1
|
Guo Q, Wang Y, Wang Q, Qian Y, Jiang Y, Dong X, Chen H, Chen X, Liu X, Yu S, Zhu J, Shan S, Wu B, Zhou W, Wang H. In the developing cerebral cortex: axonogenesis, synapse formation, and synaptic plasticity are regulated by SATB2 target genes. Pediatr Res 2023; 93:1519-1527. [PMID: 36028553 DOI: 10.1038/s41390-022-02260-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Special AT-rich sequence-binding protein 2 is essential for the development of cerebral cortex and key molecular node for the establishment of proper neural circuitry and function. Mutations in the SATB2 gene lead to SATB2-associated syndrome, which is characterized by abnormal development of skeleton and central nervous systems. METHODS We generated Satb2 knockout mouse model through CRISPR-Cas9 technology and performed RNA-seq and ChIP-seq of embryonic cerebral cortex. We conducted RT-qPCR, western blot, immunofluorescence staining, luciferase reporter assay and behavioral analysis for experimental verification. RESULTS We identified 1363 downstream effector genes of Satb2 and correlation analysis of Satb2-targeted genes and neurological disease genes showed that Satb2 contribute to cognitive and mental disorders from the early developmental stage. We found that Satb2 directly regulate the expression of Ntng1, Cdh13, Kitl, genes important for axon guidance, synaptic formation, neuron migration, and Satb2 directly activates the expression of Mef2c. We also showed that Satb2 heterozygous knockout mice showed impaired spatial learning and memory. CONCLUSIONS Taken together, our study supportsroles of Satb2 in the regulation of axonogenesis and synaptic formation at the early developmental stage and provides new insights into the complicated regulatory mechanism of Satb2 and new evidence to elucidate the pathogen of SATB2-associated syndrome. IMPACT 1363 downstream effector genes of Satb2 were classified into 5 clusters with different temporal expression patterns. We identified Plxnd1, Ntng1, Efnb2, Ephb1, Plxna2, Epha3, Plxna4, Unc5c, and Flrt2 as axon guidance molecules to regulate axonogenesis. 168 targeted genes of Satb2 were found to regulate synaptic formation in the early development of the cerebral cortex. Transcription factor Mef2c is positively regulated by Satb2, and 28 Mef2c-targeted genes can be directly regulated by Satb2. In the Morris water maze test, Satb2+/- mice showed impaired spatial learning and memory, further strengthening that Satb2 can regulate synaptic functions.
Collapse
Affiliation(s)
- Qiufang Guo
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
- Berry Genomics Co, 102206, Beijing, China
| | - Yaqiong Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Qing Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Yanyan Qian
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Yinmo Jiang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Xinran Dong
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Huiyao Chen
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Xiang Chen
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Xiuyun Liu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Sha Yu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Jitao Zhu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Shifang Shan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 200032, Shanghai, China
| | - Bingbing Wu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Wenhao Zhou
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China.
- Division of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Key Laboratory of Neonatal Diseases, Ministry of Health, 201102, Shanghai, China.
| | - Huijun Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China.
| |
Collapse
|
2
|
BARS Influences Neuronal Development by Regulation of Post-Golgi Trafficking. Cells 2022; 11:cells11081320. [PMID: 35455998 PMCID: PMC9026897 DOI: 10.3390/cells11081320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/28/2022] [Accepted: 04/08/2022] [Indexed: 02/01/2023] Open
Abstract
Neurons are highly polarized cells requiring precise regulation of trafficking and targeting of membrane proteins to generate and maintain different and specialized compartments, such as axons and dendrites. Disruption of the Golgi apparatus (GA) secretory pathway in developing neurons alters axon/dendritic formation. Therefore, detailed knowledge of the mechanisms underlying vesicles exiting from the GA is crucial for understanding neuronal polarity. In this study, we analyzed the role of Brefeldin A-Ribosylated Substrate (CtBP1-S/BARS), a member of the C-terminal-binding protein family, in the regulation of neuronal morphological polarization and the exit of membrane proteins from the Trans Golgi Network. Here, we show that BARS is expressed during neuronal development in vitro and that RNAi suppression of BARS inhibits axonal and dendritic elongation in hippocampal neuronal cultures as well as largely perturbed neuronal migration and multipolar-to-bipolar transition during cortical development in situ. In addition, using plasma membrane (PM) proteins fused to GFP and engineered with reversible aggregation domains, we observed that expression of fission dominant-negative BARS delays the exit of dendritic and axonal membrane protein-containing carriers from the GA. Taken together, these data provide the first set of evidence suggesting a role for BARS in neuronal development by regulating post-Golgi membrane trafficking.
Collapse
|
3
|
Aberrant Axo-Axonic Synaptic Reorganization in the Phosphorylated L1-CAM/Calcium Channel Subunit α2δ-1-Containing Central Terminals of Injured c-Fibers in the Spinal Cord of a Neuropathic Pain Model. eNeuro 2021; 8:ENEURO.0499-20.2021. [PMID: 33500315 PMCID: PMC8174056 DOI: 10.1523/eneuro.0499-20.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/25/2020] [Accepted: 01/08/2021] [Indexed: 12/04/2022] Open
Abstract
In the dorsal horn of the spinal cord, peripheral nerve injury induces structural and neurochemical alterations through which aberrant synaptic signals contribute to the formation of neuropathic pain. However, the role of injured primary afferent terminals in such plastic changes remain unclear. In this study, we investigated the effect of nerve injury on the morphology of cell adhesion molecule L1-CAM [total L1-CAM (tL1-CAM)]-positive primary afferent terminals and on the synaptic contact pattern in the dorsal horn. In the confocal images, the tL1-CAM-positive terminals showed morphologic changes leading to the formation of hypertrophic varicosities in the c-fiber terminal. These hypertrophic varicosities in the dorsal horn were co-labeled with phosphorylated (Ser1181) L1-CAM (pL1-CAM) and shown to store neurotransmitter peptides, but not when co-labeled with the presynaptic marker, synaptophysin. Quantitative analyses based on 3D-reconstructed confocal images revealed that peripheral nerve injury reduced dendritic synaptic contacts but promoted aberrant axo-axonic contacts on the tL1-CAM-positive hypertrophic varicosities. These tL1-CAM-positive varicosities co-expressed the injury-induced α2δ−1 subunit of the calcium channel in the dorsal horn. Administration of the anti-allodynic drug, pregabalin, inhibited accumulation of α2δ−1 and pL1-CAM associated with a reduction in hypertrophic changes of tL1-CAM-positive varicosities, and normalized injury-induced alterations in synaptic contacts in the dorsal horn. Our findings highlight the formation of aberrant spinal circuits that mediate the convergence of local neuronal signals onto injured c-fibers, suggesting that these hypertrophic varicosities may be important contributors to the pathologic mechanisms underlying neuropathic pain.
Collapse
|
4
|
Lin J, Chooi WH, Ong W, Zhang N, Bechler ME, Ffrench-Constant C, Chew SY. Oriented and sustained protein expression on biomimicking electrospun fibers for evaluating functionality of cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111407. [PMID: 33255010 DOI: 10.1016/j.msec.2020.111407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 11/25/2022]
Abstract
A proper protein orientation is often required in order to achieve specific protein-receptor interaction to elicit a desired biological response. Here, we present a Protein A-based biomimicking platform that is capable of efficiently orienting proteins for evaluating cellular behaviour. By absorbing Protein A onto aligned bio-mimicking polycaprolactone (PCL) fibers, we demonstrate that protein binding could be retained on these fibers for at least 7 days under physiologically relevant conditions. We further show that Protein A served as a molecular orientor to arrange the recombinant proteins in similar orientations. Such protein-orienting scaffolds were further verified to be biologically functional by using sensitive primary rat cortical neurons (CNs) and oligodendrocyte progenitor cells (OPCs), as model neural cells for a stringent proof of concept. Specifically, CNs that were seeded on fibers coated with Protein A and a known enhancer of neurite growth (L1 Cell Adhesion Molecular L1CAM) displayed the longest total neurite length (462.77 ± 100.79 μm, p < 0.001) as compared to the controls. Besides that, OPCs seeded on fibers coated with Protein A and Neuregulin-1 Type III (Nrg1 type III) (myelin enhancer) produced the longest myelin ensheathment length (19.8 ± 11.69 μm). These results demonstrate the efficacy of this platform for protein screening applications.
Collapse
Affiliation(s)
- Junquan Lin
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Wai Hon Chooi
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - William Ong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore; NTU Institute for Health Technologies (Health Tech NTU), Interdisciplinary Graduate School, Nanyang Technological University, Singapore 637533, Singapore
| | - Na Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Marie E Bechler
- MRC-Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH164UU, UK
| | - Charles Ffrench-Constant
- MRC-Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH164UU, UK
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.
| |
Collapse
|
5
|
Chen S, Jiang Q, Huang P, Hu C, Shen H, Schachner M, Zhao W. The L1 cell adhesion molecule affects protein kinase D1 activity in the cerebral cortex in a mouse model of Alzheimer's disease. Brain Res Bull 2020; 162:141-150. [PMID: 32540419 DOI: 10.1016/j.brainresbull.2020.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 02/05/2023]
Abstract
Alzheimer's disease (AD) is characterized by deposition of β-amyloid protein (Aβ), neurofibrillary tangles and cognitive deficits resulting from neuronal cell death. In search for the molecular underpinnings of the disease, we were interested in the relationship between Aβ, L1 cell adhesion molecule and protein kinase D1 (PKD1), which are not only implicated in neural development and functional maintenance in the adult, but are also neuroprotective under pathological conditions. Based on our observations that L1 and phosphorylated, i.e. activated, protein kinase PKD1 (pPKD1) co-localize in cultured neurons, we investigated the functional relationship between L1 and pPKD1 in the frontal lobe of an AD human cortical tissue microarray, and found increased and positively correlating levels of both molecules when compared to a non-affected human brain. Also in the APPSWE mouse model of AD, L1 and pPKD1 levels were increased in the frontal lobe. To investigate whether L1 influences PKD1-based functions in AD, cultured cortical neurons were stressed with either H2O2 or oligomeric Aβ1-42, in the presence or absence of recombinant L1 extracellular domain, and PKD1 phosphorylation was measured. As indicated by the cell viability assay, L1 maintained neuronal survival under oxidative stress and under application of oligomeric Aβ1-42, when PKD1 activity was inhibited, suggesting that L1 ameliorates some aspects of Aβ1-42 pathology in parallel with reducing PKD1 function.
Collapse
Affiliation(s)
- Shuangxi Chen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China; The First Affiliated Hospital of University of South China, University of South China, No. 69, Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China
| | - Qiong Jiang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Peizhi Huang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Chengliang Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Huifan Shen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China; Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
| | - Weijiang Zhao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China.
| |
Collapse
|
6
|
Yang WK, Chien CT. Beyond being innervated: the epidermis actively shapes sensory dendritic patterning. Open Biol 2020; 9:180257. [PMID: 30914004 PMCID: PMC6451362 DOI: 10.1098/rsob.180257] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sensing environmental cues requires well-built neuronal circuits linked to the body surface. Sensory neurons generate dendrites to innervate surface epithelium, thereby making it the largest sensory organ in the body. Previous studies have illustrated that neuronal type, physiological function and branching patterns are determined by intrinsic factors. Perhaps for effective sensation or protection, sensory dendrites bind to or are surrounded by the substrate epidermis. Recent studies have shed light on the mechanisms by which dendrites interact with their substrates. These interactions suggest that substrates can regulate dendrite guidance, arborization and degeneration. In this review, we focus on recent studies of Drosophila and Caenorhabditis elegans that demonstrate how epidermal cells can regulate dendrites in several aspects.
Collapse
Affiliation(s)
- Wei-Kang Yang
- Institute of Molecular Biology, Academia Sinica , Taipei 115 , Taiwan
| | - Cheng-Ting Chien
- Institute of Molecular Biology, Academia Sinica , Taipei 115 , Taiwan
| |
Collapse
|
7
|
Halakos EG, Connell AJ, Glazewski L, Wei S, Mason RW. Bottom up proteomics reveals novel differentiation proteins in neuroblastoma cells treated with 13-cis retinoic acid. J Proteomics 2019; 209:103491. [PMID: 31472280 DOI: 10.1016/j.jprot.2019.103491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/15/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022]
Abstract
Neuroblastoma, a cancer of the sympathetic nervous system, is the second most common pediatric cancer. A unique feature of neuroblastoma is remission in some patients due to spontaneous differentiation of metastatic tumors. 13-cis retinoic acid (13-cis RA) is currently used in the clinic to treat neuroblastoma due to its differentiation inducing effects. In this study, we used shotgun proteomics to identify proteins affected by 13-cis RA treatment in neuroblastoma SK-N-SH cells. Our results showed that 13-cis RA reduced proteins involved in extracellular matrix synthesis and organization and increased proteins involved in cell adhesion and neurofilament formation. These changes indicate that 13-cis RA induces tumor cell differentiation by decreasing extracellular matrix rigidity and increasing neurite overgrowth. Differentially-affected proteins identified in this study may be novel biomarkers of drug efficacy in the treatment of neuroblastoma. SIGNIFICANCE: As neuroblastoma can spontaneously differentiate, determining which proteins are involved in differentiation can guide development of novel treatments. 13-cis retinoic acid is currently used in the clinic as a differentiation inducer. Here we have established a proteome map of SK-N-SH cells treated with 13-cis retinoic acid. Bioinformatic analysis revealed the involvement of development, differentiation, extracellular matrix assembly, collagen biosynthesis, and neurofilament bundle association. This proteome map provides information as to which proteins are important for differentiation and identifies networks that can be targeted by drugs to treat neuroblastoma [1].
Collapse
Affiliation(s)
- Effie G Halakos
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Andrew J Connell
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Lisa Glazewski
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Robert W Mason
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
8
|
Zhao W, Tan J, Zhu T, Ou J, Li Y, Shen L, Wu H, Han L, Liu Y, Jia X, Bai T, Li H, Ke X, Zhao J, Zou X, Hu Z, Guo H, Xia K. Rare inherited missense variants of POGZ associate with autism risk and disrupt neuronal development. J Genet Genomics 2019; 46:247-257. [PMID: 31196716 DOI: 10.1016/j.jgg.2019.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 03/22/2019] [Accepted: 04/02/2019] [Indexed: 11/16/2022]
Abstract
Excess de novo likely gene-disruptive and missense variants within dozens of genes have been identified in autism spectrum disorder (ASD) and other neurodevelopmental disorders. However, many rare inherited missense variants of these high-risk genes have not been thoroughly evaluated. In this study, we analyzed the rare missense variant burden of POGZ in a large cohort of ASD patients from the Autism Clinical and Genetic Resources in China (ACGC) and further dissected the functional effect of disease-associated missense variants on neuronal development. Our results showed a significant burden of rare missense variants in ASD patients compared to the control population (P = 4.6 × 10-5, OR = 3.96), and missense variants in ASD patients showed more severe predicted functional outcomes than those in controls. Furthermore, by leveraging published large-scale sequencing data of neurodevelopmental disorders (NDDs) and sporadic case reports, we identified 8 de novo missense variants of POGZ in NDD patients. Functional analysis revealed that two inherited, but not de novo, missense variants influenced the cellular localization of POGZ and failed to rescue the defects in neurite and dendritic spine development caused by Pogz knockdown in cultured mouse primary cortical neurons. Significantly, L1CAM, an autism candidate risk gene, is differentially expressed in POGZ deficient cell lines. Reduced expression of L1cam was able to partially rescue the neurite length defects caused by Pogz knockdown. Our study showed the important roles of rare inherited missense variants of POGZ in ASD risk and neuronal development and identified the potential downstream targets of POGZ, which are important for further molecular mechanism studies.
Collapse
Affiliation(s)
- Wenjing Zhao
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Jieqiong Tan
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Tengfei Zhu
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Jianjun Ou
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ying Li
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Lu Shen
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Huidan Wu
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Lin Han
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Yanling Liu
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Xiangbin Jia
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Ting Bai
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Honghui Li
- Key Laboratory of Developmental Disorders in Children, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, 545001, China
| | - Xiaoyan Ke
- Child Mental Health Research Center, Nanjing Brain Hospital Affiliated of Nanjing Medical University, Nanjing, 210029, China
| | - Jingping Zhao
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xiaobing Zou
- Children Development Behavior Center of the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zhengmao Hu
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China
| | - Hui Guo
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China; Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Kun Xia
- Center of Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, China; School of Life Sciences and Technology, Xinjiang University, Ürümqi, 830046, China; CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, 200030, China.
| |
Collapse
|
9
|
Ye X, Qiu Y, Gao Y, Wan D, Zhu H. A Subtle Network Mediating Axon Guidance: Intrinsic Dynamic Structure of Growth Cone, Attractive and Repulsive Molecular Cues, and the Intermediate Role of Signaling Pathways. Neural Plast 2019; 2019:1719829. [PMID: 31097955 PMCID: PMC6487106 DOI: 10.1155/2019/1719829] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 01/01/2023] Open
Abstract
A fundamental feature of both early nervous system development and axon regeneration is the guidance of axonal projections to their targets in order to assemble neural circuits that control behavior. In the navigation process where the nerves grow toward their targets, the growth cones, which locate at the tips of axons, sense the environment surrounding them, including varies of attractive or repulsive molecular cues, then make directional decisions to adjust their navigation journey. The turning ability of a growth cone largely depends on its highly dynamic skeleton, where actin filaments and microtubules play a very important role in its motility. In this review, we summarize some possible mechanisms underlying growth cone motility, relevant molecular cues, and signaling pathways in axon guidance of previous studies and discuss some questions regarding directions for further studies.
Collapse
Affiliation(s)
- Xiyue Ye
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yan Qiu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yuqing Gao
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Dong Wan
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Huifeng Zhu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| |
Collapse
|
10
|
Sahu S, Zhang Z, Li R, Hu J, Shen H, Loers G, Shen Y, Schachner M. A Small Organic Compound Mimicking the L1 Cell Adhesion Molecule Promotes Functional Recovery after Spinal Cord Injury in Zebrafish. Mol Neurobiol 2018; 55:859-878. [PMID: 28070857 DOI: 10.1007/s12035-016-0254-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 10/24/2016] [Indexed: 02/05/2023]
Abstract
Tacrine is a small organic compound that was discovered to mimic the functions of the neural cell adhesion molecule L1 by promoting the cognate functions of L1 in vitro, such as neuronal survival, neuronal migration, neurite outgrowth, and myelination. Based on studies indicating that L1 enhances functional recovery in different central and peripheral nervous system disease paradigms of rodents, it deemed interesting to investigate the beneficial role of tacrine in the attractive zebrafish animal model, by evaluating functional recovery after spinal cord injury. To this aim, larval and adult zebrafish were exposed to tacrine treatment after spinal cord injury and monitored for locomotor recovery and axonal regrowth. Tacrine promoted the rapid recovery of locomotor activities in both larval and adult zebrafish, enhanced regrowth of severed axons and myelination, and reduced astrogliosis in the spinal cords. Tacrine treatment upregulated the expression of L1.1 (a homolog of the mammalian recognition molecule L1) and enhanced the L1.1-mediated intracellular signaling cascades in the injured spinal cords. These observations lead to the hope that, in combination with other therapeutic approaches, this old drug may become a useful reagent to ameliorate the deficits resulting from acute and chronic injuries of the mammalian nervous system.
Collapse
Affiliation(s)
- Sudhanshu Sahu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Zhihua Zhang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Rong Li
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Junkai Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Huifan Shen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Gabriele Loers
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Yanqin Shen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
- Medical School, Jiangnan University, 1800 Li Hu Road, Wuxi, Jiangsu, 214122, China.
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08554, USA.
| |
Collapse
|
11
|
Morales DM, Silver SA, Morgan CD, Mercer D, Inder TE, Holtzman DM, Wallendorf MJ, Rao R, McAllister JP, Limbrick DD. Lumbar Cerebrospinal Fluid Biomarkers of Posthemorrhagic Hydrocephalus of Prematurity: Amyloid Precursor Protein, Soluble Amyloid Precursor Protein α, and L1 Cell Adhesion Molecule. Neurosurgery 2017; 80:82-90. [PMID: 27571524 DOI: 10.1227/neu.0000000000001415] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 06/15/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intraventricular hemorrhage (IVH) is the most frequent, severe neurological complication of prematurity and is associated with posthemorrhagic hydrocephalus (PHH) in up to half of cases. PHH requires lifelong neurosurgical care and is associated with significant cognitive and psychomotor disability. Cerebrospinal fluid (CSF) biomarkers may provide both diagnostic information for PHH and novel insights into its pathophysiology. OBJECTIVE To explore the diagnostic ability of candidate CSF biomarkers for PHH. METHODS Concentrations of amyloid precursor protein (APP), soluble APPα (sAPPα), soluble APPβ, neural cell adhesion molecule-1 (NCAM-1), L1 cell adhesion molecule (L1CAM), tau, phosphorylated tau, and total protein (TP) were measured in lumbar CSF from neonates in 6 groups: (1) no known neurological disease (n = 33); (2) IVH grades I to II (n = 13); (3) IVH grades III to IV (n = 12); (4) PHH (n = 12); (5) ventricular enlargement without hydrocephalus (n = 10); and (6) hypoxic ischemic encephalopathy (n = 13). CSF protein levels were compared using analysis of variance, and logistic regression was performed to examine the predictive ability of each marker for PHH. RESULTS Lumbar CSF levels of APP, sAPPα, L1CAM, and TP were selectively increased in PHH compared with all other conditions (all P < .001). The sensitivity, specificity, and odds ratios of candidate CSF biomarkers for PHH were determined for APP, sAPPα, and L1CAM; cut points of 699, 514, and 113 ng/mL yielded odds ratios for PHH of 80.0, 200.0, and 68.75, respectively. CONCLUSION Lumbar CSF APP, sAPPα, L1CAM, and TP were selectively increased in PHH. These proteins, and sAPPα, in particular, hold promise as biomarkers of PHH and provide novel insight into PHH-associated neural injury and repair.
Collapse
Affiliation(s)
- Diego M Morales
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Shawgi A Silver
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Clinton D Morgan
- Depart-ment of Neurological Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Deanna Mercer
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Terri E Inder
- Department of Pediatrics, Harvard University School of Medicine, Boston, Massachusetts
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Hope Center for Neurological Disorders, Washington University School of Medi-cine, St. Louis, Missouri.,Knight Alzheimer's Disease Research Center, Washing-ton University School of Medicine, St. Louis, Missouri
| | - Michael J Wallendorf
- Division of Biostat-istics, Washington University School of Medicine, St. Louis, Missouri
| | - Rakesh Rao
- Depart-ment of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - James P McAllister
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - David D Limbrick
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri.,Hope Center for Neurological Disorders, Washington University School of Medi-cine, St. Louis, Missouri.,Depart-ment of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
12
|
Kyung JW, Cho IH, Lee S, Song WK, Ryan TA, Hoppa MB, Kim SH. Adaptor Protein 2 (AP-2) complex is essential for functional axogenesis in hippocampal neurons. Sci Rep 2017; 7:41620. [PMID: 28139716 PMCID: PMC5282494 DOI: 10.1038/srep41620] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/21/2016] [Indexed: 11/18/2022] Open
Abstract
The complexity and diversity of a neural network requires regulated elongation and branching of axons, as well as the formation of synapses between neurons. In the present study we explore the role of AP-2, a key endocytic adaptor protein complex, in the development of rat hippocampal neurons. We found that the loss of AP-2 during the early stage of development resulted in impaired axon extension and failed maturation of the axon initial segment (AIS). Normally the AIS performs two tasks in concert, stabilizing neural polarity and generating action potentials. In AP-2 silenced axons polarity is established, however there is a failure to establish action potential firing. Consequently, this impairs activity-driven Ca2+ influx and exocytosis at nerve terminals. In contrast, removal of AP-2 from older neurons does not impair axonal growth or signaling and synaptic function. Our data reveal that AP-2 has important roles in functional axogenesis by proper extension of axon as well as the formation of AIS during the early step of neurodevelopment.
Collapse
Affiliation(s)
- Jae Won Kyung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - In Ha Cho
- Department of Biology, Molecular Cellular Biology Program, Dartmouth College, Hanover, NH, 03755, USA
| | - Sukmook Lee
- Laboratory of Molecular Cancer Therapeutics, Scripps Korea Antibody Institute, Chuncheon, 24341, South Korea
| | - Woo Keun Song
- School of Life Science, Bioimaging Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, South Korea
| | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Michael B Hoppa
- Department of Biology, Molecular Cellular Biology Program, Dartmouth College, Hanover, NH, 03755, USA
| | - Sung Hyun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, South Korea.,Department of Physiology, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, 02447, South Korea
| |
Collapse
|
13
|
Morales DM, Holubkov R, Inder TE, Ahn HC, Mercer D, Rao R, McAllister JP, Holtzman DM, Limbrick DD. Cerebrospinal fluid levels of amyloid precursor protein are associated with ventricular size in post-hemorrhagic hydrocephalus of prematurity. PLoS One 2015; 10:e0115045. [PMID: 25738507 PMCID: PMC4349693 DOI: 10.1371/journal.pone.0115045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/18/2014] [Indexed: 02/03/2023] Open
Abstract
Background Neurological outcomes of preterm infants with post-hemorrhagic hydrocephalus (PHH) remain among the worst in infancy, yet there remain few instruments to inform the treatment of PHH. We previously observed PHH-associated elevations in cerebrospinal fluid (CSF) amyloid precursor protein (APP), neural cell adhesion molecule-L1 (L1CAM), neural cell adhesion molecule-1 (NCAM-1), and other protein mediators of neurodevelopment. Objective The objective of this study was to examine the association of CSF APP, L1CAM, and NCAM-1 with ventricular size as an early step toward developing CSF markers of PHH. Methods CSF levels of APP, L1CAM, NCAM-1, and total protein (TP) were measured in 12 preterm infants undergoing PHH treatment. Ventricular size was determined using cranial ultrasounds. The relationships between CSF APP, L1CAM, and NCAM-1, occipitofrontal circumference (OFC), volume of CSF removed, and ventricular size were examined using correlation and regression analyses. Results CSF levels of APP, L1CAM, and NCAM-1 but not TP paralleled treatment-related changes in ventricular size. CSF APP demonstrated the strongest association with ventricular size, estimated by frontal-occipital horn ratio (FOR) (Pearson R = 0.76, p = 0.004), followed by NCAM-1 (R = 0.66, p = 0.02) and L1CAM (R = 0.57,p = 0.055). TP was not correlated with FOR (R = 0.02, p = 0.95). Conclusions Herein, we report the novel observation that CSF APP shows a robust association with ventricular size in preterm infants treated for PHH. The results from this study suggest that CSF APP and related proteins at once hold promise as biomarkers of PHH and provide insight into the neurological consequences of PHH in the preterm infant.
Collapse
Affiliation(s)
- Diego M. Morales
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
- * E-mail:
| | - Richard Holubkov
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Terri E. Inder
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Haejun C. Ahn
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Deanna Mercer
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Rakesh Rao
- Department of Newborn Medicine, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - James P. McAllister
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - David M. Holtzman
- Department of Neurology, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
- The Hope Center for Neurological Disorders, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - David D. Limbrick
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
- The Hope Center for Neurological Disorders, Washington University in St. Louis, School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
14
|
Qi Y, Li Y, Cui SC, Zhao JJ, Liu XY, Ji CX, Sun FY, Xu P, Chen XH. Splicing factor NSSR1 reduces neuronal injury after mouse transient global cerebral ischemia. Glia 2015; 63:826-45. [PMID: 25627895 DOI: 10.1002/glia.22787] [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: 07/11/2014] [Revised: 12/08/2014] [Accepted: 12/18/2014] [Indexed: 11/06/2022]
Abstract
This study focuses on the function of NSSR1, a splicing factor, in neuronal injury in the ischemic mouse brain using the transient global cerebral ischemic mouse model and the cultured cells treated with oxygen-glucose deprivation (OGD). The results showed that the cerebral ischemia triggers the expression of NSSR1 in hippocampal astrocytes, predominantly the dephosphorylated NSSR1 proteins, and the Exon3 inclusive NCAM-L1 variant and the Exon4 inclusive CREB variant. While in the hippocampus of astrocyte-specific NSSR1 conditional knockdown (cKD) mice, where cerebral ischemia no longer triggers NSSR1 expression in astrocytes, the expression of Exon3 inclusive NCAM-L1 variant and Exon4 inclusive CREB variant were no longer triggered as well. In addition, the injury of hippocampal neurons was more severe in astrocyte-specific NSSR1 cKD mice compared with in wild-type mice after brain ischemia. Of note, the culture media harvested from the astrocytes with overexpression of NSSR1 or the Exon3 inclusive NCAM-L1 variant, or Exon4 inclusive CREB variant were all able to reduce the neuronal injury induced by OGD. The results provide the evidence demonstrating that: (1) Splicing factor NSSR1 is a new factor involved in reducing ischemic injury. (2) Ischemia induces NSSR1 expression in astrocytes, not in neurons. (3) NSSR1-mediated pathway in astrocytes is required for reducing ischemic neuronal injury. (4) NCAM-L1 and CREB are probably mediators in NSSR1-mediated pathway. In conclusion, our results suggest for the first time that NSSR1 may provide a novel mechanism for reducing neuronal injury after ischemia, probably through regulation on alternative splicing of NCAM-L1 and CREB in astrocytes.
Collapse
Affiliation(s)
- Yao Qi
- State Key Laboratory of Medical Neurobiology and Laboratory of Genomic Physiology, Institutes of Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Sanati Nezhad A, Geitmann A. The cellular mechanics of an invasive lifestyle. JOURNAL OF EXPERIMENTAL BOTANY 2013; 64:4709-28. [PMID: 24014865 DOI: 10.1093/jxb/ert254] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Invasive behaviour is the hallmark of a variety of cell types of animal, plant, and fungal origin. Here we review the purpose and mechanism of invasive growth and migration. The focus is on the physical principles governing the process, the source of invasive force, and the cellular mechanism by which the cell penetrates the substrate. The current experimental methods for measuring invasive force and the modelling approaches for studying invasive behaviour are explained, and future experimental strategies are proposed.
Collapse
Affiliation(s)
- Amir Sanati Nezhad
- McGill University and Génome Québec Innovation Centre, Biomedical Engineering Department, McGill University, Montreal, Canada
| | | |
Collapse
|
16
|
Aikawa Y. Ubiquitination within the membrane-proximal ezrin-radixin-moesin (ERM)-binding region of the L1 cell adhesion molecule. Commun Integr Biol 2013; 6:e24750. [PMID: 23986810 PMCID: PMC3737756 DOI: 10.4161/cib.24750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 04/19/2013] [Accepted: 04/19/2013] [Indexed: 12/14/2022] Open
Abstract
The dynamic turnover of the L1 cell adhesion molecule to and from the plasma membrane that is mediated through exo-and endocytic trafficking is central to axon outgrowth. Although the ubiquitination of L1 in response to incubation with an L1 antibody that mimics L1-L1 homophilic binding has been previously shown, the endocytic trafficking pathway of the ubiquitinated L1 destined for degradation is yet unclear. I have recently shown that the ubiquitinated L1 is endocytosed by Rabex-5, which is an ubiquitin-binding protein and guanine nucleotide exchange factor for Rab5, into early endosomes from the plasma membrane. Here, I speculate on the putative ubiquitination site within the membrane-proximal ezrin-binding motif in the cytoplasmic domain of L1 and discuss the regulatory role of this motif in the competition between ubiquitination and the binding of ezrin prior to L1 internalization.
Collapse
Affiliation(s)
- Yoshikatsu Aikawa
- Laboratory of Neural Membrane Biology; Graduate School of Brain Science; Doshisha University; Kyoto, Japan
| |
Collapse
|
17
|
Brand Y, Sung M, Chavez E, Wei E, Pak KK, Housley GD, Bodmer D, Ryan AF. Neural cell adhesion molecule L1 modulates type I but not type II inner ear spiral ganglion neurite outgrowth in an in vitro alternate choice assay. J Mol Neurosci 2013; 51:663-70. [PMID: 23760987 DOI: 10.1007/s12031-013-0040-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 05/27/2013] [Indexed: 12/17/2022]
Abstract
L1, a neural cell adhesion molecule of the immunoglobulin superfamily, is widely expressed in the nervous system and important in axonal outgrowth, guidance, synapse formation, and signaling. Gene deletion studies emphasize the significance of L1 during development of the central nervous system and L1 is crucial for the topographic targeting of retinal axons. In contrast to the brain and retina, the role of L1 in the inner ear is largely unknown. While previous studies have localized L1 in the developing inner ear of the chicken and mouse, its function during the innervation of the cochlea still remains largely unclear. We therefore investigated the functional role of L1 in the mammalian inner ear. Our aim was to determine whether or not L1 can modulate type I and/or type II spiral ganglion neuron outgrowth using an in vitro alternate choice assay. We found that L1, presented in stripe micropatterns, provide directional cues to neonatal rodent type I but not type II inner ear spiral ganglion neurites. The results suggest that L1 may play a role in axonal pathfinding of type I spiral ganglion dendrites toward their inner hair cell targets but not of type II toward the outer hair cells.
Collapse
Affiliation(s)
- Yves Brand
- Department of Surgery/Otolaryngology, UCSD School of Medicine, 9500 Gilman Drive MC0666, La Jolla, CA, 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Tagliavacca L, Colombo F, Racchetti G, Meldolesi J. L1CAM and its cell-surface mutants: new mechanisms and effects relevant to the physiology and pathology of neural cells. J Neurochem 2012; 124:397-409. [PMID: 22973895 PMCID: PMC3557714 DOI: 10.1111/jnc.12015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 09/05/2012] [Accepted: 09/08/2012] [Indexed: 11/27/2022]
Abstract
The L1 syndrome, a genetic disease that affects 1/30 000 newborn males, is sustained by numerous missense mutations of L1 cell adhesion molecule (L1CAM), an adhesion surface protein active also in transmembrane signaling, essential for the development and function of neurons. To investigate the cell biology of L1CAM, we employed a high RE1-silencing transcription (factor) clone of the pheochromocytoma PC12 line, defective in L1CAM expression and neurite outgrowth. The clone was transfected with wild-type L1CAM and four missense, disease-inducing point mutants encoding proteins distributed to the cell surface. The mutant-expressing cells, defective in adhesion to extracellular matrix proteins and in migration, exhibited unchanged proliferation. The nerve growth factor (NGF)-induced neurite outgrowth was re-established in defective clone cells transfected with the wild-type and the H210Q and I219T L1CAMs mutants, but not in the others. The stimulated outgrowth was confirmed in a second defective PC12 clone over-expressing the NGF receptor TrkA, treated with NGF and/or a recombinant L1CAM chimera. These results revealed a new function of L1CAM, a positive, robust and dose-dependent modulation of the TrkA receptor activated spontaneously or by NGF. The variable effects observed with the different L1CAM mutants suggest that this function contributes to the marked heterogeneity of symptoms and severity observed in the patients affected by the L1 syndrome.
Collapse
Affiliation(s)
- Luigina Tagliavacca
- Department of Neuroscience, Vita-Salute San Raffaele University and San Raffaele Institute, Milano, Italy
| | | | | | | |
Collapse
|
19
|
Steuble M, Diep TM, Schätzle P, Ludwig A, Tagaya M, Kunz B, Sonderegger P. Calsyntenin-1 shelters APP from proteolytic processing during anterograde axonal transport. Biol Open 2012; 1:761-74. [PMID: 23213470 PMCID: PMC3507217 DOI: 10.1242/bio.20121578] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 05/22/2012] [Indexed: 11/27/2022] Open
Abstract
Endocytosis of amyloid-β precursor protein (APP) is thought to represent the major source of substrate for the production of the amyloidogenic Aβ peptide by the β-secretase BACE1. The irreversible nature of proteolytic cleavage implies the existence of an efficient replenishment route for APP from its sites of synthesis to the cell surface. We recently found that APP exits the trans-Golgi network in intimate association with calsyntenin-1, a transmembrane cargo-docking protein for Kinesin-1-mediated vesicular transport. Here we characterized the function of calsyntenin-1 in neuronal APP transport using selective immunoisolation of intracellular trafficking organelles, immunocytochemistry, live-imaging, and RNAi. We found that APP is co-transported with calsyntenin-1 along axons to early endosomes in the central region of growth cones in carriers that exclude the α-secretase ADAM10. Intriguingly, calsyntenin-1/APP organelles contained BACE1, suggesting premature cleavage of APP along its anterograde path. However, we found that APP contained in calsyntenin-1/APP organelles was stable. We further analyzed vesicular trafficking of APP in cultured hippocampal neurons, in which calsyntenin-1 was reduced by RNAi. We found a markedly increased co-localization of APP and ADAM10 in axons and growth cones, along with increased proteolytic processing of APP and Aβ secretion in these neurons. This suggested that the reduced capacity for calsyntenin-1-dependent APP transport resulted in mis-sorting of APP into additional axonal carriers and, therefore, the premature encounter of unprotected APP with its ectodomain proteases. In combination, our results characterize calsyntenin-1/APP organelles as carriers for sheltered anterograde axonal transport of APP.
Collapse
Affiliation(s)
- Martin Steuble
- Department of Biochemistry, University of Zurich , Winterthurerstrasse 190, CH-8057 Zurich , Switzerland
| | | | | | | | | | | | | |
Collapse
|
20
|
Capone G, Novello G, Bavaro SL, Fasano C, Pesce Delfino A, Polito AN, Kanduc D. A qualitative description of the peptide sharing between poliovirus and Homo sapiens. Immunopharmacol Immunotoxicol 2012; 34:779-85. [PMID: 22303874 DOI: 10.3109/08923973.2012.654610] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In a companion paper, we reported that pentapeptides from human poliovirus 1, Mahoney strain, occur repeatedly in human proteins for a total of more than 18,000 overlaps. In the present study, we describe the distribution of the polio pentapeptides throughout biochemical pathways and networks characterizing functions and tissues in the human host. The present study might be of help to better define the poliovirus-host relationships as well as for designing peptide modules with anti-polio activity.
Collapse
Affiliation(s)
- Giovanni Capone
- Department of Biochemistry and Molecular Biology, Universityof Bari, Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
21
|
Nak regulates localization of clathrin sites in higher-order dendrites to promote local dendrite growth. Neuron 2011; 72:285-99. [PMID: 22017988 DOI: 10.1016/j.neuron.2011.08.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2011] [Indexed: 11/22/2022]
Abstract
VIDEO ABSTRACT During development, dendrites arborize in a field several hundred folds of their soma size, a process regulated by intrinsic transcription program and cell adhesion molecule (CAM)-mediated interaction. However, underlying cellular machineries that govern distal higher-order dendrite extension remain largely unknown. Here, we show that Nak, a clathrin adaptor-associated kinase, promotes higher-order dendrite growth through endocytosis. In nak mutants, both the number and length of higher-order dendrites are reduced, which are phenocopied by disruptions of clathrin-mediated endocytosis. Nak interacts genetically with components of the endocytic pathway, colocalizes with clathrin puncta, and is required for dendritic localization of clathrin puncta. More importantly, these Nak-containing clathrin structures preferentially localize to branching points and dendritic tips that are undergoing active growth. We present evidence that the Drosophila L1-CAM homolog Neuroglian is a relevant cargo of Nak-dependent internalization, suggesting that localized clathrin-mediated endocytosis of CAMs facilitates the extension of nearby higher-order dendrites.
Collapse
|
22
|
Tang N, Farah B, He M, Fox S, Malouf A, Littner Y, Bearer CF. Ethanol causes the redistribution of L1 cell adhesion molecule in lipid rafts. J Neurochem 2011; 119:859-67. [PMID: 21884525 DOI: 10.1111/j.1471-4159.2011.07467.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Fetal alcohol spectrum disorder is estimated to affect 1% of live births. The similarities between children with fetal alcohol syndrome and those with mutations in the gene encoding L1 cell adhesion molecule (L1) implicates L1 as a target of ethanol developmental neurotoxicity. Ethanol specifically inhibits the neurite outgrowth promoting function of L1 at pharmacologic concentrations. Emerging evidence shows that localized disruption of the lipid rafts reduces L1-mediated neurite outgrowth. We hypothesize that ethanol impairment of the association of L1 with lipid rafts is a mechanism underlying ethanol's inhibition of L1-mediated neurite outgrowth. In this study, we examine the effects of ethanol on the association of L1 and lipid rafts. We show that, in vitro, L1 but not N-cadherin shifts into lipid rafts following treatment with 25 mM ethanol. The ethanol concentrations causing this effect are similar to those inhibiting L1-mediated neurite outgrowth. Increasing chain length of the alcohol demonstrates the same cutoff as that previously shown for inhibition of L1-L1 binding. In addition, in cerebellar granule neurons in which lipid rafts are disrupted with methyl-beta-cyclodextrin, the rate of L1-mediated neurite outgrowth on L1-Fc is reduced to background rate and that this background rate is not ethanol sensitive. These data indicate that ethanol may inhibit L1-mediated neurite outgrowth by retarding L1 trafficking through a lipid raft compartment.
Collapse
Affiliation(s)
- Ningfeng Tang
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland 21209, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Winckler B, Yap CC. Endocytosis and endosomes at the crossroads of regulating trafficking of axon outgrowth-modifying receptors. Traffic 2011; 12:1099-108. [PMID: 21535338 PMCID: PMC3155643 DOI: 10.1111/j.1600-0854.2011.01213.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In neurons, many receptors must be localized correctly to axons or dendrites for proper function. During development, receptors for nerve growth and guidance are targeted to axons and localized to growth cones where receptor activation by ligands results in promotion or inhibition of axon growth. Signaling outcomes downstream of ligand binding are determined by the location, levels and residence times of receptors on the neuronal plasma membrane. Therefore, the mechanisms controlling the trafficking of these receptors are crucial to the proper wiring of circuits. Membrane proteins accumulate on the axonal surface by multiple routes, including polarized sorting in the trans Golgi network, sorting in endosomes and removal by endocytosis. Endosomes also play important roles in the signaling pathways for both growth-promoting and -inhibiting molecules: signaling endosomes derived from endocytosis are important for signaling from growth cones to cell bodies. Growth-promoting neurotrophins and growth-inhibiting Nogo-A can use EHD4/Pincher-dependent endocytosis at the growth cone for their respective retrograde signaling. In addition to retrograde transport of endosomes, anterograde transport to axons in endosomes also occurs for several receptors, including the axon outgrowth-promoting cell adhesion molecule L1/NgCAM and TrkA. L1/NgCAM also depends on EHD4/Pincher-dependent endocytosis for its axonal polarization. In this review, we will focus on receptors whose trafficking has been reported to be modulated by the EHD4/Pincher family of endosomal regulators, namely L1/NgCAM, Trk and Nogo-A. We will first summarize the pathways underlying the axonal transport of these proteins and then discuss the potential roles of EHD4/Pincher in mediating their endocytosis.
Collapse
Affiliation(s)
- Bettina Winckler
- University of Virginia Medical School, Department of Neuroscience, MR4-6115, 409 Lane Road Ext. Charlottesville, VA 22936, USA
| | - Chan Choo Yap
- University of Virginia Medical School, Department of Neuroscience, MR4-6115, 409 Lane Road Ext. Charlottesville, VA 22936, USA
| |
Collapse
|
24
|
Sinis N, Kraus A, Drakotos D, Doser M, Schlosshauer B, Müller HW, Skouras E, Bruck JC, Werdin F. Bioartificial reconstruction of peripheral nerves using the rat median nerve model. Ann Anat 2011; 193:341-6. [PMID: 21489766 DOI: 10.1016/j.aanat.2011.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 02/14/2011] [Accepted: 02/25/2011] [Indexed: 02/02/2023]
Abstract
Different bioartificial tubes were recommended for peripheral nerve reconstruction in the past. In order to replace autologous nerve grafts this materials are still under review in different animal studies. Most of them are dealing with the rodent peripheral nerves. One very popular animal model to study different materials is the rat median nerve model. With its easy excess, simple behavioral tests and reliable long term results it is attractive to many scientists in this field. This review gives an overview about the past, current and future options in this model for bioartificial nerve tubes. It summarizes the evolution of successful implantation of different materials across short nerve gaps and demonstrates the obstacles arising from long nerve gaps and the problems associated to them.
Collapse
Affiliation(s)
- Nektarios Sinis
- Department of Plastic Surgery, Marthin-Luther-Hospital Berlin, Academic Department of the Charite, Caspar-Theyss-Strasse 27–31, Grunewald, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Shieh JC, Schaar BT, Srinivasan K, Brodsky FM, McConnell SK. Endocytosis regulates cell soma translocation and the distribution of adhesion proteins in migrating neurons. PLoS One 2011; 6:e17802. [PMID: 21445347 PMCID: PMC3062553 DOI: 10.1371/journal.pone.0017802] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Accepted: 02/14/2011] [Indexed: 11/21/2022] Open
Abstract
Newborn neurons migrate from their birthplace to their final location to form a properly functioning nervous system. During these movements, young neurons must attach and subsequently detach from their substrate to facilitate migration, but little is known about the mechanisms cells use to release their attachments. We show that the machinery for clathrin-mediated endocytosis is positioned to regulate the distribution of adhesion proteins in a subcellular region just proximal to the neuronal cell body. Inhibiting clathrin or dynamin function impedes the movement of migrating neurons both in vitro and in vivo. Inhibiting dynamin function in vitro shifts the distribution of adhesion proteins to the rear of the cell. These results suggest that endocytosis may play a critical role in regulating substrate detachment to enable cell body translocation in migrating neurons.
Collapse
Affiliation(s)
- Jennifer C. Shieh
- Department of Biology, Stanford University, Stanford, California, United States of America
- Program in Neuroscience, Stanford University, Stanford, California, United States of America
| | - Bruce T. Schaar
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
| | - Karpagam Srinivasan
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Frances M. Brodsky
- Departments of Bioengineering and Therapeutic Sciences, Pharmaceutical Chemistry, and Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Susan K. McConnell
- Department of Biology, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Steuble M, Gerrits B, Ludwig A, Mateos JM, Diep TM, Tagaya M, Stephan A, Schätzle P, Kunz B, Streit P, Sonderegger P. Molecular characterization of a trafficking organelle: dissecting the axonal paths of calsyntenin-1 transport vesicles. Proteomics 2011; 10:3775-88. [PMID: 20925061 DOI: 10.1002/pmic.201000384] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Kinesin motors play crucial roles in the delivery of membranous cargo to its destination and thus for the establishment and maintenance of cellular polarization. Recently, calsyntenin-1 was identified as a cargo-docking protein for Kinesin-1-mediated axonal transport of tubulovesicular organelles along axons of central nervous system neurons. To further define the function of calsyntenin-1, we immunoisolated calsyntenin-1 organelles from murine brain homogenates and determined their proteome by MS. We found that calsyntenin-1 organelles are endowed with components of the endosomal trafficking machinery and contained the β-amyloid precursor protein (APP). Detailed biochemical analyses of calsyntenin-1 immunoisolates in conjunction with immunocytochemical colocalization studies with cultured hippocampal neurons, using endosomal marker proteins for distinct subcompartments of the endosomal pathways, indicated that neuronal axons contain at least two distinct, nonoverlapping calsyntenin-1-containing transport packages: one characterized as early-endosomal, APP positive, the other as recycling-endosomal, APP negative. We postulate that calsyntenin-1 acts as a general mediator of anterograde axonal transportation of endosomal vesicles. In this role, calsyntenin-1 may actively contribute to axonal growth and pathfinding in the developing as well as to the maintenance of neuronal polarity in the adult nervous system; further, it may actively contribute to the stabilization of APP during its anterograde axonal trajectory.
Collapse
Affiliation(s)
- Martin Steuble
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Linking L1CAM-mediated signaling to NF-κB activation. Trends Mol Med 2010; 17:178-87. [PMID: 21195665 DOI: 10.1016/j.molmed.2010.11.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 11/19/2010] [Accepted: 11/19/2010] [Indexed: 01/13/2023]
Abstract
The cell adhesion molecule L1 (L1CAM) was originally identified as a neural adhesion molecule essential for neurite outgrowth and axon guidance. Many studies have now shown that L1CAM is overexpressed in human carcinomas and associated with poor prognosis. So far, L1CAM-mediated cellular signaling has been largely attributed to an association with growth factor receptors, referred to as L1CAM-'assisted' signaling. New data demonstrate that L1CAM can signal via two additional mechanisms: 'forward' signaling via regulated intramembrane proteolysis and 'reverse' signaling via the activation of the transcription factor nuclear factor (NF)-κB. Taken together, these findings lead to a new understanding of L1CAM downstream signaling that is fundamental for the development of anti-L1CAM antibody-mediated therapeutics in human tumor cells.
Collapse
|
28
|
Franco M, Seyfried NT, Brand AH, Peng J, Mayor U. A novel strategy to isolate ubiquitin conjugates reveals wide role for ubiquitination during neural development. Mol Cell Proteomics 2010; 10:M110.002188. [PMID: 20861518 PMCID: PMC3098581 DOI: 10.1074/mcp.m110.002188] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Ubiquitination has essential roles in neuronal development and function. Ubiquitin proteomics studies on yeast and HeLa cells have proven very informative, but there still is a gap regarding neuronal tissue-specific ubiquitination. In an organism context, direct evidence for the ubiquitination of neuronal proteins is even scarcer. Here, we report a novel proteomics strategy based on the in vivo biotinylation of ubiquitin to isolate ubiquitin conjugates from the neurons of Drosophila melanogaster embryos. We confidently identified 48 neuronal ubiquitin substrates, none of which was yet known to be ubiquitinated. Earlier proteomics and biochemical studies in non-neuronal cell types had identified orthologs to some of those but not to others. The identification here of novel ubiquitin substrates, those with no known ubiquitinated ortholog, suggests that proteomics studies must be performed on neuronal cells to identify ubiquitination pathways not shared by other cell types. Importantly, several of those newly found neuronal ubiquitin substrates are key players in synaptogenesis. Mass spectrometry results were validated by Western blotting to confirm that those proteins are indeed ubiquitinated in the Drosophila embryonic nervous system and to elucidate whether they are mono- or polyubiquitinated. In addition to the ubiquitin substrates, we also identified the ubiquitin carriers that are active during synaptogenesis. Identifying endogenously ubiquitinated proteins in specific cell types, at specific developmental stages, and within the context of a living organism will allow understanding how the tissue-specific function of those proteins is regulated by the ubiquitin system.
Collapse
Affiliation(s)
- Maribel Franco
- CIC Biogune, Bizkaia Teknologi Parkea, 48160 Derio, Spain
| | | | | | | | | |
Collapse
|
29
|
Eva R, Dassie E, Caswell PT, Dick G, ffrench-Constant C, Norman JC, Fawcett JW. Rab11 and its effector Rab coupling protein contribute to the trafficking of beta 1 integrins during axon growth in adult dorsal root ganglion neurons and PC12 cells. J Neurosci 2010; 30:11654-69. [PMID: 20810886 PMCID: PMC6633432 DOI: 10.1523/jneurosci.2425-10.2010] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 07/07/2010] [Accepted: 07/09/2010] [Indexed: 01/06/2023] Open
Abstract
Integrins play an important part in axon growth, but integrin traffic in neurons is poorly understood. Expression of the tenascin-C-binding integrin alpha9 promotes axon regeneration. We have therefore studied the mechanism by which alpha9 integrin and its partner beta1 are trafficked along axons and at the growth cone using adult DRG neurons and PC12 cells. We have focused on the small GTPase Rab11 and its effector Rab coupling protein (RCP), as they are involved in the long-range trafficking of beta1 integrins in other cells. Rab11 colocalizes with alpha9 and other alpha integrins and with beta1 integrin in growth cones and axons, and immunopurified Rab11 vesicles contain alpha9 and beta1. Endocytosed beta1 integrins traffic via Rab11. However, Rab11 vesicles in axons are generally static, and alpha9 integrins undergo bouts of movement during which they leave the Rab11 compartment. In growth cones, alpha9 and beta1 overlap with RCP, particularly at the growth cone periphery. We show that beta1 integrin trafficking during neurite outgrowth involves Rab11 and RCP, and that manipulation of these molecules alters surface integrin levels and axon growth, and can be used to enhance alpha9 integrin-dependent neurite outgrowth. Our data suggest that manipulation of trafficking via Rab11 and RCP could be a useful strategy for promoting integrin-dependent axonal regeneration.
Collapse
Affiliation(s)
- Richard Eva
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom
| | - Elisa Dassie
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom
| | - Patrick T. Caswell
- Beatson Institute for Cancer Research, Bearsden, Glasgow G61 1BD, United Kingdom, and
| | - Gunnar Dick
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom
| | - Charles ffrench-Constant
- Medical Research Council Centre for Regenerative Medicine, Centre for Multiple Sclerosis Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Jim C. Norman
- Beatson Institute for Cancer Research, Bearsden, Glasgow G61 1BD, United Kingdom, and
| | - James W. Fawcett
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 2PY, United Kingdom
| |
Collapse
|
30
|
Sakurai K, Toyoshima M, Ueda H, Matsubara K, Takeda Y, Karagogeos D, Shimoda Y, Watanabe K. Contribution of the neural cell recognition molecule NB-3 to synapse formation between parallel fibers and Purkinje cells in mouse. Dev Neurobiol 2009; 69:811-24. [DOI: 10.1002/dneu.20742] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
31
|
Pruessmeyer J, Ludwig A. The good, the bad and the ugly substrates for ADAM10 and ADAM17 in brain pathology, inflammation and cancer. Semin Cell Dev Biol 2009; 20:164-74. [DOI: 10.1016/j.semcdb.2008.09.005] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 09/11/2008] [Accepted: 09/15/2008] [Indexed: 10/21/2022]
|
32
|
Hoschouer EL, Yin FQ, Jakeman LB. L1 cell adhesion molecule is essential for the maintenance of hyperalgesia after spinal cord injury. Exp Neurol 2009; 216:22-34. [PMID: 19059398 PMCID: PMC3691996 DOI: 10.1016/j.expneurol.2008.10.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Revised: 10/21/2008] [Accepted: 10/31/2008] [Indexed: 10/21/2022]
Abstract
Spinal cord injury (SCI) results in a loss of normal motor and sensory function, leading to severe disability and reduced quality of life. A large proportion of individuals with SCI also suffer from neuropathic pain symptoms. The causes of abnormal pain sensations are not well understood, but can include aberrant sprouting and reorganization of injured or spared sensory afferent fibers. L1 is a cell adhesion molecule that contributes to axonal outgrowth, guidance and fasciculation in development as well as synapse formation and plasticity throughout life. In the present study, we used L1 knockout (KO) mice to determine whether this adhesion molecule contributes to sensory dysfunction after SCI. Both wild-type (WT) and KO mice developed heat hyperalgesia following contusion injury, but the KO mice recovered normal response latencies beginning at 4 weeks post-injury. Histological analyses confirmed increased sprouting of sensory fibers containing calcitonin-gene related peptide (CGRP) in the deep dorsal horn of the lumbar spinal cord and increased numbers of interneurons expressing protein kinase C gamma (PKCgamma) in WT mice 6 weeks after injury. In contrast, L1 KO mice had less CGRP(+) fiber sprouting, but even greater numbers of PKCgamma(+) interneurons at the 6 week time point. These data demonstrate that L1 plays a role in maintenance of thermal hyperalgesia after SCI in mice, and implicate CGRP(+) fiber sprouting and the upregulation of PKCgamma expression as potential contributors to this response.
Collapse
Affiliation(s)
- Emily L. Hoschouer
- Neuroscience Graduate Studies Program
- Center for Brain and Spinal Cord Repair
- The Ohio State University Medical Center
| | - Feng Qin Yin
- Department of Physiology and Cell Biology
- Center for Brain and Spinal Cord Repair
- The Ohio State University Medical Center
| | - Lyn B. Jakeman
- Department of Physiology and Cell Biology
- Neuroscience Graduate Studies Program
- Center for Brain and Spinal Cord Repair
- The Ohio State University Medical Center
| |
Collapse
|
33
|
Mintz CD, Carcea I, McNickle DG, Dickson TC, Ge Y, Salton SR, Benson DL. ERM proteins regulate growth cone responses to Sema3A. J Comp Neurol 2008; 510:351-66. [PMID: 18651636 PMCID: PMC2691689 DOI: 10.1002/cne.21799] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Axonal growth cones initiate and sustain directed growth in response to cues in their environment. A variety of events such as receptor internalization, kinase activation, and actin rearrangement can be stimulated by guidance cues and are essential for mediating targeted growth cone behavior. Surprisingly little is known about how such disparate actions are coordinated. Our data suggest that ezrin, radixin, and moesin (ERMs), a family of highly homologous, multifunctional proteins may be able to coordinate growth cone responses to the guidance cue Semaphorin 3A (Sema3A). We show that active ERMs concentrate asymmetrically in neocortical growth cones, are rapidly and transiently inactivated by Sema3A, and are required for Sema3A-mediated growth cone collapse and guidance. The FERM domain of active ERMs regulates internalization of the Sema3A receptor, Npn1, and its coreceptor, L1CAM, while the ERM C-terminal domain binds and caps F-actin. Our data support a model in which ERMs can coordinate membrane and actin dynamics in response to Sema3A.
Collapse
Affiliation(s)
- C. David Mintz
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, NY, 10029, U.S.A
| | - Ioana Carcea
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, NY, 10029, U.S.A
| | - Daniel G. McNickle
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, NY, 10029, U.S.A
| | - Tracey C. Dickson
- Menzies Research Institute, University of Tasmania, Hobart, Tasmania 7001, Australia
| | - Yongchao Ge
- Department of Neurology, The Mount Sinai School of Medicine, New York, NY, 10029, U.S.A
| | - Stephen R.J. Salton
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, NY, 10029, U.S.A
- Department of Geriatrics, The Mount Sinai School of Medicine, New York, NY, 10029, U.S.A
| | - Deanna L. Benson
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, NY, 10029, U.S.A
| |
Collapse
|
34
|
Gómez M, Hernández ML, Pazos MR, Tolón RM, Romero J, Fernández-Ruiz J. Colocalization of CB1 receptors with L1 and GAP-43 in forebrain white matter regions during fetal rat brain development: evidence for a role of these receptors in axonal growth and guidance. Neuroscience 2008; 153:687-99. [PMID: 18400407 DOI: 10.1016/j.neuroscience.2008.02.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 01/19/2008] [Accepted: 02/05/2008] [Indexed: 12/13/2022]
Abstract
There is recent evidence supporting the notion that the cannabinoid signaling system plays a modulatory role in the regulation of cell proliferation and migration, survival of neural progenitors, neuritic elongation and guidance, and synaptogenesis. This assumption is based on the fact that cannabinoid 1-type receptors (CB(1) receptors) and their ligands emerge early in brain development and are abundantly expressed in certain brain regions that play key roles in these processes. We have recently presented in vivo evidence showing that this modulatory action might be exerted through regulating the synthesis of the cell adhesion molecule L1 that is also a key element for those processes. To further explore this issue, we conducted here immunohistochemical studies aimed at determining the cellular substrates of CB(1) receptor-L1 interactions in the rat brain during late fetal development. In this period, we previously found that the activation of CB(1) receptors increased L1 synthesis in several forebrain white matter regions but not in gray matter areas. Using double labeling studies, we observed here colocalization of both proteins in fiber tracts including the corpus callosum, the adjacent subcortical white matter, the internal capsule and the anterior commissure. Experiments conducted with cultures of fetal rat cortical nerve cells revealed that L1 is present mainly in neurons but not in glial cells. This fact, together with the results obtained in the double labeling studies, would indicate that L1 and CB(1) receptors should possibly be present in axons elongating through these white matter tracts, or, alternatively, in migrating neurons. Further experiments confirmed the presence of CB(1) receptors in elongating axons, since these receptors colocalized with growth-associated protein 43 (GAP-43), a marker of growth cones, but not with synaptophysin, a marker of active synaptic terminals, in the same forebrain white matter regions. Lastly, using cultured fetal rat cortical neurons, we also observed that the activation of cannabinoid receptors increased the levels of the full-length L1 and altered those of some active proteolytic fragments of this protein whose generation has been associated with specific steps in the process of neuritic elongation in cultured neurons. In summary, we have demonstrated that the effects caused by cannabinoid agonists on L1 are facilitated by the colocalization of this cell adhesion molecule with CB(1) receptors in several forebrain white matter regions during fetal brain development. We have provided strong evidence that this phenomenon occurs in axons elongating through these white matter tracts, and we have explored in vitro how cannabinoid receptors influence L1 levels. Considering the role played by L1 in different events related to neural development, our observations support the occurrence of a physiological mechanism by which the cannabinoid system might regulate the process of axonal growth and guidance through regulating the synthesis and function of L1.
Collapse
Affiliation(s)
- M Gómez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
35
|
Zhou X, Babu JR, da Silva S, Shu Q, Graef IA, Oliver T, Tomoda T, Tani T, Wooten MW, Wang F. Unc-51-like kinase 1/2-mediated endocytic processes regulate filopodia extension and branching of sensory axons. Proc Natl Acad Sci U S A 2007; 104:5842-7. [PMID: 17389358 PMCID: PMC1851579 DOI: 10.1073/pnas.0701402104] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The molecular mechanism and significance of endocytic processes involved in directional axon elongation are not well understood. The Unc-51 family of serine/threonine kinases was shown to be important for axon growth and was also linked to endocytosis, providing an entry point to study this problem. We found that mouse Unc-51-like kinase 1/2 (Ulk1/2) proteins are localized to vesicular structures in growth cones of mouse spinal sensory neurons. RNAi-mediated knockdown of Ulk1 and/or Ulk2 resulted in impaired endocytosis of nerve growth factor (NGF), excessive axon arborization, and severely stunted axon elongation. The evidence also indicates that Ulk1/2 mediates a non-clathrin-coated endocytosis in sensory growth cones. Interestingly, NGF can induce the interaction of Ulk1 with TrkA receptor complexes through promoting K63-polyubiquitination of Ulk1 and binding of Ulk1 to the scaffolding protein p62. These results and additional studies suggest that Ulk1/2 proteins regulate filopodia extension and neurite branching during sensory axon outgrowth, probably through regulating TrkA receptor trafficking and signaling.
Collapse
Affiliation(s)
- Xiang Zhou
- *Department of Cell Biology, Duke University Medical School, Durham, NC 27710
| | - J. Ramesh Babu
- Program in Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849
| | - Susana da Silva
- *Department of Cell Biology, Duke University Medical School, Durham, NC 27710
| | - Qing Shu
- *Department of Cell Biology, Duke University Medical School, Durham, NC 27710
| | | | - Tim Oliver
- *Department of Cell Biology, Duke University Medical School, Durham, NC 27710
| | - Toshifumi Tomoda
- Division of Neurosciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Tomomi Tani
- Research Institute for Electronic Science, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo 060-0812, Japan
| | - Marie W. Wooten
- Program in Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849
| | - Fan Wang
- *Department of Cell Biology, Duke University Medical School, Durham, NC 27710
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
36
|
Boiko T, Vakulenko M, Ewers H, Yap CC, Norden C, Winckler B. Ankyrin-dependent and -independent mechanisms orchestrate axonal compartmentalization of L1 family members neurofascin and L1/neuron-glia cell adhesion molecule. J Neurosci 2007; 27:590-603. [PMID: 17234591 PMCID: PMC6672792 DOI: 10.1523/jneurosci.4302-06.2007] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Axonal initial segments (IS) and nodes of Ranvier are functionally important membrane subdomains in which the clustering of electrogenic channels enables action potential initiation and propagation. In addition, the initial segment contributes to neuronal polarity by serving as a diffusion barrier. To study the mechanisms of axonal compartmentalization, we focused on two L1 family of cell adhesion molecules (L1-CAMs) [L1/neuron-glia cell adhesion molecule (L1/NgCAM) and neurofascin (NF)] and two neuronal ankyrins (ankB and ankG). NF and ankG accumulate specifically at the initial segment, whereas L1/NgCAM and ankB are expressed along the entire lengths of axons. We find that L1/NgCAM and NF show distinct modes of steady-state accumulation during axon outgrowth in cultured hippocampal neurons. Despite their different steady-state localizations, both L1/NgCAM and NF show slow diffusion and low detergent extractability specifically in the initial segment but fast diffusion and high detergent extractability in the distal axon. We propose that L1-CAMs do not strongly bind ankB in the distal axon because of spatial regulation of ankyrin affinity by phosphorylation. NF, conversely, is initially enriched in an ankyrin-independent manner in the axon generally and accumulates progressively in the initial segment attributable to preferential binding to ankG. Our results suggest that NF and L1/NgCAM accumulate in the axon by an ankyrin-independent pathway, but retention at the IS requires ankyrin binding.
Collapse
Affiliation(s)
- Tatiana Boiko
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908
| | - Max Vakulenko
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908
| | - Helge Ewers
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908
| | - Chan Choo Yap
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908
| | - Caren Norden
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908
| | - Bettina Winckler
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
37
|
Oblander SA, Ensslen-Craig SE, Longo FM, Brady-Kalnay SM. E-cadherin promotes retinal ganglion cell neurite outgrowth in a protein tyrosine phosphatase-mu-dependent manner. Mol Cell Neurosci 2007; 34:481-92. [PMID: 17276081 PMCID: PMC1853338 DOI: 10.1016/j.mcn.2006.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 12/01/2006] [Accepted: 12/06/2006] [Indexed: 10/23/2022] Open
Abstract
During development of the visual system, retinal ganglion cells (RGCs) require cell-cell adhesion molecules and extracellular matrix proteins for axon growth. In this study, we demonstrate that the classical cadherin, E-cadherin, is expressed in RGCs from E6 to E12 and promotes neurite outgrowth from all regions of the chick retina at E6, E8 and E10. E-cadherin is also expressed in the optic tectum. E-cadherin adhesion blocking antibodies specifically inhibit neurite outgrowth on an E-cadherin substrate. The receptor-type protein tyrosine phosphatase, PTPmu, associates with E-cadherin. In this manuscript, we demonstrate that antisense-mediated down-regulation of PTPmu, overexpression of catalytically inactive PTPmu and perturbation of endogenous PTPmu using a specific PTPmu inhibitor peptide results in a substantial reduction in neurite outgrowth on E-cadherin. Taken together, these findings demonstrate that E-cadherin is an important adhesion molecule for chick RGC neurite outgrowth and suggest that PTPmu expression and catalytic activity are required for outgrowth on an E-cadherin substrate.
Collapse
Affiliation(s)
| | | | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Susann M. Brady-Kalnay
- *Corresponding Author: Susann M. Brady-Kalnay, Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106-4960, Phone: 216-368-0330, Fax: 216-368-3055, E-mail:
| |
Collapse
|
38
|
Schultheis M, Diestel S, Schmitz B. The role of cytoplasmic serine residues of the cell adhesion molecule L1 in neurite outgrowth, endocytosis, and cell migration. Cell Mol Neurobiol 2007; 27:11-31. [PMID: 17151951 PMCID: PMC11517402 DOI: 10.1007/s10571-006-9113-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2005] [Accepted: 07/14/2006] [Indexed: 10/23/2022]
Abstract
1. The cell adhesion molecule L1 has been implicated in adhesion and migration of cells, in axon growth, guidance, and fasciculation, in myelination and synaptic plasticity. The cytoplasmic domain of neuronal L1 is highly conserved between species and has been shown to be phosphorylated at serine and tyrosine residues. 2. To investigate the significance of L1 serine phosphorylation, mutants of L1 were generated in which ser-1152, ser-1181, ser-1204, and ser-1248 were exchanged for leucine and rat B35 neuroblastoma cells were stably transfected with the L1-cDNA constructs. 3. Neurite outgrowth on poly-L-lysine (PLL) as substrate was determined either with or without differentiation into a neuronal phenotype with dbcAMP. In addition, antibody-induced endocytosis and cell migration were examined. 4. Our observations indicate that phosphorylation of single serine residues of the cytoplasmic domain of L1 contributes to neurite outgrowth through different mechanisms. Neurite growth is increased when ser-1152 or ser-1181 is replaced by a non-phosphorylatable leucine and decreased when ser-1204 or ser-1248 is mutated to leucine. Furthermore, mutation of ser-1181 to leucine results in strongly enhanced antibody-induced endocytosis of L1 and also in enhanced cell migration.
Collapse
Affiliation(s)
- M. Schultheis
- Department of Biochemistry, Institute of Animal Sciences, University of Bonn, Katzenburgweg 9a, 53115 Bonn, Germany
| | - S. Diestel
- Department of Biochemistry, Institute of Animal Sciences, University of Bonn, Katzenburgweg 9a, 53115 Bonn, Germany
| | - B. Schmitz
- Department of Biochemistry, Institute of Animal Sciences, University of Bonn, Katzenburgweg 9a, 53115 Bonn, Germany
| |
Collapse
|
39
|
Gómez M, Hernández M, Fernández-Ruiz J. The activation of cannabinoid receptors during early postnatal development reduces the expression of cell adhesion molecule L1 in the rat brain. Brain Res 2007; 1145:48-55. [PMID: 17320842 DOI: 10.1016/j.brainres.2007.01.102] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 01/16/2007] [Accepted: 01/29/2007] [Indexed: 11/16/2022]
Abstract
Cannabinoid CB(1) receptors and their ligands emerge early in brain development and are abundantly expressed in certain brain regions that play key roles in processes related to cell proliferation and migration, neuritic elongation and guidance, and synaptogenesis. This would support the notion that the cannabinoid system might play a modulatory role in the regulation of these processes. We have recently presented preliminary in vivo evidence showing that this modulatory action might be exerted, among others, through regulating the levels of several key elements in these processes, such as the L1 protein. This was observed in various white matter areas of the rat forebrain. Because these preliminary in vivo experiments focused only in fetal ages, we concentrated now in the period of early postnatal development. To this end, we analyzed the effects of the cannabinoid agonist Delta(9)-tetrahydrocannabinol (Delta(9)-THC) daily administered since the 5th day of gestation on mRNA levels for L1 in different brain structures of rat neonates at different postnatal ages (PND1, PND5 and PND12). Our results revealed that Delta(9)-THC exposure affected the levels of L1 transcripts in specific brain structures only in PND1, these effects disappearing during further days. Thus, we found reduced L1-mRNA levels in grey matter regions, such as the cerebral cortex, septum nuclei, striatum, dentate gyrus and CA3 subfield of the Ammon horn. White matter areas and subventricular zones were, however, more resistant to Delta(9)-THC exposure at this postnatal age in contrast with the previous data obtained in the fetal brain. Importantly, the effects were influenced by gender of animals, since the reductions were always more marked in females than males, also in contrast with the data reported for the fetal brain. In summary, the cannabinoid system seems to modulate the levels of L1 in several brain structures during specific periods of development [late gestation (previous data) and very early postnatal days (present data)], which correlates with the periods in which we had previously found an atypical distribution of CB(1) receptors in the developing brain. However, the magnitude of the effects of cannabinoids on L1 was influenced by two factors: gender and age of development. Considering the role played by L1 in different events related to neural development, our observations might support the occurrence of a physiological mechanism by which the cannabinoid system might regulate processes such as cell proliferation and migration, neuritic elongation and guidance, and synaptogenesis.
Collapse
MESH Headings
- Age Factors
- Aging/physiology
- Animals
- Animals, Newborn
- Brain/drug effects
- Brain/growth & development
- Brain/metabolism
- Brain Chemistry/physiology
- Cannabinoid Receptor Modulators/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cell Movement/drug effects
- Cell Movement/physiology
- Cell Proliferation/drug effects
- Down-Regulation/physiology
- Dronabinol/pharmacology
- Female
- Male
- Nerve Fibers, Myelinated/drug effects
- Nerve Fibers, Myelinated/metabolism
- Neural Cell Adhesion Molecule L1/genetics
- Neurites/drug effects
- Neurites/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/metabolism
- Sex Characteristics
- Sex Factors
- Telencephalon/drug effects
- Telencephalon/growth & development
- Telencephalon/metabolism
Collapse
Affiliation(s)
- María Gómez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040-Madrid, Spain
| | | | | |
Collapse
|
40
|
Abstract
Neural recognition molecules were discovered and characterized initially for their functional roles in cell adhesion as regulators of affinity between cells and the extracellular matrix in vitro. They were then recognized as mediators or co-receptors which trigger signal transduction mechanisms affecting cell adhesion and de-adhesion. Their involvement in contact attraction and repulsion relies on cell-intrinsic properties that are modulated by the spatial contexts of their expression at particular stages of ontogenetic development, in synaptic plasticity and during regeneration after injury. The functional roles of recognition molecules in cell proliferation and migration, determination of developmental fate, growth cone guidance, and synapse formation, stabilization and modulation have been well documented not only by in vitro, but also by in vivo studies that have been greatly aided by generation of genetically altered mice. More recently, the functions of recognition molecules have been investigated under conditions of neural repair and manipulated using a broad range of genetic and pharmacological approaches to achieve a beneficial outcome. The principal aim of most therapeutically oriented approaches has been to neutralize inhibitory factors. However, less attention has been paid to enhancing repair by stimulating the stimulatory factors. When considering potential therapeutic strategies, it is worth considering that a single recognition molecule can possess domains that are conducive or repellent and that the spatial distribution of recognition molecules can determine the overall function: Recognition molecules may be repellent for neurite outgrowth when presented as barriers or steep-concentration gradients and conducive when presented as uniform substrates. The focus of this review will be on the more recent attempts to study the conducive mechanisms with the expectation that they may be able to tip the balance from a regeneration inhospitable to a hospitable environment. It is likely that a combination of the two principles, as multifactorial as each principle may be in itself, will be of therapeutic value in humans.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Universität Hamburg, Hamburg, Germany
| | | |
Collapse
|
41
|
Miller MW, Mooney SM, Middleton FA. Transforming growth factor beta1 and ethanol affect transcription and translation of genes and proteins for cell adhesion molecules in B104 neuroblastoma cells. J Neurochem 2006; 97:1182-90. [PMID: 16686695 DOI: 10.1111/j.1471-4159.2006.03858.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Transforming growth factor (TGF) beta1 and ethanol retard the migration of young, post-mitotic neurons to the developing cerebral cortex. The coordination of this migration depends upon cell adhesion proteins (CAPs). We examined the effects of TGFbeta1 and ethanol on genes related to both TGF and CAPs. Rat B104 neuroblastoma cells were treated with TGFbeta1 (0 or 10 ng/mL) and ethanol (0 or 400 mg/dL) for 6-48 h. Total RNA was purified from each sample and analyzed using the Rat U34A GeneChip (Affymetrix). Candidate genes were those up- or down-regulated by either TGFbeta1 or ethanol. Twenty transcripts of CAPs were identified as being expressed by B104 cells and as being affected by treatment with TGFbeta1 or ethanol. The expression was verified for five representative genes (neural cell adhesion molecule, L1, and integrins alpha1, alpha7, and beta1) using assays with real-time reverse transcriptase-polymerase chain reactions. Each of these genes showed time-dependent changes. The changes were reflected in increases in protein expression that appeared within 24 or 48 h. Thus, the effects of TGFbeta1 and ethanol on CAPs parallel changes described in vivo and likely underlie changes associated with ethanol-induced alterations in neuronal migration.
Collapse
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
42
|
Tang N, He M, O’Riordan MA, Farkas C, Buck K, Lemmon V, Bearer CF. Ethanol inhibits L1 cell adhesion molecule activation of mitogen-activated protein kinases. J Neurochem 2006; 96:1480-90. [PMID: 16478533 PMCID: PMC4362514 DOI: 10.1111/j.1471-4159.2006.03649.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Inhibition of the functions of L1 cell adhesion molecule (L1) by ethanol has been implicated in the pathogenesis of the neurodevelopmental aspects of the fetal alcohol syndrome (FAS). Ethanol at pharmacological concentrations has been shown to inhibit L1-mediated neurite outgrowth of rat post-natal day 6 cerebellar granule cells (CGN). Extracellular signal-related kinases (ERK) 1/2 activation occurs following L1 clustering. Reduction in phosphoERK1/2 by inhibition of mitogen-activated protein kinase kinase (MEK) reduces neurite outgrowth of cerebellar neurons. Here, we examine the effects of ethanol on L1 activation of ERK1/2, and whether this activation occurs via activation of fibroblast growth factor receptor 1 (FGFR1). Ethanol at 25 mm markedly inhibited ERK1/2 activation by both clustering L1 with cross-linked monoclonal antibodies, or by L1-Fc chimeric proteins. Clustering L1 with subsequent ERK1/2 activation did not result in tyrosine phosphorylation of the FGFR1. In addition, inhibition of FGFR1 tyrosine kinase blocked basic fibroblast growth factor (bFGF) activation of ERK1/2, but did not affect activation of ERK1/2 by clustered L1. We conclude that ethanol disrupts the signaling pathway between L1 clustering and ERK1/2 activation, and that this occurs independently of the FGFR1 pathway in cerebellar granule cells.
Collapse
Affiliation(s)
- Ningfeng Tang
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Min He
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mary Ann O’Riordan
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chloe Farkas
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kevin Buck
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Vance Lemmon
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, Florida, USA
| | - Cynthia F. Bearer
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
43
|
Sinis N, Schaller HE, Schulte-Eversum C, Schlosshauer B, Doser M, Dietz K, Rösner H, Müller HW, Haerle M. Nerve regeneration across a 2-cm gap in the rat median nerve using a resorbable nerve conduit filled with Schwann cells. J Neurosurg 2005; 103:1067-76. [PMID: 16381194 DOI: 10.3171/jns.2005.103.6.1067] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object. In a rat model, nerve regeneration was evaluated across a 2-cm defect in the median nerve by using a resorbable artificial nerve conduit. The aim of this study was to develop an artificial, biocompatible nerve guide to induce regeneration in the peripheral nervous system.
Methods. The authors compared a nerve conduit of trimethylenecarbonate-co-epsilon-caprolactone (TMC/CL) filled with autologous Schwann cells with both an empty hollow conduit and an autologous nerve graft. Animals that did not undergo surgery served as the control group. Nerve regeneration was evaluated with the grasping test, histological analysis of the nerve, muscle weight analysis (flexor digitorum superficialis muscle), and electrophysiological examination.
After an observation period of 9 months, regeneration occurred only in animals that had received an autologous graft or a Schwann cell containing nerve conduit. No signs of regeneration were found in animals supplied with the empty conduit.
Conclusions. Results of this study reveal the important role of Schwann cells in the regeneration process across a 2-cm defect in the rat median nerve. Furthermore, Schwann cell—filled nerve conduits induced functional recovery, as demonstrated in the grasping test, that was comparable with that of the autologous graft 9 months after implantation.
Collapse
Affiliation(s)
- Nektarios Sinis
- Klinik für Hand-, Plastische-, Rekonstruktive- und Verbrennungschirurgie, Universtät Tübingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Itoh K, Shimono K, Lemmon V. Dephosphorylation and internalization of cell adhesion molecule L1 induced by theta burst stimulation in rat hippocampus. Mol Cell Neurosci 2005; 29:245-9. [PMID: 15911348 PMCID: PMC1382274 DOI: 10.1016/j.mcn.2005.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Revised: 02/13/2005] [Accepted: 02/22/2005] [Indexed: 11/20/2022] Open
Abstract
The neural cell adhesion molecule L1 may participate in initiating and maintaining synaptic changes during learning in the hippocampus. One prominent form of synaptic change in the hippocampus is long-term potentiation (LTP) that occurs following specific patterns of synaptic activity. We present evidence that Y1176 of the YRSL motif within L1 cytoplasmic domain is dephosphorylated in LTP-induced hippocampus. The dephosphorylated L1 is associated with AP-2 and AP180 that are required for clathrin-mediated internalization of L1. These data suggest that clathrin-mediated recycling of L1 at presynaptic sites is enhanced by certain kinds of neural activity, and that maintenance of LTP-induced synaptic changes is regulated by L1 recycling.
Collapse
Affiliation(s)
- Kouichi Itoh
- Laboratory of Molecular Pharmacology, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, Kagawa 769-2193, Japan
| | | | | |
Collapse
|
45
|
Horinouchi K, Nakamura Y, Yamanaka H, Watabe T, Shiosaka S. Distribution of L1cam mRNA in the adult mouse brain: In situ hybridization and Northern blot analyses. J Comp Neurol 2005; 482:386-404. [PMID: 15669056 DOI: 10.1002/cne.20398] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Previous immunohistochemical analysis revealed a wide distribution of L1cam-positive neural and nonneural structures in adult mouse brain. Although there were numerous punctate immunoreactive nerve terminals, only a few immunoreactive neuronal cell somata were present (Munakata et al. [2003] BMC Neurosci. 4:7). To explore the distribution of L1cam mRNA-containing cells, which are interpreted to be L1cam-producing cells, we performed in situ hybridization histochemistry with an antisense L1cam cRNA probe. L1cam mRNA was distributed widely from the olfactory bulb to the upper cervical cord with an uneven localization pattern in adult brain. All positive cell somata with silver grains after emulsion autoradiography were neuronal, and no grains were detected on nonneural cells in the present study. A high density of signals for neuronal L1cam mRNA was found in the thalamus, mammillary body, and hippocampus. In addition, strong hybridization signals were localized in various nuclei: main and accessory olfactory bulb, compact part of the substantia nigra, pontine gray matter, tegmental reticular nucleus, Edinger-Westphal nucleus, trigeminal motor nucleus, locus coeruleus, mesencephalic trigeminal nucleus, raphe nuclei, facial nucleus, ambiguus nucleus, dorsal motor vagal nucleus, and inferior olivary nucleus. Some long projection neurons such as the pyramidal, mitral, principal neurons of several cranial nuclei, and presumably monoaminergic cells containing noradrenalin, dopamine, and serotonin, expressed high levels of L1cam.
Collapse
Affiliation(s)
- Kazuhiro Horinouchi
- Division of Structural Cell Biology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, 630-0192 Nara, Japan
| | | | | | | | | |
Collapse
|
46
|
Loers G, Chen S, Grumet M, Schachner M. Signal transduction pathways implicated in neural recognition molecule L1 triggered neuroprotection and neuritogenesis. J Neurochem 2005; 92:1463-76. [PMID: 15748164 DOI: 10.1111/j.1471-4159.2004.02983.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The signal transduction pathways involved in adhesion molecule L1-triggered neuritogenesis and neuroprotection were investigated using the extracellular domain of mouse or human L1 in fusion with the Fc portion of human immunoglobulin G or L1 purified from mouse brain by affinity chromatography. Substrate L1-triggered neuritogenesis and neuroprotection depended on distinct but also overlapping signal transduction pathways and on the expression of L1 at the neuronal cell surface. PI3 kinase inhibitors, Src family kinase inhibitors as well as mitogen-activated protein kinase kinase inhibitors reduced both L1-triggered neuritogenesis and neuroprotection. In contrast, fibroblast growth factor receptor inhibitors, a protein kinase A inhibitor, and an inhibitor of cAMP-mediated signal transduction pathways, blocked neuritogenesis, but did not affect L1-triggered neuroprotection. Proteolytic cleavage of L1 or its interaction partners is necessary for both L1-mediated neuritogensis and neuroprotection. Furthermore, L1-triggered neuroprotection was found to be associated with increased phosphorylation of extracellular signal-regulated kinases 1/2, Akt and Bad, and inhibition of caspases. These observations suggest possibilities of differentially targeting signal transduction pathways for L1-dependent neuritogenesis and neuroprotection.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universität Hamburg, Falkenried 94, D-20251 Hamburg, Germany
| | | | | | | |
Collapse
|
47
|
Blank M, Triana-Baltzer GB, Richards CS, Berg DK. Alpha-protocadherins are presynaptic and axonal in nicotinic pathways. Mol Cell Neurosci 2004; 26:530-43. [PMID: 15276155 DOI: 10.1016/j.mcn.2004.04.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2004] [Revised: 04/12/2004] [Accepted: 04/15/2004] [Indexed: 11/25/2022] Open
Abstract
The protocadherin families pcdh-alpha, beta, and gamma have been proposed to mediate synaptic specificity via homophilic interactions. Here we report isolation of two pcdh-alpha family members from chick. We find pcdh-alpha mRNA in multiple regions of chick CNS including cerebellum, tectum, olfactory bulb, and forebrain, and in the autonomic nervous system. Immunoblots identify major components of 120 and 140 kDa both in brain and ciliary ganglion extracts. Immunohistochemistry reveals pcdh-alphas in axons and perisynaptically in preganglionic terminals, adjacent to transmitter release sites. Pcdh-alphas appear to be absent from postsynaptic sites: They are nonoverlapping with postsynaptic receptor clusters in the ganglion and are rapidly lost after ganglionic denervation. Similar pcdh-alpha patterns are found in motor axons and at neuromuscular junctions of birds and mammals, and persist into adulthood. The results indicate that pcdh-alphas are widely expressed in nicotinic cholinergic pathways and may engage in heterophilic interactions at synapses and on axons.
Collapse
Affiliation(s)
- Martina Blank
- Neurobiology Section, Division of Biological Sciences, University of California, La Jolla, San Diego, CA 92093-0357, USA
| | | | | | | |
Collapse
|