1
|
Tabuena DR, Jang SS, Grone B, Yip O, Aery Jones EA, Blumenfeld J, Liang Z, Koutsodendris N, Rao A, Ding L, Zhang AR, Hao Y, Xu Q, Yoon SY, Leon SD, Huang Y, Zilberter M. Neuronal APOE4-induced Early Hippocampal Network Hyperexcitability in Alzheimer's Disease Pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.28.555153. [PMID: 37693533 PMCID: PMC10491126 DOI: 10.1101/2023.08.28.555153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The full impact of apolipoprotein E4 (APOE4), the strongest genetic risk factor for Alzheimer's disease (AD), on neuronal and network function remains unclear. We found hippocampal region-specific network hyperexcitability in young APOE4 knock-in (E4-KI) mice which predicted cognitive deficits at old age. Network hyperexcitability in young E4-KI mice was mediated by hippocampal region-specific subpopulations of smaller and hyperexcitable neurons that were eliminated by selective removal of neuronal APOE4. Aged E4-KI mice exhibited hyperexcitable granule cells, a progressive inhibitory deficit, and E/I imbalance in the dentate gyrus, exacerbating hippocampal hyperexcitability. Single-nucleus RNA-sequencing revealed neuronal cell type-specific and age-dependent transcriptomic changes, including Nell2 overexpression in E4-KI mice. Reducing Nell2 expression in specific neuronal types of E4-KI mice with CRISPRi rescued their abnormal excitability phenotypes, implicating Nell2 overexpression as a cause of APOE4-induced hyperexcitability. These findings highlight the early transcriptomic and electrophysiological alterations underlying APOE4-induced hippocampal network dysfunction and its contribution to AD pathogenesis with aging.
Collapse
|
2
|
Genetic Mapping of Behavioral Traits Using the Collaborative Cross Resource. Int J Mol Sci 2022; 24:ijms24010682. [PMID: 36614124 PMCID: PMC9821145 DOI: 10.3390/ijms24010682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
The complicated interactions between genetic background, environment and lifestyle factors make it difficult to study the genetic basis of complex phenotypes, such as cognition and anxiety levels, in humans. However, environmental and other factors can be tightly controlled in mouse studies. The Collaborative Cross (CC) is a mouse genetic reference population whose common genetic and phenotypic diversity is on par with that of humans. Therefore, we leveraged the power of the CC to assess 52 behavioral measures associated with locomotor activity, anxiety level, learning and memory. This is the first application of the CC in novel object recognition tests, Morris water maze tasks, and fear conditioning tests. We found substantial continuous behavioral variations across the CC strains tested, and mapped six quantitative trait loci (QTLs) which influenced these traits, defining candidate genetic variants underlying these QTLs. Overall, our findings highlight the potential of the CC population in behavioral genetic research, while the identified genomic loci and genes driving the variation of relevant behavioral traits provide a foundation for further studies.
Collapse
|
3
|
Chen J, Wu J, Luo Y, Huang N. NELL2 as a potential marker of outcome in the cerebrospinal fluid of patients with tuberculous meningitis: preliminary results from a single-center observational study. Eur J Med Res 2022; 27:281. [PMID: 36494747 PMCID: PMC9733264 DOI: 10.1186/s40001-022-00921-7] [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: 09/22/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To detect the changes in Nel-like 2 (NELL2) in cerebrospinal fluid (CSF) in the outcome of tuberculous meningitis (TBM) patients and to initially evaluate its potential as a marker. METHODS We collected the clinical data of patients with suspected TBM in the First People's Hospital of Zunyi from November 2017 to January 2021 and retained their CSF. According to the selection and exclusion criteria, the TBM group (11 cases) and the control group (18 cases) were obtained. Western blotting (WB) was used to detect the level of NELL2 in the CSF of the two groups, especially the change in NELL2 before and after treatment in TBM patients. RESULTS The level of NELL2 in the TBM group was lower than that in the control group (P < 0.05), and the level of NELL2 showed an increasing trend after anti-tuberculosis treatment in the TBM group. CONCLUSIONS NELL2 in the CSF of TBM patients decreased significantly. Anti-tuberculosis treatment can improve the level of NELL2, which may become one of the potential markers of outcome in the cerebrospinal fluid of patients with tuberculous meningitis.
Collapse
Affiliation(s)
- Jianhua Chen
- grid.452884.7Department of Neurology, Third Affiliated Hospital of Zunyi Medical University, (The First People’s Hospital of Zunyi), Zunyi, 563000 China
| | - Jie Wu
- grid.452884.7Scientific Research Center, Third Affiliated Hospital of Zunyi Medical University, (The First People’s Hospital of Zunyi), Zunyi, 563000 China
| | - Yong Luo
- grid.452884.7Department of Neurology, Third Affiliated Hospital of Zunyi Medical University, (The First People’s Hospital of Zunyi), Zunyi, 563000 China
| | - Nanqu Huang
- grid.452884.7National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University, (The First People’s Hospital of Zunyi), Zunyi, 563000 China
| |
Collapse
|
4
|
Zhang Z, Lai G, Sun L. Basement-Membrane-Related Gene Signature Predicts Prognosis in WHO Grade II/III Gliomas. Genes (Basel) 2022; 13:1810. [PMID: 36292695 PMCID: PMC9602375 DOI: 10.3390/genes13101810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/29/2022] [Accepted: 10/05/2022] [Indexed: 10/17/2023] Open
Abstract
Gliomas that are classified as grade II or grade III lesions by the World Health Organization (WHO) are highly aggressive, and some may develop into glioblastomas within a short period, thus portending the conferral of a poor prognosis for patients. Previous studies have implicated basement membrane (BM)-related genes in glioma development. In this study, we constructed a prognostic model for WHO grade II/III gliomas in accordance with the risk scores of BM-related genes. Differentially expressed genes (DEGs) in the glioma samples relative to normal samples were screened from the GEO database, and five prognostically relevant BM-related genes, including NELL2, UNC5A, TNC, CSPG4, and SMOC1, were selected using Cox regression analyses for the risk score model. The median risk score was calculated, based on which high- and low-risk groups of patients were generated. The clinical information, pathological information, and risk group were combined to establish a prognostic nomogram. Both the nomogram and risk score model performed well in the independent CGGA cohort. Gene set enrichment analysis (GSEA) and immune profile, drug sensitivity, and tumor mutation burden (TMB) analyses were performed in the two risk groups. A significant enrichment of 'Autophagy-other', 'Collecting duct acid secretion', 'Glycosphingolipid biosynthesis-lacto and neolacto series', 'Valine, leucine, and isoleucine degradation', 'Vibrio cholerae infection', and other pathways were observed for patients with high risk. In addition, higher proportions of monocytes and resting CD4 memory T cells were observed in the low- and high-risk groups, respectively. In conclusion, the BM-related gene risk score model can guide the clinical management of WHO grade II and III gliomas.
Collapse
Affiliation(s)
- Zhaogang Zhang
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Guichuan Lai
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Lingling Sun
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| |
Collapse
|
5
|
Ha CM, Kim DH, Lee TH, Kim HR, Choi J, Kim Y, Kang D, Park JW, Ojeda SR, Jeong JK, Lee BJ. Transcriptional Regulatory Role of NELL2 in Preproenkephalin Gene Expression. Mol Cells 2022; 45:537-549. [PMID: 35950455 PMCID: PMC9385569 DOI: 10.14348/molcells.2022.2051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/28/2022] [Accepted: 04/03/2022] [Indexed: 12/23/2022] Open
Abstract
Preproenkephalin (PPE) is a precursor molecule for multiple endogenous opioid peptides Leu-enkephalin (ENK) and Met-ENK, which are involved in a wide variety of modulatory functions in the nervous system. Despite the functional importance of ENK in the brain, the effect of brain-derived factor(s) on PPE expression is unknown. We report the dual effect of neural epidermal growth factor (EGF)-likelike 2 (NELL2) on PPE gene expression. In cultured NIH3T3 cells, transfection of NELL2 expression vectors induced an inhibition of PPE transcription intracellularly, in parallel with downregulation of protein kinase C signaling pathways and extracellular signal-regulated kinase. Interestingly, these phenomena were reversed when synthetic NELL2 was administered extracellularly. The in vivo disruption of NELL2 synthesis resulted in an increase in PPE mRNA level in the rat brain, suggesting that the inhibitory action of intracellular NELL2 predominates the activation effect of extracellular NELL2 on PPE gene expression in the brain. Biochemical and molecular studies with mutant NELL2 structures further demonstrated the critical role of EGF-like repeat domains in NELL2 for regulation of PPE transcription. These are the first results to reveal the spatio-specific role of NELL2 in the homeostatic regulation of PPE gene expression.
Collapse
Affiliation(s)
- Chang Man Ha
- Brain Research Core Facilities and Global Relation Center of Research Strategy Office, Korea Brain Research Institute, Daegu 41068, Korea
| | - Dong Hee Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Tae Hwan Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Han Rae Kim
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Jungil Choi
- Bioenvironmental Science & Technology Division, Korea Institute of Toxicology, Jinju 52834, Korea
| | - Yoonju Kim
- Brain Research Core Facilities and Global Relation Center of Research Strategy Office, Korea Brain Research Institute, Daegu 41068, Korea
| | - Dasol Kang
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Sergio R. Ojeda
- Division of Neuroscience, Oregon National Primate Research Center/Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jin Kwon Jeong
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| |
Collapse
|
6
|
Miyaguchi M, Nakanishi Y, Maturana AD, Mizutani K, Niimi T. Conformational Change of the Hairpin-like-structured Robo2 Ectodomain Allows NELL1/2 Binding. J Mol Biol 2022; 434:167777. [DOI: 10.1016/j.jmb.2022.167777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 10/16/2022]
|
7
|
Zhao J, Wei G, Zhu J, Liu D, Qin B. Expression analysis of nel during zebrafish embryonic development. Gene Expr Patterns 2022; 45:119258. [PMID: 35691514 DOI: 10.1016/j.gep.2022.119258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/05/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022]
Abstract
Nel is a multimeric extracellular glycoprotein which predominantly expressed in the nervous system and play an important role in neural development and functions. There are three nel paralogues included nell2a, nell2b, and nell3 in zebrafish, while systematic expression analysis of the nel family is still lacking. In this study, we performed a phylogenetic analysis on 7 species, in different species the nell2a are highly conserved, as is nell2b. Then, the expression profiles of nell2a, nell2b and nell3 were detected by in situ hybridization in zebrafish embryo, and the result showed that nel genes highly enriched in the central nervous system, but distributed in different regions of the brain. In addition, nell2a is also expressed in the olfactory pit, spinal cord, otic vesicle and retina (ganglion cell layer), nell2b was detected to express in gill arches, olfactory epithelium, olfactory pit, spinal cord, photoreceptor and retina (ganglion cell layer), it should be noted that the expression of nell3 is special, was only detected at 96 hpf in the brain and spinal cord of zebrafish. Overall, our results indicate that nell2a and nell2b genes are expressed in the nervous system and eyes of zebrafish embryo, while nell3 is expressed in different regions in the nervous system. The phylogenetic analysis also shows that nell3 sequences are significantly different from nell2a and nell2b. This study provides new evidence to better understand the role of nel in zebrafish embryo development.
Collapse
Affiliation(s)
- Jinxiang Zhao
- The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Guanyun Wei
- School of Life Science, Nantong Laboratory of Development and Diseases; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jiang Zhu
- The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Dong Liu
- School of Life Science, Nantong Laboratory of Development and Diseases; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| | - Bing Qin
- The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China; Suqian Clinical College of Xuzhou Medical University, Suqian, China.
| |
Collapse
|
8
|
Shaker MR, Kahtan A, Prasad R, Lee JH, Pietrogrande G, Leeson HC, Sun W, Wolvetang EJ, Slonchak A. Neural Epidermal Growth Factor-Like Like Protein 2 Is Expressed in Human Oligodendroglial Cell Types. Front Cell Dev Biol 2022; 10:803061. [PMID: 35265611 PMCID: PMC8899196 DOI: 10.3389/fcell.2022.803061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/06/2022] [Indexed: 01/14/2023] Open
Abstract
Neural epidermal growth factor-like like 2 (NELL2) is a cytoplasmic and secreted glycosylated protein with six epidermal growth factor-like domains. In animal models, NELL2 is predominantly expressed in neural tissues where it regulates neuronal differentiation, polarization, and axon guidance, but little is known about the role of NELL2 in human brain development. In this study, we show that rostral neural stem cells (rNSC) derived from human-induced pluripotent stem cell (hiPSC) exhibit particularly strong NELL2 expression and that NELL2 protein is enriched at the apical side of neural rosettes in hiPSC-derived brain organoids. Following differentiation of human rostral NSC into neurons, NELL2 remains robustly expressed but changes its subcellular localization from >20 small cytoplasmic foci in NSC to one–five large peri-nuclear puncta per neuron. Unexpectedly, we discovered that in human brain organoids, NELL2 is readily detectable in the oligodendroglia and that the number of NELL2 puncta increases as oligodendrocytes mature. Artificial intelligence-based machine learning further predicts a strong association of NELL2 with multiple human white matter diseases, suggesting that NELL2 may possess yet unexplored roles in regulating oligodendrogenesis and/or myelination during human cortical development and maturation.
Collapse
Affiliation(s)
- Mohammed R Shaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Amna Kahtan
- St Cloud Technical & Community College, St Cloud, MN, United States
| | - Renuka Prasad
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Ju-Hyun Lee
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Giovanni Pietrogrande
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Hannah C Leeson
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Andrii Slonchak
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
9
|
Wilmerding A, Bouteille L, Rinaldi L, Caruso N, Graba Y, Delfini MC. HOXB8 Counteracts MAPK/ERK Oncogenic Signaling in a Chicken Embryo Model of Neoplasia. Int J Mol Sci 2021; 22:8911. [PMID: 34445617 PMCID: PMC8396257 DOI: 10.3390/ijms22168911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/18/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
HOX transcription factors are members of an evolutionarily conserved family of proteins required for the establishment of the anteroposterior body axis during bilaterian development. Although they are often deregulated in cancers, the molecular mechanisms by which they act as oncogenes or tumor suppressor genes are only partially understood. Since the MAPK/ERK signaling pathway is deregulated in most cancers, we aimed at apprehending if and how the Hox proteins interact with ERK oncogenicity. Using an in vivo neoplasia model in the chicken embryo consisting in the overactivation of the ERK1/2 kinases in the trunk neural tube, we analyzed the consequences of the HOXB8 gain of function at the morphological and transcriptional levels. We found that HOXB8 acts as a tumor suppressor, counteracting ERK-induced neoplasia. The HOXB8 tumor suppressor function relies on a large reversion of the oncogenic transcriptome induced by ERK. In addition to showing that the HOXB8 protein controls the transcriptional responsiveness to ERK oncogenic signaling, our study identified new downstream targets of ERK oncogenic activation in an in vivo context that could provide clues for therapeutic strategies.
Collapse
Affiliation(s)
- Axelle Wilmerding
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Lauranne Bouteille
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Lucrezia Rinaldi
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
- Beth Israel Deaconess Medical Center, Department of Medicine and the Cancer Center, Division of Hematology, Harvard Initiative of RNA Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nathalie Caruso
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Yacine Graba
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| | - Marie-Claire Delfini
- Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), 13288 Marseille, France; (A.W.); (L.B.); (L.R.); (N.C.)
| |
Collapse
|
10
|
Szeto B, Valentini C, Aksit A, Werth EG, Goeta S, Brown LM, Olson ES, Kysar JW, Lalwani AK. Impact of Systemic versus Intratympanic Dexamethasone Administration on the Perilymph Proteome. J Proteome Res 2021; 20:4001-4009. [PMID: 34291951 DOI: 10.1021/acs.jproteome.1c00322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glucocorticoids are the first-line treatment for sensorineural hearing loss, but little is known about the mechanism of their protective effect or the impact of route of administration. The recent development of hollow microneedles enables safe and reliable sampling of perilymph for proteomic analysis. Using these microneedles, we investigate the effect of intratympanic (IT) versus intraperitoneal (IP) dexamethasone administration on guinea pig perilymph proteome. Guinea pigs were treated with IT dexamethasone (n = 6), IP dexamethasone (n = 8), or untreated for control (n = 8) 6 h prior to aspiration. The round window membrane (RWM) was accessed via a postauricular approach, and hollow microneedles were used to perforate the RWM and aspirate 1 μL of perilymph. Perilymph samples were analyzed by liquid chromatography-mass spectrometry-based label-free quantitative proteomics. Mass spectrometry raw data files have been deposited in an international public repository (MassIVE proteomics repository at https://massive.ucsd.edu/) under data set # MSV000086887. In the 22 samples of perilymph analyzed, 632 proteins were detected, including the inner ear protein cochlin, a perilymph marker. Of these, 14 proteins were modulated by IP, and three proteins were modulated by IT dexamethasone. In both IP and IT dexamethasone groups, VGF nerve growth factor inducible was significantly upregulated compared to control. The remaining adjusted proteins modulate neurons, inflammation, or protein synthesis. Proteome analysis facilitated by the use of hollow microneedles shows that route of dexamethasone administration impacts changes seen in perilymph proteome. Compared to IT administration, the IP route was associated with greater changes in protein expression, including proteins involved in neuroprotection, inflammatory pathway, and protein synthesis. Our findings show that microneedles can mediate safe and effective intracochlear sampling and hold promise for inner ear diagnostics.
Collapse
Affiliation(s)
- Betsy Szeto
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States
| | - Chris Valentini
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States
| | - Aykut Aksit
- Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| | - Emily G Werth
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Shahar Goeta
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Lewis M Brown
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Elizabeth S Olson
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States.,Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Jeffrey W Kysar
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States.,Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| | - Anil K Lalwani
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States.,Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| |
Collapse
|
11
|
NELL2 modulates cell proliferation and apoptosis via ERK pathway in the development of benign prostatic hyperplasia. Clin Sci (Lond) 2021; 135:1591-1608. [PMID: 34195782 DOI: 10.1042/cs20210476] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023]
Abstract
Benign prostatic hyperplasia (BPH) is a quite common illness but its etiology and mechanism remain unclear. Neural epidermal growth factor-like like 2 (NELL2) plays multifunctional roles in neural cell growth and is strongly linked to the urinary tract disease. Current study aims to determine the expression, functional activities and underlying mechanism of NELL2 in BPH. Human prostate cell lines and tissues from normal human and BPH patients were utilized. Immunohistochemical staining, immunofluorescent staining, RT-polymerase chain reaction (PCR) and Western blotting were performed. We further generated cell models with NELL2 silenced or overexpressed. Subsequently, proliferation, cycle, and apoptosis of prostate cells were determined by cell counting kit-8 (CCK-8) assay and flow cytometry analysis. The epithelial-mesenchymal transition (EMT) and fibrosis process were also analyzed. Our study revealed that NELL2 was up-regulated in BPH samples and localized in the stroma and the epithelium compartments of human prostate tissues. NELL2 deficiency induced a mitochondria-dependent cell apoptosis, and inhibited cell proliferation via phosphorylating extracellular signal-regulated kinase 1/2 (ERK1/2) activation. Additionally, suppression of ERK1/2 with U0126 incubation could significantly reverse NELL2 deficiency triggered cell apoptosis. Consistently, overexpression of NELL2 promoted cell proliferation and inhibited cell apoptosis. However, NELL2 interference was observed no effect on EMT and fibrosis process. Our novel data demonstrated that up-regulation of NELL2 in the enlarged prostate could contribute to the development of BPH through enhancing cell proliferation and inhibited a mitochondria-dependent cell apoptosis via the ERK pathway. The NELL2-ERK system might represent an important target to facilitate the development of future therapeutic approaches in BPH.
Collapse
|
12
|
Beccari L, Jaquier G, Lopez-Delisle L, Rodriguez-Carballo E, Mascrez B, Gitto S, Woltering J, Duboule D. Dbx2 regulation in limbs suggests interTAD sharing of enhancers. Dev Dyn 2021; 250:1280-1299. [PMID: 33497014 PMCID: PMC8451760 DOI: 10.1002/dvdy.303] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND During tetrapod limb development, the HOXA13 and HOXD13 transcription factors are critical for the emergence and organization of the autopod, the most distal aspect where digits will develop. Since previous work had suggested that the Dbx2 gene is a target of these factors, we set up to analyze in detail this potential regulatory interaction. RESULTS We show that HOX13 proteins bind to mammalian-specific sequences at the vicinity of the Dbx2 locus that have enhancer activity in developing digits. However, the functional inactivation of the DBX2 protein did not elicit any particular phenotype related to Hox genes inactivation in digits, suggesting either redundant or compensatory mechanisms. We report that the neighboring Nell2 and Ano6 genes are also expressed in distal limb buds and are in part controlled by the same Dbx2 enhancers despite being localized into two different topologically associating domains (TADs) flanking the Dbx2 locus. CONCLUSIONS We conclude that Hoxa13 and Hoxd genes cooperatively activate Dbx2 expression in developing digits through binding to mammalian specific regulatory sequences in the Dbx2 neighborhood. Furthermore, these enhancers can overcome TAD boundaries in either direction to co-regulate a set of genes located in distinct chromatin domains.
Collapse
Affiliation(s)
- Leonardo Beccari
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, University Claude Bernard Lyon1, Lyon, France
| | - Gabriel Jaquier
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | | | - Eddie Rodriguez-Carballo
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Department of Molecular Biology, University of Geneva, Geneva, Switzerland
| | - Bénédicte Mascrez
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Sandra Gitto
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Joost Woltering
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Denis Duboule
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.,School of Life Sciences, Federal School of Technology (EPFL), Lausanne, Switzerland.,Collège de France, Paris, France
| |
Collapse
|
13
|
Kim HR, Kim DH, An JY, Kang D, Park JW, Hwang EM, Seo EJ, Jang IH, Ha CM, Lee BJ. NELL2 Function in Axon Development of Hippocampal Neurons. Mol Cells 2020; 43:581-589. [PMID: 32597395 PMCID: PMC7332358 DOI: 10.14348/molcells.2020.0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/06/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023] Open
Abstract
Neurons have multiple dendrites and single axon. This neuronal polarity is gradually established during early processes of neuronal differentiation: generation of multiple neurites (stages 1-2); differentiation (stage 3) and maturation (stages 4-5) of an axon and dendrites. In this study, we demonstrated that the neuron-specific n-glycosylated protein NELL2 is important for neuronal polarization and axon growth using cultured rat embryonic hippocampal neurons. Endogenous NELL2 expression was gradually increased in parallel with the progression of developmental stages of hippocampal neurons, and overexpression of NELL2 stimulated neuronal polarization and axon growth. In line with these results, knockdown of NELL2 expression resulted in deterioration of neuronal development, including inhibition of neuronal development progression, decreased axon growth and increased axon branching. Inhibitor against extracellular signal-regulated kinase (ERK) dramatically inhibited NELL2-induced progression of neuronal development and axon growth. These results suggest that NELL2 is an important regulator for the morphological development for neuronal polarization and axon growth.
Collapse
Affiliation(s)
- Han Rae Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 4460, Korea
- Present address: Department of Pharmacology and Physiology, School of Medicine & Health Sciences, The George Washington University, Washington, D.C. 20037, USA
- These authors contributed equally to this work.
| | - Dong Hee Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 4460, Korea
- These authors contributed equally to this work.
| | - Ji Young An
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 4460, Korea
- These authors contributed equally to this work.
| | - Dasol Kang
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 4460, Korea
| | - Jeong Woo Park
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 4460, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 079, Korea
| | - Eun Jin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan 50612, Korea
| | - Il Ho Jang
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan 50612, Korea
| | - Chang Man Ha
- Research Division and Brain Research Core Facilities of Korea Brain Research Institute, Daegu 1068, Korea,
| | - Byung Ju Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 4460, Korea
| |
Collapse
|
14
|
Lee VM, Hernandez S, Giang B, Chabot C, Hernandez J, de Bellard ME. Molecular Events Controlling Cessation of Trunk Neural Crest Migration and Onset of Differentiation. Front Cell Dev Biol 2020; 8:199. [PMID: 32318567 PMCID: PMC7147452 DOI: 10.3389/fcell.2020.00199] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/09/2020] [Indexed: 12/30/2022] Open
Abstract
Neural crest cells (NCC) migrate extensively in vertebrate embryos to populate diverse derivatives including ganglia of the peripheral nervous system. Little is known about the molecular mechanisms that lead migrating trunk NCC to settle at selected sites in the embryo, ceasing their migration and initiating differentiation programs. To identify candidate genes involved in these processes, we profiled genes up-regulated in purified post-migratory compared with migratory NCC using a staged, macroarrayed cDNA library. A secondary screen of in situ hybridization revealed that many genes are specifically enhanced in neural crest-derived ganglia, including macrophage migration inhibitory factor (MIF), a ligand for CXCR4 receptor. Through in vivo and in vitro assays, we found that MIF functions as a potent chemoattractant for NCC. These results provide a molecular profile of genes expressed concomitant with gangliogenesis, thus, offering new markers and potential regulatory candidates involved in cessation of migration and onset of differentiation.
Collapse
Affiliation(s)
- Vivian M Lee
- Universal Cells Inc., Seattle, WA, United States
| | - Sergio Hernandez
- Biology Department, California State University Northridge, Northridge, CA, United States
| | - Belle Giang
- Moorpark College, Moorpark, CA, United States
| | - Chris Chabot
- Biology Department, California State University Northridge, Northridge, CA, United States
| | | | - Maria Elena de Bellard
- Biology Department, California State University Northridge, Northridge, CA, United States
| |
Collapse
|
15
|
Nakamoto C, Durward E, Horie M, Nakamoto M. Nell2 regulates the contralateral-versus-ipsilateral visual projection as a domain-specific positional cue. Development 2019; 146:dev.170704. [PMID: 30745429 DOI: 10.1242/dev.170704] [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: 08/15/2018] [Accepted: 01/29/2019] [Indexed: 01/15/2023]
Abstract
In mammals with binocular vision, retinal ganglion cell (RGC) axons from each eye project to eye-specific domains in the contralateral and ipsilateral dorsal lateral geniculate nucleus (dLGN), underpinning disparity-based stereopsis. Although domain-specific axon guidance cues that discriminate contralateral and ipsilateral RGC axons have long been postulated as a key mechanism for development of the eye-specific retinogeniculate projection, the molecular nature of such cues has remained elusive. Here, we show that the extracellular glycoprotein Nell2 (neural epidermal growth factor-like-like 2) is expressed in the dorsomedial region of the dLGN, which ipsilateral RGC axons terminate in and contralateral axons avoid. In Nell2 mutant mice, contralateral RGC axons abnormally invaded the ipsilateral domain of the dLGN, and ipsilateral axons terminated in partially fragmented patches, forming a mosaic pattern of contralateral and ipsilateral axon-termination zones. In vitro, Nell2 exerted inhibitory effects on contralateral, but not ipsilateral, RGC axons. These results provide evidence that Nell2 acts as a domain-specific positional label in the dLGN that discriminates contralateral and ipsilateral RGC axons, and that it plays essential roles in the establishment of the eye-specific retinogeniculate projection.
Collapse
Affiliation(s)
- Chizu Nakamoto
- Aberdeen Developmental Biology Group, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Elaine Durward
- Aberdeen Developmental Biology Group, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Masato Horie
- Department of CNS Research, Otsuka Pharmaceutical, 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Masaru Nakamoto
- Aberdeen Developmental Biology Group, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
16
|
Yamamoto N, Kashiwagi M, Ishihara M, Kojima T, Maturana AD, Kuroda S, Niimi T. Robo2 contains a cryptic binding site for neural EGFL-like (NELL) protein 1/2. J Biol Chem 2019; 294:4693-4703. [PMID: 30700556 DOI: 10.1074/jbc.ra118.005819] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/14/2019] [Indexed: 12/15/2022] Open
Abstract
The signaling pathways that are mediated by Slit ligands and their Roundabout (Robo) family of receptors play multifunctional roles in the development of the nervous system and other organs. A recent study identified neural epidermal growth factor-like (NEL)-like 2 (NELL2) as a novel ligand for Robo3. In this study, we carried out a comprehensive analysis of the interaction between NELL1 and the Robo family of receptors and demonstrated that Robo2 contains a cryptic binding site for both NELL1 and NELL2. NELL1/2 binds to the first fibronectin type III (FNIII) domain of Robo2 but not to intact Robo2. Mutation analysis revealed that several amino acids within the first FNIII domain are critical for NELL1 binding to Robo2 but not to Robo1. The Robo2 deletion mutants without the fourth immunoglobulin domain and single amino acid substitution mutants that can influence the architecture of the ectodomain facilitated binding to NELL1/2. Acidic conditions increased the binding affinity of Robo2 for NELL1. These results suggest that Robo2 functions as a receptor for NELL1/2, particularly under circumstances where Robo2 undergoes proteolytic digestion. If this is not the case, conformational changes of the ectodomain of Robo2 may unmask the binding site for NELL1/2.
Collapse
Affiliation(s)
- Naoka Yamamoto
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Manabu Kashiwagi
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Manami Ishihara
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Takaaki Kojima
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Andrés D Maturana
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Shun'ichi Kuroda
- the Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan
| | - Tomoaki Niimi
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| |
Collapse
|
17
|
Jeong JK, Kim JG, Kim HR, Lee TH, Park JW, Lee BJ. A Role of Central NELL2 in the Regulation of Feeding Behavior in Rats. Mol Cells 2017; 40:186-194. [PMID: 28301916 PMCID: PMC5386956 DOI: 10.14348/molcells.2017.2278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 02/21/2017] [Accepted: 03/06/2017] [Indexed: 12/02/2022] Open
Abstract
A brain-enriched secreting signal peptide, NELL2, has been suggested to play multiple roles in the development, survival, and activity of neurons in mammal. We investigated here a possible involvement of central NELL2 in regulating feeding behavior and metabolism. In situ hybridization and an im-munohistochemical approach were used to determine expression of NELL2 as well as its colocalization with proopiomelanocortin (POMC) and neuropeptide Y (NPY) in the rat hypothalamus. To investigate the effect of NELL2 on feeding behavior, 2 nmole of antisense NELL2 oligodeoxynucleotide was administered into the lateral ventricle of adult male rat brains for 6 consecutive days, and changes in daily body weight, food, and water intake were monitored. Metabolic state-dependent NELL2 expression in the hypothalamus was tested in vivo using a fasting model. NELL2 was noticeably expressed in the hypothalamic nuclei controlling feeding behavior. Furthermore, all arcuatic POMC and NPY positive neurons produced NELL2. The NELL2 gene expression in the hypothalamus was up-regulated by fasting. However, NELL2 did not affect POMC and NPY gene expression in the hypothalamus. A blockade of NELL2 production in the hypothalamus led to a reduction in daily food intake, followed by a loss in body weight without a change in daily water intake in normal diet condition. NELL2 did not affect short-term hunger dependent appetite behavior. Our data suggests that hypothalamic NELL2 is associated with appetite behavior, and thus central NELL2 could be a new therapeutic target for obesity.
Collapse
Affiliation(s)
- Jin Kwon Jeong
- Department of Pharmacology and Physiology, School of Medicine & Health Sciences, The George Washington University,
USA
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012,
Korea
| | - Han Rae Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Tae Hwan Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| |
Collapse
|
18
|
de Ramon Francàs G, Zuñiga NR, Stoeckli ET. The spinal cord shows the way - How axons navigate intermediate targets. Dev Biol 2016; 432:43-52. [PMID: 27965053 DOI: 10.1016/j.ydbio.2016.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/26/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022]
Abstract
Functional neural circuits depend on the establishment of specific connections between neurons and their target cells. To this end, many axons have to travel long distances to reach their target cells during development. Studies addressing the molecular mechanisms of axon guidance have to overcome the complexity of subpopulation-specific requirements with respect to pathways, guidance cues, and target recognition. Compared to the brain, the relatively simple structure of the spinal cord provides an advantage for experimental studies of axon guidance mechanisms. Therefore, the so far best understood model for axon guidance is the dI1 population of dorsal interneurons of the spinal cord. They extend their axons ventrally towards the floor plate. After midline crossing, they turn rostrally along the contralateral floor-plate border. Despite the fact that the trajectory of dI1 axons seems to be rather simple, the number of axon guidance molecules involved in the decisions taken by these axons is bewildering. Because guidance molecules and mechanisms are conserved throughout the developing nervous system, we can generalize what we have learned about the navigation of the floor plate as an intermediate target for commissural axons to the brain.
Collapse
Affiliation(s)
- Gemma de Ramon Francàs
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Nikole R Zuñiga
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Esther T Stoeckli
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
19
|
Yuen-Chi Lau R, Qian X, Po KT, Li LM, Guo X. Response of Rat Tibia to Prolonged Unloading Under the Influence of Electrical Stimulation at the Dorsal Root Ganglion. Neuromodulation 2016; 20:284-289. [DOI: 10.1111/ner.12488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/18/2016] [Accepted: 07/26/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Roy Yuen-Chi Lau
- Department of Rehabilitation Sciences; the Hong Kong Polytechnic University; Hong Kong SAR China
| | - Xing Qian
- School of Aerospace; Tsinghua University; People's Republic of China
| | - Kai-Ting Po
- Department of Rehabilitation Sciences; the Hong Kong Polytechnic University; Hong Kong SAR China
| | - Lu-Ming Li
- School of Aerospace; Tsinghua University; People's Republic of China
| | - Xia Guo
- Department of Rehabilitation Sciences; the Hong Kong Polytechnic University; Hong Kong SAR China
| |
Collapse
|
20
|
Yang Y, Mu J, Chen G, Zhan Y, Zhong J, Wei Y, Cheng K, Qin B, You H, Xie P. iTRAQ-based quantitative proteomic analysis of cerebrospinal fluid reveals NELL2 as a potential diagnostic biomarker of tuberculous meningitis. Int J Mol Med 2015; 35:1323-32. [PMID: 25760060 DOI: 10.3892/ijmm.2015.2131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 02/17/2015] [Indexed: 11/06/2022] Open
Abstract
Tuberculous meningitis (TBM) is a serious complication of tuberculosis that affects the central nervous system. As TBM may result in permanent sequelae and death, rapid, accurate diagnostic tests using novel biomarkers are required for the early diagnosis and treatment of TBM. A quantitative proteomic study was therefore performed to identify differential proteins in the cerebrospinal fluid (CSF) obtained from TBM patients (n=12) and healthy controls (n=12). CSF samples were labelled with iTRAQ™ and analyzed by LC-MS/MS. Gene ontology and Pathway analysis were conducted using DAVID bioinformatics resources. Neural epidermal growth factor-like like 2 (NELL2) with the largest fold-change value was selected for validation by western blotting. Proteomic phenotyping revealed over-representation in two inflammation-associated processes, complement and coagulation cascades as well as cell adhesion molecules. Western blotting showed a significant decrease in NELL2 levels in TBM subjects compared to healthy controls. The AUC analysis revealed NELL2 was able to distinguish TBM subjects from healthy controls with 83.3% sensitivity and 75% specificity. In conclusion, the results showed that CSF NELL2 is a potential diagnostic biomarker for TBM. Further evaluation of these findings in larger studies including anti-tuberculosis medicated and unmedicated patient cohorts with other intracranial infectious diseases is required for clinical translation.
Collapse
Affiliation(s)
- Yongtao Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Jun Mu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Guanghui Chen
- Institute of Neuroscience, Chongqing Medical University, Chongqing, P.R. China
| | - Yuan Zhan
- Institute of Neuroscience, Chongqing Medical University, Chongqing, P.R. China
| | - Jiaju Zhong
- Institute of Neuroscience, Chongqing Medical University, Chongqing, P.R. China
| | - Youdong Wei
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Ke Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Bin Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Hongmin You
- Institute of Neuroscience, Chongqing Medical University, Chongqing, P.R. China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
21
|
Lau YC, Qian X, Po KT, Li LM, Guo X. Electrical stimulation at the dorsal root ganglion preserves trabecular bone mass and microarchitecture of the tibia in hindlimb-unloaded rats. Osteoporos Int 2015; 26:481-8. [PMID: 25212672 DOI: 10.1007/s00198-014-2866-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/19/2014] [Indexed: 01/14/2023]
Abstract
SUMMARY This study seeks to investigate the effect of electrical stimulation (ES) at dorsal root ganglion (DRG) on disuse bone loss in a rat model. Hindlimb unloading for 14 days resulted in significant bone loss in rat tibia while rats with ES at DRG showed a significant reduced bone loss INTRODUCTION Mechanical unloading induces osteoporosis in both human and animals. Previous studies demonstrated that electrical stimulation (ES) to dorsal root ganglion (DRG) could trigger secretion of calcitonin gene-related peptide (CGRP) which plays an important role in bone modeling and remodeling. This study seeks to investigate the effect of ES to DRG on disuse bone loss in a rat model. METHODS Twenty-four rats were randomly assigned in three experimental groups: cage control (CC), hindlimb unloading (HU), and hindlimb unloading with ES (HUES). ES was applied via implantable micro-electrical stimulators (IMES) to right DRGs at vertebral levels L4-L6 in HUES group. RESULTS Hindlimb unloading for 14 days resulted in 25.9% decrease in total bone mineral content (BMC), 29.2% decrease in trabecular BMD and trabecular microarchitecture and connectivity were significantly deteriorated in the proximal tibia metaphysis in HU group, while rats with ES at DRG showed significant reduced bone loss that there was 3.8% increase in total BMC, 2.3% decrease in trabecular BMD, and significant improvement in trabecular microarchitecture. There was a concurrent enhancement of expression of CGRP in stimulated DRGs. CONCLUSIONS The results confirm the effect of ES at DRG on enhancing CGRP expression and suggest potential applications of IMES for the prevention and treatment of disuse bone loss.
Collapse
Affiliation(s)
- Y-C Lau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | | | | | | | | |
Collapse
|
22
|
Newbern JM. Molecular control of the neural crest and peripheral nervous system development. Curr Top Dev Biol 2015; 111:201-31. [PMID: 25662262 DOI: 10.1016/bs.ctdb.2014.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A transient and unique population of multipotent stem cells, known as neural crest cells (NCCs), generate a bewildering array of cell types during vertebrate development. An attractive model among developmental biologists, the study of NCC biology has provided a wealth of knowledge regarding the cellular and molecular mechanisms important for embryogenesis. Studies in numerous species have defined how distinct phases of NCC specification, proliferation, migration, and survival contribute to the formation of multiple functionally distinct organ systems. NCC contributions to the peripheral nervous system (PNS) are well known. Critical developmental processes have been defined that provide outstanding models for understanding how extracellular stimuli, cell-cell interactions, and transcriptional networks cooperate to direct cellular diversification and PNS morphogenesis. Dissecting the complex extracellular and intracellular mechanisms that mediate the formation of the PNS from NCCs may have important therapeutic implications for neurocristopathies, neuropathies, and certain forms of cancer.
Collapse
Affiliation(s)
- Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA.
| |
Collapse
|
23
|
Lee DY, Kim E, Lee YS, Ryu H, Park JY, Hwang EM. The cytosolic splicing variant of NELL2 inhibits PKCβ1 in glial cells. Biochem Biophys Res Commun 2014; 454:459-64. [PMID: 25450684 DOI: 10.1016/j.bbrc.2014.10.110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/21/2014] [Indexed: 11/29/2022]
Abstract
NELL2 is an abundant glycoprotein containing EGF-like domain in the neural tissues where it has multiple physiological functions by interacting with protein kinase C (PKC). There are two different splicing variant forms of NELL2 identified so far. One is secreted NELL2 (sNELL2) which is a neuron-specific variant and the other is cytosolic NELL2 (cNELL2) which is non-secreted splicing variant of NELL2. Although cNELL2 structure was well characterized, the expression pattern or the cellular function of cNELL2 is not fully determined. In this study, we found that cNELL2 specifically interacts with PKCβ isotypes and inhibits PKCβ1 through direct binding to the N-terminal pseudosubstrate domain of PKCβ1. Here, we also demonstrate that cNELL2 is predominantly expressed and has inhibitory effects on the PKC downstream signaling pathways in astrocytes thereby establishing cNELL2 as an endogenous inhibitor of PKCβ1 in glia.
Collapse
Affiliation(s)
- Da Yong Lee
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 305-806, Republic of Korea
| | - Eunju Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Young-Sun Lee
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Hwani Ryu
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 136-703, Republic of Korea.
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea; Neuroscience Program, University of Science and Technology (UST), Daejeon 305-350, Republic of Korea.
| |
Collapse
|
24
|
Durruthy-Durruthy R, Gottlieb A, Hartman BH, Waldhaus J, Laske RD, Altman R, Heller S. Reconstruction of the mouse otocyst and early neuroblast lineage at single-cell resolution. Cell 2014; 157:964-78. [PMID: 24768691 PMCID: PMC4051200 DOI: 10.1016/j.cell.2014.03.036] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/03/2014] [Accepted: 03/12/2014] [Indexed: 01/24/2023]
Abstract
The otocyst harbors progenitors for most cell types of the mature inner ear. Developmental lineage analyses and gene expression studies suggest that distinct progenitor populations are compartmentalized to discrete axial domains in the early otocyst. Here, we conducted highly parallel quantitative RT-PCR measurements on 382 individual cells from the developing otocyst and neuroblast lineages to assay 96 genes representing established otic markers, signaling-pathway-associated transcripts, and novel otic-specific genes. By applying multivariate cluster, principal component, and network analyses to the data matrix, we were able to readily distinguish the delaminating neuroblasts and to describe progressive states of gene expression in this population at single-cell resolution. It further established a three-dimensional model of the otocyst in which each individual cell can be precisely mapped into spatial expression domains. Our bioinformatic modeling revealed spatial dynamics of different signaling pathways active during early neuroblast development and prosensory domain specification.
Collapse
Affiliation(s)
- Robert Durruthy-Durruthy
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Assaf Gottlieb
- Departments of Bioengineering and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Byron H Hartman
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jörg Waldhaus
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Roman D Laske
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Russ Altman
- Departments of Bioengineering and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefan Heller
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
25
|
Kim DH, Kim HR, Choi EJ, Kim DY, Kim KK, Kim BS, Park JW, Lee BJ. Neural epidermal growth factor-like like protein 2 (NELL2) promotes aggregation of embryonic carcinoma P19 cells by inducing N-cadherin expression. PLoS One 2014; 9:e85898. [PMID: 24465772 PMCID: PMC3897553 DOI: 10.1371/journal.pone.0085898] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/03/2013] [Indexed: 11/18/2022] Open
Abstract
NELL2 was first identified as a mammalian homolog of chick NEL (Neural EGF-like) protein. It is almost exclusively expressed in neurons of the rat brain and has been suggested to play a role in neural differentiation. However, there is still no clear evidence for the detailed function of NELL2 in the differentiation of neurons. In this study, we identified NELL2 function during neural differentiation of mouse embryonic carcinoma P19 cells. Endogenous expression of NELL2 in the P19 cells increased in parallel with the neuronal differentiation induced by retinoic acid (RA). We found that the mouse NELL2 promoter contains RA response elements (RAREs) and that treatment with RA increased NELL2 promoter activity. Transfection of P19 cells with NELL2 expression vectors induced a dramatic increase in cell aggregation, resulting in the facilitation of neural differentiation. Moreover, NELL2 significantly increased N-cadherin expression in the P19 cell. These data suggest that NELL2 plays an important role in the regulation of neuronal differentiation via control of N-cadherin expression and cell aggregation.
Collapse
Affiliation(s)
- Dong Hee Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Han Rae Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Eun Jung Choi
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Dong Yeol Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Kwang Kon Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Byung Sam Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Jeong Woo Park
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
| | - Byung Ju Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, South Korea
- * E-mail:
| |
Collapse
|
26
|
Ha CM, Hwang EM, Kim E, Lee DY, Chang S, Lee BJ, Hong SG, Park JY. The molecular mechanism of NELL2 movement and secretion in hippocampal progenitor HiB5 cells. Mol Cells 2013; 36:527-33. [PMID: 24352699 PMCID: PMC3887960 DOI: 10.1007/s10059-013-0216-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 11/26/2022] Open
Abstract
Neural epidermal growth factor-like protein-like 2 (NELL2) is a secreted glycoprotein that is predominantly expressed in the nervous system, but little is known about the intracellular movement and secretion mechanism of this protein. By monitoring the localization and movements of enhanced green fluorescent protein (EGFP)-labeled NELL2 in living cultured hippocampal neuroprogenitor HiB5 cells, we determined the subcellular localization of NELL2 and its intracellular movement and secretion mechanism. Cterminal EGFP-fused NELL2 showed a typical expression pattern of secreted proteins, especially with respect to its localization in the endoplasmic reticulum, Golgi apparatus, and punctate structures. Vesicles containing NELL2 exhibited bidirectional movement in HiB5 cells. The majority of the vesicles (70.1%) moved in an anterograde direction with an average velocity of 0.454 μm/s, whereas some vesicles (28.7%) showed retrograde movement with an average velocity of 0.302 μm/s. The movement patterns of NELL2 vesicles were dependent upon the presence of microtubules in HiB5 cells. Anterograde movement of NELL2 did not lead to a detectable accumulation of NELL2 in the peripheral region of the cell, indicating that it was secreted into the culture medium. We also showed that the N-terminal 29 amino acids of NELL2 were important for secretion of this protein. Taken together, these results strongly suggest that the N-terminal region of NELL2 determines both the pattern of its intracellular expression and transport of NELL2 vesicles by high-velocity movement. Therefore, NELL2 may affect the cellular activity of cells in a paracrine or autocrine manner.
Collapse
Affiliation(s)
- Chang Man Ha
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju 660-290,
Korea
- Convergence Brain Research Department, Korea Brain Research Institute (KBRI), Daegu 700-010,
Korea
| | - Eun Mi Hwang
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju 660-290,
Korea
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791,
Korea
| | - Eunju Kim
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju 660-290,
Korea
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791,
Korea
| | - Da Yong Lee
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791,
Korea
| | - Sunghoe Chang
- Department of Biomedical Sciences, Neuroscience Research Institute, Biomembrane Plasticity Research Center, Seoul National University College of Medicine, Seoul 110-799,
Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749,
Korea
| | - Seong-Geun Hong
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju 660-290,
Korea
| | - Jae-Yong Park
- Department of Physiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju 660-290,
Korea
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 136-791,
Korea
| |
Collapse
|
27
|
Nakamoto C, Kuan SL, Findlay AS, Durward E, Ouyang Z, Zakrzewska ED, Endo T, Nakamoto M. Nel positively regulates the genesis of retinal ganglion cells by promoting their differentiation and survival during development. Mol Biol Cell 2013; 25:234-44. [PMID: 24258025 PMCID: PMC3890344 DOI: 10.1091/mbc.e13-08-0453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
For correct functioning of the nervous system, the appropriate number and complement of neuronal cell types must be produced during development. However, the molecular mechanisms that regulate the production of individual classes of neurons are poorly understood. In this study, we investigate the function of the thrombospondin-1-like glycoprotein, Nel (neural epidermal growth factor [EGF]-like), in the generation of retinal ganglion cells (RGCs) in chicks. During eye development, Nel is strongly expressed in the presumptive retinal pigment epithelium and RGCs. Nel overexpression in the developing retina by in ovo electroporation increases the number of RGCs, whereas the number of displaced amacrine cells decreases. Conversely, knockdown of Nel expression by transposon-mediated introduction of RNA interference constructs results in decrease in RGC number and increase in the number of displaced amacrine cells. Modifications of Nel expression levels do not appear to affect proliferation of retinal progenitor cells, but they significantly alter the progression rate of RGC differentiation from the central retina to the periphery. Furthermore, Nel protects RGCs from apoptosis during retinal development. These results indicate that Nel positively regulates RGC production by promoting their differentiation and survival during development.
Collapse
Affiliation(s)
- Chizu Nakamoto
- Aberdeen Developmental Biology Group, School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Mohr R, Neckel P, Zhang Y, Stachon S, Nothelfer K, Schaeferhoff K, Obermayr F, Bonin M, Just L. Molecular and cell biological effects of 3,5,3′-triiodothyronine on progenitor cells of the enteric nervous system in vitro. Stem Cell Res 2013; 11:1191-205. [DOI: 10.1016/j.scr.2013.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 07/15/2013] [Accepted: 08/01/2013] [Indexed: 01/18/2023] Open
|
29
|
Kim DH, Roh YG, Lee HH, Lee SY, Kim SI, Lee BJ, Leem SH. The E2F1 oncogene transcriptionally regulates NELL2 in cancer cells. DNA Cell Biol 2013; 32:517-23. [PMID: 23829315 DOI: 10.1089/dna.2013.1974] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
NELL2 was first identified as a mammalian homolog of the chicken NEL protein. It was expressed in neurons and has been suggested to play a role in cell survival. However, no clear evidence has yet been available for functions of NELL2. In this study, we found two E2F1 binding sites located in the NELL2 promoter region. We examined the expression of NELL2 and E2F1 in human breast cancer cells (MDA-MB231, MCF7) and bladder cancer cells (5637, UC5). In MDA-MB231 and 5637, the expression levels of NELL2 and E2F1 were higher. To examine the interaction between E2F1 and NELL2, the binding activity was checked by a promoter assay and chromatin immunoprecipitation. From the results, we suggest that NELL2 is a novel target gene of E2F1, which is a key regulator of cell proliferation. We reveal that expression of NELL2 is regulated by E2F1, specifically, mRNA and protein levels of NELL2 are elevated upon activation of exogenous E2F1. Moreover, cells overexpressing NELL2 increased their invasive ability and an enhancement of the effect was observed when NELL2 and E2F1 were coexpressed in MDA-MB231 cells. Therefore, we suggest a novel activity for NELL2 in cancer progression through the regulation of E2F1.
Collapse
Affiliation(s)
- Dong Hee Kim
- Department of Biological Science, College of Natural Sciences, Dong-A University, Busan, South Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Rebl A, Verleih M, Köllner B, Korytář T, Goldammer T. Duplicated NELL2 genes show different expression patterns in two rainbow trout strains after temperature and pathogen challenge. Comp Biochem Physiol B Biochem Mol Biol 2012; 163:65-73. [DOI: 10.1016/j.cbpb.2012.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/30/2012] [Accepted: 05/02/2012] [Indexed: 12/13/2022]
|
31
|
Munemasa Y, Chang CS, Kwong JMK, Kyung H, Kitaoka Y, Caprioli J, Piri N. The neuronal EGF-related gene Nell2 interacts with Macf1 and supports survival of retinal ganglion cells after optic nerve injury. PLoS One 2012; 7:e34810. [PMID: 22496866 PMCID: PMC3319615 DOI: 10.1371/journal.pone.0034810] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 03/05/2012] [Indexed: 12/20/2022] Open
Abstract
Nell2 is a neuron-specific protein containing six epidermal growth factor-like domains. We have identified Nell2 as a retinal ganglion cell (RGC)-expressed gene by comparing mRNA profiles of control and RGC-deficient rat retinas. The aim of this study was to analyze Nell2 expression in wild-type and optic nerve axotomized retinas and evaluate its potential role in RGCs. Nell2-positive in situ and immunohistochemical signals were localized to irregularly shaped cells in the ganglion cell layer (GCL) and colocalized with retrogradely-labeled RGCs. No Nell2-positive cells were detected in 2 weeks optic nerve transected (ONT) retinas characterized with approximately 90% RGC loss. RT-PCR analysis showed a dramatic decrease in the Nell2 mRNA level after ONT compared to the controls. Immunoblot analysis of the Nell2 expression in the retina revealed the presence of two proteins with approximate MW of 140 and 90 kDa representing glycosylated and non-glycosylated Nell2, respectively. Both products were almost undetectable in retinal protein extracts two weeks after ONT. Proteome analysis of Nell2-interacting proteins carried out with MALDI-TOF MS (MS) identified microtubule-actin crosslinking factor 1 (Macf1), known to be critical in CNS development. Strong Macf1 expression was observed in the inner plexiform layer and GCL where it was colocalizied with Thy-1 staining. Since Nell2 has been reported to increase neuronal survival of the hippocampus and cerebral cortex, we evaluated the effect of Nell2 overexpression on RGC survival. RGCs in the nasal retina were consistently more efficiently transfected than in other areas (49% vs. 13%; n = 5, p<0.05). In non-transfected or pEGFP-transfected ONT retinas, the loss of RGCs was approximately 90% compared to the untreated control. In the nasal region, Nell2 transfection led to the preservation of approximately 58% more cells damaged by axotomy compared to non-transfected (n = 5, p<0.01) or pEGFP-transfected controls (n = 5, p<0.01).
Collapse
Affiliation(s)
- Yasunari Munemasa
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Department Ophthalmology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Chang-Sheng Chang
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jacky M. K. Kwong
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Haksu Kyung
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Yasushi Kitaoka
- Department Ophthalmology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Joseph Caprioli
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Natik Piri
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Abstract
Autonomic neuron development is controlled by a network of transcription factors, which is induced by bone morphogenetic protein signalling in neural crest progenitor cells. This network intersects with a transcriptional program in migratory neural crest cells that pre-specifies autonomic neuron precursor cells. Recent findings demonstrate that the transcription factors acting in the initial specification and differentiation of sympathetic neurons are also important for the proliferation of progenitors and immature neurons during neurogenesis. Elimination of Phox2b, Hand2 and Gata3 in differentiated neurons affects the expression of subtype-specific and/or generic neuronal properties or neuron survival. Taken together, transcription factors previously shown to act in initial neuron specification and differentiation display a much broader spectrum of functions, including control of neurogenesis and the maintenance of subtype characteristics and survival of mature neurons.
Collapse
Affiliation(s)
- Hermann Rohrer
- Research Group Developmental Neurobiology, Max-Planck-Institute for Brain Research, 60528 Frankfurt/Main, Germany.
| |
Collapse
|
33
|
Hwangpo TA, Jordan JD, Premsrirut PK, Jayamaran G, Licht JD, Iyengar R, Neves SR. G Protein-regulated inducer of neurite outgrowth (GRIN) modulates Sprouty protein repression of mitogen-activated protein kinase (MAPK) activation by growth factor stimulation. J Biol Chem 2012; 287:13674-85. [PMID: 22383529 DOI: 10.1074/jbc.m111.320705] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gα(o/i) interacts directly with GRIN (G protein-regulated inducer of neurite outgrowth). Using the yeast two-hybrid system, we identified Sprouty2 as an interacting partner of GRIN. Gα(o) and Sprouty2 bind to overlapping regions of GRIN, thus competing for GRIN binding. Imaging experiments demonstrated that Gα(o) expression promoted GRIN translocation to the plasma membrane, whereas Sprouty2 expression failed to do so. Given the role of Sprouty2 in the regulation of growth factor-mediated MAPK activation, we examined the contribution of the GRIN-Sprouty2 interaction to CB1 cannabinoid receptor regulation of FGF receptor signaling. In Neuro-2A cells, a system that expresses all of the components endogenously, modulation of GRIN levels led to regulation of MAPK activation. Overexpression of GRIN potentiated FGF activation of MAPK and decreased tyrosine phosphorylation of Sprouty2. Pretreatment with G(o/i)-coupled CB1 receptor agonist attenuated subsequent FGF activation of MAPK. Decreased expression of GRIN both diminished FGF activation of MAPK and blocked CB1R attenuation of MAPK activation. These observations indicate that Gα(o) interacts with GRIN and outcompetes GRIN from bound Sprouty. Free Sprouty then in turn inhibits growth factor signaling. Thus, here we present a novel mechanism of how G(o/i)-coupled receptors can inhibit growth factor signaling to MAPK.
Collapse
Affiliation(s)
- Tracy Anh Hwangpo
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Nakamura R, Nakamoto C, Obama H, Durward E, Nakamoto M. Structure-function analysis of Nel, a thrombospondin-1-like glycoprotein involved in neural development and functions. J Biol Chem 2011; 287:3282-91. [PMID: 22157752 DOI: 10.1074/jbc.m111.281485] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nel (neural epidermal growth factor (EGF)-like molecule) is a multimeric, multimodular extracellular glycoprotein with heparin-binding activity and structural similarities to thrombospondin-1. Nel is predominantly expressed in the nervous system and has been implicated in neuronal proliferation and differentiation, retinal axon guidance, synaptic functions, and spatial learning. The Nel protein contains an N-terminal thrombospondin-1 (TSP-N) domain, five cysteine-rich domains, and six EGF-like domains. However, little is known about the functions of specific domains of the Nel protein. In this study, we have performed structure-function analysis of Nel, by using a series of expression constructs for different regions of the Nel protein. Our studies demonstrate that the TSP-N domain is responsible for homo-multimer formation of Nel and its heparin-binding activity. In vivo, Nel and related Nell1 are expressed in several regions of the mouse central nervous system with partly overlapping patterns. When they are expressed in the same cells in vitro, Nel and Nell1 can form hetero-multimers through the TSP-N domain, but they do not hetero-oligomerize with thrombospondin-1. Whereas both the TSP-N domain and cysteine-rich domains can bind to retinal axons in vivo, only the latter causes growth cone collapse in cultured retinal axons, suggesting that cysteine-rich domains interact with and activate an inhibitory axon guidance receptor. These results suggest that Nel interacts with a range of molecules through its different domains and exerts distinct functions.
Collapse
Affiliation(s)
- Ritsuko Nakamura
- Aberdeen Developmental Biology Group, School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, United Kingdom
| | | | | | | | | |
Collapse
|
35
|
Type 2 helper T-cell cytokines induce morphologic and molecular characteristics of atopic dermatitis in human skin equivalent. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2091-9. [PMID: 21514424 DOI: 10.1016/j.ajpath.2011.01.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 12/13/2010] [Accepted: 01/25/2011] [Indexed: 01/18/2023]
Abstract
Both the immune system and the epidermis likely have an important role in the pathogenesis of atopic dermatitis (AD). The objective of the present study was to develop a human skin equivalent model exhibiting morphologic and molecular characteristics of AD in a controlled manner. Skin equivalents generated from normal adult human keratinocytes were stimulated with type 2 T-helper cell (Th2) cytokines IL-4 and IL-13, and morphologic features and gene expression of the epidermis were studied. Th2 cytokines induced intercellular edema similar to spongiotic changes observed in lesional AD as assessed at histopathologic analysis and electron microscopy. Furthermore, genes known to be specifically expressed in epidermis of patients with AD such as CAII and NELL2 were induced. In contrast, expression of psoriasis-associated genes such as elafin and hBD2 was not changed. Th2 cytokines caused DNA fragmentation in the keratinocytes, which could be inhibited by the caspase inhibitor Z-VAD, which suggests that apoptosis was induced. In addition, up-regulation of the death receptor Fas was observed in keratinocytes after Th2 cytokine stimulation. IL-4 and IL-13 induced phosphorylation of the signaling molecule STAT6. It was concluded that the skin equivalent model described herein may be useful in investigation of the epidermal aspects of AD and for study of drugs that act at the level of keratinocyte biology.
Collapse
|
36
|
Vernes SC, Oliver PL, Spiteri E, Lockstone HE, Puliyadi R, Taylor JM, Ho J, Mombereau C, Brewer A, Lowy E, Nicod J, Groszer M, Baban D, Sahgal N, Cazier JB, Ragoussis J, Davies KE, Geschwind DH, Fisher SE. Foxp2 regulates gene networks implicated in neurite outgrowth in the developing brain. PLoS Genet 2011; 7:e1002145. [PMID: 21765815 PMCID: PMC3131290 DOI: 10.1371/journal.pgen.1002145] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 05/07/2011] [Indexed: 11/19/2022] Open
Abstract
Forkhead-box protein P2 is a transcription factor that has been associated with intriguing aspects of cognitive function in humans, non-human mammals, and song-learning birds. Heterozygous mutations of the human FOXP2 gene cause a monogenic speech and language disorder. Reduced functional dosage of the mouse version (Foxp2) causes deficient cortico-striatal synaptic plasticity and impairs motor-skill learning. Moreover, the songbird orthologue appears critically important for vocal learning. Across diverse vertebrate species, this well-conserved transcription factor is highly expressed in the developing and adult central nervous system. Very little is known about the mechanisms regulated by Foxp2 during brain development. We used an integrated functional genomics strategy to robustly define Foxp2-dependent pathways, both direct and indirect targets, in the embryonic brain. Specifically, we performed genome-wide in vivo ChIP-chip screens for Foxp2-binding and thereby identified a set of 264 high-confidence neural targets under strict, empirically derived significance thresholds. The findings, coupled to expression profiling and in situ hybridization of brain tissue from wild-type and mutant mouse embryos, strongly highlighted gene networks linked to neurite development. We followed up our genomics data with functional experiments, showing that Foxp2 impacts on neurite outgrowth in primary neurons and in neuronal cell models. Our data indicate that Foxp2 modulates neuronal network formation, by directly and indirectly regulating mRNAs involved in the development and plasticity of neuronal connections.
Collapse
Affiliation(s)
- Sonja C. Vernes
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter L. Oliver
- Medical Research Council Functional Genetics Unit, University of Oxford, Oxford, United Kingdom
| | - Elizabeth Spiteri
- Program in Neurogenetics, Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Helen E. Lockstone
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Rathi Puliyadi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Jennifer M. Taylor
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Joses Ho
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Cedric Mombereau
- INSERM Institute du Fer à Moulin, University Pierre and Marie Curie, UMR-S 839, Paris, France
| | - Ariel Brewer
- INSERM Institute du Fer à Moulin, University Pierre and Marie Curie, UMR-S 839, Paris, France
| | - Ernesto Lowy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Jérôme Nicod
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Matthias Groszer
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- INSERM Institute du Fer à Moulin, University Pierre and Marie Curie, UMR-S 839, Paris, France
| | - Dilair Baban
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Natasha Sahgal
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Jean-Baptiste Cazier
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Jiannis Ragoussis
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Kay E. Davies
- Medical Research Council Functional Genetics Unit, University of Oxford, Oxford, United Kingdom
| | - Daniel H. Geschwind
- Program in Neurogenetics, Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
- Semel Institute and Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Simon E. Fisher
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
37
|
Choi EJ, Kim DH, Kim JG, Kim DY, Kim JD, Seol OJ, Jeong CS, Park JW, Choi MY, Kang SG, Costa ME, Ojeda SR, Lee BJ. Estrogen-dependent transcription of the NEL-like 2 (NELL2) gene and its role in protection from cell death. J Biol Chem 2010; 285:25074-84. [PMID: 20538601 PMCID: PMC2915743 DOI: 10.1074/jbc.m110.100545] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 06/01/2010] [Indexed: 11/06/2022] Open
Abstract
NELL2 (neural tissue-specific epidermal growth factor-like repeat domain-containing protein) is a secreted glycoprotein that is predominantly expressed in neural tissues. We reported previously that NELL2 mRNA abundance in brain is increased by estrogen (E2) treatment and that NELL2 is involved in the E2-dependent organization of a sexually dimorphic nucleus in the preoptic area. In this study we cloned the mouse NELL2 promoter and found it to contain two half-E2 response elements. Electrophoretic mobility shift assays and promoter assays showed that E2 and its receptors (ERalpha and ERbeta) stimulated NELL2 transcription by binding to the two half-E2 response elements. Hippocampal neuroprogenitor HiB5 cells expressing recombinant NELL2 showed increased cell survival under cell death-inducing conditions. Blockade of endogenous synthesis of NELL2 in HiB5 cells abolished the cell survival effect of E2 and resulted in a decrease in phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2). These data suggest that the NELL2 gene is trans-activated by E2 and contributes to mediating the survival promoting effects of E2 via intracellular signaling pathway of ERK.
Collapse
Affiliation(s)
- Eun Jung Choi
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Dong Hee Kim
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Jae Geun Kim
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Dong Yeol Kim
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Jung Dae Kim
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Ok Ju Seol
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Choon Soo Jeong
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Jeong Woo Park
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Min Young Choi
- the Department of Life Science and Applied Life Science, Gyeongsang National University, 900 Gajwa-dong, Jinju 660-701, South Korea
| | - Sung Goo Kang
- the School of Biotechnology and Biomedical Sciences, Inje University, Kimhae 621-749, South Korea, and
| | - Maria E. Costa
- the Division of Neuroscience, Oregon National Primate Research Center/Oregon Health and Science University, Beaverton, Oregon 97006
| | - Sergio R. Ojeda
- the Division of Neuroscience, Oregon National Primate Research Center/Oregon Health and Science University, Beaverton, Oregon 97006
| | - Byung Ju Lee
- From the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| |
Collapse
|
38
|
Horvat-Gordon M, Praul C, Ramachandran R, Bartell P, Leach, R. Use of microarray analysis to study gene expression in the avian epiphyseal growth plate. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2010; 5:12-23. [DOI: 10.1016/j.cbd.2009.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 08/31/2009] [Accepted: 08/31/2009] [Indexed: 12/15/2022]
|
39
|
Identification and characterization of a truncated isoform of NELL2. Biochem Biophys Res Commun 2010; 391:529-34. [DOI: 10.1016/j.bbrc.2009.11.092] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 11/13/2009] [Indexed: 11/20/2022]
|
40
|
Kamsteeg M, Jansen P, Van Vlijmen-Willems I, Van Erp P, Rodijk-Olthuis D, Van Der Valk P, Feuth T, Zeeuwen P, Schalkwijk J. Molecular diagnostics of psoriasis, atopic dermatitis, allergic contact dermatitis and irritant contact dermatitis. Br J Dermatol 2009; 162:568-78. [DOI: 10.1111/j.1365-2133.2009.09547.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Zhang T, Berrocal JG, Frizzell KM, Gamble MJ, DuMond ME, Krishnakumar R, Yang T, Sauve AA, Kraus WL. Enzymes in the NAD+ salvage pathway regulate SIRT1 activity at target gene promoters. J Biol Chem 2009; 284:20408-17. [PMID: 19478080 DOI: 10.1074/jbc.m109.016469] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In mammals, nicotinamide phosphoribosyltransferase (NAMPT) and nicotinamide mononucleotide adenylyltransferase 1 (NMNAT-1) constitute a nuclear NAD(+) salvage pathway which regulates the functions of NAD(+)-dependent enzymes such as the protein deacetylase SIRT1. One of the major functions of SIRT1 is to regulate target gene transcription through modification of chromatin-associated proteins. However, little is known about the molecular mechanisms by which NAD(+) biosynthetic enzymes regulate SIRT1 activity to control gene transcription in the nucleus. In this study we show that stable short hairpin RNA-mediated knockdown of NAMPT or NMNAT-1 in MCF-7 breast cancer cells reduces total cellular NAD(+) levels and alters global patterns of gene expression. Furthermore, we show that SIRT1 plays a key role in mediating the gene regulatory effects of NAMPT and NMNAT-1. Specifically, we found that SIRT1 binds to the promoters of genes commonly regulated by NAMPT, NMNAT-1, and SIRT1 and that SIRT1 histone deacetylase activity is regulated by NAMPT and NMNAT-1 at these promoters. Most significantly, NMNAT-1 interacts with, and is recruited to target gene promoters by SIRT1. Collectively, our results reveal a mechanism for the direct control of SIRT1 deacetylase activity at a set of target gene promoters by NMNAT-1. This mechanism, in collaboration with NAMPT-dependent regulation of nuclear NAD(+) production, establishes an important pathway for transcription regulation by NAD(+).
Collapse
Affiliation(s)
- Tong Zhang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
In vitro guidance of retinal axons by a tectal lamina-specific glycoprotein Nel. Mol Cell Neurosci 2009; 41:113-9. [PMID: 19249368 DOI: 10.1016/j.mcn.2009.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 12/31/2008] [Accepted: 02/06/2009] [Indexed: 11/20/2022] Open
Abstract
Nel is a glycoprotein containing five chordin-like and six epidermal growth factor-like domains and is strongly expressed in the nervous system. In this study, we have examined expression patterns and in vitro functions of Nel in the chicken retinotectal system. We have found that in the developing tectum, expression of Nel is localized in specific laminae that retinal axons normally do not enter, including the border between the retinorecipient and non-retinorecipient laminae. Nel-binding activity is detected on retinal axons both in vivo and in vitro, suggesting that retinal axons express a receptor for Nel. In vitro, Nel inhibits retinal axon outgrowth and induces growth cone collapse and axon retraction. These results indicate that Nel acts as an inhibitory guidance cue for retinal axons, and suggest its roles in the establishment of the lamina-specificity in the retinotectal projection.
Collapse
|
43
|
Fassunke J, Majores M, Tresch A, Niehusmann P, Grote A, Schoch S, Becker AJ. Array analysis of epilepsy-associated gangliogliomas reveals expression patterns related to aberrant development of neuronal precursors. ACTA ACUST UNITED AC 2008; 131:3034-50. [PMID: 18819986 DOI: 10.1093/brain/awn233] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gangliogliomas, the most frequent neoplasms in patients with pharmacoresistant focal epilepsies, are characterized by histological combinations of glial and dysplastic neuronal elements, a highly differentiated phenotype and rare gene mutations. Their molecular basis and relationship to other low-grade brain tumours are not completely understood. Systematic investigations of altered gene expression in gangliogliomas have been hampered by their cellular complexity, the lack of suitable control tissue and of sensitive expression profiling approaches. Here, we have used discrete microdissected ganglioglioma and adjacent control brain tissue obtained from the neurosurgical access to the tumour of identical patients (n = 6) carefully matched for equivalent glial and neuronal elements in an amount sufficient for oligonucleotide microarray hybridization without repetitive amplification. Multivariate statistical analysis identified a rich profile of genes with altered expression in gangliogliomas. Many differentially expressed transcripts related to intra- and intercellular signalling including protein kinase C and its target NELL2 in identical ganglioglioma cell components as determined by real-time quantitative RT-PCR (qRT-PCR) and in situ hybridization. We observed the LIM-domain-binding 2 (LDB2) transcript, critical for brain development during embryogenesis, as one of the strongest reduced mRNAs in gangliogliomas. Subsequent qRT-PCR in dysembryoplastic neuroepithelial tumours (n = 7) revealed partial expression similarities as well as marked differences from gangliogliomas. The demonstrated gene expression profile differentiates gangliogliomas from other low-grade primary brain tumours. shRNA-mediated silencing of LDB2 resulted in substantially aberrant dendritic arborization in cultured developing primary hippocampal neurons. The present data characterize novel molecular mechanisms operating in gangliogliomas that contribute to the development of dysplastic neurons and an aberrant neuronal network.
Collapse
Affiliation(s)
- Jana Fassunke
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Hwang EM, Kim DG, Lee BJ, Choi J, Kim E, Park N, Kang D, Han J, Choi WS, Hong SG, Park JY. Alternative splicing generates a novel non-secretable cytosolic isoform of NELL2. Biochem Biophys Res Commun 2007; 353:805-11. [PMID: 17196548 DOI: 10.1016/j.bbrc.2006.12.115] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Accepted: 12/11/2006] [Indexed: 11/21/2022]
Abstract
NELL2 is as a neuron-specific secreted glycoprotein. The present study provides evidence of an alternatively spliced variant of the rat NELL2 gene that yields cytosolic NELL2 (cNELL2). cNELL2 was initially detected in the thymus and subsequently found to be ubiquitously expressed in many other tissues. The absence of the sequences corresponding to the third exon, which contains the terminal portion of the signal peptide, accounts for the uniform distribution of cNELL2 throughout the cytoplasm. This is in contrast to NELL2, which is preferentially located at distinct subcellular structures involved in the secretary process, such as endoplasmic reticulum and Golgi apparatus. Western blot analysis showed that cNELL2 was not present in the medium but only in lysates, while NELL2 was detected as a glycosylated larger form in both lysates and media. Immunoprecipitation analysis revealed that cNELL2 interacts with PKCbeta1. These results suggest that cNELL2 is involved in PKCbeta1-mediated intracellular signaling.
Collapse
Affiliation(s)
- Eun Mi Hwang
- Department of Neurobiology, Institute of Health Science, and Medical Research Center for Neural Dysfunction, Gyeongsang National University College of Medicine, 90 Chilam-Dong, Jinju 660-751, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Eng SR, Dykes IM, Lanier J, Fedtsova N, Turner EE. POU-domain factor Brn3a regulates both distinct and common programs of gene expression in the spinal and trigeminal sensory ganglia. Neural Dev 2007; 2:3. [PMID: 17239249 PMCID: PMC1796875 DOI: 10.1186/1749-8104-2-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2006] [Accepted: 01/19/2007] [Indexed: 12/05/2022] Open
Abstract
Background General somatic sensation is conveyed to the central nervous system at cranial levels by the trigeminal ganglion (TG), and at spinal levels by the dorsal root ganglia (DRG). Although these ganglia have similar functions, they have distinct embryological origins, in that both contain neurons originating from the neural crest, while only the TG includes cells derived from the placodal ectoderm. Results Here we use microarray analysis of E13.5 embryos to demonstrate that the developing DRG and TG have very similar overall patterns of gene expression. In mice lacking the POU-domain transcription factor Brn3a, the DRG and TG exhibit many common changes in gene expression, but a subset of Brn3a target genes show increased expression only in the TG. In the wild-type TG these Brn3a-repressed genes are silent, yet their promoter regions exhibit histone H3-acetylation levels similar to constitutively transcribed gene loci. This increased H3-acetylation is not observed in the DRG, suggesting that chromatin modifications play a role in cell-specific target gene regulation by Brn3a. Conclusion These results demonstrate that one developmental role of Brn3a is to repress potential differences in gene expression between sensory neurons generated at different axial levels, and to regulate a convergent program of developmental gene expression, in which functionally similar populations of neurons are generated from different embryological substrates.
Collapse
Affiliation(s)
- S Raisa Eng
- Department of Psychiatry, University of California, San Diego and VA San Diego Healthcare System, Gilman Drive, La Jolla, CA 92093-0603, USA
| | - Iain M Dykes
- Department of Psychiatry, University of California, San Diego and VA San Diego Healthcare System, Gilman Drive, La Jolla, CA 92093-0603, USA
| | - Jason Lanier
- Department of Psychiatry, University of California, San Diego and VA San Diego Healthcare System, Gilman Drive, La Jolla, CA 92093-0603, USA
| | - Natalia Fedtsova
- Department of Psychiatry, University of California, San Diego and VA San Diego Healthcare System, Gilman Drive, La Jolla, CA 92093-0603, USA
| | - Eric E Turner
- Department of Psychiatry, University of California, San Diego and VA San Diego Healthcare System, Gilman Drive, La Jolla, CA 92093-0603, USA
| |
Collapse
|
46
|
Nelson BR, Hartman BH, Georgi SA, Lan MS, Reh TA. Transient inactivation of Notch signaling synchronizes differentiation of neural progenitor cells. Dev Biol 2007; 304:479-98. [PMID: 17280659 PMCID: PMC1979095 DOI: 10.1016/j.ydbio.2007.01.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 12/23/2006] [Accepted: 01/02/2007] [Indexed: 11/19/2022]
Abstract
In the developing nervous system, the balance between proliferation and differentiation is critical to generate the appropriate numbers and types of neurons and glia. Notch signaling maintains the progenitor pool throughout this process. While many components of the Notch pathway have been identified, the downstream molecular events leading to neural differentiation are not well understood. We have taken advantage of a small molecule inhibitor, DAPT, to block Notch activity in retinal progenitor cells, and analyzed the resulting molecular and cellular changes over time. DAPT treatment causes a massive, coordinated differentiation of progenitors that produces cell types appropriate for their developmental stage. Transient exposure of retina to DAPT for specific time periods allowed us to define the period of Notch inactivation that is required for a permanent commitment to differentiate. Inactivation of Notch signaling revealed a cascade of proneural bHLH transcription factor gene expression that correlates with stages in progenitor cell differentiation. Microarray/QPCR analysis confirms the changes in Notch signaling components, and reveals new molecular targets for investigating neuronal differentiation. Thus, transient inactivation of Notch signaling synchronizes progenitor cell differentiation, and allows for a systematic analysis of key steps in this process.
Collapse
Affiliation(s)
- Branden R. Nelson
- Department of Biological Structure, University of Washington, Seattle, WA 98195
| | - Byron H. Hartman
- Department of Biological Structure, University of Washington, Seattle, WA 98195
| | - Sean A. Georgi
- Neurobiology and Behavior Program, University of Washington, Seattle, WA 98195
| | - Michael S. Lan
- The Research Institute for Children, Children's Hospital, New Orleans, LA 70118
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, WA 98195
- Neurobiology and Behavior Program, University of Washington, Seattle, WA 98195
- Author for correspondence: Dr. T.A. Reh, Department of Biological Structure, Box 357420, University of Washington, Seattle, WA 98195, , phone 206-543-8043, fax 206-543-1524
| |
Collapse
|
47
|
Matsuyama S, Doe N, Kurihara N, Tanizawa K, Kuroda S, Iso H, Horie M. Spatial learning of mice lacking a neuron-specific epidermal growth factor family protein, NELL2. J Pharmacol Sci 2005; 98:239-43. [PMID: 15988128 DOI: 10.1254/jphs.fp0050211] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
NELL2 is a neuron-specific thrombospondin-1-like extracellular protein containing six epidermal growth factor-like domains. We previously disrupted the NELL2 gene in mice by gene targeting and showed that long-term potentiation is enhanced in vivo in the dentate gyrus of NELL2-deficient mice. To further elucidate the physiological roles of NELL2, we performed a behavioral characterization of NELL2(-/-) and their heterozygous control mice. NELL2-deficient mice exhibited learning impairment in the Morris water maze task. However, we observed no difference in passive avoidance learning between NELL2(-/-) and NELL2(+/-) mice. These observations suggest that NELL2 plays an important role in hippocampus-dependent spatial learning and that emotional learning does not depend critically on NELL2.
Collapse
Affiliation(s)
- Shogo Matsuyama
- Division of Molecular Pharmacology and Pharmacogenomics, Department of Genome Sciences, Kobe University Graduate School of Medicine, Hyogo, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Thippeswamy T, McKay JS, Quinn J, Morris R. Either nitric oxide or nerve growth factor is required for dorsal root ganglion neurons to survive during embryonic and neonatal development. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 154:153-64. [PMID: 15707669 DOI: 10.1016/j.devbrainres.2004.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2004] [Revised: 10/20/2004] [Accepted: 10/21/2004] [Indexed: 12/24/2022]
Abstract
During early embryonic (E12) development almost all dorsal root ganglion (DRG) neurons express the neuronal isoform of nitric oxide synthase (nNOS). At this stage, the axons of these neurons are rudimentary and have not made contact with peripheral tissue targets. As their axons establish contact with peripheral targets such as the skin, the number of neurons expressing nNOS decrease that correspond to increased immunoreactivity for nerve growth factor (NGF) in the skin, and its high affinity receptor, tyrosine kinase A (trkA) in both skin and DRG neurons. During late postnatal development, very few DRG neurons express nNOS; however, axotomy or NGF deprivation of cultured DRG neurons induce nNOS and NOS blockade causes neuronal death. In contrast, NGF-deprived embryonic and neonatal DRG neurons die by apoptosis, while NOS blockade has no effect. Overall, these observations suggest that NGF and nitric oxide (NO) interact during embryonic and postnatal development to facilitate neuronal selection and survival. The roles of NO, NGF and its receptor trkA in DRG neurons during different stages of development are discussed.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Apoptosis/physiology
- Axotomy/methods
- Cell Count/methods
- Cell Survival/drug effects
- Cell Survival/physiology
- Cells, Cultured
- Embryo, Mammalian
- Enzyme Inhibitors/pharmacology
- Female
- Ganglia, Spinal/cytology
- Ganglia, Spinal/embryology
- Ganglia, Spinal/growth & development
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/physiology
- Immunohistochemistry/methods
- In Situ Nick-End Labeling/methods
- Male
- Models, Biological
- NG-Nitroarginine Methyl Ester/pharmacology
- Nerve Growth Factor/metabolism
- Neural Inhibition/drug effects
- Neurons/drug effects
- Neurons/metabolism
- Nitric Oxide/metabolism
- Pregnancy
- Proto-Oncogene Proteins c-jun/metabolism
- Rats
- Rats, Wistar
- Receptor, trkA/metabolism
Collapse
|