adult flies. In the adult brain, reporter expression was observed throughout several brain regions, including the mushroom body (MBs), antennal lobe (AL) and optic lobe neurons, which is consistent with endogenous DNRX expression via antibody staining. Interestingly, DNRX was also expressed in clock neurons. Meanwhile, we found that DNRX expression in the MBs was required for olfactory learning and memory.
Collapse
|
29
|
Wan D, Zhang ZC, Zhang X, Li Q, Han J. X chromosome-linked intellectual disability protein PQBP1 associates with and regulates the translation of specific mRNAs. Hum Mol Genet 2015; 24:4599-614. [DOI: 10.1093/hmg/ddv191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/19/2015] [Indexed: 01/08/2023] Open
|
30
|
Li T, Tian Y, Li Q, Chen H, Lv H, Xie W, Han J. The Neurexin/N-Ethylmaleimide-sensitive Factor (NSF) Interaction Regulates Short Term Synaptic Depression. J Biol Chem 2015; 290:17656-17667. [PMID: 25953899 DOI: 10.1074/jbc.m115.644583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Indexed: 11/06/2022] Open
Abstract
Although Neurexins, which are cell adhesion molecules localized predominantly to the presynaptic terminals, are known to regulate synapse formation and synaptic transmission, their roles in the regulation of synaptic vesicle release during repetitive nerve stimulation are unknown. Here, we show that nrx mutant synapses exhibit rapid short term synaptic depression upon tetanic nerve stimulation. Moreover, we demonstrate that the intracellular region of NRX is essential for synaptic vesicle release upon tetanic nerve stimulation. Using a yeast two-hybrid screen, we find that the intracellular region of NRX interacts with N-ethylmaleimide-sensitive factor (NSF), an enzyme that mediates soluble NSF attachment protein receptor (SNARE) complex disassembly and plays an important role in synaptic vesicle release. We further map the binding sites of each molecule and demonstrate that the NRX/NSF interaction is critical for both the distribution of NSF at the presynaptic terminals and SNARE complex disassembly. Our results reveal a previously unknown role of NRX in the regulation of short term synaptic depression upon tetanic nerve stimulation and provide new mechanistic insights into the role of NRX in synaptic vesicle release.
Collapse
Affiliation(s)
- Tao Li
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096
| | - Yao Tian
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096
| | - Qian Li
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096
| | - Huiying Chen
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096
| | - Huihui Lv
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096
| | - Wei Xie
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS 226001, China
| | - Junhai Han
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS 226001, China.
| |
Collapse
|
31
|
Hu W, Wang T, Wang X, Han J. Ih channels control feedback regulation from amacrine cells to photoreceptors. PLoS Biol 2015; 13:e1002115. [PMID: 25831426 PMCID: PMC4382183 DOI: 10.1371/journal.pbio.1002115] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/23/2015] [Indexed: 12/02/2022] Open
Abstract
In both vertebrates and invertebrates, photoreceptors’ output is regulated by feedback signals from interneurons that contribute to several important visual functions. Although synaptic feedback regulation of photoreceptors is known to occur in Drosophila, many questions about the underlying molecular mechanisms and physiological implementation remain unclear. Here, we systematically investigated these questions using a broad range of experimental methods. We isolated two Ih mutant fly lines that exhibit rhythmic photoreceptor depolarization without light stimulation. We discovered that Ih channels regulate glutamate release from amacrine cells by modulating calcium channel activity. Moreover, we showed that the eye-enriched kainate receptor (EKAR) is expressed in photoreceptors and receives the glutamate signal released from amacrine cells. Finally, we presented evidence that amacrine cell feedback regulation helps maintain light sensitivity in ambient light. Our findings suggest plausible molecular underpinnings and physiological effects of feedback regulation from amacrine cells to photoreceptors. These results provide new mechanistic insight into how synaptic feedback regulation can participate in network processing by modulating neural information transfer and circuit excitability. A systematic study of the Drosophila visual system clarifies the molecular mechanisms and physiological effects of feedback regulation of photoreceptors by amacrine cells, essential for maintaining light sensitivity. Feedback regulation is a common feature of neural circuits during the process of acquiring information. Therefore, it is important to understand how this phenomenon occurs. Using the primary visual system of the fruit fly Drosophila melanogaster as a model, we systematically investigated the molecular mechanisms and the physiological implementation of feedback regulation from amacrine cells (second order neurons that are present in the lamina) to photoreceptors. We isolated two fly lines with mutations in the gene that encodes for the ion channel known as Ih, whose photoreceptors exhibited rhythmic depolarizations in the absence of light stimulation. We demonstrated that Ih channels function in amacrine cells to regulate the release of the neurotransmitter glutamate by modulating the activity of the voltage-gated calcium channel, Cac. We further found that the glutamate signal released by amacrine cells is sensed and transduced by glutamate receptors expressed by the photoreceptors. Finally, we showed that this feedback regulation is critical for maintaining light sensitivity in the presence of ambient light. Our results suggest that regulation of synaptic feedback in a neuronal network modulates information transfer and circuit excitability.
Collapse
Affiliation(s)
- Wen Hu
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Tingting Wang
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiao Wang
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Junhai Han
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- * E-mail:
| |
Collapse
|
32
|
Abstract
Sleep is an essential and evolutionarily conserved behavior that is closely related to synaptic function. However, whether neuroligins (Nlgs), which are cell adhesion molecules involved in synapse formation and synaptic transmission, are involved in sleep is not clear. Here, we show that Drosophila Nlg4 (DNlg4) is highly expressed in large ventral lateral clock neurons (l-LNvs) and that l-LNv-derived DNlg4 is essential for sleep regulation. GABA transmission is impaired in mutant l-LNv, and sleep defects in dnlg4 mutant flies can be rescued by genetic manipulation of GABA transmission. Furthermore, dnlg4 mutant flies exhibit a severe reduction in GABAA receptor RDL clustering, and DNlg4 associates with RDLs in vivo. These results demonstrate that DNlg4 regulates sleep through modulating GABA transmission in l-LNvs, which provides the first known link between a synaptic adhesion molecule and sleep in Drosophila.
Collapse
|
33
|
Xiong B, Bellen HJ. Rhodopsin homeostasis and retinal degeneration: lessons from the fly. Trends Neurosci 2013; 36:652-60. [PMID: 24012059 DOI: 10.1016/j.tins.2013.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 08/02/2013] [Accepted: 08/12/2013] [Indexed: 11/16/2022]
Abstract
Rhodopsins (Rh) are G protein-coupled receptors that function as light-sensors in photoreceptors. In humans, Rh mutations cause retinitis pigmentosa (RP), a degenerative disease that ultimately results in blindness. Studies in Drosophila have provided many insights into basic Rh biology and have identified pathways that lead to retinal degeneration. It has been shown that, because Rh is very abundant in photoreceptors, its accumulation in numerous organelles induces severe stress and results in degeneration of these cells. Moreover, genetic lesions that affect proper activation of membrane-bound Rh lead to disruption in Ca(2+) homeostasis which also causes photoreceptor degeneration. We review here the molecular signals involved in Rh homeostasis and the mechanisms underlying retinal degeneration in flies, and discuss possible links to human diseases.
Collapse
Affiliation(s)
- Bo Xiong
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|