1
|
Rocha Caldas G, do Amaral L, Munhoz Rodrigues D, Mayrink de Miranda A, Aparecida Guinaim Dos Santos N, Machado Rocha L, Tame Parreira RL, Cardozo Dos Santos A, Kenupp Bastos J. Brazilian Green Propolis' Artepillin C and Its Acetylated Derivative Activate the NGF-Signaling Pathways and Induce Neurite Outgrowth in NGF-Deprived PC12 Cells. Chem Biodivers 2023; 20:e202301294. [PMID: 37953436 DOI: 10.1002/cbdv.202301294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/14/2023]
Abstract
Artepillin C is the most studied compound in Brazilian Green Propolis and, along with its acetylated derivative, displays neurotrophic activity on PC12 cells. Specific inhibitors of the trkA receptor (K252a), PI3K/Akt (LY294002), and MAPK/ERK (U0126) signaling pathways were used to investigate the neurotrophic mechanism. The expression of proteins involved in axonal and synaptic plasticity (GAP-43 and Synapsin I) was assessed by western blotting. Additionally, physicochemical properties, pharmacokinetics, and drug-likeness were evaluated by the SwissADME web tool. Both compounds induced neurite outgrowth by activating the NGF-signaling pathways but through different neuronal proteins. Furthermore, in silico analyses showed interesting physicochemical and pharmacokinetic properties of these compounds. Therefore, these compounds could play an important role in axonal and synaptic plasticity and should be further investigated.
Collapse
Affiliation(s)
- Gabriel Rocha Caldas
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Lilian do Amaral
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Débora Munhoz Rodrigues
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Aline Mayrink de Miranda
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Leandro Machado Rocha
- Natural Products Technology Laboratory-Fluminense Federal University, Niterói, RJ, Brazil
| | | | | | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
2
|
Miny L, Maisonneuve BGC, Quadrio I, Honegger T. Modeling Neurodegenerative Diseases Using In Vitro Compartmentalized Microfluidic Devices. Front Bioeng Biotechnol 2022; 10:919646. [PMID: 35813998 PMCID: PMC9263267 DOI: 10.3389/fbioe.2022.919646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
Abstract
The human brain is a complex organ composed of many different types of cells interconnected to create an organized system able to efficiently process information. Dysregulation of this delicately balanced system can lead to the development of neurological disorders, such as neurodegenerative diseases (NDD). To investigate the functionality of human brain physiology and pathophysiology, the scientific community has been generated various research models, from genetically modified animals to two- and three-dimensional cell culture for several decades. These models have, however, certain limitations that impede the precise study of pathophysiological features of neurodegeneration, thus hindering therapeutical research and drug development. Compartmentalized microfluidic devices provide in vitro minimalistic environments to accurately reproduce neural circuits allowing the characterization of the human central nervous system. Brain-on-chip (BoC) is allowing our capability to improve neurodegeneration models on the molecular and cellular mechanism aspects behind the progression of these troubles. This review aims to summarize and discuss the latest advancements of microfluidic models for the investigations of common neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Louise Miny
- NETRI, Lyon, France
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
| | | | - Isabelle Quadrio
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
- Laboratory of Neurobiology and Neurogenetics, Department of Biochemistry and Molecular Biology, Lyon University Hospital, Bron, France
| | | |
Collapse
|
3
|
Yang S, Cheng J, Man C, Jiang L, Long G, Zhao W, Zheng D. Effects of exogenous nerve growth factor on the expression of BMP-9 and VEGF in the healing of rabbit mandible fracture with local nerve injury. J Orthop Surg Res 2021; 16:74. [PMID: 33478541 PMCID: PMC7818757 DOI: 10.1186/s13018-021-02220-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/11/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Mandibular fracture healing is a complex process involving nerves and growth factors. Nerve growth factor (NGF) not only facilitates the maintenance of sympathetic neurite growth but also stimulates other growth factors that can promote the essential osteogenesis and angiogenesis for fracture healing. Therefore, it is necessary to analyze the combined effects of NGF, bone morphogenic protein-9 (BMP-9), and vascular endothelial growth factor (VEGF) to accelerate the healing of mandible fractures. METHODS The models of mandible fracture with local nerve injury established in 48 rabbits were randomly divided into nerve growth factor group (NGF group), gelatin sponge group (GS group), blank group, and intact group. The recovery of nerve reflex was assessed by observing the number of rabbits with lower lip responses to acupuncture. The fracture healing was observed with visual and CBCT, and then callus tissues from the mandibular fracture area were collected for hematoxylin and eosin (HE) staining observation, and the expression of BMP-9 and VEGF in callus at different stages was detected by quantitative real-time PCR (qRT-PCR). RESULTS Needling reaction in the lower lip showed the number of animals with nerve reflex recovery was significantly higher in the NGF group than that in the GS and blank groups at the 2nd and 4th weeks after the operation. The combined results of macroscopic observation, CBCT examination, and histological analysis showed that a large number of osteoblasts and some vascular endothelial cells were found around the trabecular bone in the NGF group and the amount of callus formation and reconstruction was better than that in the GS group at the 2nd week after the operation. The qRT-PCR results indicated that the expression levels of BMP-9 and VEGF in the four groups reached the highest values at the 2nd week, while the expression levels of both in the NGF group were significantly higher than that in the GS group. CONCLUSION The exogenous NGF could accelerate the healing of mandible fractures. This work will provide a new foundation and theoretical basis for clarifying the mechanism of fracture healing, thereby promoting fracture healing and reducing the disability rate of patients.
Collapse
Affiliation(s)
- Sen Yang
- Oral Maxillofacial Trauma and Orthognathic Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiao Cheng
- Oral Maxillofacial Trauma and Orthognathic Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| | - Cheng Man
- Oral Maxillofacial Trauma and Orthognathic Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Lian Jiang
- Oral Maxillofacial Trauma and Orthognathic Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Guogeng Long
- Oral Maxillofacial Trauma and Orthognathic Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Wenjun Zhao
- Oral Maxillofacial Trauma and Orthognathic Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Dexin Zheng
- Oral Maxillofacial Trauma and Orthognathic Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
4
|
Del Rio JA, Ferrer I. Potential of Microfluidics and Lab-on-Chip Platforms to Improve Understanding of " prion-like" Protein Assembly and Behavior. Front Bioeng Biotechnol 2020; 8:570692. [PMID: 33015021 PMCID: PMC7506036 DOI: 10.3389/fbioe.2020.570692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Human aging is accompanied by a relevant increase in age-associated chronic pathologies, including neurodegenerative and metabolic diseases. The appearance and evolution of numerous neurodegenerative diseases is paralleled by the appearance of intracellular and extracellular accumulation of misfolded proteins in affected brains. In addition, recent evidence suggests that most of these amyloid proteins can behave and propagate among neural cells similarly to infective prions. In order to improve understanding of the seeding and spreading processes of these "prion-like" amyloids, microfluidics and 3D lab-on-chip approaches have been developed as highly valuable tools. These techniques allow us to monitor changes in cellular and molecular processes responsible for amyloid seeding and cell spreading and their parallel effects in neural physiology. Their compatibility with new optical and biochemical techniques and their relative availability have increased interest in them and in their use in numerous laboratories. In addition, recent advances in stem cell research in combination with microfluidic platforms have opened new humanized in vitro models for myriad neurodegenerative diseases affecting different cellular targets of the vascular, muscular, and nervous systems, and glial cells. These new platforms help reduce the use of animal experimentation. They are more reproducible and represent a potential alternative to classical approaches to understanding neurodegeneration. In this review, we summarize recent progress in neurobiological research in "prion-like" protein using microfluidic and 3D lab-on-chip approaches. These approaches are driven by various fields, including chemistry, biochemistry, and cell biology, and they serve to facilitate the development of more precise human brain models for basic mechanistic studies of cell-to-cell interactions and drug discovery.
Collapse
Affiliation(s)
- Jose A Del Rio
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases (Ciberned), Barcelona, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Isidre Ferrer
- Center for Networked Biomedical Research on Neurodegenerative Diseases (Ciberned), Barcelona, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
5
|
Regulation of human glioma cell migration, tumor growth, and stemness gene expression using a Lck targeted inhibitor. Oncogene 2018; 38:1734-1750. [PMID: 30353164 PMCID: PMC6462869 DOI: 10.1038/s41388-018-0546-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 10/03/2018] [Accepted: 10/06/2018] [Indexed: 11/28/2022]
Abstract
Migration of human glioma cells (hGCs) within the brain parenchyma makes glioblastoma one of the most aggressive and lethal tumors. Studies of the cellular and molecular mechanisms underlying hGC migration are hindered by the limitations of existing glioma models. Here we developed a dorsal root ganglion axon-oligodendrocyte-hGC co-culture to study in real time the migration and interaction of hGCs with their microenvironment. hGCs interact with myelinated and non-myelinated axons through the formation of pseudopodia. Isolation of pseudopodia-localized polysome-bound RNA reveals transcripts of Lck, Paxillin, Crk-II, and Rac1 that undergo local translation. Inhibition of Lck phosphorylation using a small-molecule inhibitor (Lck-I), blocks the phosphorylation of Paxillin and Crk-II, the formation of pseudopodia and the migration of hGCs. In vivo intraventricular administration of the Lck-I using an orthotopic xenograft glioma model, results in statistically significant inhibition of tumor size and significant down-regulation of Nanog-targeted genes, which are associated with glioblastoma patient survival. Moreover, treatment of human glioma stem cells (hGSCs) with Lck-I, results in significant inhibition of self-renewal and tumor-sphere formation. The involvement of Lck in different levels of glioma malignant progression, such as migration, tumor growth, and regulation of cancer stemness, makes Lck a potentially important therapeutic target for human glioblastomas.
Collapse
|
6
|
Alvites R, Rita Caseiro A, Santos Pedrosa S, Vieira Branquinho M, Ronchi G, Geuna S, Varejão AS, Colette Maurício A. Peripheral nerve injury and axonotmesis: State of the art and recent advances. COGENT MEDICINE 2018. [DOI: 10.1080/2331205x.2018.1466404] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Ana Rita Caseiro
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto (REQUIMTE/LAQV), R. Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sílvia Santos Pedrosa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Mariana Vieira Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Giulia Ronchi
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Stefano Geuna
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Artur S.P. Varejão
- CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Ospedale San Luigi, 10043 Orbassano, Turin, Italy
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| |
Collapse
|
7
|
Nerve regeneration techniques respecting the special characteristics of the inferior alveolar nerve. J Craniomaxillofac Surg 2016; 44:1381-6. [PMID: 27435058 DOI: 10.1016/j.jcms.2016.06.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/21/2016] [Accepted: 06/27/2016] [Indexed: 11/21/2022] Open
Abstract
PURPOSE The aim of this study was to examine the in situ regeneration of the inferior alveolar nerve (IAN) in its bony channel, using autologous tissue in combination with a recombinant human nerve growth factor (rhNGF). MATERIALS AND METHODS A total of 20 New Zealand rabbits were randomly divided into five groups. Following dissection of the IAN, the animals underwent reconstruction either with muscle tissue (groups 1 and 2) or with fat tissue (groups 3 and 4). In group 5 (control), the dissected nerve was resected and reconstructed by placement of the reversed autologous segment. After 2 and 4 weeks, 1 mL rhNGF was locally injected in groups 1 and 3. Nerve function was monitored by measuring the jaw-opening reflex using electromyography for a period of 24 weeks. RESULTS Regeneration of the nerve was achieved in all groups, but preoperative threshold values were not achieved. Comparing the experimental groups to the control, there was a significant difference in favor of the autologous nerve reconstruction. Differences between the experimental groups remained statistically not significant. CONCLUSION Regeneration of the IAN with autologous tissue is possible, but without achieving preoperative thresholds. Additional injection of a growth factor seems to improve the speed of regeneration for fat and muscle grafts.
Collapse
|
8
|
Turney SG, Ahmed M, Chandrasekar I, Wysolmerski RB, Goeckeler ZM, Rioux RM, Whitesides GM, Bridgman PC. Nerve growth factor stimulates axon outgrowth through negative regulation of growth cone actomyosin restraint of microtubule advance. Mol Biol Cell 2016; 27:500-17. [PMID: 26631553 PMCID: PMC4751601 DOI: 10.1091/mbc.e15-09-0636] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/19/2023] Open
Abstract
Nerve growth factor (NGF) promotes growth, differentiation, and survival of sensory neurons in the mammalian nervous system. Little is known about how NGF elicits faster axon outgrowth or how growth cones integrate and transform signal input to motor output. Using cultured mouse dorsal root ganglion neurons, we found that myosin II (MII) is required for NGF to stimulate faster axon outgrowth. From experiments inducing loss or gain of function of MII, specific MII isoforms, and vinculin-dependent adhesion-cytoskeletal coupling, we determined that NGF causes decreased vinculin-dependent actomyosin restraint of microtubule advance. Inhibition of MII blocked NGF stimulation, indicating the central role of restraint in directed outgrowth. The restraint consists of myosin IIB- and IIA-dependent processes: retrograde actin network flow and transverse actin bundling, respectively. The processes differentially contribute on laminin-1 and fibronectin due to selective actin tethering to adhesions. On laminin-1, NGF induced greater vinculin-dependent adhesion-cytoskeletal coupling, which slowed retrograde actin network flow (i.e., it regulated the molecular clutch). On fibronectin, NGF caused inactivation of myosin IIA, which negatively regulated actin bundling. On both substrates, the result was the same: NGF-induced weakening of MII-dependent restraint led to dynamic microtubules entering the actin-rich periphery more frequently, giving rise to faster elongation.
Collapse
Affiliation(s)
- Stephen G Turney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Mostafa Ahmed
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Indra Chandrasekar
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Robert B Wysolmerski
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Zoe M Goeckeler
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Robert M Rioux
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - George M Whitesides
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Paul C Bridgman
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
9
|
NGF in Early Embryogenesis, Differentiation, and Pathology in the Nervous and Immune Systems. Curr Top Behav Neurosci 2015; 29:125-152. [PMID: 26695167 DOI: 10.1007/7854_2015_420] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The physiology of NGF is extremely complex, and although the study of this neurotrophin began more than 60 years ago, it is far from being concluded. NGF, its precursor molecule pro-NGF, and their different receptor systems (i.e., TrkA, p75NTR, and sortilin) have key roles in the development and adult physiology of both the nervous and immune systems. Although the NGF receptor system and the pathways activated are similar for all types of cells sensitive to NGF, the effects exerted during embryonic differentiation and in committed mature cells are strikingly different and sometimes opposite. Bearing in mind the pleiotropic effects of NGF, alterations in its expression and synthesis, as well as variations in the types of receptor available and in their respective levels of expression, may have profound effects and play multiple roles in the development and progression of several diseases. In recent years, the use of NGF or of inhibitors of its receptors has been prospected as a therapeutic tool in a variety of neurological diseases and injuries. In this review, we outline the different roles played by the NGF system in various moments of nervous and immune system differentiation and physiology, from embryonic development to aging. The data collected over the past decades indicate that NGF activities are highly integrated among systems and are necessary for the maintenance of homeostasis. Further, more integrated and multidisciplinary studies should take into consideration these multiple and interactive aspects of NGF physiology in order to design new therapeutic strategies based on the manipulation of NGF and its intracellular pathways.
Collapse
|
10
|
Dinh ND, Chiang YY, Hardelauf H, Waide S, Janasek D, West J. Preparation of neuronal co-cultures with single cell precision. J Vis Exp 2014. [PMID: 24894871 DOI: 10.3791/51389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Microfluidic embodiments of the Campenot chamber have attracted great interest from the neuroscience community. These interconnected co-culture platforms can be used to investigate a variety of questions, spanning developmental and functional neurobiology to infection and disease propagation. However, conventional systems require significant cellular inputs (many thousands per compartment), inadequate for studying low abundance cells, such as primary dopaminergic substantia nigra, spiral ganglia, and Drosophilia melanogaster neurons, and impractical for high throughput experimentation. The dense cultures are also highly locally entangled, with few outgrowths (<10%) interconnecting the two cultures. In this paper straightforward microfluidic and patterning protocols are described which address these challenges: (i) a microfluidic single neuron arraying method, and (ii) a water masking method for plasma patterning biomaterial coatings to register neurons and promote outgrowth between compartments. Minimalistic neuronal co-cultures were prepared with high-level (>85%) intercompartment connectivity and can be used for high throughput neurobiology experiments with single cell precision.
Collapse
Affiliation(s)
- Ngoc-Duy Dinh
- Leibniz-Institut für Analytische Wissenschaften, ISAS
| | - Ya-Yu Chiang
- Leibniz-Institut für Analytische Wissenschaften, ISAS; Department of Biochemical Engineering, University College London
| | | | - Sarah Waide
- Leibniz-Institut für Analytische Wissenschaften, ISAS
| | - Dirk Janasek
- Leibniz-Institut für Analytische Wissenschaften, ISAS
| | - Jonathan West
- Leibniz-Institut für Analytische Wissenschaften, ISAS; Institute for Life Sciences, University of Southampton;
| |
Collapse
|
11
|
Siddique R, Vyas A, Thakor N, Brushart TM. A two-compartment organotypic model of mammalian peripheral nerve repair. J Neurosci Methods 2014; 232:84-92. [PMID: 24837281 DOI: 10.1016/j.jneumeth.2014.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 05/05/2014] [Accepted: 05/06/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Schwann cells in the distal stump of transected nerve upregulate growth factors that support regeneration on a modality-specific basis. It is unclear, however, which of these preferentially support motor axon regeneration. Identification of these factors will require a model that can isolate growth factor effects to growing axons while reproducing the complex three-dimensional structure of peripheral nerve. NEW METHOD A two-compartment PDMS base is topped by a collagen-coated membrane that supports a spinal cord cross-section above one compartment. Fluorescent motoneurons in this section reinnervate a segment of peripheral nerve that directs axons through a water-tight barrier to the second compartment, where nerve repair is performed. RESULTS Motoneurons remain healthy for several weeks. The axons they project through the water-tight barrier survive transection and cross a nerve repair in substantial numbers to reinnervate an additional nerve segment. Fluidic isolation of the two compartments was confirmed with a dye leakage test, and the physiologic integrity of the system was tested by retrograde labeling of only those motor neurons to which tracer was exposed and by limitation of toxin effects to a single compartment. COMPARISON WITH EXISTING METHODS Nerve repair cannot be modeled in monolayer cell culture. Our previous organotypic model accurately modeled nerve repair, but did not allow individual control of motoneuron and growth cone environments. CONCLUSIONS This model isolates treatment effects to growing axons while reproducing the complex three-dimensional structure of peripheral nerve. Additionally, it facilitates surgical manipulation of tissues and high-resolution imaging.
Collapse
Affiliation(s)
- Rezina Siddique
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Alka Vyas
- Department of Orthopaedic Surgery, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| | - Nitish Thakor
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.,SINAPSE Institute, National University of Singapore, Singapore
| | - Thomas M Brushart
- Department of Orthopaedic Surgery, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA.,Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| |
Collapse
|
12
|
Li Y, Yang M, Huang Z, Chen X, Maloney MT, Zhu L, Liu J, Yang Y, Du S, Jiang X, Wu JY. AxonQuant: A Microfluidic Chamber Culture-Coupled Algorithm That Allows High-Throughput Quantification of Axonal Damage. Neurosignals 2014; 22:14-29. [PMID: 24603552 DOI: 10.1159/000358092] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 12/18/2013] [Indexed: 11/19/2022] Open
Abstract
Published methods for imaging and quantitatively analyzing morphological changes in neuronal axons have serious limitations because of their small sample sizes, and their time-consuming and nonobjective nature. Here we present an improved microfluidic chamber design suitable for fast and high-throughput imaging of neuronal axons. We developed the AxonQuant algorithm, which is suitable for automatic processing of axonal imaging data. This microfluidic chamber-coupled algorithm allows calculation of an 'axonal continuity index' that quantitatively measures axonal health status in a manner independent of neuronal or axonal density. This method allows quantitative analysis of axonal morphology in an automatic and nonbiased manner. Our method will facilitate large-scale high-throughput screening for genes or therapeutic compounds for neurodegenerative diseases involving axonal damage. When combined with imaging technologies utilizing different gene markers, this method will provide new insights into the mechanistic basis for axon degeneration. Our microfluidic chamber culture-coupled AxonQuant algorithm will be widely useful for studying axonal biology and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yang Li
- School of Electronic Science and Engineering, Nanjing University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Repair of the Peripheral Nerve-Remyelination that Works. Brain Sci 2013; 3:1182-97. [PMID: 24961524 PMCID: PMC4061866 DOI: 10.3390/brainsci3031182] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/07/2013] [Accepted: 07/19/2013] [Indexed: 12/15/2022] Open
Abstract
In this review we summarize the events known to occur after an injury in the peripheral nervous system. We have focused on the Schwann cells, as they are the most important cells for the repair process and facilitate axonal outgrowth. The environment created by this cell type is essential for the outcome of the repair process. The review starts with a description of the current state of knowledge about the initial events after injury, followed by Wallerian degeneration, and subsequent regeneration. The importance of surgical repair, carried out as soon as possible to increase the chances of a good outcome, is emphasized throughout the review. The review concludes by describing the target re-innervation, which today is one of the most serious problems for nerve regeneration. It is clear, compiling this data, that even though regeneration of the peripheral nervous system is possible, more research in this area is needed in order to perfect the outcome.
Collapse
|
14
|
Culture of primary rat hippocampal neurons: design, analysis, and optimization of a microfluidic device for cell seeding, coherent growth, and solute delivery. Biomed Microdevices 2013; 15:97-108. [PMID: 22965807 DOI: 10.1007/s10544-012-9691-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We present the design, analysis, construction, and culture results of a microfluidic device for the segregation and chemical stimulation of primary rat hippocampal neurons. Our device is designed to achieve spatio-temporal solute delivery to discrete sections of neurons with mitigated mechanical stress. We implement a geometric guidance technique to direct axonal processes of the neurons into specific areas of the device to achieve solute segregation along routed cells. Using physicochemical modeling, we predict flows, concentration profiles, and mechanical stresses within pertiment sections of the device. We demonstrate cell viability and growth within the closed device over a period of 11 days. Additionally, our modeling methodology may be generalized and applied to other device geometries.
Collapse
|
15
|
Saijilafu, Zhang BY, Zhou FQ. Signaling pathways that regulate axon regeneration. Neurosci Bull 2013; 29:411-20. [PMID: 23846598 DOI: 10.1007/s12264-013-1357-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/25/2013] [Indexed: 10/26/2022] Open
Abstract
Neurons in the mammalian central nervous system (CNS) cannot regenerate axons after injury. in contrast, neurons in the mammalian peripheral nervous system and in some non-mammalian models, such as C. elegans and Drosophila, are able to regrow axons. Understanding the molecular mechanisms by which these neurons support axon regeneration will help us find ways to enhance mammalian CNS axon regeneration. Here, recent studies in which signaling pathways regulating naturally-occurring axon regeneration that have been identified are reviewed, focusing on how these pathways control gene expression and growth-cone function during axon regeneration.
Collapse
Affiliation(s)
- Saijilafu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
16
|
Dinh ND, Chiang YY, Hardelauf H, Baumann J, Jackson E, Waide S, Sisnaiske J, Frimat JP, van Thriel C, Janasek D, Peyrin JM, West J. Microfluidic construction of minimalistic neuronal co-cultures. LAB ON A CHIP 2013; 13:1402-12. [PMID: 23403713 DOI: 10.1039/c3lc41224e] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
In this paper we present compartmentalized neuron arraying (CNA) microfluidic circuits for the preparation of neuronal networks using minimal cellular inputs (10-100-fold less than existing systems). The approach combines the benefits of microfluidics for precision single cell handling with biomaterial patterning for the long term maintenance of neuronal arrangements. A differential flow principle was used for cell metering and loading along linear arrays. An innovative water masking technique was developed for the inclusion of aligned biomaterial patterns within the microfluidic environment. For patterning primary neurons the technique involved the use of meniscus-pinning micropillars to align a water mask for plasma stencilling a poly-amine coating. The approach was extended for patterning the human SH-SY5Y neuroblastoma cell line using a poly(ethylene glycol) (PEG) back-fill and for dopaminergic LUHMES neuronal precursors by the further addition of a fibronectin coating. The patterning efficiency Epatt was >75% during lengthy in chip culture, with ∼85% of the outgrowth channels occupied by neurites. Neurons were also cultured in next generation circuits which enable neurite guidance into all outgrowth channels for the formation of extensive inter-compartment networks. Fluidic isolation protocols were developed for the rapid and sustained treatment of the different cellular and sub-cellular compartments. In summary, this research demonstrates widely applicable microfluidic methods for the construction of compartmentalized brain models with single cell precision. These minimalistic ex vivo tissue constructs pave the way for high throughput experimentation to gain deeper insights into pathological processes such as Alzheimer and Parkinson Diseases, as well as neuronal development and function in health.
Collapse
Affiliation(s)
- Ngoc-Duy Dinh
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., 44227 Dortmund, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Park JW, Kim HJ, Kang MW, Jeon NL. Advances in microfluidics-based experimental methods for neuroscience research. LAB ON A CHIP 2013; 13:509-521. [PMID: 23306275 DOI: 10.1039/c2lc41081h] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The application of microfluidics to neuroscience applications has always appealed to neuroscientists because of the capability to control the cellular microenvironment in both a spatial and temporal manner. Recently, there has been rapid development of biological micro-electro-mechanical systems (BioMEMS) for both fundamental and applied neuroscience research. In this review, we will discuss the applications of BioMEMS to various topics in the field of neuroscience. The purpose of this review is to summarise recent advances in the components and design of the BioMEMS devices, in vitro disease models, electrophysiology and neural stem cell research. We envision that microfluidics will play a key role in future neuroscience research, both fundamental and applied research.
Collapse
Affiliation(s)
- Jae Woo Park
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Korea
| | | | | | | |
Collapse
|
18
|
Mok SA, Lund K, Lapointe P, Campenot RB. A HaloTag® method for assessing the retrograde axonal transport of the p75 neurotrophin receptor and other proteins in compartmented cultures of rat sympathetic neurons. J Neurosci Methods 2013; 214:91-104. [PMID: 23348044 DOI: 10.1016/j.jneumeth.2013.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 10/19/2012] [Accepted: 01/08/2013] [Indexed: 11/30/2022]
Abstract
We have adapted HaloTag® (HT) technology for use in compartmented cultures of rat sympathetic neurons in order to provide a technique that can be broadly applied to studies of the retrograde transport of molecules that play roles in neurotrophin signaling. Transfected neurons expressing HT protein alone, HT protein fused to the p75 neurotrophin receptor (p75NTR) or HT protein fused to tubulin α-1B were maintained in compartmented cultures in which cell bodies and proximal axons of rat sympathetic neurons reside in proximal compartments and their distal axons extend into distal compartments. HT ligand containing a fluorescent tetramethylrhodamine (TMR) label was applied either in the distal compartments or the proximal compartments, and the transport of labeled proteins was assayed by gel fluorescence imaging and TMR immunoblot. HT protein expressed alone displayed little or no retrograde transport. HT protein fused to either the intracellular C-terminus or the extracellular N-terminus of p75NTR was retrogradely transported. The retrograde transport of p75NTR was augmented when the distal axons were provided with nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) or antibodies to BDNF. The anterograde transport of HT protein fused to the N-terminus of tubulin α-1B was also demonstrated. We conclude that retrograde transport of HT fusion proteins provides a powerful and novel approach in studies of axonal transport.
Collapse
Affiliation(s)
- Sue-Ann Mok
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | | | | | | |
Collapse
|
19
|
A role for Schwann cell-derived neuregulin-1 in remyelination. Nat Neurosci 2012; 16:48-54. [PMID: 23222914 DOI: 10.1038/nn.3281] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/15/2012] [Indexed: 02/06/2023]
Abstract
After peripheral nerve injury, axons regenerate and become remyelinated by resident Schwann cells. However, myelin repair never results in the original myelin thickness, suggesting insufficient stimulation by neuronal growth factors. Upon testing this hypothesis, we found that axonal neuregulin-1 (NRG1) type III and, unexpectedly, also NRG1 type I restored normal myelination when overexpressed in transgenic mice. This led to the observation that Wallerian degeneration induced de novo NRG1 type I expression in Schwann cells themselves. Mutant mice lacking a functional Nrg1 gene in Schwann cells are fully myelinated but exhibit impaired remyelination in adult life. We suggest a model in which loss of axonal contact triggers denervated Schwann cells to transiently express NRG1 as an autocrine/paracrine signal that promotes Schwann cell differentiation and remyelination.
Collapse
|
20
|
Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice. Sci Rep 2012. [PMID: 23205266 PMCID: PMC3510466 DOI: 10.1038/srep00898] [Citation(s) in RCA: 282] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pathological studies on Parkinson's disease (PD) patients suggest that PD pathology progresses from the enteric nervous system (ENS) and the olfactory bulb into the central nervous system. We have previously shown that environmental toxins acting locally on the ENS mimic this PD-like pathology progression pattern in mice. Here, we show for the first time that the resection of the autonomic nerves stops this progression. Moreover, our results show that an environmental toxin (i.e. rotenone) promotes the release of alpha-synuclein by enteric neurons and that released enteric alpha-synuclein is up-taken by presynaptic sympathetic neurites and retrogradely transported to the soma, where it accumulates. These results strongly suggest that pesticides can initiate the progression of PD pathology and that this progression is based on the transneuronal and retrograde axonal transport of alpha-synuclein. If confirmed in patients, this study would have crucial implications in the strategies used to prevent and treat PD.
Collapse
|
21
|
Cengiz N, Oztürk G, Erdoğan E, Him A, Oğuz EK. Consequences of neurite transection in vitro. J Neurotrauma 2012; 29:2465-74. [PMID: 20121423 DOI: 10.1089/neu.2009.0947] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In order to quantify degenerative and regenerative changes and analyze the contribution of multiple factors to the outcome after neurite transection, we cultured adult mouse dorsal root ganglion neurons, and with a precise laser beam, we transected the nerve fibers they extended. Cell preparations were continuously visualized for 24 h with time-lapse microscopy. More distal cuts caused a more elongated field of degeneration, while thicker neurites degenerated faster than thinner ones. Transected neurites degenerated more if the uncut neurites of the same neuron simultaneously degenerated. If any of these uncut processes regenerated, the transected neurites underwent less degeneration. Regeneration of neurites was limited to distal cuts. Unipolar neurons had shorter regeneration than multipolar ones. Branching slowed the regenerative process, while simultaneous degeneration of uncut neurites increased it. Proximal lesions, small neuronal size, and extensive and rapid neurite degeneration were predictive of death of an injured neuron, which typically displayed necrotic rather than apoptotic form. In conclusion, this in vitro model proved useful in unmasking many new aspects and correlates of mechanically-induced neurite injury.
Collapse
Affiliation(s)
- Nurettin Cengiz
- Department of Histology and Embryology, Yüzüncü Yil University Medical School, Van, Turkey
| | | | | | | | | |
Collapse
|
22
|
Yang IH, Gary D, Malone M, Dria S, Houdayer T, Belegu V, McDonald JW, Thakor N. Axon myelination and electrical stimulation in a microfluidic, compartmentalized cell culture platform. Neuromolecular Med 2012; 14:112-8. [PMID: 22527791 DOI: 10.1007/s12017-012-8170-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 02/02/2012] [Indexed: 02/07/2023]
Abstract
Axon demyelination contributes to the loss of sensory and motor function following injury or disease in the central nervous system. Numerous reports have demonstrated that myelination can be achieved in neuron/oligodendrocyte co-cultures. However, the ability to selectively treat neuron or oligodendrocyte (OL) cell bodies in co-cultures improves the value of these systems when designing mechanism-based therapeutics. We have developed a microfluidic-based compartmentalized culture system to achieve segregation of neuron and OL cell bodies while simultaneously allowing the formation of myelin sheaths. Our microfluidic platform allows for a high replicate number, minimal leakage, and high flexibility. Using a custom built lid, fit with platinum electrodes for electrical stimulation (10-Hz pulses at a constant 3 V with ~190 kΩ impedance), we employed the microfluidic platform to achieve activity-dependent myelin segment formation. Electrical stimulation of dorsal root ganglia resulted in a fivefold increase in the number of myelinated segments/mm² when compared to unstimulated controls (19.6 ± 3.0 vs. 3.6 ± 2.3 MBP+ segments/mm²). This work describes the modification of a microfluidic, multi-chamber system so that electrical stimulation can be used to achieve increased levels of myelination while maintaining control of the cell culture microenvironment.
Collapse
Affiliation(s)
- In Hong Yang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
McCall J, Weidner N, Blesch A. Neurotrophic factors in combinatorial approaches for spinal cord regeneration. Cell Tissue Res 2012; 349:27-37. [PMID: 22526621 DOI: 10.1007/s00441-012-1388-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 02/23/2012] [Indexed: 01/09/2023]
Abstract
Axonal regeneration is inhibited by a plethora of different mechanisms in the adult central nervous system (CNS). While neurotrophic factors have been shown to stimulate axonal growth in numerous animal models of nervous system injury, a lack of suitable growth substrates, an insufficient activation of neuron-intrinsic regenerative programs, and extracellular inhibitors of regeneration limit the efficacy of neurotrophic factor delivery for anatomical and functional recovery after spinal cord injury. Thus, growth-stimulating factors will likely have to be combined with other treatment approaches to tap into the full potential of growth factor therapy for axonal regeneration. In addition, the temporal and spatial distribution of growth factors have to be tightly controlled to achieve biologically active concentrations, to allow for the chemotropic guidance of axons, and to prevent adverse effects related to the widespread distribution of neurotrophic factors. Here, we will review the rationale for combinatorial treatments in axonal regeneration and summarize some recent progress in promoting axonal regeneration in the injured CNS using such approaches.
Collapse
Affiliation(s)
- Julianne McCall
- Spinal Cord Injury Center, Heidelberg University Hospital, Schlierbacher Landstrasse 200 a, 69118 Heidelberg, Germany
| | | | | |
Collapse
|
24
|
Liu CM, Hur EM, Zhou FQ. Coordinating Gene Expression and Axon Assembly to Control Axon Growth: Potential Role of GSK3 Signaling. Front Mol Neurosci 2012; 5:3. [PMID: 22347166 PMCID: PMC3272657 DOI: 10.3389/fnmol.2012.00003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 01/09/2012] [Indexed: 12/23/2022] Open
Abstract
Axon growth requires the coordinated regulation of gene expression in the neuronal soma, local protein translation in the axon, anterograde transport of synthesized raw materials along the axon, and assembly of cytoskeleton and membranes in the nerve growth cone. Glycogen synthase kinase 3 (GSK3) signaling has recently been shown to play key roles in the regulation of axonal transport and cytoskeletal assembly during axon growth. GSK3 signaling is also known to regulate gene expression via controlling the functions of many transcription factors, suggesting that GSK3 may be an important regulator of gene transcription supporting axon growth. We review signaling pathways that control local axon assembly at the growth cone and gene expression in the soma during developmental or regenerative axon growth and discuss the potential involvement of GSK3 signaling in these processes, with a particular focus on how GSK3 signaling modulates the function of axon growth-associated transcription factors.
Collapse
Affiliation(s)
- Chang-Mei Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine Baltimore, MD, USA
| | | | | |
Collapse
|
25
|
Elliott T. Cross-Talk Induces Bifurcations in Nonlinear Models of Synaptic Plasticity. Neural Comput 2012; 24:455-522. [DOI: 10.1162/neco_a_00224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Linear models of synaptic plasticity provide a useful starting-point for examining the dynamics of neuronal development and learning, but their inherent problems are well known. Models of synaptic plasticity that embrace the demands of biological realism are therefore typically nonlinear. Viewed from a more abstract perspective, nonlinear models of synaptic plasticity are a subset of nonlinear dynamical systems. As such, they may therefore exhibit bifurcations under the variation of control parameters, including noise and errors in synaptic updates. One source of noise or error is the cross-talk that occurs during otherwise Hebbian plasticity. Under cross-talk, stimulation of a set of synapses can induce or modify plasticity in adjacent, unstimulated synapses. Here, we analyze two nonlinear models of developmental synaptic plasticity and a model of independent component analysis in the presence of a simple model of cross-talk. We show that cross-talk does indeed induce bifurcations in these models, entirely destroying their ability to acquire either developmentally or learning-related patterns of fixed points. Importantly, the critical level of cross-talk required to induce bifurcations in these models is very sensitive to the statistics of the afferents’ activities and the number of afferents synapsing on a postsynaptic cell. In particular, the critical level can be made arbitrarily small. Because bifurcations are inevitable in nonlinear models, our results likely apply to many nonlinear models of synaptic plasticity, although the precise details vary by model. Hence, many nonlinear models of synaptic plasticity are potentially fatally compromised by the toxic influence of cross-talk and other sources of noise and errors more generally. We conclude by arguing that biologically realistic models of synaptic plasticity must be robust against noise-induced bifurcations and that biological systems may have evolved strategies to circumvent their possible dangers.
Collapse
Affiliation(s)
- Terry Elliott
- Department of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, U.K
| |
Collapse
|
26
|
Strakova J, Demizieux L, Campenot RB, Vance DE, Vance JE. Involvement of CTP:phosphocholine cytidylyltransferase-β2 in axonal phosphatidylcholine synthesis and branching of neurons. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:617-25. [DOI: 10.1016/j.bbalip.2011.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 06/06/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
|
27
|
Hellman AN, Vahidi B, Kim HJ, Mismar W, Steward O, Jeon NL, Venugopalan V. Examination of axonal injury and regeneration in micropatterned neuronal culture using pulsed laser microbeam dissection. LAB ON A CHIP 2010; 10:2083-92. [PMID: 20532390 PMCID: PMC3380453 DOI: 10.1039/b927153h] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
We describe the integrated use of pulsed laser microbeam irradiation and microfluidic cell culture methods to examine the dynamics of axonal injury and regeneration in vitro. Microfabrication methods are used to place high purity dissociated central nervous system neurons in specific regions that allow the axons to interact with permissive and inhibitory substrates. Acute injury to neuron bundles is produced via the delivery of single 180 ps duration, lambda = 532 nm laser pulses. Laser pulse energies of 400 nJ and 800 nJ produce partial and complete transection of the axons, respectively, resulting in elliptical lesions 25 mum and 50 mum in size. The dynamics of the resulting degeneration and regrowth of proximal and distal axonal segments are examined for up to 8 h using time-lapse microscopy. We find the proximal and distal dieback distances from the site of laser microbeam irradiation to be roughly equal for both partial and complete transection of the axons. In addition, distinct growth cones emerge from the proximal neurite segments within 1-2 h post-injury, followed by a uniform front of regenerating axons that originate from the proximal segment and traverse the injury site within 8 h. We also examine the use of EGTA to chelate the extracellular calcium and potentially reduce the severity of the axonal degeneration following injury. While we find the addition of EGTA to reduce the severity of the initial dieback, it also hampers neurite repair and interferes with the formation of neuronal growth cones to traverse the injury site. This integrated use of laser microbeam dissection within a micropatterned cell culture system to produce precise zones of neuronal injury shows potential for high-throughput screening of agents to promote neuronal regeneration.
Collapse
Affiliation(s)
- Amy N. Hellman
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
- Department of Chemical Engineering & Materials Science, University of California, Irvine, CA 92697
- Laser Microbeam and Medical Program, Beckman Laser Institute, University of California, Irvine, CA 92697
| | - Behrad Vahidi
- Department of Biomedical Engineering, University of California, Irvine, CA 92697
- Department of Mechanical & Aerospace Engineering, Seoul National University, Seoul 151-742, KOREA
| | - Hyung Joon Kim
- Department of Biomedical Engineering, University of California, Irvine, CA 92697
| | - Wael Mismar
- Department of Biomedical Engineering, University of California, Irvine, CA 92697
| | - Oswald Steward
- Reeve-Irvine Research Center for Spinal Cord Injury, University of California, Irvine, CA 92697
| | - Noo Li Jeon
- Department of Biomedical Engineering, University of California, Irvine, CA 92697
- Department of Mechanical & Aerospace Engineering, Seoul National University, Seoul 151-742, KOREA
| | - Vasan Venugopalan
- Department of Chemical Engineering & Materials Science, University of California, Irvine, CA 92697
- Laser Microbeam and Medical Program, Beckman Laser Institute, University of California, Irvine, CA 92697
- Department of Biomedical Engineering, University of California, Irvine, CA 92697
- Correspondence:
| |
Collapse
|
28
|
Park KK, Liu K, Hu Y, Kanter JL, He Z. PTEN/mTOR and axon regeneration. Exp Neurol 2010; 223:45-50. [DOI: 10.1016/j.expneurol.2009.12.032] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 12/28/2009] [Accepted: 12/30/2009] [Indexed: 12/18/2022]
|
29
|
Campenot RB, Lund K, Mok SA. Production of compartmented cultures of rat sympathetic neurons. Nat Protoc 2009; 4:1869-87. [DOI: 10.1038/nprot.2009.210] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
30
|
Hernández-Ochoa EO, Prosser BL, Wright NT, Contreras M, Weber DJ, Schneider MF. Augmentation of Cav1 channel current and action potential duration after uptake of S100A1 in sympathetic ganglion neurons. Am J Physiol Cell Physiol 2009; 297:C955-70. [PMID: 19657060 DOI: 10.1152/ajpcell.00140.2009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
S100A1, a 21-kDa dimeric Ca2+-binding protein of the EF-hand type, is expressed in cardiomyocytes and is an important regulator of heart function. During ischemia, cardiomyocytes secrete S100A1 to the extracellular space. Although the effects of extracellular S100A1 have been documented in cardiomyocytes, it is unclear whether S100A1 exerts modulatory effects on other tissues in proximity with cardiac cells. Therefore, we sought to investigate the effects of exogenous S100A1 on Ca2+ signals and electrical properties of superior cervical ganglion (SCG) neurons. Immunostaining and Western blot assays indicated no endogenous S100A1 in SCG neurons. Cultured SCG neurons took up S100A1 when it was present in the extracellular medium. Inside the cell exogenous S100A1 localized in a punctate pattern throughout the cytoplasm but was excluded from the nuclei. S100A1 partially colocalized with markers for both receptor- and non-receptor-mediated endocytosis, indicating that in SCG neurons multiple endocytotic pathways are involved in S100A1 internalization. In compartmentalized SCG cultures, axonal projections were capable of uptake and transport of S100A1 toward the neuronal somas. Exogenous S100A1 applied either extra- or intracellularly enhanced Cav1 channel currents in a PKA-dependent manner, prolonged action potentials, and amplified action potential-induced Ca2+ transients. NMR chemical shift perturbation of Ca2+-S100A1 in the presence of a peptide from the regulatory subunit of PKA verifies that S100A1 directly interacts with PKA, and that this interaction likely occurs in the hydrophobic binding pocket of Ca2+-S100A1. Our results suggest the hypothesis that in sympathetic neurons exogenous S100A1 may lead to an increase of sympathetic output.
Collapse
Affiliation(s)
- Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | |
Collapse
|
31
|
NGF uptake and retrograde signaling mechanisms in sympathetic neurons in compartmented cultures. Results Probl Cell Differ 2009; 48:141-58. [PMID: 19343309 DOI: 10.1007/400_2009_7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Many neurons depend for their survival on retrograde signals to their cell bodies generated by nerve growth factor (NGF) or other neurotrophins at their axon terminals. Apoptosis resulting from the loss of retrograde NGF signaling contributes to the elimination of excess and misconnected neurons during development and to the death of neurons during the course of neurodegenerative diseases. Possible mechanisms of retrograde signaling include (1) retrograde transport of signaling endosomes, carrying NGF bound to activated TrkA, (2) retrograde transport of signaling molecules downstream of TrkA, and (3) retrograde propagation of a phosphorylation signal without transport of signaling molecules. Evidence is also described, which indicates that two or more retrograde signaling mechanisms exist to regulate neuronal survival, including recent evidence that withdrawal of NGF from distal axons produces a retrograde apoptotic signal, which is transported to the cell bodies, where it initiates the apoptotic program, leading to the death of the neuron.
Collapse
|
32
|
Damon DH. TH and NPY in sympathetic neurovascular cultures: role of LIF and NT-3. Am J Physiol Cell Physiol 2007; 294:C306-12. [PMID: 18032527 DOI: 10.1152/ajpcell.00214.2007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The sympathetic nervous system is an important determinant of vascular function. The effects of the sympathetic nervous system are mediated via release of neurotransmitters and neuropeptides from postganglionic sympathetic neurons. The present study tests the hypothesis that vascular smooth muscle cells (VSM) maintain adrenergic neurotransmitter/neuropeptide expression in the postganglionic sympathetic neurons that innervate them. The effects of rat aortic and tail artery VSM (AVSM and TAVSM, respectively) on neuropeptide Y (NPY) and tyrosine hydroxylase (TH) were assessed in cultures of dissociated sympathetic neurons. AVSM decreased TH (39 +/- 12% of control) but did not affect NPY. TAVSM decreased TH (76 +/- 10% of control) but increased NPY (153 +/- 20% of control). VSM expressed leukemia inhibitory factor (LIF) and neurotrophin-3 (NT-3), which are known to modulate NPY and TH expression. Sympathetic neurons innervating blood vessels expressed LIF and NT-3 receptors. Inhibition of LIF inhibited the effect of AVSM on TH. Inhibition of neurotrophin-3 (NT-3) decreased TH and NPY in neurons grown in the presence of TAVSM. These data suggest that vascular-derived LIF decreases TH and vascular-derived NT-3 increases or maintains NPY and TH expression in postganglionic sympathetic neurons. NPY and TH in vascular sympathetic nerves are likely to modulate NPY and/or norepinephrine release from these nerves and are thus likely to affect blood flow and blood pressure. The present studies suggest a novel mechanism whereby VSM would modulate sympathetic control of vascular function.
Collapse
Affiliation(s)
- Deborah H Damon
- Dept. of Pharmacology, Univ. of Vermont, 89 Beaumont Ave., Given Bldg., Burlington, VT 05405, USA.
| |
Collapse
|
33
|
Carter JM, Demizieux L, Campenot RB, Vance DE, Vance JE. Phosphatidylcholine biosynthesis via CTP:phosphocholine cytidylyltransferase 2 facilitates neurite outgrowth and branching. J Biol Chem 2007; 283:202-212. [PMID: 17981805 DOI: 10.1074/jbc.m706531200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Hallmarks of neuronal differentiation are neurite sprouting, extension, and branching. We previously showed that increased expression of CTP:phosphocholine cytidylyltransferase beta2 (CTbeta2), an isoform of a key phosphatidylcholine (PC) biosynthetic enzyme, accompanies neurite outgrowth (Carter, J. M., Waite, K. A., Campenot, R. B., Vance, J. E., and Vance, D. E. (2003) J. Biol. Chem. 278, 44988-44994). CTbeta2 mRNA is highly expressed in the brain. We show that CTbeta2 is abundant in axons of rat sympathetic neurons and retinal ganglion cells. We used RNA silencing to decrease CTbeta2 expression in PC12 cells differentiated by nerve growth factor. In CTbeta2-silenced cells, numbers of primary and secondary neurites were markedly reduced, suggesting that CTbeta2 facilitates neurite outgrowth and branching. However, the length of individual neurites was significantly increased, and the total amount of neuronal membrane was unchanged. Neurite branching of PC12 cells is known to be inhibited by activation of Akt and promoted by the Akt inhibitor LY294002. Our experiments showed that LY294002 increases neurite sprouting and branching in control PC12 cells but not in CTbeta2-deficient cells. CTbeta2 was not phosphorylated in vitro by Akt. However, inhibition of Cdk5 by roscovitine blocked CTbeta2 phosphorylation and reduced neurite outgrowth and branching. These results highlight the importance of CTbeta2 in neurons for promoting neurite outgrowth and branching and represent the first identification of a lipid biosynthetic enzyme that facilitates these functions.
Collapse
Affiliation(s)
- Jodi M Carter
- Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta T6G 2S2, Canada; Department of Biochemistry, Edmonton, Alberta T6G 2S2, Canada
| | - Laurent Demizieux
- Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta T6G 2S2, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | | | - Dennis E Vance
- Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta T6G 2S2, Canada; Department of Biochemistry, Edmonton, Alberta T6G 2S2, Canada
| | - Jean E Vance
- Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta T6G 2S2, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2S2, Canada.
| |
Collapse
|
34
|
Saavedra L, Mohamed A, Ma V, Kar S, de Chaves EP. Internalization of beta-amyloid peptide by primary neurons in the absence of apolipoprotein E. J Biol Chem 2007; 282:35722-32. [PMID: 17911110 DOI: 10.1074/jbc.m701823200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Extracellular accumulation of beta-amyloid peptide (Abeta) has been linked to the development of Alzheimer disease. The importance of intraneuronal Abeta has been recognized more recently. Although considerable evidence indicates that extracellular Abeta contributes to the intracellular pool of Abeta, the mechanisms involved in Abeta uptake by neurons are poorly understood. We examined the molecular mechanisms involved in Abeta-(1-42) internalization by primary neurons in the absence of apolipoprotein E. We demonstrated that Abeta-(1-42) is more efficiently internalized by axons than by cell bodies of sympathetic neurons, suggesting that Abeta-(1-42) uptake might be mediated by proteins enriched in the axons. Although the acetylcholine receptor alpha7nAChR, previously suggested to be involved in Abeta internalization, is enriched in axons, our results indicate that it does not mediate Abeta-(1-42) internalization. Moreover, receptors of the low density lipoprotein receptor family are not essential for Abeta-(1-42) uptake in the absence of apolipoprotein E because receptor-associated protein had no effect on Abeta uptake. By expressing the inactive dynamin mutant dynK44A and the clathrin hub we found that Abeta-(1-42) internalization is independent of clathrin but dependent on dynamin, which suggests an endocytic pathway involving caveolae/lipid rafts. Confocal microscopy studies showing that Abeta did not co-localize with the early endosome marker EEA1 further support a clathrin-independent mechanism. The lack of co-localization of Abeta with caveolin in intracellular vesicles and the normal uptake of Abeta by neurons that do not express caveolin indicate that Abeta does not require caveolin either. Instead partial co-localization of Abeta-(1-42) with cholera toxin subunit B and sensitivity to reduction of cellular cholesterol and sphingolipid levels suggest a caveolae-independent, raft-mediated mechanism. Understanding the molecular events involved in neuronal Abeta internalization might identify potential therapeutic targets for Alzheimer disease.
Collapse
Affiliation(s)
- Lucila Saavedra
- Department of Pharmacology, University of Alberta, Edmonton, Canada
| | | | | | | | | |
Collapse
|
35
|
Blackmore M, Letourneau PC. Protein synthesis in distal axons is not required for axon growth in the embryonic spinal cord. Dev Neurobiol 2007; 67:976-86. [PMID: 17506497 DOI: 10.1002/dneu.20395] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
It is now well established that new proteins are synthesized in the distal segments of elongating axons, where they may play an essential role in some guidance decisions. It remains unclear, however, whether distal protein synthesis also plays an essential role in axon growth per se. Previous in vitro experiments have shown that blocking protein synthesis in distal axons has no effect on the rate of axonal advance. However, because these experiments were performed in vitro and over a relatively short time period, the role of distal protein synthesis over longer periods and in a native tissue environment remained untested. Here, we tested whether protein synthesis in distal axons plays an essential role in the elongation of descending axons in the embryonic spinal cord. We developed an in situ model of the brainstem-spinal projection of the embryonic chick, and developed a split-chamber method in which inhibitors of proteins synthesis could be applied independently to cell bodies in the brainstem or to distal axons in the spinal cord. When protein synthesis was blocked in distal axons, axon growth remained robust for 2 days, which is the length of the experiment. However, when protein synthesis was blocked only in the brainstem, axonal elongation in the spinal cord ceased within 6 h. These data showed that protein synthesis in the distal axon is not essential to continue the advance of axons. Rather, essential proteins are synthesized more proximally and then transported rapidly to the distal axon.
Collapse
Affiliation(s)
- Murray Blackmore
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
36
|
O'Brien JJ, Nathanson NM. Retrograde activation of STAT3 by leukemia inhibitory factor in sympathetic neurons. J Neurochem 2007; 103:288-302. [PMID: 17608645 DOI: 10.1111/j.1471-4159.2007.04736.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Leukemia inhibitory factor (LIF) is a member of the interleukin-6 family of cytokines and signals through the glycoprotein 130 and LIF receptor beta subunits. Binding of cytokines to these subunits activates multiple signaling cascades, including the Janus kinase (Jak)/signal transducers and activators of transcription (STAT) pathway. We used compartmentalized cultures of sympathetic neurons and immunocytochemical analyses of STAT3 to examine the mechanisms involved in retrograde signaling of LIF from distal neurites (DN) to cell bodies. Addition of LIF to the DN of these neurons triggers the activation and nuclear translocation of STAT3. Inhibition of Jak activity in the cell bodies prevented LIF-induced retrograde activation of STAT3, while block of Jak activity in the DN had no effect on the appearance of activated STAT3 in the nucleus. These results show that the transport of activated Jak is not the main mechanism mediating retrograde signaling. Although there is an increase in phosphorylated STAT3 in the neurites after distal stimulation, the transport of activated STAT3 is not necessary for retrograde signaling. Our results are consistent with a signaling endosome model for retrograde signaling, in which the LIF/glycoprotein 130/LIF receptor/Jak complex is internalized and transported to activate STAT3 in the cell body.
Collapse
Affiliation(s)
- Jennifer J O'Brien
- Department of Pharmacology, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
37
|
Low LK, Cheng HJ. Axon pruning: an essential step underlying the developmental plasticity of neuronal connections. Philos Trans R Soc Lond B Biol Sci 2007; 361:1531-44. [PMID: 16939973 PMCID: PMC1664669 DOI: 10.1098/rstb.2006.1883] [Citation(s) in RCA: 197] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Regressive events play a key role in modifying neural connectivity in early development. An important regressive event is the pruning of neuronal processes. Pruning is a strategy often used to selectively remove exuberant neuronal branches and connections in the immature nervous system to ensure the proper formation of functional circuitry. In the following review, we discuss our present understanding of the cellular and molecular mechanisms that regulate the pruning of axons during neuronal development as well as in neurological diseases. The evidence suggests that there are several similarities between the mechanisms that are involved in developmental axon pruning and axon elimination in disease. In summary, these findings provide researchers with a unique perspective on how developmental plasticity is achieved and how to develop strategies to treat complex neurological diseases.
Collapse
|
38
|
Abstract
Neurotrophins are a small family of dimeric secretory proteins in vertebrate neurons with a broad spectrum of functions. They are generated as pro-proteins with a functionality that is distinct from the proteolytically processed form. The cellular responses of neurotrophins are mediated by three different types of receptor proteins, the receptor tyrosine kinases of the Trk family, the neurotrophin receptor p75(NTR), which is a member of the tumor necrosis factor receptor (TNFR) superfamily, and sortilin, previously characterized as neurotensin receptor. Recent studies have revealed an intriguing pattern: neurotrophins can elicit opposing signals utilising their variable configuration and different receptor types.
Collapse
Affiliation(s)
- Rüdiger Schweigreiter
- Biocenter Innsbruck, Division of Neurobiochemistry, Medical University Innsbruck, Fritz-Pregl-Strasse 3, A-6020 Innsbruck Innsbruck, Austria.
| |
Collapse
|
39
|
Zhou FQ, Snider WD. Intracellular control of developmental and regenerative axon growth. Philos Trans R Soc Lond B Biol Sci 2006; 361:1575-92. [PMID: 16939976 PMCID: PMC1664665 DOI: 10.1098/rstb.2006.1882] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Accepted: 06/19/2006] [Indexed: 01/10/2023] Open
Abstract
Axon growth is a highly regulated process that requires stimulating signals from extracellular factors. The extracellular signals are then transduced to regulate coordinately gene expression and local axon assembly. Growth factors, especially neurotrophins that act via receptor tyrosine kinases, have been heavily studied as extracellular factors that stimulate axon growth. Downstream of receptor tyrosine kinases, recent studies have suggested that phosphatidylinositol-3 kinase (PI3K) regulates local assembly of axonal cytoskeleton, especially microtubules, via glycogen synthase kinase 3beta (GSK-3beta) and multiple microtubule binding proteins. The role of extracellular signal regulated kinase (ERK) signalling in regulation of local axon assembly is less clear, but may involve the regulation of local protein translation. Gene expression during axon growth is regulated by transcription factors, among which cyclic AMP response element binding protein and nuclear factors of activated T-cells (NFATs) are known to be required for neurotrophin (NT)-induced axon extension. In addition to growth factors, extracellular matrix molecules and neuronal activity contribute importantly to control axon growth. Increasingly, evidence suggests that these influences act to enhance growth via coordinating with growth factor signalling. Finally, evidence is emerging that developmental versus regenerative axon growth may be mediated by distinct signalling pathways, both at the level of gene transcription and at the level of local axon assembly.
Collapse
Affiliation(s)
- Feng-Quan Zhou
- Department of Orthopedic Surgery, The Johns Hopkins University School of MedicineBaltimore, MD 21287, USA
- Department of Neuroscience, The Johns Hopkins University School of MedicineBaltimore, MD 21205, USA
| | - William D Snider
- UNC-Neuroscience Center, University of North Carolina at Chapel Hill8109 Neuroscience Research Building, 103 Mason Farm Road, Chapel Hill, NC 27599-7250, USA
| |
Collapse
|
40
|
Mok SA, Campenot RB. A nerve growth factor-induced retrograde survival signal mediated by mechanisms downstream of TrkA. Neuropharmacology 2006; 52:270-8. [PMID: 16949623 DOI: 10.1016/j.neuropharm.2006.07.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2006] [Revised: 07/03/2006] [Accepted: 07/14/2006] [Indexed: 11/16/2022]
Abstract
Considerable evidence suggests that mammalian neurons are always poised to destroy themselves by apoptosis but are blocked by retrograde survival signals triggered in their axon terminals by neurotrophic factors secreted by the target cells they innervate. Studies with nerve growth factor (NGF) and its receptor, TrkA, form the basis of the prevalent theory of retrograde signaling. According to this theory, retrograde survival signals travel to the cell bodies in the form of endosomes produced at the axon terminals with internalized NGF in their lumens bound to phosphorylated TrkA in their membranes. The inhibition of TrkA phosphorylation in the cell bodies of sympathetic neurons in compartmented cultures by K252a blocked retrograde NGF signaling in some studies in accord with this theory, but other studies do not show a block. We report that local block of TrkA phosphorylation in the cell bodies and proximal axons with another kinase inhibitor, Gö6976 (25nM), did not block the survival signal from NGF at distal axons, while Gö6976 at the distal axons completely blocked the retrograde survival signal. These results suggest that downstream signals activated by phosphorylated TrkA in the distal axons carry the retrograde survival signals to the cell bodies, possibly via a downstream type of signaling endosome not necessarily transporting NGF or phosphorylated TrkA. Unlike Gö6976, K252a exerted a survival effect on its own when applied to cell bodies/proximal axons or distal axons of completely NGF-deprived neurons. The latter effect suggests that downstream retrograde survival signals can arise from alterations in one or more kinase activities in the distal axons without activation of TrkA by NGF.
Collapse
Affiliation(s)
- Sue A Mok
- Department of Cell Biology, University of Alberta, 5-14 Medical Sciences Building, Edmonton, Alberta T6G 2H7, Canada
| | | |
Collapse
|
41
|
Abstract
Compared with other organs, the brain is highly enriched in cholesterol. Essentially all cholesterol in the brain is synthesized within the brain; the blood-brain barrier prevents the import of plasma lipoproteins into the brain. Consequently, the brain operates an independent lipoprotein transport system in which glial cells produce ApoE (apolipoprotein E)-containing lipoproteins that are thought to deliver cholesterol to neurons for axonal growth and repair. We have shown that ApoE-containing lipoproteins generated by glial cells stimulate axon extension. ApoE associated with lipoprotein particles, and a receptor of the low-density lipoprotein receptor family, are required for stimulation of axon growth. NPC (Niemann-Pick type C) disease is a severe neurological disorder caused by mutations in the NPC1 or NPC2 gene. A hallmark of this disease is impaired transport of cholesterol out of late endosomes/lysosomes and the accumulation of cholesterol in these organelles. Although cholesterol accumulates in cell bodies of neurons from NPC1-deficient mice, the cholesterol content of axons is reduced. The presence of NPC1 in endosomal structures in nerve terminals, and the finding of aberrant synaptic vesicles, suggest that defects in synaptic vesicle recycling contribute to neurological abnormalities characteristic of NPC disease. We have also shown that ApoE-containing lipoproteins produced by glial cells from NCP1-deficient mice are of normal composition and stimulate axon extension.
Collapse
Affiliation(s)
- J E Vance
- Canadian Institutes for Health Research Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton.
| | | | | |
Collapse
|
42
|
Melli G, Keswani SC, Fischer A, Chen W, Höke A. Spatially distinct and functionally independent mechanisms of axonal degeneration in a model of HIV-associated sensory neuropathy. ACTA ACUST UNITED AC 2006; 129:1330-8. [PMID: 16537566 DOI: 10.1093/brain/awl058] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Sensory polyneuropathies are the most frequent neurological complication of human immunodeficiency virus (HIV) infection. Distal symmetric polyneuropathy (DSP), associated with HIV infection, is characterized by length-dependent axonal degeneration of sensory fibres. In rodent dorsal root ganglia (DRG) cultures, HIV viral envelope protein gp120 results in neurotoxicity and axonal degeneration. Since it is unknown whether the axonal degeneration is a consequence of neuronal death or whether it is due to a direct toxic effect on axons, we investigated gp120-induced axonal toxicity using compartmentalized cultures of sensory neurons. Our results show that gp120 causes neuronal apoptosis and axonal degeneration through two, independent and spatially separated mechanisms of action: (i) an indirect insult to cell bodies, requiring the presence of Schwann cells, results in neuronal apoptotic death and subsequent axonal degeneration; (ii) a direct, local toxicity exerted on axons through activation of mitochondrial caspase pathway that is independent of cell body. This local axonal toxicity is mediated through binding of gp120 to axonal chemokine receptors and can be prevented by chemokine receptor blockers. In conclusion, we propose a novel pathway of axonal degeneration mediated by gp120 that is dependent on local activation of caspases in the axon. This observation suggests that axonal protection is a relevant therapeutic target for HIV-associated sensory neuropathy. Furthermore, chemokine receptor inhibitors, which are currently being developed as HIV entry inhibitor drugs, may also have a therapeutic role in HIV-associated peripheral neuropathies by preventing axonal degeneration.
Collapse
Affiliation(s)
- Giorgia Melli
- Department of Neurology, Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | |
Collapse
|
43
|
Singh KK, Miller FD. Activity regulates positive and negative neurotrophin-derived signals to determine axon competition. Neuron 2005; 45:837-45. [PMID: 15797546 DOI: 10.1016/j.neuron.2005.01.049] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2004] [Revised: 09/23/2004] [Accepted: 01/14/2005] [Indexed: 11/29/2022]
Abstract
Developmental axon competition plays a key role in sculpting neural circuitry. Here, we have asked how activity and neurotrophins could interact to select one axon over another. Using compartmented cultures of sympathetic neurons, we show that, in the presence of NGF, local depolarization confers a competitive growth advantage on the depolarized axon collaterals and at the same time disadvantages the growth of unstimulated axons from the same and competing neurons. Depolarization mediates the competitive advantage by activating a CaMKII-MEK pathway, which converges to enhance local NGF-mediated downstream growth signals. Patterned electrical stimulation also acts via this pathway to enhance NGF-promoted axonal growth. In contrast, the competitive disadvantage is due to BDNF secreted from and acting on the unstimulated, competing axons through p75NTR. Thus, activity regulates both positive and negative neurotrophin-derived signaling cascades to confer a competitive growth advantage on one axon versus another, thereby providing a cellular mechanism for developmental axon selection.
Collapse
Affiliation(s)
- Karun K Singh
- Department of Developmental Biology, Institute of Medical Sciences, University of Toronto, Toronto, M5G 1X8, Canada
| | | |
Collapse
|
44
|
Wittig JH, Ryan AF, Asbeck PM. A reusable microfluidic plate with alternate-choice architecture for assessing growth preference in tissue culture. J Neurosci Methods 2005; 144:79-89. [PMID: 15848242 DOI: 10.1016/j.jneumeth.2004.10.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2004] [Revised: 10/15/2004] [Accepted: 10/15/2004] [Indexed: 11/30/2022]
Abstract
We present the design of a chamber to evaluate in vitro how species and concentrations of soluble molecules control features of cell growth-potentially including cell proliferation, cell motility, process extension, and process termination. We have created a reusable cell culture plate that integrates a microfluidic media delivery network with standard cell culture environment. The microfluidic network delivers a stream of cell culture media with a step-like concentration gradient down a 50-100 microm wide microchannel called the presentation region. Migrating cells or growing cell processes freely choose between the two distinct chemical environments in the presentation region, but they are forced to exclusively choose either one environment or the other when they grow past a physical barrier acting as a decision point. Our fabrication technique requires little specialized equipment, and can be carried out in approximately 4 days per plate. We demonstrate the effectiveness of our plates as neurites from spiral ganglion explants preferentially grow in media containing neurotrophin-3 (NT-3) as opposed to media without NT-3. Our design could be used without modification to study dissociated cell responses to soluble growth cues, and for behavioral screening of small motile organisms.
Collapse
Affiliation(s)
- John H Wittig
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA 92093-0407, USA.
| | | | | |
Collapse
|
45
|
Abstract
Cholesterol is highly enriched in the brain compared to other tissues. Essentially all cholesterol in the brain is synthesized endogenously since plasma lipoproteins are unable to cross the blood-brain barrier. Cholesterol is transported within the central nervous system in the form of apolipoprotein E-containing lipoprotein particles that are secreted mainly by glial cells. Cholesterol is excreted from the brain in the form of 24-hydroxycholesterol. Apolipoprotein E and cholesterol have been implicated in the formation of amyloid plaques in Alzheimer's disease. In addition, the progressive neurodegenerative disorder Niemann-Pick C disease is characterized by defects in intracellular trafficking of cholesterol.
Collapse
Affiliation(s)
- Jean E Vance
- Department of Medicine, Canadian Institutes for Health Research, Group on the Molecular and Cell Biology of Lipids, 332 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada T6G 2S2.
| | | | | |
Collapse
|
46
|
Zhou FQ, Zhou J, Dedhar S, Wu YH, Snider WD. NGF-induced axon growth is mediated by localized inactivation of GSK-3beta and functions of the microtubule plus end binding protein APC. Neuron 2004; 42:897-912. [PMID: 15207235 DOI: 10.1016/j.neuron.2004.05.011] [Citation(s) in RCA: 433] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2003] [Revised: 03/26/2004] [Accepted: 05/10/2004] [Indexed: 12/15/2022]
Abstract
Little is known about how nerve growth factor (NGF) signaling controls the regulated assembly of microtubules that underlies axon growth. Here we demonstrate that a tightly regulated and localized activation of phosphatidylinositol 3-kinase (PI3K) at the growth cone is essential for rapid axon growth induced by NGF. This spatially activated PI3K signaling is conveyed downstream through a localized inactivation of glycogen synthase kinase 3beta (GSK-3beta). These two spatially coupled kinases control axon growth via regulation of a microtubule plus end binding protein, adenomatous polyposis coli (APC). Our results demonstrate that NGF signals are transduced to the axon cytoskeleton via activation of a conserved cell polarity signaling pathway.
Collapse
Affiliation(s)
- Feng-Quan Zhou
- UNC-Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
47
|
Hayashi H, Karten B, Vance DE, Campenot RB, Maue RA, Vance JE. Methods for the study of lipid metabolism in neurons. Anal Biochem 2004; 331:1-16. [PMID: 15245991 DOI: 10.1016/j.ab.2004.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2004] [Indexed: 11/21/2022]
Affiliation(s)
- Hideki Hayashi
- Group on Molecualr and Cell Biology of Lipids and Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Kulbatski I, Cook DJ, Tator CH. Calcium entry through L-type calcium channels is essential for neurite regeneration in cultured sympathetic neurons. J Neurotrauma 2004; 21:357-74. [PMID: 15115609 DOI: 10.1089/089771504322972130] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Previous work showed that a post-neuritotomy rise in [Ca2+]i is required for regeneration. We tested the following hypotheses in cultured sympathetic neurons: (1) blocking L-type channels at the time of injury inhibits regeneration; (2) enhancing Ca2+ entry through L-type Ca2+ channels enhances regeneration; (3) L-type Ca2+ channel distribution is predominantly on the soma and proximal neurites of uninjured and injured neurons. To visualize L-type Ca2+ channels and block Ca2+ influx, the fluorescent dihydropyridine antagonist, DM-BODIPY, was used. Our results show that regeneration is markedly inhibited by the antagonist when administered 20 min. prior to injury, in the presence or absence of nerve growth factor (NGF) (p < 0.0001). Severe degeneration of proximal and distal neurites was seen 48 h after injury. Regeneration was minimally inhibited by the antagonist when administered 5 min after injury (p < 0.05), but not inhibited when administered 2 or 24 h after injury (p > 0.05). We found that L-type channels are distributed ubiquitously on the soma and neurites of uninjured and injured cells, and on regenerating neurites. The addition of the L-type channel agonist, BayK8644, (1 microM) 20 min prior to injury enhanced neurite length at 24 h post-injury (p = 0.002). Blocking L-type channels did not affect the viability of uninjured or injured cells. For the first time, it has been shown that Ca2+ entry through L-type Ca2+ channels is essential for post-neuritotomy sympathetic neurite regeneration, and that this effect shows a strict temporal dependency. We also demonstrated that regeneration can be enhanced by increasing Ca2+ influx through L-type channels.
Collapse
Affiliation(s)
- Iris Kulbatski
- Toronto Western Research Institute, University of Toronto, and Toronto Western Hospital, Toronto, Ontario, Canada
| | | | | |
Collapse
|
49
|
Cook DJ, Kulbatski I, Tator CH. Inosine reverses the inhibitory effects of the L-type Ca2+ channel antagonist, DM-BODIPY-dihydropyridine, on neuritogenesis in an in vitro rat superior cervical ganglia axotomy model. Neurosci Lett 2004; 358:75-8. [PMID: 15026152 DOI: 10.1016/j.neulet.2003.12.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2003] [Revised: 12/09/2003] [Accepted: 12/10/2003] [Indexed: 11/28/2022]
Abstract
It has recently been demonstrated that L-type calcium channel antagonism with the fluorescent dihydropyridine DM-BODIPY-dihydropyridine (DMBD) inhibits neurite regeneration in rat superior cervical ganglia (SCG). The neuritogenic effects of inosine have been described in various models and the mechanism is thought to be N-kinase dependent. Because of the final common pathway between calcium dependent and N-kinase dependent neurite regeneration it was hypothesized that inosine would increase regeneration in normally regenerating SCG and reverse the inhibitory effects of DMBD on regenerating SCG. An in vitro model of rat SCG injury, where mature neurites are transected and observed at 2 and 24 h, was used to assess the effects of inosine on DMBD treated neurons. Results demonstrate a significant inhibition of growth in DMBD treated cultures, significantly increased growth in the inosine + DMBD treated SCG over DMBD treated cells and significantly increased growth in the inosine alone treated group over control cells. There is also evidence that inosine + DMBD treatment promotes linear growth of neurites. The implications of the findings are discussed.
Collapse
Affiliation(s)
- Douglas J Cook
- Division of Cellular and Molecular Biology, Toronto Western Research Institute, 399 Bathurst Street, Toronto, Ontario M5T 2S8, Canada
| | | | | |
Collapse
|
50
|
Abstract
The development of dendrites is a crucial step in the formation of cortical circuitry. The morphogen brain-derived neurotrophic factor (BDNF) may mediate the effects of activity on dendritic morphology since its expression and release are thought to be activity-dependent. Using two-photon microscopy, the autocrine and paracrine effects of BDNF on dendritic morphology were assessed. Overexpression of BDNF profoundly altered the form and stability of basal dendritic arbors via an autocrine mechanism. Paracrine BDNF also altered dendritic branching, though in a highly local fashion. BDNF is capable of acting as an intercellular morphogen, and could hypothetically shape dendritic arbors to best fit the developing structure and function of the pre-synaptic circuit.
Collapse
|