1
|
Sugawara T, Matsu-Ura H, Inagaki R, Kawamura T, Tanaka M, Hara Y, Saito K, Fukaya M, Moriguchi S, Sakagami H. EFA6A, a Guanine Nucleotide Exchange Factor for Arf6, Regulates Developmental Stage-Dependent Spine Morphogenesis, Synaptic Plasticity, and Long-Term Memory in the Hippocampus. Mol Neurobiol 2025:10.1007/s12035-025-05009-x. [PMID: 40335791 DOI: 10.1007/s12035-025-05009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
EFA6A is a guanine nucleotide exchange factor for ADP ribosylation factor 6 (Arf6), a small GTPase involved in membrane trafficking and actin cytoskeleton remodeling. While EFA6A-Arf6 signaling has been shown to regulate dendritic spine formation and maintenance in cultured neurons, its role in higher brain functions remains unclear in vivo. Here, we generated mice lacking two EFA6A splicing isoforms, EFA6A and EFA6As, to examine their role in regulating spine morphology and hippocampus-dependent learning and memory. The loss of EFA6A and EFA6As caused reduced dendritic spine density in developing CA1 pyramidal neurons, whereas dendritic spines aberrantly increased in adults. Furthermore, the mutant mice also showed impaired maintenance of long-term potentiation (LTP) at Schaffer collateral-CA1 synapses in the hippocampus and memory retention in the passive avoidance test. These findings provide the first in vivo evidence that the EFA6A isoforms, EFA6A and EFA6As, collectively regulate spine formation bidirectionally in a developmental stage-dependent manner, which is likely to underlie hippocampal synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Takeyuki Sugawara
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hikaru Matsu-Ura
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Ryo Inagaki
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Taichi Kawamura
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Manabu Tanaka
- Bio-Imaging Center, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshinobu Hara
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Koji Saito
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
- Department of Anatomical Science, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Shigeki Moriguchi
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
2
|
Donati M, Goutas D, Pissaloux D, Olivares S, Kervarrec T, Nosek D, Goto K, Lemahieu J, Loontiens S, Van der Meulen J, Mansour B, Perrone G, Macagno N, Gerami P, Kazakov DV, De la Fouchardiere A. Clinical, Morphologic, and Genomic Findings in Spitz Tumors With RET Fusion: A Series of 31 Cases. Mod Pathol 2025; 38:100740. [PMID: 39986469 DOI: 10.1016/j.modpat.2025.100740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
RET-fused Spitz neoplasms represent a rare and poorly characterized category of Spitz tumors. Here we describe the clinical, histologic, and molecular findings of 31 Spitz neoplasms with RET fusion diagnosed as Spitz nevus (n = 16), atypical Spitz tumors (n = 13), and Spitz melanoma (n = 2). The lesions mainly occurred in children and young adults of both sexes with a predilection for the extremities. Microscopically, they were mainly symmetrical compound melanocytic neoplasms with a dome-shaped/slightly raised silhouette predominantly composed of epithelioid, spindled, and/or smaller nevoid melanocytes arranged in confluent nests. Dyscohesive melanocytes within the nests in the upper part of the lesions, prominent Kamino bodies, giant multinucleated melanocytes, variable pigmentation, and increased vascularity with vascular ectasia were frequent features. RNA sequencing detected 9 different 5' (N-terminus) fusion partners, including KIF5B (n = 8), LMNA (n = 7), CCDC6 (n = 6), OPTN (n = 3), MYO5A (n = 2), and NCOA4, ERC1, MYH9, AGAP3 (n = 1). Of these, OPTN::RET and AGAP3::RET represent novel fusions, and 3 further 5' fusion partners, namely NCOA4, ERC1, and MYH9, have never been reported in Spitz tumors. Although as a whole group, the tumors showed a heterogeneous histopathologic presentation, correlation of the morphologic features and the 5' fusion partners demonstrated certain associations. Nevoid melanocytes were exclusively encountered in cases with KIF5B fusion partner. Neuroid-like appearances with intersecting fascicles of spindled cells typified both MYO5A-fused cases. Epithelioid melanocyte population dominated cases with LMNA and CCDC6 fusion partners. Transepidermal elimination/floating intraepidermal nests of pigmented spindled and epithelioid melanocytes were observed in the OPTN subgroup. The remaining cases with less frequent 5' fusion partners manifested in general more atypical histopathologic features, including nuclear pleomorphism, high mitotic count, atypical mitoses, and sheet-like growth pattern. Melanoma fluorescence in situ hybridization probe kit targeting RREB1, MYC, CDKN2A, and CCND1, was negative for copy number variation in 4 cases tested, including 2 cases with complete p16 nuclear loss on immunohistochemistry. Array comparative genomic hybridization was performed in 3 lesions and detected numerous segmental chromosomal imbalances in 2 of them that were diagnosed as Spitz melanoma. DNA and RNA sequencing detected several further genomic alterations, including POU2F3 overexpression in 3 highly pigmented lesions. Further studies are needed to confirm possible correlations between the microscopic features and a particular fusion partner (or additional genetic events) in RET-fused Spitz neoplasms.
Collapse
Affiliation(s)
- Michele Donati
- Department of Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Department of Pathology, Università Campus Bio-Medico di Roma, Roma, Italy.
| | - Dimitrios Goutas
- First Department of Pathology, The National and Kapodistrian University of Athens, Greece
| | - Daniel Pissaloux
- Department of Biopathology, Centre Leon Berard, Lyon, France; Department of Research, University of Lyon, Universite Claude Bernard Lyon 1, Cancer Research Centre of Lyon, Lyon, France
| | - Shantel Olivares
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Thibault Kervarrec
- Department of Pathology, University Hospital of Tours, Tours, France; 'Biologie des infections à polyomavirus' Team, UMR1282 INRAE, University of Tours, Tours, France
| | - Daniel Nosek
- Department of Pathology, Umeå University, Umeå, Sweden
| | - Keisuke Goto
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Tokyo, Japan; Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; Department of Dermato-Oncology/Dermatology, National Hospital Organization Kagoshima, Medical Center, Kagoshima, Japan
| | | | - Siebe Loontiens
- Molecular Diagnostics, Ghent University Hospital, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Biomolecular Medicine, University of Ghent, Ghent, Belgium
| | - Joni Van der Meulen
- Molecular Diagnostics, Ghent University Hospital, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Biomolecular Medicine, University of Ghent, Ghent, Belgium
| | - Boulos Mansour
- Department of Pathology, Ospedale Israelitico, Rome, Italy
| | - Giuseppe Perrone
- Department of Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Department of Pathology, Università Campus Bio-Medico di Roma, Roma, Italy
| | - Nicolas Macagno
- Marseille Medical Genetics, Institut MarMaRa, Aix-Marseille University, INSERM, Marseille, France
| | - Pedram Gerami
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Dmitry V Kazakov
- IDP Institut für Dermatohistopathologie, Pathologie Institut Enge, Zürich, Switzerland
| | - Arnaud De la Fouchardiere
- Department of Biopathology, Centre Leon Berard, Lyon, France; Department of Research, University of Lyon, Universite Claude Bernard Lyon 1, Cancer Research Centre of Lyon, Lyon, France
| |
Collapse
|
3
|
Xiao J, Feng C, Zhu T, Zhang X, Chen X, Li Z, You J, Wang Q, Zhuansun D, Meng X, Wang J, Xiang L, Yu X, Zhou B, Tang W, Tou J, Wang Y, Yang H, Yu L, Liu Y, Jiang X, Ren H, Yu M, Chen Q, Yin Q, Liu X, Xu Z, Wu D, Yu D, Wu X, Yang J, Xiong B, Chen F, Hao X, Feng J. Rare and common genetic variants underlying the risk of Hirschsprung's disease. Hum Mol Genet 2025; 34:586-598. [PMID: 39817569 DOI: 10.1093/hmg/ddae205] [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: 08/14/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025] Open
Abstract
Hirschsprung's disease (HSCR) is a congenital enteric neuropathic disorder characterized by high heritability (>80%) and polygenic inheritance (>20 genes). The previous genome-wide association studies (GWAS) identified several common variants associated with HSCR and demonstrated increased predictive performance for HSCR risk in Europeans using a genetic risk score, there remains a notable gap in knowledge regarding Chinese populations. We conducted whole exome sequencing in a HSCR case cohort in Chinese. By using the common controls (505 controls from 1KG EAS and 10 588 controls from ChinaMAP), we conducted GWAS for the common variants in the exome and gene-based association for rare variants. We further validated the associated variants and genes in replicated samples and in vitro and vivo experiments. We identified one novel gene PLK5 by GWAS and suggested 45 novel putative genes based the gene-based test. By using genetic variant at RET and PLK5, we constructed a genetic risk score that could identify the individuals with very high genetic risk for HSCR. Compared with patients with zero or one risk allele from the three variants, the risk for HSCR was 36.61 times higher with six alleles. In addition, we delineated a HSCR risk gene landscape that encompasses 57 genes, which explains 88.5% and 54.5% of HSCR in Chinese and European, respectively. In summary, this study improved the understanding of genetic architecture of HSCR and provided a risk prediction approach for HSCR in the Chinese.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Chenzhao Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Tianqi Zhu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xuan Zhang
- Department of Pediatric Surgery, Pingshan District Maternal & Child Healthcare Hospital of Shenzhen, No. 6 Longtian South Road, Longtian Subdistrict, Pingshan District, Shenzhen, Guangdong 518122, China
| | - Xuyong Chen
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Zejian Li
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jingyi You
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Qiong Wang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Didi Zhuansun
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xinyao Meng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jing Wang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Lei Xiang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xiaosi Yu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Bingyan Zhou
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Road, Gulou District, Nanjing, Jiangsu 210008, China
| | - Jinfa Tou
- Department of General Surgery, Children's Hospital, Zhejiang University School of Medicine, No. 3333 Binsheng Road, Binjiang District, Hangzhou, Zhejiang 310003, China
| | - Yi Wang
- Department of General and Neonatal Surgery, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong District, Chongqing 400014, China
| | - Heying Yang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, No. 1 Renmin Road, Erqi District, Henan 450052, China
| | - Lei Yu
- Department of Neonatal Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Hong Kong Road, Jiang'an District, Wuhan, Hubei 430030, China
| | - Yuanmei Liu
- Department of Pediatric Surgery, The Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563000, China
| | - Xuewu Jiang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Shantou University Medical College, No. 69, Dongxia North Road, Jinping District, Shantou, Guangdong 515041, China
| | - Hongxia Ren
- Department of Neonatal Surgery, Children's Hospital of Shanxi, No. 13 Xinminbei Street, Xinhualing district, Taiyuan, Shanxi 030013, China
| | - Mei Yu
- Department of Pediatric Surgery, Guiyang Maternal and Child Health Hospital, No. 63 Ruijin South Road, Nanming district, Guiyang, Guizhou 550002, China
| | - Qi Chen
- Department of Pediatric Surgery, The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfuqian Street, Erqi District, Zhengzhou 450052, Henan, China
| | - Qiang Yin
- Department of General Surgery, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan 515041, China
| | - Xiang Liu
- Department of Pediatric Surgery, Anhui Provincial Children's Hospital, No. 39 Wangjiang East Road, Wuhu Road Subdistrict, Hefei, Anhui 230051, China
| | - Zhilin Xu
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, No. 199 Dazhi Street, Nangang district, Harbin, Heilongjiang 150001, China
| | - Dianming Wu
- Department of Pediatric Surgery, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Gulou District, Fuzhou, Fujian 350001, China
| | - Donghai Yu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xiaojuan Wu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jixin Yang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Bo Xiong
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Qiaokou District, Wuhan, Hubei 430030, China
| | - Feng Chen
- Department of Pediatric Surgery, Union Hospital, Fujian Medical University, No. 29, Xinquan Road, Gulou District, Fuzhou, Fujian 350001, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College Huazhong University of Science and Technology, No. 13 Hangkong Road, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jiexiong Feng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| |
Collapse
|
4
|
Zhao T, Yang J, Wang M, Liu J. Rare BRAF gene fusions in metastatic early-onset colon cancer: A case report. Heliyon 2024; 10:e40753. [PMID: 39687148 PMCID: PMC11648153 DOI: 10.1016/j.heliyon.2024.e40753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Gene fusions offer new therapeutic options for patients with metastatic colon cancer (CC). BRAF gene fusions are infrequent somatic mutations found in CC with prognostic and promising targeted therapies. CC diagnosed before the age of 50 was regarded as early-onset CC (EOCC). The incidence of EOCC is increasing, yet there is a clear unmet need to improve the management of EOCC. Herein, we selectively reported a case of metastatic EOCC with rare BRAF gene fusions. The right-sided tumors were radically resected. Next-generation sequencing (NGS) was performed on formalin-fixed paraffin-embedded tissues to eliminate gene variations. Histologically, the colonic hepatic flexure showed focal mucinous adenocarcinoma changes along with high-grade intraepithelial neoplasia. The results of histopathological examination belonged to pT1bN1bM0 ⅢA stage. Targeted DNA sequencing revealed AGAP3::BRAF (A10;B9) fusion and BRAF::AGAP3 (B8;A11) fusion were simultaneously detected in this case. Microsatellite instability-high (MSI-H) and RAS/BRAF V600E mutations were not detected. During a limited 1.5-year follow-up period, neither a confirmed local recurrence nor a distant organ metastasis occurred in this case. We propose that BRAF fusion variations can occur in metastatic EOCC.
Collapse
Affiliation(s)
- Tingting Zhao
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, China
| | - Junting Yang
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, China
| | - Meirong Wang
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, China
| | - Jie Liu
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
5
|
Dhanasiri AK, Siciliani D, Kortner TM, Krogdahl Å. Epigenetic changes in pyloric caeca of Atlantic salmon fed diets containing increasing levels of lipids and choline. Epigenetics 2024; 19:2305079. [PMID: 38281164 PMCID: PMC10824149 DOI: 10.1080/15592294.2024.2305079] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024] Open
Abstract
An earlier study of ours investigating the effect of dietary lipid levels on the choline requirement of Atlantic salmon showed increasing severity of intestinal steatosis with increasing lipid levels. As choline is involved in epigenetic regulation by being the key methyl donor, pyloric caeca samples from the study were analysed for epigenetic effects of dietary lipid and choline levels. The diets varied in lipid levels between 16% and 28%, and choline levels between 1.9 and 2.3 g/kg. The diets were fed for 8 weeks to Atlantic salmon of 25 g of initial weight. Using reduced representation bisulfite sequencing (RRBS), this study revealed that increasing dietary lipid levels induced methylation differences in genes involved in membrane transport and signalling pathways, and in microRNAs important for the regulation of lipid homoeostasis. Increasing choline levels also affected genes involved in fatty acid biosynthesis and transport, lipolysis, and lipogenesis, as well as important immune genes. Our observations confirmed that choline is involved in epigenetic regulation in Atlantic salmon, as has been reported for higher vertebrates. This study showed the need for the inclusion of biomarkers of epigenetic processes in studies that must be conducted to define optimal choline levels in diets for Atlantic salmon.
Collapse
Affiliation(s)
- Anusha K.S. Dhanasiri
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Daphne Siciliani
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Trond M. Kortner
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Åshild Krogdahl
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| |
Collapse
|
6
|
Fornezza S, Delvecchio VS, Harvey WT, Dishuck PC, Eichler EE, Giannuzzi G. AGAP duplicons associate with structural diversity at Chromosome 10q11.22. Genome Res 2024; 34:1487-1499. [PMID: 39322278 PMCID: PMC11534156 DOI: 10.1101/gr.279454.124] [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: 04/11/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
The 10q11.22 chromosomal region is a duplication-rich interval of the human genome and one of the last to be fully assembled. It carries copy number-variable genes associated with intellectual disability, bipolar disorder, and obesity. In this study, we characterized the structural diversity at this locus by analyzing 64 haploid assemblies produced by the Human Pangenome Reference Consortium. We identified 11 alternative haplotypes that differ in the copy number and/or orientation of large genomic segments, ranging from hundreds of kilobase pairs (kbp) to over one megabase pair (Mbp). We uncovered a 2.4 Mbp size difference between the shortest and longest haplotypes. Breakpoint analysis revealed that genomic instability results from nonallelic homologous recombination between segmental duplication (SD) pairs with varying similarity (94.4%-99.6%). Nonetheless, these pairs generally recombine at positions where their identity is higher (>99.6%). Recurrent inversions occur with different breakpoints within the same inverted SD pair. Inversion polymorphisms shuffle the entire SD arrangement, creating new predispositions to copy-number variations. The SD architecture is associated with a catarrhine-specific subgroup of the AGAP gene family, which likely triggered the accumulation of SDs at this locus over the past 25 million years of human evolution. Our results reveal extensive structural diversity and genomic instability at the 10q11.22 locus, and expand the general understanding of the mutational mechanisms behind SD-mediated rearrangements.
Collapse
Affiliation(s)
| | | | - William T Harvey
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Philip C Dishuck
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, USA
| | | |
Collapse
|
7
|
Li XK, Huang YH, Zhao R, Cao WQ, Lu L, Han JQ, Zhou Y, Zhang X, Wu WA, Tao JJ, Wei W, Zhang WK, Chen SY, Ma B, Zhao H, Yin CC, Zhang JS. Membrane protein MHZ3 regulates the on-off switch of ethylene signaling in rice. Nat Commun 2024; 15:5987. [PMID: 39013913 PMCID: PMC11252128 DOI: 10.1038/s41467-024-50290-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
Ethylene regulates plant growth, development, and stress adaptation. However, the early signaling events following ethylene perception, particularly in the regulation of ethylene receptor/CTRs (CONSTITUTIVE TRIPLE RESPONSE) complex, remains less understood. Here, utilizing the rapid phospho-shift of rice OsCTR2 in response to ethylene as a sensitive readout for signal activation, we revealed that MHZ3, previously identified as a stabilizer of ETHYLENE INSENSITIVE 2 (OsEIN2), is crucial for maintaining OsCTR2 phosphorylation. Genetically, both functional MHZ3 and ethylene receptors prove essential for OsCTR2 phosphorylation. MHZ3 physically interacts with both subfamily I and II ethylene receptors, e.g., OsERS2 and OsETR2 respectively, stabilizing their association with OsCTR2 and thereby maintaining OsCTR2 activity. Ethylene treatment disrupts the interactions within the protein complex MHZ3/receptors/OsCTR2, reducing OsCTR2 phosphorylation and initiating downstream signaling. Our study unveils the dual role of MHZ3 in fine-tuning ethylene signaling activation, providing insights into the initial stages of the ethylene signaling cascade.
Collapse
Affiliation(s)
- Xin-Kai Li
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Hua Huang
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Rui Zhao
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wu-Qiang Cao
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Long Lu
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jia-Qi Han
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yang Zhou
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xun Zhang
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wen-Ai Wu
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian-Jun Tao
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Wei
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wan-Ke Zhang
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shou-Yi Chen
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Biao Ma
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - He Zhao
- The Sainsbury Laboratory, University of East Anglia, Norwich, UK.
| | - Cui-Cui Yin
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jin-Song Zhang
- Key Lab of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
8
|
Xie B, Liang J, Jiang J, Zeng T, Liu L, Xie D, Zhu G, Xiong L, Zhang K, Liu D, Gong J, Chen X, Lai R, Xie H. Zebrafish myo7aa affects congenital hearing by regulating Rho-GTPase signaling. Front Mol Neurosci 2024; 17:1405109. [PMID: 39081296 PMCID: PMC11287254 DOI: 10.3389/fnmol.2024.1405109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/31/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction myo7aa, the homolog of the human Usher 1B syndrome pathogenic gene, myo7A, plays an important role in stereociliary development and maintenance, therefore, is critical for hearing and balance. However, the molecular mechanisms that myo7aa regulate hearing and balance still need to be studied. Methods In this study, we generated two independent zebrafish myo7aa knockout lines using CRISPR/Cas9 technology. To investigate the effects of myo7aa on hearing, YO-PRO-1 staining and startle response assay were used. To gain insight into the specific molecular mechanisms by which myo7aa affects hearing, transcriptome sequencing and bioinformatics analysis were employed. Results Our study showed that hair cells of myo7aa-/- zebrafish can not take up YO-PRO-1 fluorescent dye and are insensitive to acoustic stimulation in myo7aa-/- zebrafish compared to wild type. Genes related to the Rho GTPase signaling pathway, such as arhgap33, dab2ip, and arghef40, are significantly down-regulated in myo7aa-/- zebrafish embryos at 3 dpf. GTP and ATP compensation can partially rescue the hair cell defects in myo7aa knockout zebrafish. Discussion Our findings suggest that zebrafish myo7aa affects congenital hearing by regulating Rho GTPase signaling, and loss of myo7aa leads to abnormal Rho GTPase signaling and impairs hair cell function. myo7aa, myo7A, arhgap33, dab2ip, arghef40 and myo7aa-/- fonts in the abstract are italicized. -/- is a superscript format.
Collapse
Affiliation(s)
- Binling Xie
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jiaxin Liang
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jifan Jiang
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Ting Zeng
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Ling Liu
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Dinghua Xie
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ganghua Zhu
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lei Xiong
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Kanjia Zhang
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, Key Laboratory of Neuroregeneration of Jiangsu and MOE, School of Life Sciences, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Jie Gong
- Nantong Laboratory of Development and Diseases, Key Laboratory of Neuroregeneration of Jiangsu and MOE, School of Life Sciences, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Xiangding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ruosha Lai
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huaping Xie
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
9
|
Chien YC, Yoon GM. Subcellular dynamics of ethylene signaling drive plant plasticity to growth and stress: Spatiotemporal control of ethylene signaling in Arabidopsis. Bioessays 2024; 46:e2400043. [PMID: 38571390 DOI: 10.1002/bies.202400043] [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: 02/22/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
Volatile compounds, such as nitric oxide and ethylene gas, play a vital role as signaling molecules in organisms. Ethylene is a plant hormone that regulates a wide range of plant growth, development, and responses to stress and is perceived by a family of ethylene receptors that localize in the endoplasmic reticulum. Constitutive Triple Response 1 (CTR1), a Raf-like protein kinase and a key negative regulator for ethylene responses, tethers to the ethylene receptors, but undergoes nuclear translocation upon activation of ethylene signaling. This ER-to-nucleus trafficking transforms CTR1 into a positive regulator for ethylene responses, significantly enhancing stress resilience to drought and salinity. The nuclear trafficking of CTR1 demonstrates that the spatiotemporal control of ethylene signaling is essential for stress adaptation. Understanding the mechanisms governing the spatiotemporal control of ethylene signaling elements is crucial for unraveling the system-level regulatory mechanisms that collectively fine-tune ethylene responses to optimize plant growth, development, and stress adaptation.
Collapse
Affiliation(s)
- Yuan-Chi Chien
- Department of Botany and Plant Pathology and Center for Plant Biology, Purdue University, West Lafayette, Indiana, USA
| | - Gyeong Mee Yoon
- Department of Botany and Plant Pathology and Center for Plant Biology, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
10
|
Kauwe G, Pareja-Navarro KA, Yao L, Chen JH, Wong I, Saloner R, Cifuentes H, Nana AL, Shah S, Li Y, Le D, Spina S, Grinberg LT, Seeley WW, Kramer JH, Sacktor TC, Schilling B, Gan L, Casaletto KB, Tracy TE. KIBRA repairs synaptic plasticity and promotes resilience to tauopathy-related memory loss. J Clin Invest 2024; 134:e169064. [PMID: 38299587 PMCID: PMC10836803 DOI: 10.1172/jci169064] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/21/2023] [Indexed: 02/02/2024] Open
Abstract
Synaptic plasticity is obstructed by pathogenic tau in the brain, representing a key mechanism that underlies memory loss in Alzheimer's disease (AD) and related tauopathies. Here, we found that reduced levels of the memory-associated protein KIdney/BRAin (KIBRA) in the brain and increased KIBRA protein levels in cerebrospinal fluid are associated with cognitive impairment and pathological tau levels in disease. We next defined a mechanism for plasticity repair in vulnerable neurons using the C-terminus of the KIBRA protein (CT-KIBRA). We showed that CT-KIBRA restored plasticity and memory in transgenic mice expressing pathogenic human tau; however, CT-KIBRA did not alter tau levels or prevent tau-induced synapse loss. Instead, we found that CT-KIBRA stabilized the protein kinase Mζ (PKMζ) to maintain synaptic plasticity and memory despite tau-mediated pathogenesis. Thus, our results distinguished KIBRA both as a biomarker of synapse dysfunction and as the foundation for a synapse repair mechanism to reverse cognitive impairment in tauopathy.
Collapse
Affiliation(s)
- Grant Kauwe
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Lei Yao
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jackson H. Chen
- Buck Institute for Research on Aging, Novato, California, USA
| | - Ivy Wong
- Buck Institute for Research on Aging, Novato, California, USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Helen Cifuentes
- Buck Institute for Research on Aging, Novato, California, USA
| | - Alissa L. Nana
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Samah Shah
- Buck Institute for Research on Aging, Novato, California, USA
| | - Yaqiao Li
- Gladstone Institutes, San Francisco, Califoria, USA
| | - David Le
- Gladstone Institutes, San Francisco, Califoria, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neurosciences, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neurosciences, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Todd C. Sacktor
- The Robert F. Furchgott Center of Neural and Behavioral Science, Departments of Physiology and Pharmacology, Anesthesiology, and Neurology, State University of New York Health Sciences University, Brooklyn, New York, USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Tara E. Tracy
- Buck Institute for Research on Aging, Novato, California, USA
| |
Collapse
|
11
|
Jiang S, Han X, Liu T, He Y, Zhao Z, Liu T, Cheng S, Zhang J, Duan L, Liu Y, Cheng T, Liu Y, Ye Q, Gao S. HUNK inhibits cargo uptake and lysosomal traffic in the caveolar pathway via the AGAP3/ARF6. Sci Bull (Beijing) 2024; 69:173-178. [PMID: 38071109 DOI: 10.1016/j.scib.2023.11.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 01/16/2024]
Affiliation(s)
- Siyuan Jiang
- Zhongda Hospital, Medical School, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Xiaoqi Han
- Medical School of Guizhou University, Guiyang 550025, China; Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Tihui Liu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Ying He
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Zidong Zhao
- Zhongda Hospital, Medical School, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Tongfeng Liu
- Medical School of Guizhou University, Guiyang 550025, China
| | - Shuwen Cheng
- Nanjing University Medical School, Nanjing 210046, China
| | - Jihang Zhang
- Zhongda Hospital, Medical School, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China; Medical School of Guizhou University, Guiyang 550025, China
| | - Liqiang Duan
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Yajuan Liu
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Tianyou Cheng
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing 100850, China
| | - Shan Gao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China.
| |
Collapse
|
12
|
Artimagnella O, Mazzon E, Salamone S, Pollastro F, Gugliandolo A, Chiricosta L. Cannabinerol (CBNR) Influences Synaptic Genes Associated with Cytoskeleton and Ion Channels in NSC-34 Cell Line: A Transcriptomic Study. Biomedicines 2024; 12:189. [PMID: 38255294 PMCID: PMC10813620 DOI: 10.3390/biomedicines12010189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Cannabinoids are receiving great attention as a novel approach in the treatment of cognitive and motor disabilities, which characterize neurological disorders. To date, over 100 phytocannabinoids have been extracted from Cannabis sativa, and some of them have shown neuroprotective properties and the capacity to influence synaptic transmission. In this study, we investigated the effects of a less-known phytocannabinoid, cannabinerol (CBNR), on neuronal physiology. Using the NSC-34 motor-neuron-like cell line and next-generation sequencing analysis, we discovered that CBNR influences synaptic genes associated with synapse organization and specialization, including genes related to the cytoskeleton and ion channels. Specifically, the calcium, sodium, and potassium channel subunits (Cacna1b, Cacna1c, Cacnb1, Grin1, Scn8a, Kcnc1, Kcnj9) were upregulated, along with genes related to NMDAR (Agap3, Syngap1) and calcium (Cabp1, Camkv) signaling. Moreover, cytoskeletal and cytoskeleton-associated genes (Actn2, Ina, Trio, Marcks, Bsn, Rtn4, Dgkz, Htt) were also regulated by CBNR. These findings highlight the important role played by CBNR in the regulation of synaptogenesis and synaptic transmission, suggesting the need for further studies to evaluate the neuroprotective role of CBNR in the treatment of synaptic dysfunctions that characterize motor disabilities in many neurological disorders.
Collapse
Affiliation(s)
- Osvaldo Artimagnella
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy (E.M.)
| |
Collapse
|
13
|
Højgaard K, Szöllősi B, Henningsen K, Minami N, Nakanishi N, Kaadt E, Tamura M, Morris RGM, Takeuchi T, Elfving B. Novelty-induced memory consolidation is accompanied by increased Agap3 transcription: a cross-species study. Mol Brain 2023; 16:69. [PMID: 37749596 PMCID: PMC10521532 DOI: 10.1186/s13041-023-01056-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023] Open
Abstract
Novelty-induced memory consolidation is a well-established phenomenon that depends on the activation of a locus coeruleus-hippocampal circuit. It is associated with the expression of activity-dependent genes that may mediate initial or cellular memory consolidation. Several genes have been identified to date, however, to fully understand the mechanisms of memory consolidation, additional candidates must be identified. In this cross-species study, we used a contextual novelty-exploration paradigm to identify changes in gene expression in the dorsal hippocampus of both mice and rats. We found that changes in gene expression following contextual novelty varied between the two species, with 9 genes being upregulated in mice and 3 genes in rats. Comparison across species revealed that ArfGAP with a GTPase domain, an ankyrin repeat and PH domain 3 (Agap3) was the only gene being upregulated in both, suggesting a potentially conserved role for Agap3. AGAP3 is known to regulate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptor trafficking in the synapse, which suggests that increased transcription of Agap3 may be involved in maintaining functional plasticity. While we identified several genes affected by contextual novelty exploration, we were unable to fully reverse these changes using SCH 23390, a dopamine D1/D5 receptor antagonist. Further research on the role of AGAP3 in novelty-induced memory consolidation could lead to better understanding of this process and guide future research.
Collapse
Affiliation(s)
- Kristoffer Højgaard
- Translational Neuropsychiatry Unit, Department of Clinical medicine, Aarhus University, Aarhus N, DK8200, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, DK8000, Denmark
| | - Bianka Szöllősi
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, DK8000, Denmark
| | - Kim Henningsen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, DK8000, Denmark
| | - Natsumi Minami
- Neuroscience Research Unit, Mitsubishi Tanabe Pharma Corporation, Kanagawa, 227-0033, Japan
| | - Nobuhiro Nakanishi
- Data Science Department, Mitsubishi Tanabe Pharma Corporation, Kanagawa, 227-0033, Japan
| | - Erik Kaadt
- Translational Neuropsychiatry Unit, Department of Clinical medicine, Aarhus University, Aarhus N, DK8200, Denmark
| | - Makoto Tamura
- Neuroscience Research Unit, Mitsubishi Tanabe Pharma Corporation, Kanagawa, 227-0033, Japan
- NeuroDiscovery Lab, Mitsubishi Tanabe Pharma Holdings America Inc, Cambridge, MA, 02139, USA
| | - Richard G M Morris
- Laboratory for Cognitive Neuroscience, Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Tomonori Takeuchi
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, DK8000, Denmark.
- Center for Proteins in Memory - PROMEMO, Department of Biomedicine, Danish National Research Foundation, Aarhus University, Aarhus C, DK8000, Denmark.
- Gftd DeSci, Gftd DAO, Tokyo, 162-0044, Japan.
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical medicine, Aarhus University, Aarhus N, DK8200, Denmark.
| |
Collapse
|
14
|
Harris HA, Friedman C, Starling AP, Dabelea D, Johnson SL, Fuemmeler BF, Jima D, Murphy SK, Hoyo C, Jansen PW, Felix JF, Mulder R. An epigenome-wide association study of child appetitive traits and DNA methylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.17.549289. [PMID: 37503194 PMCID: PMC10370073 DOI: 10.1101/2023.07.17.549289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Childhood appetitive traits are consistently associated with obesity risk, and yet their etiology is poorly understood. Appetitive traits are complex phenotypes which are hypothesized to be influenced by both genetic and environmental factors, as well as their interactions. Early-life epigenetic processes, such as DNA methylation (DNAm), may be involved in the developmental programming of appetite regulation in childhood. In the current study, we meta-analyzed epigenome-wide association studies (EWASs) of cord blood DNAm and early-childhood appetitive traits. Data were from two independent cohorts: the Generation R Study (n=1,086, Rotterdam, the Netherlands) and the Healthy Start study (n=236, Colorado, USA). DNAm at autosomal methylation sites in cord blood was measured using the Illumina Infinium HumanMethylation450 BeadChip. Parents reported on their child's food responsiveness, emotional undereating, satiety responsiveness and food fussiness using the Children's Eating Behaviour Questionnaire at age 4-5 years. Multiple regression models were used to examine the association of DNAm (predictor) at the individual site- and regional-level (using DMRff) with each appetitive trait (outcome), adjusting for covariates. Bonferroni-correction was applied to adjust for multiple testing. There were no associations of DNAm and any appetitive trait at the individual site-level. However, at the regional level, we identified 45 associations of DNAm with food responsiveness, 7 associations of DNAm with emotional undereating, 13 associations of DNAm with satiety responsiveness, and 9 associations of DNAm with food fussiness. This study shows that DNAm in the newborn may partially explain variation in appetitive traits expressed in early childhood and provides preliminary support for early programming of child appetitive traits through DNAm. Investigating differential DNAm associated with appetitive traits could be an important first step in identifying biological pathways underlying the development of these behaviors.
Collapse
Affiliation(s)
- Holly A. Harris
- Department of Child & Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Erasmus University Rotterdam, Department of Psychology, Education & Child Studies, Rotterdam, the Netherlands
| | - Chloe Friedman
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anne P. Starling
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Susan L. Johnson
- Department of Pediatrics, Section of Nutrition, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bernard F. Fuemmeler
- Virginia Commonwealth University, Mase Comprehensive Cancer Center, Richmond, VA, USA
| | - Dereje Jima
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Susan K. Murphy
- Duke University Medical Center, Department of Obstetrics and Gynecology, Reproductive Sciences, Durham, NC, USA
| | - Cathrine Hoyo
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Pauline W. Jansen
- Department of Child & Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Erasmus University Rotterdam, Department of Psychology, Education & Child Studies, Rotterdam, the Netherlands
| | - Janine F. Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Rosa Mulder
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
15
|
Kauwe G, Pareja-Navarro KA, Yao L, Chen JH, Wong I, Saloner R, Cifuentes H, Nana AL, Shah S, Li Y, Le D, Spina S, Grinberg LT, Seeley WW, Kramer JH, Sacktor TC, Schilling B, Gan L, Casaletto KB, Tracy TE. KIBRA repairs synaptic plasticity and promotes resilience to tauopathy-related memory loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.543777. [PMID: 37398236 PMCID: PMC10312627 DOI: 10.1101/2023.06.12.543777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Synaptic plasticity is obstructed by pathogenic tau in the brain, representing a key mechanism that underlies memory loss in Alzheimer's disease (AD) and related tauopathies. Here, we define a mechanism for plasticity repair in vulnerable neurons using the C-terminus of the KIdney/BRAin (KIBRA) protein (CT-KIBRA). We show that CT-KIBRA restores plasticity and memory in transgenic mice expressing pathogenic human tau; however, CT-KIBRA did not alter tau levels or prevent tau-induced synapse loss. Instead, we find that CT-KIBRA binds to and stabilizes protein kinase Mζ (PKMζ) to maintain synaptic plasticity and memory despite tau-mediated pathogenesis. In humans we find that reduced KIBRA in brain and increased KIBRA in cerebrospinal fluid are associated with cognitive impairment and pathological tau levels in disease. Thus, our results distinguish KIBRA both as a novel biomarker of synapse dysfunction in AD and as the foundation for a synapse repair mechanism to reverse cognitive impairment in tauopathy.
Collapse
Affiliation(s)
- Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA USA
| | | | - Lei Yao
- Buck Institute for Research on Aging, Novato, CA USA
| | | | - Ivy Wong
- Buck Institute for Research on Aging, Novato, CA USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | | | - Alissa L. Nana
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Samah Shah
- Buck Institute for Research on Aging, Novato, CA USA
| | - Yaqiao Li
- Gladstone Institutes, San Francisco, CA USA
| | - David Le
- Gladstone Institutes, San Francisco, CA USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
- Weill Institute for Neurosciences, Department of Pathology, University of California, San Francisco USA
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
- Weill Institute for Neurosciences, Department of Pathology, University of California, San Francisco USA
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Todd C. Sacktor
- The Robert F. Furchgott Center of Neural and Behavioral Science, Departments of Physiology and Pharmacology, Anesthesiology, and Neurology, State University of New York Health Sciences University, Brooklyn, NY USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Tara E. Tracy
- Buck Institute for Research on Aging, Novato, CA USA
| |
Collapse
|
16
|
Desai SS, Ravindran F, Panchal A, Ojha N, Jadhav S, Choudhary B. Whole transcriptome sequencing reveals HOXD11-AGAP3, a novel fusion transcript in the Indian acute leukemia cohort. Front Genet 2023; 14:1100587. [PMID: 37113989 PMCID: PMC10126405 DOI: 10.3389/fgene.2023.1100587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/06/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction: Acute leukemia is a heterogeneous disease with distinct genotypes and complex karyotypes leading to abnormal proliferation of hematopoietic cells. According to GLOBOCAN reports, Asia accounts for 48.6% of leukemia cases, and India reports ~10.2% of all leukemia cases worldwide. Previous studies have shown that the genetic landscape of AML in India is significantly different from that in the western population by WES. Methods: We have sequenced and analyzed 9 acute myeloid leukemia (AML) transcriptome samples in the present study. We performed fusion detection in all the samples and categorized the patients based on cytogenetic abnormalities, followed by a differential expression analysis and WGCNA analysis. Finally, Immune profiles were obtained using CIBERSORTx. Results: We found a novel fusion HOXD11-AGAP3 in 3 patients, BCR-ABL1 in 4, and KMT2A-MLLT3 in one patient. Categorizing the patients based on their cytogenetic abnormalities and performing a differential expression analysis, followed by WGCNA analysis, we observed that in the HOXD11-AGAP3 group, correlated co-expression modules were enriched with genes from pathways like Neutrophil degranulation, Innate Immune system, ECM degradation, and GTP hydrolysis. Additionally, we obtained HOXD11-AGAP3-specific overexpression of chemokines CCL28 and DOCK2. Immune profiling using CIBRSORTx revealed differences in the immune profiles across all the samples. We also observed HOXD11-AGAP3-specific elevated expression of lincRNA HOTAIRM1 and its interacting partner HOXA2. Discussion: The findings highlight population-specific HOXD11-AGAP3, a novel cytogenetic abnormality in AML. The fusion led to alterations in immune system represented by CCL28 and DOCK2 over-expression. Interestingly, in AML, CCL28 is known prognostic marker. Additionally, non-coding signatures (HOTAIRM1) were observed specific to the HOXD11-AGAP3 fusion transcript which are known to be implicated in AML.
Collapse
Affiliation(s)
- Sagar Sanjiv Desai
- Department of Biotechnology and Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
- Graduate Student Registered Under Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Febina Ravindran
- Department of Biotechnology and Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| | - Amey Panchal
- Cancer Centre, Healthcare Global Enterprises Ltd., Bangalore, India
| | - Nishit Ojha
- Cancer Centre, Healthcare Global Enterprises Ltd., Bangalore, India
| | - Sachin Jadhav
- Cancer Centre, Healthcare Global Enterprises Ltd., Bangalore, India
| | - Bibha Choudhary
- Department of Biotechnology and Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| |
Collapse
|
17
|
Park HL, Seo DH, Lee HY, Bakshi A, Park C, Chien YC, Kieber JJ, Binder BM, Yoon GM. Ethylene-triggered subcellular trafficking of CTR1 enhances the response to ethylene gas. Nat Commun 2023; 14:365. [PMID: 36690618 PMCID: PMC9870993 DOI: 10.1038/s41467-023-35975-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
The phytohormone ethylene controls plant growth and stress responses. Ethylene-exposed dark-grown Arabidopsis seedlings exhibit dramatic growth reduction, yet the seedlings rapidly return to the basal growth rate when ethylene gas is removed. However, the underlying mechanism governing this acclimation of dark-grown seedlings to ethylene remains enigmatic. Here, we report that ethylene triggers the translocation of the Raf-like protein kinase CONSTITUTIVE TRIPLE RESPONSE1 (CTR1), a negative regulator of ethylene signaling, from the endoplasmic reticulum to the nucleus. Nuclear-localized CTR1 stabilizes the ETHYLENE-INSENSITIVE3 (EIN3) transcription factor by interacting with and inhibiting EIN3-BINDING F-box (EBF) proteins, thus enhancing the ethylene response and delaying growth recovery. Furthermore, Arabidopsis plants with enhanced nuclear-localized CTR1 exhibited improved tolerance to drought and salinity stress. These findings uncover a mechanism of the ethylene signaling pathway that links the spatiotemporal dynamics of cellular signaling components to physiological responses.
Collapse
Affiliation(s)
- Hye Lin Park
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, 47907, USA
- Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
| | - Dong Hye Seo
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, 47907, USA
- Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
- Department of Systems Biology, Yonsei University, Seoul, 03722, Korea
| | - Han Yong Lee
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, 47907, USA
- Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
- Department of Biology, Chosun University, Gwangju, 61452, Korea
| | - Arkadipta Bakshi
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996, USA
- Department of Botany, UW-Madison, Madison, WI, USA
| | - Chanung Park
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, 47907, USA
- Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
| | - Yuan-Chi Chien
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, 47907, USA
- Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
| | - Joseph J Kieber
- Department of Biology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Brad M Binder
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Gyeong Mee Yoon
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, 47907, USA.
- Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
18
|
Paoli C, Burel‐Vandenbos F, Coulomb‐l'Hermine A, Cros J, Pondrom M, Kubiniek V, Pedeutour F, Dadone‐Montaudié B. AGAP3: A novel BRAF fusion partner in pediatric pancreatic-type acinar cell carcinoma. Genes Chromosomes Cancer 2022; 61:734-739. [PMID: 35949061 PMCID: PMC9804258 DOI: 10.1002/gcc.23091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/27/2022] [Accepted: 08/07/2022] [Indexed: 01/05/2023] Open
Abstract
Most available molecular data on pancreatic acinar cell carcinoma (PACC) are provided by studies of adult cases. BRAF, RAF1, or RET rearrangements have been described in approximately 30% of cases. To the best of our knowledge, only seven cases with molecular data have been reported in pediatric PACC. We report here the comprehensive study of a pancreatic-type ACC from a 6-year-old patient. We detected an AGAP3::BRAF fusion. This result showing a BRAF rearrangement demonstrates a molecular link between adult and pediatric PACC. Moreover, it identifies AGAP3, a gene located at 7q36.1 that encodes a major component of the N-methyl-d-aspartate (NMDA) receptor signaling complex, as a partner gene of BRAF. The variability of BRAF partners is consistent with a driver role of BRAF alterations in PACC. The identification of such alterations is noteworthy for considering the use of MEK inhibitors in metastatic cases. We did not detect associated genomic instability. The better outcome of pediatric cases might be related to their stable genomic background.
Collapse
Affiliation(s)
- Charlotte Paoli
- Central Laboratory of Pathology, Department of Pathology and Molecular OncologyUniversity Hospital of Nice‐Côte d'Azur UniversityNiceFrance
| | - Fanny Burel‐Vandenbos
- Central Laboratory of Pathology, Department of Pathology and Molecular OncologyUniversity Hospital of Nice‐Côte d'Azur UniversityNiceFrance
| | | | - Jérôme Cros
- Department of PathologyBeaujon HospitalClichyFrance
| | - Morgane Pondrom
- Department of Pediatric Oncology and HematologyUniversity Hospital of Nice‐Côte d'Azur UniversityNiceFrance
| | - Valérie Kubiniek
- Laboratory of Solid Tumor Genetics, Department of Pathology and Molecular OncologyUniversity Hospital of Nice‐Côte d'Azur UniversityNiceFrance,Laboratory of Solid Tumor GeneticsInstitute for Research on Cancer and Aging of Nice (IRCAN)NiceFrance
| | - Florence Pedeutour
- Laboratory of Solid Tumor Genetics, Department of Pathology and Molecular OncologyUniversity Hospital of Nice‐Côte d'Azur UniversityNiceFrance,Laboratory of Solid Tumor GeneticsInstitute for Research on Cancer and Aging of Nice (IRCAN)NiceFrance
| | - Bérengère Dadone‐Montaudié
- Central Laboratory of Pathology, Department of Pathology and Molecular OncologyUniversity Hospital of Nice‐Côte d'Azur UniversityNiceFrance,Laboratory of Solid Tumor Genetics, Department of Pathology and Molecular OncologyUniversity Hospital of Nice‐Côte d'Azur UniversityNiceFrance,Laboratory of Solid Tumor GeneticsInstitute for Research on Cancer and Aging of Nice (IRCAN)NiceFrance
| |
Collapse
|
19
|
Zhao K, Zhang H, Wu Y, Liu J, Li X, Lin J. Integrated analysis and identification of hub genes as novel biomarkers for Alzheimer’s disease. Front Aging Neurosci 2022; 14:901972. [PMID: 36110430 PMCID: PMC9468260 DOI: 10.3389/fnagi.2022.901972] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is an intractable and progressive neurodegenerative disorder that can lead to severe cognitive decline, impaired speech, short-term memory loss, and finally an inability to function in daily life. For patients, their families, and even all of society, AD can impart great emotional pressure and economic costs. Therefore, this study aimed to investigate potential diagnostic biomarkers of AD. Using the Gene Expression Omnibus (GEO) database, the expression profiles of genes were extracted from the GSE5281, GSE28146, and GSE48350 microarray datasets. Then, immune-related genes were identified by the intersections of differentially expressed genes (DEGs). Functional enrichment analyses, including Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, Disease Ontology (DO), and Gene Set Enrichment Analysis (GSEA), were performed. Subsequently, random forest models and least absolute shrinkage and selection operator regression were used to further screen hub genes, which were then validated using receiver operating characteristic (ROC) curve analysis. Finally, 153 total immune-related DEGs were identified in relation to AD. DO analysis of these immune-related DEGs showed that they were enriched in “lung disease,” “reproductive system disease,” and “atherosclerosis.” Single GSEA of hub genes showed that they were particularly enriched in “oxidative phosphorylation.” ROC analysis of AGAP3 yielded an area under the ROC curve of 0.878 for GSE5281, 0.727 for GSE28146, and 0.635 for GSE48350. Moreover, immune infiltration analysis demonstrated that AGAP3 was related to follicular helper T cells, naïve CD4 T cells, naïve B cells, memory B cells, macrophages M0, macrophages M1, macrophages M2, resting natural killer (NK) cells, activated NK cells, monocytes, neutrophils, eosinophils, and activated mast cells. These results indicate that identifying immune-related DEGs might enhance the current understanding of the development and prognosis of AD. Furthermore, AGAP3 not only plays a vital role in AD progression and diagnosis but could also serve as a valuable target for further research on AD.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Neurology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Hui Zhang
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Yinyan Wu
- Department of Neurology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Jianzhi Liu
- Department of Neurology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Xuezhong Li
- Department of Neurology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Jianyang Lin
- Department of General Surgery, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
- *Correspondence: Jianyang Lin,
| |
Collapse
|
20
|
Viard J, Loe-Mie Y, Daudin R, Khelfaoui M, Plancon C, Boland A, Tejedor F, Huganir RL, Kim E, Kinoshita M, Liu G, Haucke V, Moncion T, Yu E, Hindie V, Bléhaut H, Mircher C, Herault Y, Deleuze JF, Rain JC, Simonneau M, Lepagnol-Bestel AM. Chr21 protein-protein interactions: enrichment in proteins involved in intellectual disability, autism, and late-onset Alzheimer's disease. Life Sci Alliance 2022; 5:e202101205. [PMID: 35914814 PMCID: PMC9348576 DOI: 10.26508/lsa.202101205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Down syndrome (DS) is caused by human chromosome 21 (HSA21) trisomy. It is characterized by a poorly understood intellectual disability (ID). We studied two mouse models of DS, one with an extra copy of the <i>Dyrk1A</i> gene (189N3) and the other with an extra copy of the mouse Chr16 syntenic region (Dp(16)1Yey). RNA-seq analysis of the transcripts deregulated in the embryonic hippocampus revealed an enrichment in genes associated with chromatin for the 189N3 model, and synapses for the Dp(16)1Yey model. A large-scale yeast two-hybrid screen (82 different screens, including 72 HSA21 baits and 10 rebounds) of a human brain library containing at least 10<sup>7</sup> independent fragments identified 1,949 novel protein-protein interactions. The direct interactors of HSA21 baits and rebounds were significantly enriched in ID-related genes (<i>P</i>-value < 2.29 × 10<sup>-8</sup>). Proximity ligation assays showed that some of the proteins encoded by HSA21 were located at the dendritic spine postsynaptic density, in a protein network at the dendritic spine postsynapse. We located HSA21 DYRK1A and DSCAM, mutations of which increase the risk of autism spectrum disorder (ASD) 20-fold, in this postsynaptic network. We found that an intracellular domain of DSCAM bound either DLGs, which are multimeric scaffolds comprising receptors, ion channels and associated signaling proteins, or DYRK1A. The DYRK1A-DSCAM interaction domain is conserved in <i>Drosophila</i> and humans. The postsynaptic network was found to be enriched in proteins associated with ARC-related synaptic plasticity, ASD, and late-onset Alzheimer's disease. These results highlight links between DS and brain diseases with a complex genetic basis.
Collapse
Affiliation(s)
- Julia Viard
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Yann Loe-Mie
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Rachel Daudin
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Malik Khelfaoui
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Christine Plancon
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Anne Boland
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Francisco Tejedor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Universidad Miguel Hernandez-Campus de San Juan, San Juan, Spain
| | - Richard L Huganir
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Science, Nagoya University Graduate School of Science, Nagoya, Japan
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Institut für Molekulare Pharmakologie (FMP) and Freie Universität Berlin, Berlin, Germany
| | | | - Eugene Yu
- Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, State University of New York at Buffalo, Buffalo, NY, USA
| | | | | | | | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- INSERM, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
- PHENOMIN, Institut Clinique de la Souris, ICS, GIE CERBM, CNRS, INSERM, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Jean-François Deleuze
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | | | - Michel Simonneau
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Université Paris-Saclay, CNRS, ENS Paris-Saclay, CentraleSupélec, LuMIn, Gif sur Yvette, France
- Department of Biology, Ecole Normale Supérieure Paris-Saclay Université Paris-Saclay, Gif sur Yvette, France
| | | |
Collapse
|
21
|
Differential Methylation Profile in Fragile X Syndrome-Prone Offspring Mice after in Utero Exposure to Lactobacillus Reuteri. Genes (Basel) 2022; 13:genes13081300. [PMID: 35893036 PMCID: PMC9331364 DOI: 10.3390/genes13081300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 01/13/2023] Open
Abstract
Environmental factors such as diet, gut microbiota, and infections have proven to have a significant role in epigenetic modifications. It is known that epigenetic modifications may cause behavioral and neuronal changes observed in neurodevelopmental disabilities, including fragile X syndrome (FXS) and autism (ASD). Probiotics are live microorganisms that provide health benefits when consumed, and in some cases are shown to decrease the chance of developing neurological disorders. Here, we examined the epigenetic outcomes in offspring mice after feeding of a probiotic organism, Lactobacillus reuteri (L. reuteri), to pregnant mother animals. In this study, we tested a cohort of Western diet-fed descendant mice exhibiting a high frequency of behavioral features and lower FMRP protein expression similar to what is observed in FXS in humans (described in a companion manuscript in this same GENES special topic issue). By investigating 17,735 CpG sites spanning the whole mouse genome, we characterized the epigenetic profile in two cohorts of mice descended from mothers treated and non-treated with L. reuteri to determine the effect of prenatal probiotic exposure on the prevention of FXS-like symptoms. We found several genes involved in different neurological pathways being differentially methylated (p ≤ 0.05) between the cohorts. Among the key functions, synaptogenesis, neurogenesis, synaptic modulation, synaptic transmission, reelin signaling pathway, promotion of specification and maturation of neurons, and long-term potentiation were observed. The results of this study are relevant as they could lead to a better understanding of the pathways involved in these disorders, to novel therapeutics approaches, and to the identification of potential biomarkers for early detection of these conditions.
Collapse
|
22
|
Mogenet A, Barlesi F, Besse B, Michiels S, Karimi M, Tran-Dien A, Girard N, Mazieres J, Audigier-Valette C, Locatelli-Sanchez M, Kamal M, Gestraud P, Hamza A, Jacquet A, Jimenez M, Yara S, Greillier L, Bertucci F, Planchard D, Soria JC, Bieche I, Tomasini P. Molecular profiling of non-small-cell lung cancer patients with or without brain metastases included in the randomized SAFIR02-LUNG trial and association with intracranial outcome. Lung Cancer 2022; 169:31-39. [DOI: 10.1016/j.lungcan.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/13/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
|
23
|
Fan LL, Sheng Y, Wang CY, Li YL, Liu JS. Case Report: Congenital Brain Dysplasia, Developmental Delay and Intellectual Disability in a Patient With a 7q35-7q36.3 Deletion. Front Genet 2021; 12:761003. [PMID: 34925452 PMCID: PMC8671813 DOI: 10.3389/fgene.2021.761003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
7q terminal deletion syndrome is a rare condition presenting with multiple congenital malformations, including abnormal brain and facial structures, developmental delay, intellectual disability, abnormal limbs, and sacral anomalies. At least 40 OMIM genes located in the 7q34-7q36.3 region act as candidate genes for these phenotypes, of which SHH, EN2, KCNH2, RHEB, HLXB9, EZH2, MNX1 and LIMR1 may be the most important. In this study, we discuss the case of a 2.5-year-old male patient with multiple malformations, congenital brain dysplasia, developmental delay, and intellectual disability. A high-resolution genome-wide single nucleotide polymorphism array and real-time polymerase chain reaction were performed to detect genetic lesions. A de novo 9.4 Mb deletion in chromosome region 7q35-7q36.3 (chr7:147,493,985-156,774,460) was found. This chromosome region contains 68 genes, some of which are candidate genes for each phenotype. To the best of our knowledge, this is a rare case report of 7q terminal deletion syndrome in a Chinese patient. Our study identifies a rare phenotype in terms of brain structure abnormalities and cerebellar sulcus widening in patients with deletion in 7q35-7q36.3.
Collapse
Affiliation(s)
- Liang-Liang Fan
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha, China.,Departments of Reproductive Genetics, HeBei General Hospital, ShiJiaZhuang, China.,Department of Cell Biology, The School of Life Sciences, Central South University, Changsha, China
| | - Yue Sheng
- Department of Cell Biology, The School of Life Sciences, Central South University, Changsha, China
| | - Chen-Yu Wang
- Department of Cell Biology, The School of Life Sciences, Central South University, Changsha, China
| | - Ya-Li Li
- Departments of Reproductive Genetics, HeBei General Hospital, ShiJiaZhuang, China
| | - Ji-Shi Liu
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cell Biology, The School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
24
|
Branch CL, Semenov GA, Wagner DN, Sonnenberg BR, Pitera AM, Bridge ES, Taylor SA, Pravosudov VV. The genetic basis of spatial cognitive variation in a food-caching bird. Curr Biol 2021; 32:210-219.e4. [PMID: 34735793 DOI: 10.1016/j.cub.2021.10.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/15/2021] [Accepted: 10/14/2021] [Indexed: 01/02/2023]
Abstract
Spatial cognition is used by most organisms to navigate their environment. Some species rely particularly heavily on specialized spatial cognition to survive, suggesting that a heritable component of cognition may be under natural selection. This idea remains largely untested outside of humans, perhaps because cognition in general is known to be strongly affected by learning and experience.1-4 We investigated the genetic basis of individual variation in spatial cognition used by non-migratory food-caching birds to recover food stores and survive harsh montane winters. Comparing the genomes of wild, free-living birds ranging from best to worst in their performance on a spatial cognitive task revealed significant associations with genes involved in neuron growth and development and hippocampal function. These results identify candidate genes associated with differences in spatial cognition and provide a critical link connecting individual variation in spatial cognition with natural selection.
Collapse
Affiliation(s)
- Carrie L Branch
- Cornell Lab of Ornithology, Cornell University, Ithaca, NY 14850, USA.
| | - Georgy A Semenov
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO 80309, USA
| | - Dominique N Wagner
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO 80309, USA
| | - Benjamin R Sonnenberg
- Ecology, Evolution, and Conservation Biology Graduate Program, University of Nevada, Reno, NV 89557, USA
| | - Angela M Pitera
- Ecology, Evolution, and Conservation Biology Graduate Program, University of Nevada, Reno, NV 89557, USA
| | - Eli S Bridge
- Ecology and Evolutionary Biology, University of Oklahoma, Norman, OK 73019, USA
| | - Scott A Taylor
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO 80309, USA
| | - Vladimir V Pravosudov
- Ecology, Evolution, and Conservation Biology Graduate Program, University of Nevada, Reno, NV 89557, USA.
| |
Collapse
|
25
|
Pérez RF, Tejedor JR, Santamarina-Ojeda P, Martínez VL, Urdinguio RG, Villamañán L, Candiota AP, Sarró NMV, Barradas M, Fernandez-Marcos PJ, Serrano M, Fernández AF, Fraga MF. Conservation of Aging and Cancer Epigenetic Signatures across Human and Mouse. Mol Biol Evol 2021; 38:3415-3435. [PMID: 33871658 PMCID: PMC8321527 DOI: 10.1093/molbev/msab112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aging and cancer are two interrelated processes, with aging being a major risk factor for the development of cancer. Parallel epigenetic alterations have been described for both, although differences, especially within the DNA hypomethylation scenario, have also been recently reported. Although many of these observations arise from the use of mouse models, there is a lack of systematic comparisons of human and mouse epigenetic patterns in the context of disease. However, such comparisons are significant as they allow to establish the extent to which some of the observed similarities or differences arise from pre-existing species-specific epigenetic traits. Here, we have used reduced representation bisulfite sequencing to profile the brain methylomes of young and old, tumoral and nontumoral brain samples from human and mouse. We first characterized the baseline epigenomic patterns of the species and subsequently focused on the DNA methylation alterations associated with cancer and aging. Next, we described the functional genomic and epigenomic context associated with the alterations, and finally, we integrated our data to study interspecies DNA methylation levels at orthologous CpG sites. Globally, we found considerable differences between the characteristics of DNA methylation alterations in cancer and aging in both species. Moreover, we describe robust evidence for the conservation of the specific cancer and aging epigenomic signatures in human and mouse. Our observations point toward the preservation of the functional consequences of these alterations at multiple levels of genomic regulation. Finally, our analyses reveal a role for the genomic context in explaining disease- and species-specific epigenetic traits.
Collapse
Affiliation(s)
- Raúl F Pérez
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain.,Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain.,Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain.,Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Juan Ramón Tejedor
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain.,Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain.,Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain.,Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Pablo Santamarina-Ojeda
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain.,Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain.,Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain.,Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Virginia López Martínez
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain.,Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain.,Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain.,Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Rocío G Urdinguio
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain.,Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain.,Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain.,Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Lucía Villamañán
- Unitat de Bioquímica de Biociències, Departament de Bioquímica i Biologia Molecular, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Unitat de Bioquímica de Biociències, Departament de Bioquímica i Biologia Molecular, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - N Mí Vidal Sarró
- Servicio Anatomía Patológica, Hospital Universitari de Bellvitge-IDIBELL, Hospitalet de Llobregat, Spain
| | - Marta Barradas
- Metabolic Syndrome Group-BIOPROMET, Madrid Institute for Advanced Studies-IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Pablo Jose Fernandez-Marcos
- Metabolic Syndrome Group-BIOPROMET, Madrid Institute for Advanced Studies-IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Manuel Serrano
- Tumour Suppression Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Agusín F Fernández
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain.,Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain.,Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain.,Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Mario F Fraga
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain.,Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.,Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain.,Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain.,Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| |
Collapse
|
26
|
Nagashima S, Ito N, Kobayashi R, Shiiba I, Shimura H, Fukuda T, Hagihara H, Miyakawa T, Inatome R, Yanagi S. Forebrain-specific deficiency of the GTPase CRAG/Centaurin-γ3 leads to immature dentate gyri and hyperactivity in mice. J Biol Chem 2021; 296:100620. [PMID: 33811862 PMCID: PMC8099661 DOI: 10.1016/j.jbc.2021.100620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 11/26/2022] Open
Abstract
Mouse models of various neuropsychiatric disorders, such as schizophrenia, often display an immature dentate gyrus, characterized by increased numbers of immature neurons and neuronal progenitors and a dearth of mature neurons. We previously demonstrated that the CRMP5-associated GTPase (CRAG), a short splice variant of Centaurin-γ3/AGAP3, is highly expressed in the dentate gyrus. CRAG promotes cell survival and antioxidant defense by inducing the activation of serum response factors at promyelocytic leukemia protein bodies, which are nuclear stress-responsive domains, during neuronal development. However, the physiological role of CRAG in neuronal development remains unknown. Here, we analyzed the role of CRAG using dorsal forebrain-specific CRAG/Centaurin-γ3 knockout mice. The mice revealed maturational abnormality of the hippocampal granule cells, including increased doublecortin-positive immature neurons and decreased calbindin-positive mature neurons, a typical phenotype of immature dentate gyri. Furthermore, the mice displayed hyperactivity in the open-field test, a common measure of exploratory behavior, suggesting that these mice may serve as a novel model for neuropsychiatric disorder associated with hyperactivity. Thus, we conclude that CRAG is required for the maturation of neurons in the dentate gyrus, raising the possibility that its deficiency might promote the development of psychiatric disorders in humans.
Collapse
Affiliation(s)
- Shun Nagashima
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan.
| | - Naoki Ito
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan; Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Reiki Kobayashi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Isshin Shiiba
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan; Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Hiroki Shimura
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Toshifumi Fukuda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Hideo Hagihara
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Ryoko Inatome
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan; Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan.
| |
Collapse
|
27
|
ADAP1/Centaurin-α1 Negatively Regulates Dendritic Spine Function and Memory Formation in the Hippocampus. eNeuro 2021; 8:ENEURO.0111-20.2020. [PMID: 33139322 PMCID: PMC7808333 DOI: 10.1523/eneuro.0111-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022] Open
Abstract
ADAP1/Centaurin-α1 (CentA1) functions as an Arf6 GTPase-activating protein highly enriched in the brain. Previous studies demonstrated the involvement of CentA1 in brain function as a regulator of dendritic differentiation and a potential mediator of Alzheimer’s disease (AD) pathogenesis. To better understand the neurobiological functions of CentA1 signaling in the brain, we developed Centa1 knock-out (KO) mice. The KO animals showed neither brain development nor synaptic ultrastructure deficits in the hippocampus. However, they exhibited significantly higher density and enhanced structural plasticity of dendritic spines in the CA1 region of the hippocampus compared with non-transgenic (NTG) littermates. Moreover, the deletion of Centa1 improved performance in the object-in-place (OIP) spatial memory task. These results suggest that CentA1 functions as a negative regulator of spine density and plasticity, and of hippocampus-dependent memory formation. Thus, CentA1 and its downstream signaling may serve as a potential therapeutic target to prevent memory decline associated with aging and brain disorders.
Collapse
|
28
|
Graves AR, Roth RH, Tan HL, Zhu Q, Bygrave AM, Lopez-Ortega E, Hong I, Spiegel AC, Johnson RC, Vogelstein JT, Tward DJ, Miller MI, Huganir RL. Visualizing synaptic plasticity in vivo by large-scale imaging of endogenous AMPA receptors. eLife 2021; 10:66809. [PMID: 34658338 PMCID: PMC8616579 DOI: 10.7554/elife.66809] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 10/16/2021] [Indexed: 02/06/2023] Open
Abstract
Elucidating how synaptic molecules such as AMPA receptors mediate neuronal communication and tracking their dynamic expression during behavior is crucial to understand cognition and disease, but current technological barriers preclude large-scale exploration of molecular dynamics in vivo. We have developed a suite of innovative methodologies that break through these barriers: a new knockin mouse line with fluorescently tagged endogenous AMPA receptors, two-photon imaging of hundreds of thousands of labeled synapses in behaving mice, and computer vision-based automatic synapse detection. Using these tools, we can longitudinally track how the strength of populations of synapses changes during behavior. We used this approach to generate an unprecedentedly detailed spatiotemporal map of synapses undergoing changes in strength following sensory experience. More generally, these tools can be used as an optical probe capable of measuring functional synapse strength across entire brain areas during any behavioral paradigm, describing complex system-wide changes with molecular precision.
Collapse
Affiliation(s)
- Austin R Graves
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States,Kavli Neuroscience Discovery InstituteBaltimoreUnited States
| | - Richard H Roth
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Han L Tan
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Qianwen Zhu
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Elena Lopez-Ortega
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ingie Hong
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Alina C Spiegel
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States,Kavli Neuroscience Discovery InstituteBaltimoreUnited States
| | - Richard C Johnson
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Joshua T Vogelstein
- Kavli Neuroscience Discovery InstituteBaltimoreUnited States,Center for Imaging Science, Johns Hopkins University School of EngineeringBaltimoreUnited States,Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Daniel J Tward
- Kavli Neuroscience Discovery InstituteBaltimoreUnited States,Center for Imaging Science, Johns Hopkins University School of EngineeringBaltimoreUnited States,Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Michael I Miller
- Kavli Neuroscience Discovery InstituteBaltimoreUnited States,Center for Imaging Science, Johns Hopkins University School of EngineeringBaltimoreUnited States,Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States,Kavli Neuroscience Discovery InstituteBaltimoreUnited States
| |
Collapse
|
29
|
Rozés-Salvador V, González-Billault C, Conde C. The Recycling Endosome in Nerve Cell Development: One Rab to Rule Them All? Front Cell Dev Biol 2020; 8:603794. [PMID: 33425908 PMCID: PMC7793921 DOI: 10.3389/fcell.2020.603794] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Endocytic recycling is an intracellular process that returns internalized molecules back to the plasma membrane and plays crucial roles not only in the reuse of receptor molecules but also in the remodeling of the different components of this membrane. This process is required for a diversity of cellular events, including neuronal morphology acquisition and functional regulation, among others. The recycling endosome (RE) is a key vesicular component involved in endocytic recycling. Recycling back to the cell surface may occur with the participation of several different Rab proteins, which are master regulators of membrane/protein trafficking in nerve cells. The RE consists of a network of interconnected and functionally distinct tubular subdomains that originate from sorting endosomes and transport their cargoes along microtubule tracks, by fast or slow recycling pathways. Different populations of REs, particularly those formed by Rab11, Rab35, and Arf6, are associated with a myriad of signaling proteins. In this review, we discuss the cumulative evidence suggesting the existence of heterogeneous domains of REs, controlling different aspects of neurogenesis, with a particular focus on the commonalities and singularities of these REs and their contribution to nerve development and differentiation in several animal models.
Collapse
Affiliation(s)
- Victoria Rozés-Salvador
- Instituto de Investigación Médica Mercedes y Martín Ferreyra INIMEC-CONICET-UNC, Córdoba, Argentina.,Instituto de Ciencias Básicas, Universidad Nacional de Villa María, Córdoba, Argentina
| | - Christian González-Billault
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile.,Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, United States
| | - Cecilia Conde
- Instituto de Investigación Médica Mercedes y Martín Ferreyra INIMEC-CONICET-UNC, Córdoba, Argentina
| |
Collapse
|
30
|
Schürmann B, Bermingham DP, Kopeikina KJ, Myczek K, Yoon S, Horan KE, Kelly CJ, Martin-de-Saavedra MD, Forrest MP, Fawcett-Patel JM, Smith KR, Gao R, Bach A, Burette AC, Rappoport JZ, Weinberg RJ, Martina M, Penzes P. A novel role for the late-onset Alzheimer's disease (LOAD)-associated protein Bin1 in regulating postsynaptic trafficking and glutamatergic signaling. Mol Psychiatry 2020; 25:2000-2016. [PMID: 30967682 PMCID: PMC6785379 DOI: 10.1038/s41380-019-0407-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022]
Abstract
Postsynaptic trafficking plays a key role in regulating synapse structure and function. While spiny excitatory synapses can be stable throughout adult life, their morphology and function is impaired in Alzheimer's disease (AD). However, little is known about how AD risk genes impact synaptic function. Here we used structured superresolution illumination microscopy (SIM) to study the late-onset Alzheimer's disease (LOAD) risk factor BIN1, and show that this protein is abundant in postsynaptic compartments, including spines. While postsynaptic Bin1 shows colocalization with clathrin, a major endocytic protein, it also colocalizes with the small GTPases Rab11 and Arf6, components of the exocytic pathway. Bin1 participates in protein complexes with Arf6 and GluA1, and manipulations of Bin1 lead to changes in spine morphology, AMPA receptor surface expression and trafficking, and AMPA receptor-mediated synaptic transmission. Our data provide new insights into the mesoscale architecture of postsynaptic trafficking compartments and their regulation by a major LOAD risk factor.
Collapse
Affiliation(s)
- Britta Schürmann
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Daniel P. Bermingham
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Katherine J. Kopeikina
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kristoffer Myczek
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Katherine E. Horan
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Crystle J. Kelly
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Marc P. Forrest
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Katharine R. Smith
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ruoqi Gao
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anthony Bach
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Joshua Z. Rappoport
- Center for Advanced Microscopy and Nikon Imaging Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Marco Martina
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
31
|
Perdigão C, Barata MA, Araújo MN, Mirfakhar FS, Castanheira J, Guimas Almeida C. Intracellular Trafficking Mechanisms of Synaptic Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2020; 14:72. [PMID: 32362813 PMCID: PMC7180223 DOI: 10.3389/fncel.2020.00072] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease characterized by progressive memory loss. Although AD neuropathological hallmarks are extracellular amyloid plaques and intracellular tau tangles, the best correlate of disease progression is synapse loss. What causes synapse loss has been the focus of several researchers in the AD field. Synapses become dysfunctional before plaques and tangles form. Studies based on early-onset familial AD (eFAD) models have supported that synaptic transmission is depressed by β-amyloid (Aβ) triggered mechanisms. Since eFAD is rare, affecting only 1% of patients, research has shifted to the study of the most common late-onset AD (LOAD). Intracellular trafficking has emerged as one of the pathways of LOAD genes. Few studies have assessed the impact of trafficking LOAD genes on synapse dysfunction. Since endocytic traffic is essential for synaptic function, we reviewed Aβ-dependent and independent mechanisms of the earliest synaptic dysfunction in AD. We have focused on the role of intraneuronal and secreted Aβ oligomers, highlighting the dysfunction of endocytic trafficking as an Aβ-dependent mechanism of synapse dysfunction in AD. Here, we reviewed the LOAD trafficking genes APOE4, ABCA7, BIN1, CD2AP, PICALM, EPH1A, and SORL1, for which there is a synaptic link. We conclude that in eFAD and LOAD, the earliest synaptic dysfunctions are characterized by disruptions of the presynaptic vesicle exo- and endocytosis and of postsynaptic glutamate receptor endocytosis. While in eFAD synapse dysfunction seems to be triggered by Aβ, in LOAD, there might be a direct synaptic disruption by LOAD trafficking genes. To identify promising therapeutic targets and biomarkers of the earliest synaptic dysfunction in AD, it will be necessary to join efforts in further dissecting the mechanisms used by Aβ and by LOAD genes to disrupt synapses.
Collapse
Affiliation(s)
- Catarina Perdigão
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Mariana A Barata
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Margarida N Araújo
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Farzaneh S Mirfakhar
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jorge Castanheira
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cláudia Guimas Almeida
- Laboratory Neuronal Trafficking in Aging, CEDOC Chronic Diseases Research Center, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
32
|
Tan HL, Roth RH, Graves AR, Cudmore RH, Huganir RL. Lamina-specific AMPA receptor dynamics following visual deprivation in vivo. eLife 2020; 9:e52420. [PMID: 32125273 PMCID: PMC7053996 DOI: 10.7554/elife.52420] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/10/2020] [Indexed: 11/19/2022] Open
Abstract
Regulation of AMPA receptor (AMPAR) expression is central to synaptic plasticity and brain function, but how these changes occur in vivo remains elusive. Here, we developed a method to longitudinally monitor the expression of synaptic AMPARs across multiple cortical layers in awake mice using two-photon imaging. We observed that baseline AMPAR expression in individual spines is highly dynamic with more dynamics in primary visual cortex (V1) layer 2/3 (L2/3) neurons than V1 L5 neurons. Visual deprivation through binocular enucleation induces a synapse-specific and depth-dependent change of synaptic AMPARs in V1 L2/3 neurons, wherein deep synapses are potentiated more than superficial synapses. The increase is specific to L2/3 neurons and absent on apical dendrites of L5 neurons, and is dependent on expression of the AMPAR-binding protein GRIP1. Our study demonstrates that specific neuronal connections, across cortical layers and even within individual neurons, respond uniquely to changes in sensory experience.
Collapse
Affiliation(s)
- Han L Tan
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Richard H Roth
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Austin R Graves
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Robert H Cudmore
- Department of Physiology and Membrane Biology, University of California School of MedicineDavisUnited States
| | - Richard L Huganir
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
33
|
Stangl C, Post JB, van Roosmalen MJ, Hami N, Verlaan-Klink I, Vos HR, van Es RM, Koudijs MJ, Voest EE, Snippert HJG, Kloosterman WP. Diverse BRAF Gene Fusions Confer Resistance to EGFR-Targeted Therapy via Differential Modulation of BRAF Activity. Mol Cancer Res 2020; 18:537-548. [PMID: 31911540 DOI: 10.1158/1541-7786.mcr-19-0529] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 11/13/2019] [Accepted: 01/02/2020] [Indexed: 11/16/2022]
Abstract
Fusion genes can be oncogenic drivers in a variety of cancer types and represent potential targets for targeted therapy. The BRAF gene is frequently involved in oncogenic gene fusions, with fusion frequencies of 0.2%-3% throughout different cancers. However, BRAF fusions rarely occur in the same gene configuration, potentially challenging personalized therapy design. In particular, the impact of the wide variety of fusion partners on the oncogenic role of BRAF during tumor growth and drug response is unknown. Here, we used patient-derived colorectal cancer organoids to functionally characterize and cross-compare BRAF fusions containing various partner genes (AGAP3, DLG1, and TRIM24) with respect to cellular behavior, downstream signaling activation, and response to targeted therapies. We demonstrate that 5' fusion partners mainly promote canonical oncogenic BRAF activity by replacing the auto-inhibitory N-terminal region. In addition, the 5' partner of BRAF fusions influences their subcellular localization and intracellular signaling capacity, revealing distinct subsets of affected signaling pathways and altered gene expression. Presence of the different BRAF fusions resulted in varying sensitivities to combinatorial inhibition of MEK and the EGF receptor family. However, all BRAF fusions conveyed resistance to targeted monotherapy against the EGF receptor family, suggesting that BRAF fusions should be screened alongside other MAPK pathway alterations to identify patients with metastatic colorectal cancer to exclude from anti-EGFR-targeted treatment. IMPLICATIONS: Although intracellular signaling and sensitivity to targeted therapies of BRAF fusion genes are influenced by their 5' fusion partner, we show that all investigated BRAF fusions confer resistance to clinically relevant EGFR inhibition.
Collapse
Affiliation(s)
- Christina Stangl
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands.,Division of Molecular Oncology, Netherlands Cancer Institute, and Oncode Institute, Amsterdam, the Netherlands
| | - Jasmin B Post
- Molecular Cancer Research, Center for Molecular Medicine, and Oncode Institute, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Markus J van Roosmalen
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands.,Princess Máxima Center for Pediatric Oncology and Oncode Institute, Utrecht, the Netherlands
| | - Nizar Hami
- Molecular Cancer Research, Center for Molecular Medicine, and Oncode Institute, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Ingrid Verlaan-Klink
- Molecular Cancer Research, Center for Molecular Medicine, and Oncode Institute, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Harmjan R Vos
- Molecular Cancer Research, Center for Molecular Medicine, and Oncode Institute, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Robert M van Es
- Molecular Cancer Research, Center for Molecular Medicine, and Oncode Institute, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Marco J Koudijs
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands.,Center for Personalized Cancer Treatment, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Emile E Voest
- Division of Molecular Oncology, Netherlands Cancer Institute, and Oncode Institute, Amsterdam, the Netherlands.
| | - Hugo J G Snippert
- Molecular Cancer Research, Center for Molecular Medicine, and Oncode Institute, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - W P Kloosterman
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands. .,Cyclomics, Utrecht, the Netherlands.,Frame Cancer Therapeutics, Amsterdam, the Netherlands
| |
Collapse
|
34
|
Modulation of the MAPKs pathways affects Aβ-induced cognitive deficits in Alzheimer's disease via activation of α7nAChR. Neurobiol Learn Mem 2020; 168:107154. [PMID: 31904546 DOI: 10.1016/j.nlm.2019.107154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 11/17/2019] [Accepted: 12/31/2019] [Indexed: 01/21/2023]
Abstract
Cognitive impairment in Alzheimer's disease (AD) is characterized by being deficient at learning and memory. Aβ1-42 oligomers have been shown to impair rodent cognitive function. We previously demonstrated that activation of α7nAChR, inhibition of p38 or JNK could alleviate Aβ-induced memory deficits in Y maze test. In this study, we investigated whether the effects of α7nAChR and MAPKs on Y maze test is reproducible with a hippocampus-dependent spatial memory test such as Morris water maze. We also assessed the possible co-existence of hippocampus-independent recognition memory dysfunction using a novel object recognition test and an alternative and stress free hippocampus-dependent recognition memory test such as the novel place recognition. Besides, previous research from our lab has shown that MAPKs pathways regulate Aβ internalization through mediating α7nAChR. In our study, whether MAPKs pathways exert their functions in cognition by modulating α7nAChR through regulating glutamate receptors and synaptic protein, remain little known. Our results showed that activation of α7nAChR restored spatial memory, novel place recognition memory, and short-term and long-term memory in novel object recognition. Inhibition of p38 restored spatial memory and short-term and long-term memory in novel object recognition. Inhibition of ERK restored short-term memory in novel object recognition and novel place recognition memory. Inhibition of JNK restored spatial memory, short-term memory in novel object recognition and novel place recognition memory. Beside this, the activation of α7nAChR, inhibition of p38 or JNK restored Aβ-induced levels of NMDAR1, NMDAR2A, NMDAR2B, GluR1, GluR2 and PSD95 in Aβ-injected mice without influencing synapsin 1. In addition, these treatments also recovered the expression of acetylcholinesterase (AChE). Finally, we found that the inhibition of p38 or JNK resulted in the upregulation of α7nAChR mRNA levels in the hippocampus. Our results indicated that inhibition of p38 or JNK MAPKs could alleviate Aβ-induced spatial memory deficits through regulating activation of α7nAChR via recovering memory-related proteins. Moreover, p38, ERK and JNK MAPKs exert different functions in spatial and recognition memory.
Collapse
|
35
|
Lin L, Lyu Q, Kwan PY, Zhao J, Fan R, Chai A, Lai CSW, Chan YS, Shen X, Lai KO. The epilepsy and intellectual disability-associated protein TBC1D24 regulates the maintenance of excitatory synapses and animal behaviors. PLoS Genet 2020; 16:e1008587. [PMID: 32004315 PMCID: PMC7015432 DOI: 10.1371/journal.pgen.1008587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 02/12/2020] [Accepted: 12/29/2019] [Indexed: 12/27/2022] Open
Abstract
Perturbation of synapse development underlies many inherited neurodevelopmental disorders including intellectual disability (ID). Diverse mutations on the human TBC1D24 gene are strongly associated with epilepsy and ID. However, the physiological function of TBC1D24 in the brain is not well understood, and there is a lack of genetic mouse model that mimics TBC1D24 loss-of-function for the study of animal behaviors. Here we report that TBC1D24 is present at the postsynaptic sites of excitatory synapses, where it is required for the maintenance of dendritic spines through inhibition of the small GTPase ARF6. Mice subjected to viral-mediated knockdown of TBC1D24 in the adult hippocampus display dendritic spine loss, deficits in contextual fear memory, as well as abnormal behaviors including hyperactivity and increased anxiety. Interestingly, we show that the protein stability of TBC1D24 is diminished by the disease-associated missense mutation that leads to F251L amino acid substitution. We further generate the F251L knock-in mice, and the homozygous mutants show increased neuronal excitability, spontaneous seizure and pre-mature death. Moreover, the heterozygous F251L knock-in mice survive into adulthood but display dendritic spine defects and impaired memory. Our findings therefore uncover a previously uncharacterized postsynaptic function of TBC1D24, and suggest that impaired dendritic spine maintenance contributes to the pathophysiology of individuals harboring TBC1D24 gene mutations. The F251L knock-in mice represent a useful animal model for investigation of the mechanistic link between TBC1D24 loss-of-function and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lianfeng Lin
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Quanwei Lyu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Pui-Yi Kwan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Junjun Zhao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Ruolin Fan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Anping Chai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Ying-Shing Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Xuting Shen
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Kwok-On Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
36
|
Petersen A, Brown JC, Gerges NZ. BRAG1/IQSEC2 as a regulator of small GTPase-dependent trafficking. Small GTPases 2020; 11:1-7. [PMID: 29363391 PMCID: PMC6959296 DOI: 10.1080/21541248.2017.1361898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 10/18/2022] Open
Abstract
Precise trafficking events, such as those that underlie synaptic transmission and plasticity, require complex regulation. G-protein signaling plays an essential role in the regulation of membrane and protein trafficking. However, it is not well understood how small GTPases and their regulatory proteins coordinate such specific events. Our recent publication focused on a highly abundant synaptic GEF, BRAG1, whose physiologic relevance was unknown. We find that BRAG1s GEF activity is required for activity-dependent trafficking of AMPARs. Moreover, BRAG1 bidirectionally regulates synaptic transmission in a manner independent of this activity. In addition to the GEF domain, BRAG1 contains several functional domains whose roles are not yet understood but may mediate protein-protein interactions and regulatory effects necessary for its role in regulation of AMPAR trafficking. In this commentary, we explore the potential for BRAG1 to provide specificity of small GTPase signaling, coordinating activity-dependent activation of small GTPase activity with signaling and scaffolding molecules involved in trafficking through its GEF activity and other functional domains.
Collapse
Affiliation(s)
- Amber Petersen
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Joshua C. Brown
- Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, USA
| | - Nashaat Z. Gerges
- Department of Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Biopharmaceutical Sciences, School of Pharmacy, The Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
37
|
Roth RH, Cudmore RH, Tan HL, Hong I, Zhang Y, Huganir RL. Cortical Synaptic AMPA Receptor Plasticity during Motor Learning. Neuron 2019; 105:895-908.e5. [PMID: 31901303 DOI: 10.1016/j.neuron.2019.12.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 10/30/2019] [Accepted: 12/04/2019] [Indexed: 02/04/2023]
Abstract
Modulation of synaptic strength through trafficking of AMPA receptors (AMPARs) is a fundamental mechanism underlying synaptic plasticity, learning, and memory. However, the dynamics of AMPAR trafficking in vivo and its correlation with learning have not been resolved. Here, we used in vivo two-photon microscopy to visualize surface AMPARs in mouse cortex during the acquisition of a forelimb reaching task. Daily training leads to an increase in AMPAR levels at a subset of spatially clustered dendritic spines in the motor cortex. Surprisingly, we also observed increases in spine AMPAR levels in the visual cortex. There, synaptic potentiation depends on the availability of visual input during motor training, and optogenetic inhibition of visual cortex activity impairs task performance. These results indicate that motor learning induces widespread cortical synaptic potentiation by increasing the net trafficking of AMPARs into spines, including in non-motor brain regions.
Collapse
Affiliation(s)
- Richard H Roth
- Solomon H. Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Robert H Cudmore
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA 95616, USA
| | - Han L Tan
- Solomon H. Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- Solomon H. Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yong Zhang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission of the P.R. China, IDG/McGovern Institute for Brain Research at PKU, Peking University, Beijing 100191, China
| | - Richard L Huganir
- Solomon H. Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
38
|
Critical role of CRAG, a splicing variant of centaurin-γ3/AGAP3, in ELK1-dependent SRF activation at PML bodies. Sci Rep 2019; 9:20107. [PMID: 31882856 PMCID: PMC6934726 DOI: 10.1038/s41598-019-56559-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 12/10/2019] [Indexed: 11/08/2022] Open
Abstract
CRMP-5-associated GTPase (CRAG), a short splicing variant of centaurin-γ3/AGAP3, is predominantly expressed in the developing brain. We previously demonstrated that CRAG, but not centaurin-γ3, translocates to the nucleus and activates the serum response factor (SRF)-c-Fos pathway in cultured neuronal cells. However, the physiological relevance of CRAG in vivo is unknown. Here, we found that CRAG/centaurin-γ3-knockout mice showed intensively suppressed kainic acid-induced c-fos expression in the hippocampus. Analyses of molecular mechanisms underlying CRAG-mediated SRF activation revealed that CRAG has an essential role in GTPase activity, interacts with ELK1 (a co-activator of SRF), and activates SRF in an ELK1-dependent manner. Furthermore, CRAG and ELK1 interact with promyelocytic leukaemia bodies through SUMO-interacting motifs, which is required for SRF activation. These results suggest that CRAG plays a critical role in ELK1-dependent SRF-c-fos activation at promyelocytic leukaemia bodies in the developing brain.
Collapse
|
39
|
Zeng HL, Yu FL, Zhang Z, Yang Q, Jin S, He X, Chen X, Shen Y, Cheng L, Guo L, Xu F. Quantitative proteomics study of host response to virulent and attenuated pseudorabies virus infection in mouse brain. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:307-315. [DOI: 10.1016/j.bbapap.2017.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/16/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022]
|
40
|
Mao X, Fan C, Yu X, Chen B, Jin F. DDEFL1 correlated with Rho GTPases activity in breast cancer. Oncotarget 2017; 8:112487-112497. [PMID: 29348842 PMCID: PMC5762527 DOI: 10.18632/oncotarget.22095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/22/2017] [Indexed: 01/13/2023] Open
Abstract
DDEFL1 is related to maintaining a limiting amount of ARF6 in GTP-loaded form by accelerating its GTP hydrolysis activity, which has been implicated in hepatocellular cancer pathogenesis and lung cancer development. We investigated DDEFL1 expression in breast cancer and paired normal breast tissues by immunohistochemistry and found that DDEFL1 expression was significantly associated with tumor size, lymph node metastasis, high content of elastosis and TNM stage but not with menopausal status or age. We detected the mRNA and protein expression of DDEFL1 in breast cancer cell lines by Western blotting and quantitative real-time PCR (qRT-PCR). DDEFL1 was obvious in MDA-MB-435s and MDA-MB-231 but very weak in ZR-75-1. Further experiments were conducted to evaluate the effect of DDEFL1 small interfering RNA (siRNA) transfection on the biological behavior of MDA-MB-231. After transfection, the effects of DDEFL1 inhibition on expression of mRNA and protein were also analyzed by Western blotting and qRT-PCR. Increased apoptosis and invasive ability, decreased cellular proliferation was found in MDA-MB-231 with successful DDEFL1 siRNA transient transfection (p < 0.05). Western blotting and qRT-PCR results showed that the DDEFL1 inhibition up-regulated Caspase-3, Apaf-1, cytochrome c, and Bax expression and down-regulated Bcl-2 expression. The DDEFL1 inhibition also down-regulated the mRNA and protein expression of Rho, CDC42 and Rac1. Our study provided a functional linkage through DDEFL1 with breast cancer biological behaviours by Rho GTPases. Possible implication of our main finding for the DDEFL1 role in breast cancer and the downstream signaling pathways for the treatment of breast cancer.
Collapse
Affiliation(s)
- Xiaoyun Mao
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Chuifeng Fan
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, China
| | - Xinmiao Yu
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Bo Chen
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Feng Jin
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| |
Collapse
|
41
|
Nakayama K, Ohashi R, Shinoda Y, Yamazaki M, Abe M, Fujikawa A, Shigenobu S, Futatsugi A, Noda M, Mikoshiba K, Furuichi T, Sakimura K, Shiina N. RNG105/caprin1, an RNA granule protein for dendritic mRNA localization, is essential for long-term memory formation. eLife 2017; 6. [PMID: 29157358 PMCID: PMC5697933 DOI: 10.7554/elife.29677] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/22/2017] [Indexed: 12/18/2022] Open
Abstract
Local regulation of synaptic efficacy is thought to be important for proper networking of neurons and memory formation. Dysregulation of global translation influences long-term memory in mice, but the relevance of the regulation specific for local translation by RNA granules remains elusive. Here, we demonstrate roles of RNG105/caprin1 in long-term memory formation. RNG105 deletion in mice impaired synaptic strength and structural plasticity in hippocampal neurons. Furthermore, RNG105-deficient mice displayed unprecedentedly severe defects in long-term memory formation in spatial and contextual learning tasks. Genome-wide profiling of mRNA distribution in the hippocampus revealed an underlying mechanism: RNG105 deficiency impaired the asymmetric somato-dendritic localization of mRNAs. Particularly, RNG105 deficiency reduced the dendritic localization of mRNAs encoding regulators of AMPAR surface expression, which was consistent with attenuated homeostatic AMPAR scaling in dendrites and reduced synaptic strength. Thus, RNG105 has an essential role, as a key regulator of dendritic mRNA localization, in long-term memory formation. Messages pass from one nerve cell to the next across gaps called synapses. The first neuron releases chemical signals from the end of its long, thin nerve fiber. The second receives the message at receptors on branching structures known as dendrites. Each connection has a corresponding bump called a dendritic spine. As animals learn, these can grow larger, strengthening the connection. This is the basis of how memories form. To strengthen a synapse, the cell must transport the materials to the dendritic spine. The cell makes copies of the genetic instructions to strengthen the synapse in the form of messenger RNA (often shortened to mRNA). But, this happens in the body of the cell, a long way from the dendrites themselves. The mRNA travels from the cell body to the dendrites in collections of molecules referred to as ‘RNA granules’. One of the key components of the RNA granule system is a protein called RNG105/caprin1. Now, Nakayama, Ohashi et al. have engineered mice to delete the gene for RNG105/caprin1, revealing its effect on memory. Mice lacking RNG105/caprin1 struggled to make long-term memories. Unlike their normal counterparts, these mutant mice did not become accustomed to new environments or objects. They also found it more challenging to learn the position of a hidden platform in a water-based maze. Lastly, over time, the mutant mice forgot to be fearful of a dark chamber where they had received a small electric shock. Memories form in a part of the brain called the hippocampus and the dendritic spines in this region were smaller in mice lacking RNG105/caprin1. Furthermore, when the nerve cells from this part of the brain were grown in Petri dishes, they did not respond normally to stimulation. The dendritic spines of normal cells increased in size, but those on the cells lacking RNG105/caprin1 got smaller compared to normal cells. A closer look revealed that the distribution of mRNA in brain cells from mice lacking RNG105/caprin1 differed from that of normal mice. Some pieces of genetic information failed to make it from the cell body to the dendrites. This included mRNA involved in making regulators of a component of dendritic spines called the AMPA receptor. The AMPA receptor detects the chemical messenger, glutamate, and is crucial for memory formation. These findings further our understanding of long-term memory and open the way for future research into human disease. Mutations in RNA granule components, including RNG105/caprin1, have links to conditions such as amyotrophic lateral sclerosis (ALS) and autism spectrum disorder (ASD). Further investigation could reveal new targets for drug treatment.
Collapse
Affiliation(s)
- Kei Nakayama
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Japan.,Department of Basic Biology, SOKENDAI, Okazaki, Japan.,Okazaki Institute for Integrative Bioscience, Okazaki, Japan
| | - Rie Ohashi
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Japan.,Department of Basic Biology, SOKENDAI, Okazaki, Japan
| | - Yo Shinoda
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan.,School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akihiro Fujikawa
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Japan
| | - Shuji Shigenobu
- Department of Basic Biology, SOKENDAI, Okazaki, Japan.,Functional Genomics Facility, National Institute for Basic Biology, Okazaki, Japan
| | - Akira Futatsugi
- Department of Basic Medical Science, Kobe City College of Nursing, Hyogo, Japan
| | - Masaharu Noda
- Department of Basic Biology, SOKENDAI, Okazaki, Japan.,Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, Brain Science Institute, Wako, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Nobuyuki Shiina
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Japan.,Department of Basic Biology, SOKENDAI, Okazaki, Japan.,Okazaki Institute for Integrative Bioscience, Okazaki, Japan
| |
Collapse
|
42
|
The functions of Reelin in membrane trafficking and cytoskeletal dynamics: implications for neuronal migration, polarization and differentiation. Biochem J 2017; 474:3137-3165. [PMID: 28887403 DOI: 10.1042/bcj20160628] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023]
Abstract
Reelin is a large extracellular matrix protein with relevant roles in mammalian central nervous system including neurogenesis, neuronal polarization and migration during development; and synaptic plasticity with its implications in learning and memory, in the adult. Dysfunctions in reelin signaling are associated with brain lamination defects such as lissencephaly, but also with neuropsychiatric diseases like autism, schizophrenia and depression as well with neurodegeneration. Reelin signaling involves a core pathway that activates upon reelin binding to its receptors, particularly ApoER2 (apolipoprotein E receptor 2)/LRP8 (low-density lipoprotein receptor-related protein 8) and very low-density lipoprotein receptor, followed by Src/Fyn-mediated phosphorylation of the adaptor protein Dab1 (Disabled-1). Phosphorylated Dab1 (pDab1) is a hub in the signaling cascade, from which several other downstream pathways diverge reflecting the different roles of reelin. Many of these pathways affect the dynamics of the actin and microtubular cytoskeleton, as well as membrane trafficking through the regulation of the activity of small GTPases, including the Rho and Rap families and molecules involved in cell polarity. The complexity of reelin functions is reflected by the fact that, even now, the precise mode of action of this signaling cascade in vivo at the cellular and molecular levels remains unclear. This review addresses and discusses in detail the participation of reelin in the processes underlying neurogenesis, neuronal migration in the cerebral cortex and the hippocampus; and the polarization, differentiation and maturation processes that neurons experiment in order to be functional in the adult brain. In vivo and in vitro evidence is presented in order to facilitate a better understanding of this fascinating system.
Collapse
|
43
|
Kulkarni A, Al-Hraishawi H, Simhadri S, Hirshfield KM, Chen S, Pine S, Jeyamohan C, Sokol L, Ali S, Teo ML, White E, Rodriguez-Rodriguez L, Mehnert JM, Ganesan S. BRAF Fusion as a Novel Mechanism of Acquired Resistance to Vemurafenib in BRAFV600E Mutant Melanoma. Clin Cancer Res 2017; 23:5631-5638. [PMID: 28539463 DOI: 10.1158/1078-0432.ccr-16-0758] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/11/2017] [Accepted: 05/16/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Many patients with BRAFV600E mutant melanoma treated with BRAF inhibitors experience a rapid response, but ultimately develop resistance. Insight into the mechanism of resistance is critical for development of more effective treatment strategies.Experimental Design: Comprehensive genomic profiling of serial biopsies was performed in a patient with a BRAFV600E mutant metastatic melanoma who developed resistance to vemurafenib. An AGAP3-BRAF fusion gene, identified in the vemurafenib-resistant tumor, was expressed in BRAFV600E melanoma cell lines, and its effect on drug sensitivity was evaluated.Results: Clinical resistance to vemurafenib in a melanoma harboring a BRAFV600E mutation was associated with acquisition of an AGAP3-BRAF fusion gene. Expression of the AGAP3-BRAF fusion in BRAFV600E mutant melanoma cells induced vemurafenib resistance; however, these cells remained relatively sensitive to MEK inhibitors. The patient experienced clinical benefit following treatment with the combination of a BRAF and a MEK inhibitor. Rebiopsy of the tumor at a later time point, after BRAF and MEK inhibitors had been discontinued, showed loss of the AGAP3-BRAF fusion gene. Mixing experiments suggest that cells harboring both BRAFV600E and AGAP3-BRAF only have a fitness advantage over parental BRAFV600E cells during active treatment with a BRAF inhibitor.Conclusions: We report acquisition of a BRAF fusion as a novel mechanism of acquired resistance to vemurafenib in a patient with melanoma harboring a BRAFV600E mutation. The acquisition and regression of clones harboring this fusion during the presence and absence of a BRAF inhibitor are consistent with rapidly evolving clonal dynamics in melanoma. Clin Cancer Res; 23(18); 5631-8. ©2017 AACR.
Collapse
Affiliation(s)
- Atul Kulkarni
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | | | - Srilatha Simhadri
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Kim M Hirshfield
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Suzie Chen
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey.,Rutgers Ernest Mario School of Pharmacy, Piscataway Township, New Jersey
| | - Sharon Pine
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | | | - Levi Sokol
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Siraj Ali
- Foundation Medicine, Inc. Cambridge, Massachusetts
| | - Man Lung Teo
- Central Comprehensive Cancer Centre, Central District, Hong Kong
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Lorna Rodriguez-Rodriguez
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department Obstetrics/Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Janice M Mehnert
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey. .,Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey.,Developmental Therapeutics/Phase I Program, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey. .,Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
44
|
Guénette S, Strecker P, Kins S. APP Protein Family Signaling at the Synapse: Insights from Intracellular APP-Binding Proteins. Front Mol Neurosci 2017; 10:87. [PMID: 28424586 PMCID: PMC5371672 DOI: 10.3389/fnmol.2017.00087] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/13/2017] [Indexed: 12/17/2022] Open
Abstract
Understanding the molecular mechanisms underlying amyloid precursor protein family (APP/APP-like proteins, APLP) function in the nervous system can be achieved by studying the APP/APLP interactome. In this review article, we focused on intracellular APP interacting proteins that bind the YENPTY internalization motif located in the last 15 amino acids of the C-terminal region. These proteins, which include X11/Munc-18-interacting proteins (Mints) and FE65/FE65Ls, represent APP cytosolic binding partners exhibiting different neuronal functions. A comparison of FE65 and APP family member mutant mice revealed a shared function for APP/FE65 protein family members in neurogenesis and neuronal positioning. Accumulating evidence also supports a role for membrane-associated APP/APLP proteins in synapse formation and function. Therefore, it is tempting to speculate that APP/APLP C-terminal interacting proteins transmit APP/APLP-dependent signals at the synapse. Herein, we compare our current knowledge of the synaptic phenotypes of APP/APLP mutant mice with those of mice lacking different APP/APLP interaction partners and discuss the possible downstream effects of APP-dependent FE65/FE65L or X11/Mint signaling on synaptic vesicle release, synaptic morphology and function. Given that the role of X11/Mint proteins at the synapse is well-established, we propose a model highlighting the role of FE65 protein family members for transduction of APP/APLP physiological function at the synapse.
Collapse
Affiliation(s)
| | - Paul Strecker
- Department of Biology, Division of Human Biology, University of KaiserslauternKaiserslautern, Germany
| | - Stefan Kins
- Department of Biology, Division of Human Biology, University of KaiserslauternKaiserslautern, Germany
| |
Collapse
|
45
|
Fukaya M, Ohta S, Hara Y, Tamaki H, Sakagami H. Distinct subcellular localization of alternative splicing variants of EFA6D, a guanine nucleotide exchange factor for Arf6, in the mouse brain. J Comp Neurol 2016; 524:2531-52. [PMID: 27241101 DOI: 10.1002/cne.24048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/31/2016] [Accepted: 05/24/2016] [Indexed: 11/07/2022]
Abstract
EFA6D (guanine nucleotide exchange factor for ADP-ribosylation factor 6 [Arf6]D) is also known as EFA6R, Psd3, and HCA67. It is the fourth member of the EFA6 family with guanine nucleotide exchange activity for Arf6, a small guanosine triphosphatase (GTPase) that regulates endosomal trafficking and actin cytoskeleton remodeling. We propose a classification and nomenclature of 10 EFA6D variants deposited in the GenBank database as EFA6D1a, 1b, 1c, 1d, 1s, 2a, 2b, 2c, 2d, and 2s based on the combination of N-terminal and C-terminal insertions. Polymerase chain reaction analysis showed the expression of all EFA6D variants except for variants a and d in the adult mouse brain. Immunoblotting analysis with novel variant-specific antibodies showed the endogenous expression of EFA6D1b, EFA6D1c, and EFA6D1s at the protein level, with the highest expression being EFA6D1s, in the brain. Immunoblotting analysis of forebrain subcellular fractions showed the distinct subcellular distribution of EFA6D1b/c and EFA6D1s. The immunohistochemical analysis revealed distinct but overlapping immunoreactive patterns between EFA6D1b/c and EFA6D1s in the mouse brain. In immunoelectron microscopic analyses of the hippocampal CA3 region, EFA6D1b/c was present predominantly in the mossy fiber axons of dentate granule cells, whereas EFA6D1s was present abundantly in the cell bodies, dendritic shafts, and spines of hippocampal pyramidal cells. These results provide the first anatomical evidence suggesting the functional diversity of EFA6D variants, particularly EFA6D1b/c and EFA6D1s, in neurons. J. Comp. Neurol. 524:2531-2552, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Shingo Ohta
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshinobu Hara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hideaki Tamaki
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
46
|
Tracy TE, Sohn PD, Minami SS, Wang C, Min SW, Li Y, Zhou Y, Le D, Lo I, Ponnusamy R, Cong X, Schilling B, Ellerby LM, Huganir RL, Gan L. Acetylated Tau Obstructs KIBRA-Mediated Signaling in Synaptic Plasticity and Promotes Tauopathy-Related Memory Loss. Neuron 2016; 90:245-60. [PMID: 27041503 DOI: 10.1016/j.neuron.2016.03.005] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 01/17/2016] [Accepted: 02/22/2016] [Indexed: 11/27/2022]
Abstract
Tau toxicity has been implicated in the emergence of synaptic dysfunction in Alzheimer's disease (AD), but the mechanism by which tau alters synapse physiology and leads to cognitive decline is unclear. Here we report abnormal acetylation of K274 and K281 on tau, identified in AD brains, promotes memory loss and disrupts synaptic plasticity by reducing postsynaptic KIdney/BRAin (KIBRA) protein, a memory-associated protein. Transgenic mice expressing human tau with lysine-to-glutamine mutations to mimic K274 and K281 acetylation (tauKQ) exhibit AD-related memory deficits and impaired hippocampal long-term potentiation (LTP). TauKQ reduces synaptic KIBRA levels and disrupts activity-induced postsynaptic actin remodeling and AMPA receptor insertion. The LTP deficit was rescued by promoting actin polymerization or by KIBRA expression. In AD patients with dementia, we found enhanced tau acetylation is linked to loss of KIBRA. These findings suggest a novel mechanism by which pathogenic tau causes synaptic dysfunction and cognitive decline in AD pathogenesis.
Collapse
Affiliation(s)
- Tara E Tracy
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Peter Dongmin Sohn
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 91458, USA
| | - S Sakura Minami
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Sang-Won Min
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Yaqiao Li
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | - David Le
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | - Iris Lo
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | | | - Xin Cong
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Lisa M Ellerby
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 91458, USA.
| |
Collapse
|
47
|
Elagabani MN, Briševac D, Kintscher M, Pohle J, Köhr G, Schmitz D, Kornau HC. Subunit-selective N-Methyl-d-aspartate (NMDA) Receptor Signaling through Brefeldin A-resistant Arf Guanine Nucleotide Exchange Factors BRAG1 and BRAG2 during Synapse Maturation. J Biol Chem 2016; 291:9105-18. [PMID: 26884337 DOI: 10.1074/jbc.m115.691717] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Indexed: 11/06/2022] Open
Abstract
The maturation of glutamatergic synapses in the CNS is regulated by NMDA receptors (NMDARs) that gradually change from a GluN2B- to a GluN2A-dominated subunit composition during postnatal development. Here we show that NMDARs control the activity of the small GTPase ADP-ribosylation factor 6 (Arf6) by consecutively recruiting two related brefeldin A-resistant Arf guanine nucleotide exchange factors, BRAG1 and BRAG2, in a GluN2 subunit-dependent manner. In young cortical cultures, GluN2B and BRAG1 tonically activated Arf6. In mature cultures, Arf6 was activated through GluN2A and BRAG2 upon NMDA treatment, whereas the tonic Arf6 activation was not detectable any longer. This shift in Arf6 regulation and the associated drop in Arf6 activity were reversed by a knockdown of BRAG2. Given their sequential recruitment during development, we examined whether BRAG1 and BRAG2 influence synaptic currents in hippocampal CA1 pyramidal neurons using patch clamp recordings in acute slices from mice at different ages. The number of AMPA receptor (AMPAR) miniature events was reduced by depletion of BRAG1 but not by depletion of BRAG2 during the first 2 weeks after birth. In contrast, depletion of BRAG2 during postnatal weeks 4 and 5 reduced the number of AMPAR miniature events and compromised the quantal sizes of both AMPAR and NMDAR currents evoked at Schaffer collateral synapses. We conclude that both Arf6 activation through GluN2B-BRAG1 during early development and the transition from BRAG1- to BRAG2-dependent Arf6 signaling induced by the GluN2 subunit switch are critical for the development of mature glutamatergic synapses.
Collapse
Affiliation(s)
- Mohammad Nael Elagabani
- From the Neuroscience Research Center (NWFZ) and Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and
| | - Dušica Briševac
- From the Neuroscience Research Center (NWFZ) and Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and
| | | | - Jörg Pohle
- the Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Georg Köhr
- the Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | | | - Hans-Christian Kornau
- From the Neuroscience Research Center (NWFZ) and Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and
| |
Collapse
|
48
|
Tagliatti E, Fadda M, Falace A, Benfenati F, Fassio A. Arf6 regulates the cycling and the readily releasable pool of synaptic vesicles at hippocampal synapse. eLife 2016; 5. [PMID: 26731518 PMCID: PMC4764570 DOI: 10.7554/elife.10116] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 01/04/2016] [Indexed: 01/29/2023] Open
Abstract
Recycling of synaptic vesicles (SVs) is a fundamental step in the process of neurotransmission. Endocytosed SV can travel directly into the recycling pool or recycle through endosomes but little is known about the molecular actors regulating the switch between these SV recycling routes. ADP ribosylation factor 6 (Arf6) is a small GTPase known to participate in constitutive trafficking between plasma membrane and early endosomes. Here, we have morphologically and functionally investigated Arf6-silenced hippocampal synapses and found an activity dependent accumulation of synaptic endosome-like organelles and increased release-competent docked SVs. These features were phenocopied by pharmacological blockage of Arf6 activation. The data reveal an unexpected role for this small GTPase in reducing the size of the readily releasable pool of SVs and in channeling retrieved SVs toward direct recycling rather than endosomal sorting. We propose that Arf6 acts at the presynapse to define the fate of an endocytosed SV. DOI:http://dx.doi.org/10.7554/eLife.10116.001 Communication between neurons takes place at cell-to-cell contacts called synapses. Each synapse is formed between one neuron that sends the message, and another neuron that receives it. The neuron before the synapse – called the presynaptic neuron – contains packets called synaptic vesicles, which are full of chemical messengers ready to be released upon activity. Accurate communication between neurons relies on the exact composition, and organized trafficking, of the synaptic vesicles when the neuron is active. Synapses also contain bigger structures, called endosomal structures, which may represent an intermediate station in which synaptic vesicle composition is controlled. However, the trafficking of synaptic vesicles through the endosomal structures is poorly understood. Now, Tagliatti, Fadda et al. have revealed that a protein called Arf6 plays an important role in presynaptic neurons. The experiments involved rat neurons grown in the laboratory, and showed that Arf6 controls both the number of synaptic vesicles ready to be released and the trafficking of synaptic vesicles via endosomal structures in active neurons. The next step following on from these findings is to understand how Arf6 exerts its effects and how this protein is regulated in the presynaptic neuron. DOI:http://dx.doi.org/10.7554/eLife.10116.002
Collapse
Affiliation(s)
- Erica Tagliatti
- Center of Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Manuela Fadda
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Antonio Falace
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Fabio Benfenati
- Center of Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Anna Fassio
- Center of Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
49
|
Epitope Fingerprinting for Recognition of the Polyclonal Serum Autoantibodies of Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:267989. [PMID: 26417591 PMCID: PMC4568325 DOI: 10.1155/2015/267989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 02/18/2015] [Indexed: 02/06/2023]
Abstract
Autoantibodies (aAb) associated with Alzheimer's disease (AD) have not been sufficiently characterized and their exact involvement is undefined. The use of information technology and computerized analysis with phage display technology was used, in the present research, to map the epitope of putative self-antigens in AD patients. A 12-mer random peptide library, displayed on M13 phages, was screened using IgG from AD patients with two repetitions. Seventy-one peptides were isolated; however, only 10 were positive using the Elisa assay technique (Elisa Index > 1). The results showed that the epitope regions of the immunoreactive peptides, identified by phage display analysis, were on the exposed surfaces of the proteins. The putative antigens MAST1, Enah, MAO-A, X11/MINT1, HGF, SNX14, ARHGAP 11A, APC, and CENTG3, which have been associated with AD or have functions in neural tissue, may indicate possible therapeutic targets.
Collapse
|
50
|
Abstract
Integrins are adhesion and survival molecules involved in axon growth during CNS development, as well as axon regeneration after injury in the peripheral nervous system (PNS). Adult CNS axons do not regenerate after injury, partly due to a low intrinsic growth capacity. We have previously studied the role of integrins in axon growth in PNS axons; in the present study, we investigate whether integrin mechanisms involved in PNS regeneration may be altered or lacking from mature CNS axons by studying maturing CNS neurons in vitro. In rat cortical neurons, we find that integrins are present in axons during initial growth but later become restricted to the somato-dendritic domain. We investigated how this occurs and whether it can be altered to enhance axonal growth potential. We find a developmental change in integrin trafficking; transport becomes predominantly retrograde throughout axons, but not dendrites, as neurons mature. The directionality of transport is controlled through the activation state of ARF6, with developmental upregulation of the ARF6 GEF ARNO enhancing retrograde transport. Lowering ARF6 activity in mature neurons restores anterograde integrin flow, allows transport into axons, and increases axon growth. In addition, we found that the axon initial segment is partly responsible for exclusion of integrins and removal of this structure allows integrins into axons. Changing posttranslational modifications of tubulin with taxol also allows integrins into the proximal axon. The experiments suggest that the developmental loss of regenerative ability in CNS axons is due to exclusion of growth-related molecules due to changes in trafficking.
Collapse
|