1
|
Nawrotek K, Chyb M, Gatkowska J, Rudnicka K, Michlewska S, Jóźwiak P. Effect of sodium L-lactate on bioactive properties of chitosan-hydroxyapatite/polycaprolactone conduits for peripheral nerve tissue engineering. Int J Biol Macromol 2024; 281:136254. [PMID: 39366606 DOI: 10.1016/j.ijbiomac.2024.136254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Biomaterials and synthetic polymers have been widely used to replicate the regenerative microenvironment of the peripheral nervous system. Chitosan-based conduits have shown promise in the regeneration of nerve injuries. However, to mimic the regenerative microenvironment, the scaffold structure should possess bioactive properties. This can be achieved by the incorporation of biomolecules (e.g., proteins, peptides) or trophic factors that should preferably be aligned and/or released with controlled kinetics to activate the process of positive axon chemotaxis. In this study, sodium L-lactate has been used to enhance the bioactive properties of chitosan-hydroxyapatite/polycaprolactone electrodeposits. Next, two methods have been developed to incorporate NGF-loaded microspheres - Method 1 involves entrapment and co-deposition of NGF-loaded microspheres, while Method 2 is based on absorption of NGF-loaded microspheres. The study shows that modification of chitosan-hydroxyapatite/polycaprolactone conduits by sodium L-lactate significantly improves their bioactive, biological, and physicochemical properties. The obtained implants are cytocompatible, enhancing the neurite regeneration process by stimulating its elongation. The absorption of NGF-loaded microspheres into the conduit structure may be considered more favorable for the stimulation of axonal elongation compared to entrapment, as it allows for trophic factor dose-dependent controlled release. The developed conduits possess properties essential for the successful treatment of peripheral nerve discontinuities.
Collapse
Affiliation(s)
- Katarzyna Nawrotek
- Lodz University of Technology, Faculty of Process and Environmental Engineering, Department of Environmental Engineering, Wolczanska 213, 93-005 Lodz, Poland; Lodz University of Technology, International Centre for Research on Innovative Bio-based Materials, 2/22 Stefanowskiego, 90-537, Poland; Warsaw University of Technology, Centre for Advanced Materials and Technology (CEZAMAT), 19 Poleczki, 02-822 Warsaw, Poland.
| | - Maciej Chyb
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences Banacha 12/16, 90-237 Lodz, Poland.
| | - Justyna Gatkowska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland.
| | - Karolina Rudnicka
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Immunology and Infectious Biology, 90-237 Lodz, Poland.
| | - Sylwia Michlewska
- University of Lodz, Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, Banacha 12/16, 90-237 Lodz, Poland.
| | - Piotr Jóźwiak
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Invertebrates Zoology and Hydrobiology, Banacha 12/16, 90-324 Lodz, Poland.
| |
Collapse
|
2
|
Qiu Z, Minegishi T, Aoki D, Abe K, Baba K, Inagaki N. Adhesion-clutch between DCC and netrin-1 mediates netrin-1-induced axonal haptotaxis. Front Mol Neurosci 2024; 17:1307755. [PMID: 38375502 PMCID: PMC10875621 DOI: 10.3389/fnmol.2024.1307755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/08/2024] [Indexed: 02/21/2024] Open
Abstract
The growth cone, a motile structure located at the tip of growing axons, senses extracellular guidance cues and translates them into directional forces that drive axon outgrowth and guidance. Axon guidance directed by chemical cues on the extracellular adhesive substrate is termed haptotaxis. Recent studies reported that netrin-1 on the substrate functions as a haptotactic axon guidance cue. However, the mechanism mediating netrin-1-induced axonal haptotaxis remains unclear. Here, we demonstrate that substrate-bound netrin-1 induces axonal haptotaxis by facilitating physical interactions between the netrin-1 receptor, DCC, and the adhesive substrates. DCC serves as an adhesion receptor for netrin-1. The clutch-linker molecule shootin1a interacted with DCC, linking it to actin filament retrograde flow at the growth cone. Speckle imaging analyses showed that DCC underwent either grip (stop) or retrograde slip on the adhesive substrate. The grip state was more prevalent on netrin-1-coated substrate compared to the control substrate polylysine, thereby transmitting larger traction force on the netrin-1-coated substrate. Furthermore, disruption of the linkage between actin filament retrograde flow and DCC by shootin1 knockout impaired netrin-1-induced axonal haptotaxis. These results suggest that the directional force for netrin-1-induced haptotaxis is exerted on the substrates through the adhesion-clutch between DCC and netrin-1 which occurs asymmetrically within the growth cone.
Collapse
Affiliation(s)
| | | | | | | | | | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Nara, Japan
| |
Collapse
|
3
|
Sinclair P, Kabbani N. Ionotropic and metabotropic responses by alpha 7 nicotinic acetylcholine receptors. Pharmacol Res 2023; 197:106975. [PMID: 38032294 DOI: 10.1016/j.phrs.2023.106975] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) belong to a superfamily of cys-loop receptors characterized by the assembly of five subunits into a multi-protein channel complex. Ligand binding to nAChRs activates rapid allosteric transitions of the receptor leading to channel opening and ion flux in neuronal and non-neuronal cell. Thus, while ionotropic properties of nAChRs are well recognized, less is known about ligand-mediated intracellular metabotropic signaling responses. Studies in neural and non-neural cells confirm ionotropic and metabotropic channel responses following ligand binding. In this review we summarize evidence on the existence of ionotropic and metabotropic signaling responses by homopentameric α7 nAChRs in various cell types. We explore how coordinated calcium entry through the ion channel and calcium release from nearby stores gives rise to signaling important for the modulation of cytoskeletal motility and cell growth. Amino acid residues for intracellular protein binding within the α7 nAChR support engagement in metabotropic responses including signaling through heterotrimeric G proteins in neural and immune cells. Understanding the dual properties of ionotropic and metabotropic nAChR responses is essential in advancing drug development for the treatment of various human disease.
Collapse
Affiliation(s)
| | - Nadine Kabbani
- Interdisciplinary Program in Neuroscience, Fairfax, VA, USA; School of Systems Biology, George Mason University, Fairfax, VA, USA.
| |
Collapse
|
4
|
Yang X, Tohda C. Diosgenin restores memory function via SPARC-driven axonal growth from the hippocampus to the PFC in Alzheimer's disease model mice. Mol Psychiatry 2023; 28:2398-2411. [PMID: 37085711 PMCID: PMC10611574 DOI: 10.1038/s41380-023-02052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023]
Abstract
Central nervous system axons have minimal capacity to regenerate in adult brains, hindering memory recovery in Alzheimer's disease (AD). Although recent studies have shown that damaged axons sprouted in adult and AD mouse brains, long-distance axonal re-innervation to their targets has not been achieved. We selectively visualized axon-growing neurons in the neural circuit for memory formation, from the hippocampus to the prefrontal cortex, and showed that damaged axons successfully extended to their native projecting area in mouse models of AD (5XFAD) by administration of an axonal regenerative agent, diosgenin. In vivo transcriptome analysis detected the expression profile of axon-growing neurons directly isolated from the hippocampus of 5XFAD mice. Secreted protein acidic and rich in cysteine (SPARC) was the most expressed gene in axon-growing neurons. Neuron-specific overexpression of SPARC via adeno-associated virus serotype 9 delivery in the hippocampus recovered memory deficits and axonal projection to the prefrontal cortex in 5XFAD mice. DREADDs (Designer receptors exclusively activated by designer drugs) analyses revealed that SPARC overexpression-induced axonal growth in the 5XFAD mouse brain directly contributes to memory recovery. Elevated levels of SPARC on axonal membranes interact with extracellular rail-like collagen type I to promote axonal remodeling along their original tracings in primary cultured hippocampal neurons. These findings suggest that SPARC-driven axonal growth in the brain may be a promising therapeutic strategy for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ximeng Yang
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
5
|
Wit CB, Hiesinger PR. Neuronal filopodia: From stochastic dynamics to robustness of brain morphogenesis. Semin Cell Dev Biol 2023; 133:10-19. [PMID: 35397971 DOI: 10.1016/j.semcdb.2022.03.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 12/30/2022]
Abstract
Brain development relies on dynamic morphogenesis and interactions of neurons. Filopodia are thin and highly dynamic membrane protrusions that are critically required for neuronal development and neuronal interactions with the environment. Filopodial interactions are typically characterized by non-deterministic dynamics, yet their involvement in developmental processes leads to stereotypic and robust outcomes. Here, we discuss recent advances in our understanding of how filopodial dynamics contribute to neuronal differentiation, migration, axonal and dendritic growth and synapse formation. Many of these advances are brought about by improved methods of live observation in intact developing brains. Recent findings integrate known and novel roles ranging from exploratory sensors and decision-making agents to pools for selection and mechanical functions. Different types of filopodial dynamics thereby reveal non-deterministic subcellular decision-making processes as part of genetically encoded brain development.
Collapse
Affiliation(s)
- Charlotte B Wit
- Devision of Neurobiology, Institute of Biology, Freie Universität Berlin, Berlin, Germany
| | - P Robin Hiesinger
- Devision of Neurobiology, Institute of Biology, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Duan G, Li C, Yan X, Yang S, Wang S, Sun X, Zhao L, Song T, Pan Y, Wang X. Construction of a mineralized collagen nerve conduit for peripheral nerve injury repair. Regen Biomater 2022; 10:rbac089. [PMID: 36683739 PMCID: PMC9847629 DOI: 10.1093/rb/rbac089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/12/2022] [Accepted: 10/26/2022] [Indexed: 01/19/2023] Open
Abstract
A new nerve guidance conduits (NGCs) named MC@Col containing Type I collagen (Col) and mineralized collagen (MC) was developed, enhancing mechanical and degradation behavior. The physicochemical properties, the mechanical properties and in vitro degradation behavior were all evaluated. The adhesion and proliferation of Schwann cells (SCs) were observed. In the in vivo experiment, MC@Col NGC and other conduits including Col, chitosan (CST) and polycaprolactone (PCL) conduit were implanted to repair a 10-mm-long Sprague-Dawley rat's sciatic nerve defect. Histological analyses, morphological analyses, electrophysiological analyses and further gait analyses were all evaluated after implantation in 12 weeks. The strength and degradation performance of the MC@Col NGC were improved by the addition of MC in comparison with pure Col NGC. In vitro cytocompatibility evaluation revealed that the SCs had good viability, attachment and proliferation in the MC@Col. In in vivo results, the regenerative outcomes of MC@Col NGC were close to those by an autologous nerve graft in some respects, but superior to those by Col, CST and PCL conduits. The MC@Col NGC exhibited good mechanical performance as well as biocompatibility to bridge nerve gap and guide nerve regeneration, thus showing great promising potential as a new type of conduit in clinical applications.
Collapse
Affiliation(s)
- Guman Duan
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China,Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Chengli Li
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China,Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xiaoqing Yan
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China,Department of Orthopedics, Beijing Changping District Hospital, Beijing 102202, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Shuo Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Tianxi Song
- Beijing Allgens Medical Science and Technology Co., Ltd, Beijing 100176, China
| | - Yongwei Pan
- Correspondence address. Tel: 86-10-62782966, E-mail: (X.W.); (Y.P.)
| | - Xiumei Wang
- Correspondence address. Tel: 86-10-62782966, E-mail: (X.W.); (Y.P.)
| |
Collapse
|
7
|
Mohanan AG, Gunasekaran S, Jacob RS, Omkumar RV. Role of Ca2+/Calmodulin-Dependent Protein Kinase Type II in Mediating Function and Dysfunction at Glutamatergic Synapses. Front Mol Neurosci 2022; 15:855752. [PMID: 35795689 PMCID: PMC9252440 DOI: 10.3389/fnmol.2022.855752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/21/2022] [Indexed: 01/25/2023] Open
Abstract
Glutamatergic synapses harbor abundant amounts of the multifunctional Ca2+/calmodulin-dependent protein kinase type II (CaMKII). Both in the postsynaptic density as well as in the cytosolic compartment of postsynaptic terminals, CaMKII plays major roles. In addition to its Ca2+-stimulated kinase activity, it can also bind to a variety of membrane proteins at the synapse and thus exert spatially restricted activity. The abundance of CaMKII in glutamatergic synapse is akin to scaffolding proteins although its prominent function still appears to be that of a kinase. The multimeric structure of CaMKII also confers several functional capabilities on the enzyme. The versatility of the enzyme has prompted hypotheses proposing several roles for the enzyme such as Ca2+ signal transduction, memory molecule function and scaffolding. The article will review the multiple roles played by CaMKII in glutamatergic synapses and how they are affected in disease conditions.
Collapse
Affiliation(s)
- Archana G. Mohanan
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sowmya Gunasekaran
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - Reena Sarah Jacob
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - R. V. Omkumar
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- *Correspondence: R. V. Omkumar,
| |
Collapse
|
8
|
Abstract
The establishment of a functioning neuronal network is a crucial step in neural development. During this process, neurons extend neurites-axons and dendrites-to meet other neurons and interconnect. Therefore, these neurites need to migrate, grow, branch and find the correct path to their target by processing sensory cues from their environment. These processes rely on many coupled biophysical effects including elasticity, viscosity, growth, active forces, chemical signaling, adhesion and cellular transport. Mathematical models offer a direct way to test hypotheses and understand the underlying mechanisms responsible for neuron development. Here, we critically review the main models of neurite growth and morphogenesis from a mathematical viewpoint. We present different models for growth, guidance and morphogenesis, with a particular emphasis on mechanics and mechanisms, and on simple mathematical models that can be partially treated analytically.
Collapse
Affiliation(s)
- Hadrien Oliveri
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK.
| |
Collapse
|
9
|
Nakagawa-Tamagawa N, Kirino E, Sugao K, Nagata H, Tagawa Y. Involvement of Calcium-Dependent Pathway and β Subunit-Interaction in Neuronal Migration and Callosal Projection Deficits Caused by the Cav1.2 I1166T Mutation in Developing Mouse Neocortex. Front Neurosci 2021; 15:747951. [PMID: 34955712 PMCID: PMC8692569 DOI: 10.3389/fnins.2021.747951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Gain-of-function mutations in the L-type Ca2+ channel Cav1.2 cause Timothy syndrome (TS), a multisystem disorder associated with neurologic symptoms, including autism spectrum disorder (ASD), seizures, and intellectual disability. Cav1.2 plays key roles in neural development, and its mutation can affect brain development and connectivity through Ca2+-dependent and -independent mechanisms. Recently, a gain-of-function mutation, I1166T, in Cav1.2 was identified in patients with TS-like disorder. Its channel properties have been analyzed in vitro but in vivo effects of this mutation on brain development remain unexplored. Methods:In utero electroporation was performed on ICR mice at embryonic day 15 to express GFP, wild-type, and mutant Cav1.2 channels into cortical layer 2/3 excitatory neurons in the primary somatosensory area. The brain was fixed at postnatal days 14–16, sliced, and scanned using confocal microscopy. Neuronal migration of electroporated neurons was examined in the cortex of the electroporated hemisphere, and callosal projection was examined in the white matter and contralateral hemisphere. Results: Expression of the I1166T mutant in layer 2/3 neurons caused migration deficits in approximately 20% of electroporated neurons and almost completely diminished axonal arborization in the contralateral hemisphere. Axonal projection in the white matter was not affected. We introduced second mutations onto Cav1.2 I1166T; L745P mutation blocks Ca2+ influx through Cav1.2 channels and inhibits the Ca2+-dependent pathway, and the W440A mutation blocks the interaction of the Cav1.2 α1 subunit to the β subunit. Both second mutations recovered migration and projection. Conclusion: This study demonstrated that the Cav1.2 I1166T mutation could affect two critical steps during cerebrocortical development, migration and axonal projection, in the mouse brain. This is mediated through Ca2+-dependent pathway downstream of Cav1.2 and β subunit-interaction.
Collapse
Affiliation(s)
- Nao Nakagawa-Tamagawa
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.,Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Saitama, Japan
| | - Emi Kirino
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kohtaroh Sugao
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Hidetaka Nagata
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Yoshiaki Tagawa
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
10
|
Carmona A, Chen S, Domart F, Choquet D, Ortega R. Imaging the structural organization of chemical elements in growth cones of developing hippocampal neurons. Metallomics 2021; 14:6462920. [PMID: 34910190 DOI: 10.1093/mtomcs/mfab073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/06/2021] [Indexed: 11/14/2022]
Abstract
During neurodevelopment, neurons form growth cones, F-actin rich extensions located at the distal end of the neurites. Growth cones allow dendrites and axons to build synaptic connections through a process of neurite guidance whose mechanisms have not been fully elucidated. Calcium is an important element in this process by inducing F-actin reorganization. We hypothesized that other biologically active elements might be involved in the growth cone-mediated neurite guidance mechanisms. We performed super resolution and confocal microscopy of F-actin, followed by synchrotron X-ray fluorescence microscopy of phosphorous, sulfur, chlorine, potassium, calcium, iron and zinc on growth cones from primary rat hippocampal neurons. We identified two main patterns of element organization. First, active growth cones presenting an asymmetric distribution of Ca co-localized with the cytoskeleton protein F-actin. In active growth cones, we found that the distributions of P, S, Cl, K and Zn are correlated with Ca. This correlation is lost in the second pattern, quiescent growth cones, exhibiting a spread elemental distribution. These results suggest that Ca is not the only element required in the F-actin rich active regions of growth cones. In addition, highly concentrated Fe spots of sub-micrometer size were observed in calcium-rich areas of active growth cones. These results reveal the need for biological active elements in growth cones during neural development and may help explain why early life deficiencies of elements, such as Fe or Zn, induce learning and memory deficits in children.
Collapse
Affiliation(s)
| | - Si Chen
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Florelle Domart
- Univ. Bordeaux, CNRS, CENBG, UMR 5797, 33170 Gradignan, France.,Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Daniel Choquet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France.,Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, 33000 Bordeaux, France
| | - Richard Ortega
- Univ. Bordeaux, CNRS, CENBG, UMR 5797, 33170 Gradignan, France
| |
Collapse
|
11
|
Abstract
Calcium (Ca2+) is a unique mineral that serves as both a nutrient and a signal in all eukaryotes. To maintain Ca2+ homeostasis for both nutrition and signaling purposes, the toolkit for Ca2+ transport has expanded across kingdoms of eukaryotes to encode specific Ca2+ signals referred to as Ca2+ signatures. In parallel, a large array of Ca2+-binding proteins has evolved as specific sensors to decode Ca2+ signatures. By comparing these coding and decoding mechanisms in fungi, animals, and plants, both unified and divergent themes have emerged, and the underlying complexity will challenge researchers for years to come. Considering the scale and breadth of the subject, instead of a literature survey, in this review we focus on a conceptual framework that aims to introduce to readers to the principles and mechanisms of Ca2+ signaling. We finish with several examples of Ca2+-signaling pathways, including polarized cell growth, immunity and symbiosis, and systemic signaling, to piece together specific coding and decoding mechanisms in plants versus animals. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sheng Luan
- Department of Plant and Microbial Biology, University of California, Berkeley, California 94720, USA;
| | - Chao Wang
- Department of Plant and Microbial Biology, University of California, Berkeley, California 94720, USA;
| |
Collapse
|
12
|
Kapr J, Petersilie L, Distler T, Lauria I, Bendt F, Sauter CM, Boccaccini AR, Rose CR, Fritsche E. Human Induced Pluripotent Stem Cell-Derived Neural Progenitor Cells Produce Distinct Neural 3D In Vitro Models Depending on Alginate/Gellan Gum/Laminin Hydrogel Blend Properties. Adv Healthc Mater 2021; 10:e2100131. [PMID: 34197049 PMCID: PMC11468953 DOI: 10.1002/adhm.202100131] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/01/2021] [Indexed: 12/12/2022]
Abstract
Stable and predictive neural cell culture models are a necessary premise for many research fields. However, conventional 2D models lack 3D cell-material/-cell interactions and hence do not reflect the complexity of the in vivo situation properly. Here two alginate/gellan gum/laminin (ALG/GG/LAM) hydrogel blends are presented for the fabrication of human induced pluripotent stem cell (hiPSC)-based 3D neural models. For hydrogel embedding, hiPSC-derived neural progenitor cells (hiNPCs) are used either directly or after 3D neural pre-differentiation. It is shown that stiffness and stress relaxation of the gel blends, as well as the cell differentiation strategy influence 3D model development. The embedded hiNPCs differentiate into neurons and astrocytes within the gel blends and display spontaneous intracellular calcium signals. Two fit-for-purpose models valuable for i) applications requiring a high degree of complexity, but less throughput, such as disease modeling and long-term exposure studies and ii) higher throughput applications, such as acute exposures or substance screenings are proposed. Due to their wide range of applications, adjustability, and printing capabilities, the ALG/GG/LAM based 3D neural models are of great potential for 3D neural modeling in the future.
Collapse
Affiliation(s)
- Julia Kapr
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Laura Petersilie
- Institute of NeurobiologyHeinrich Heine UniversityDüsseldorf40225Germany
| | - Thomas Distler
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐University Erlangen‐NurembergErlangen91054Germany
| | - Ines Lauria
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Farina Bendt
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Clemens M. Sauter
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Aldo R. Boccaccini
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐University Erlangen‐NurembergErlangen91054Germany
| | - Christine R. Rose
- Institute of NeurobiologyHeinrich Heine UniversityDüsseldorf40225Germany
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
- Medical FacultyHeinrich Heine UniversityDüsseldorf40225Germany
| |
Collapse
|
13
|
Li H, Lou R, Xu X, Xu C, Yu Y, Xu Y, Hu L, Xiang Y, Lin X, Tang S. The variations in human orphan G protein-coupled receptor QRFPR affect PI3K-AKT-mTOR signaling. J Clin Lab Anal 2021; 35:e23822. [PMID: 34018631 DOI: 10.1002/jcla.23822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND QRFPR is a recently identified member of the G protein-coupled receptor and is an orphan receptor for 26Rfa, which plays important role in the regulation of many physiological functions. METHODS Here, we employed whole exome sequencing (WES) to examine the patients with intellectual disability (ID) and difficulty in feeding. We performed SIFT and PolyPhen2 predictions for the variants. The structure model was built from scratch by I-TASSER. Here, results derived from a number of cell-based functional assays, including shRNA experiment, intracellular Ca2+ measurement, the expression of PI3 K-AKT-mTOR, and phosphorylation. The functional effect of QRFPR variants on PI3K-AKT-mTOR signaling was evaluated in vitro transfection experiments. RESULT Here, we identified two QRFPR variants at c.202 T>C (p.Y68H) and c.1111C>T (p.R371W) in 2 unrelated individuals. Structural analysis revealed that p.Y68H and p.R371W variants may affect the side chain structure of adjacent amino acids causing reduced binding of QRFPR to 26Rfa. The results show that QRFPR stimulated by 26Rfa leading to the transient rise of intracellular Ca2+ . The QRFPR variations p.Y68H and p.R371 W can reduce the mobilization of intracellular Ca2+ . The phosphorylation levels of the PI3K, Akt, and mTOR were significantly up- or downregulated by QRFPR overexpression or silencing, respectively. The QRFPR variations inhibited PI3K-AKT-mTOR signaling, resulting in downregulation of p-mTOR. CONCLUSIONS Our findings suggest that QRFPR acts as important role in neurodevelopment, and the effects of QRFPR are likely to be mediated by the Ca2+ -dependent PI3K-AKT-mTOR pathways. Importantly, these findings provide a foundation for future elucidation of GPCR-mediated signaling and the physiological implications.
Collapse
Affiliation(s)
- Huanzheng Li
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China.,Human Aging Research Institute, Nanchang University, Nanchang, China
| | - Ran Lou
- Department of Acupuncture, Wenzhou Central Hospital, Wenzhou, China
| | - Xueqin Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Chenyang Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yuan Yu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yunzhi Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Lin Hu
- Department of Blood Transfusion, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanbao Xiang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xuan Lin
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Shaohua Tang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Kaltezioti V, Foskolou IP, Lavigne MD, Ninou E, Tsampoula M, Fousteri M, Margarity M, Politis PK. Prox1 inhibits neurite outgrowth during central nervous system development. Cell Mol Life Sci 2021; 78:3443-3465. [PMID: 33247761 PMCID: PMC11072475 DOI: 10.1007/s00018-020-03709-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022]
Abstract
During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.
Collapse
Affiliation(s)
- Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Iosifina P Foskolou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matthieu D Lavigne
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Elpinickie Ninou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Maria Fousteri
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Marigoula Margarity
- Laboratory of Human and Animal Physiology, Department of Biology, School of Natural Sciences, University of Patras, 26500, Rio Achaias, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece.
| |
Collapse
|
15
|
Sun L, Wei H. Ryanodine Receptors: A Potential Treatment Target in Various Neurodegenerative Disease. Cell Mol Neurobiol 2020; 41:1613-1624. [PMID: 32833122 DOI: 10.1007/s10571-020-00936-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023]
Abstract
Progressive neuronal demise is a key contributor to the key pathogenic event implicated in many different neurodegenerative disorders (NDDs). There are several therapeutic strategies available; however, none of them are particularly effective. Targeted neuroprotective therapy is one such therapy, which seems a compelling option, yet remains challenging due to the internal heterogeneity of the mechanisms underlying various NDDs. An alternative method to treat NDDs is to exploit common modalities involving molecularly distinct subtypes and thus develop specialized drugs with broad-spectrum characteristics. There is mounting evidence which supports for the theory that dysfunctional ryanodine receptors (RyRs) disrupt intracellular Ca2+ homeostasis, contributing to NDDs significantly. This review aims to provide direct and indirect evidence on the intersection of NDDs and RyRs malfunction, and to shed light on novel strategies to treat RyRs-mediated disease, modifying pharmacological therapies such as the potential therapeutic role of dantrolene, a RyRs antagonist.
Collapse
Affiliation(s)
- Liang Sun
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA
- Department of Anesthesiology, Peking University People's Hospital, Beijing, 100044, China
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
16
|
Urbina FL, Gupton SL. SNARE-Mediated Exocytosis in Neuronal Development. Front Mol Neurosci 2020; 13:133. [PMID: 32848598 PMCID: PMC7427632 DOI: 10.3389/fnmol.2020.00133] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
The formation of the nervous system involves establishing complex networks of synaptic connections between proper partners. This developmental undertaking requires the rapid expansion of the plasma membrane surface area as neurons grow and polarize, extending axons through the extracellular environment. Critical to the expansion of the plasma membrane and addition of plasma membrane material is exocytic vesicle fusion, a regulated mechanism driven by soluble N-ethylmaleimide-sensitive factor attachment proteins receptors (SNAREs). Since their discovery, SNAREs have been implicated in several critical neuronal functions involving exocytic fusion in addition to synaptic transmission, including neurite initiation and outgrowth, axon specification, axon extension, and synaptogenesis. Decades of research have uncovered a rich variety of SNARE expression and function. The basis of SNARE-mediated fusion, the opening of a fusion pore, remains an enigmatic event, despite an incredible amount of research, as fusion is not only heterogeneous but also spatially small and temporally fast. Multiple modes of exocytosis have been proposed, with full-vesicle fusion (FFV) and kiss-and-run (KNR) being the best described. Whereas most in vitro work has reconstituted fusion using VAMP-2, SNAP-25, and syntaxin-1; there is much to learn regarding the behaviors of distinct SNARE complexes. In the past few years, robust heterogeneity in the kinetics and fate of the fusion pore that varies by cell type have been uncovered, suggesting a paradigm shift in how the modes of exocytosis are viewed is warranted. Here, we explore both classic and recent work uncovering the variety of SNAREs and their importance in the development of neurons, as well as historical and newly proposed modes of exocytosis, their regulation, and proteins involved in the regulation of fusion kinetics.
Collapse
Affiliation(s)
- Fabio L. Urbina
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- UNC Neuroscience Center, Chapel Hill, NC, United States
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, United States
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
17
|
Ardhani R, Ana ID, Tabata Y. Gelatin hydrogel membrane containing carbonate hydroxyapatite for nerve regeneration scaffold. J Biomed Mater Res A 2020; 108:2491-2503. [PMID: 32418269 DOI: 10.1002/jbm.a.37000] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/11/2020] [Accepted: 04/19/2020] [Indexed: 12/26/2022]
Abstract
A scaffold that mimics physicochemical structure of nerve and supplies calcium ions in axonal environment is an attractive alternative for nerve regeneration, especially when applied in critical nerve defect. Various scaffold material, design, including their combination with several growth-induced substances and cells application have been being investigated and used in the area of nerve tissue engineering. However, the development remains challenges today because they are still far from ideal concerning their stability, reproducibility, including complicated handling related to the poor mechanical strength. In view of the current basis, in this study, the introduction of carbonated hydroxyapatite (CHA) as promising candidate to increase mechanical properties of nerve scaffold is reported. The incorporation of CHA was not only expected to provide better mechanical properties of the scaffold. Under physiological condition, CHA is known to be the most stable phases of calcium phosphate compound. Therefore, CHA was expected to provide controlled release calcium for better axonal environment and promote fasten nerve regeneration. This study shows that CHA incorporated gelatin membrane has ideal microstructure to prevent fibrous tissue ingrowth into the injury site, while retaining its capability to survive nerve tissue by allowing adequate glucose and specific proteins diffusion. The provided Ca2+ release to the environment promoted neuronal growth, without suppressing acetylcholine esterase release activity. Neurite elongation was dramatically higher in the gelatin membrane incorporated with CHA. Introduction of CHA into gelatin membrane represents a new generation medical device for nerve reconstruction, with CHA was considered as a promising factor.
Collapse
Affiliation(s)
- Retno Ardhani
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
18
|
Syntaphilin-Mediated Docking of Mitochondria at the Growth Cone Is Dispensable for Axon Elongation In Vivo. eNeuro 2019; 6:ENEURO.0026-19.2019. [PMID: 31481398 PMCID: PMC6751374 DOI: 10.1523/eneuro.0026-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 08/15/2019] [Accepted: 08/22/2019] [Indexed: 11/25/2022] Open
Abstract
Mitochondria are abundantly detected at the growth cone, the dynamic distal tip of developing axons that directs growth and guidance. It is, however, poorly understood how mitochondrial dynamics relate to growth cone behavior in vivo, and which mechanisms are responsible for anchoring mitochondria at the growth cone during axon pathfinding. Here, we show that in retinal axons elongating along the optic tract in zebrafish, mitochondria accumulate in the central area of the growth cone and are occasionally observed in filopodia extending from the growth cone periphery. Mitochondrial behavior at the growth cone in vivo is dynamic, with mitochondrial positioning and anterograde transport strongly correlating with growth cone behavior and axon outgrowth. Using novel zebrafish mutant lines that lack the mitochondrial anchoring proteins Syntaphilin a and b, we further show that Syntaphilins contribute to mitochondrial immobilization at the growth cone. Syntaphilins are, however, not required for proper growth cone morphology and axon growth in vivo, indicating that Syntaphilin-mediated anchoring of mitochondria at the growth cone plays only a minor role in elongating axons.
Collapse
|
19
|
Johnson A, Nasser TIN, Spencer GE. Inhibition of Rho GTPases in Invertebrate Growth Cones Induces a Switch in Responsiveness to Retinoic Acid. Biomolecules 2019; 9:biom9090460. [PMID: 31500289 PMCID: PMC6769630 DOI: 10.3390/biom9090460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
During development, growth cones are essential for axon pathfinding by sensing numerous guidance cues in their environment. Retinoic acid, the metabolite of vitamin A, is important for neurite outgrowth during vertebrate development, but may also play a role in axon guidance, though little is known of the cellular mechanisms involved. Our previous studies showed that retinoid-induced growth cone turning of invertebrate motorneurons requires local protein synthesis and calcium influx. However, the signalling pathways that link calcium influx to cytoskeletal dynamics involved in retinoid-mediated growth cone turning are not currently known. The Rho GTPases, Cdc42 and Rac, are known regulators of the growth cone cytoskeleton. Here, we demonstrated that inhibition of Cdc42 or Rac not only prevented growth cone turning toward retinoic acid but could also induce a switch in growth cone responsiveness to chemorepulsion or growth cone collapse. However, the effects of Cdc42 or Rac inhibition on growth cone responsiveness differed, depending on whether the turning was induced by the all-trans or 9-cis retinoid isomer. The effects also differed depending on whether the growth cones maintained communication with the cell body. These data strongly suggest that Cdc42 and Rac are downstream effectors of retinoic acid during growth cone guidance.
Collapse
Affiliation(s)
- Alysha Johnson
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St Catharines, ON L2S 3A1, Canada
| | - Tamara I N Nasser
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St Catharines, ON L2S 3A1, Canada
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
20
|
Formicola N, Vijayakumar J, Besse F. Neuronal ribonucleoprotein granules: Dynamic sensors of localized signals. Traffic 2019; 20:639-649. [PMID: 31206920 DOI: 10.1111/tra.12672] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
Membrane-less organelles, because of their capacity to dynamically, selectively and reversibly concentrate molecules, are very well adapted for local information processing and rapid response to environmental fluctuations. These features are particularly important in the context of neuronal cells, where synapse-specific activation, or localized extracellular cues, induce signaling events restricted to specialized axonal or dendritic subcompartments. Neuronal ribonucleoprotein (RNP) particles, or granules, are nonmembrane bound macromolecular condensates that concentrate specific sets of mRNAs and regulatory proteins, promoting their long-distance transport to axons or dendrites. Neuronal RNP granules also have a dual function in regulating the translation of associated mRNAs: while preventing mRNA translation at rest, they fuel local protein synthesis upon activation. As revealed by recent work, rapid and reversible switches between these two functional modes are triggered by modifications of the networks of interactions underlying RNP granule assembly. Such flexible properties also come with a cost, as neuronal RNP granules are prone to transition into pathological aggregates in response to mutations, aging, or cellular stresses, further emphasizing the need to better understand the mechanistic principles governing their dynamic assembly and regulation in living systems.
Collapse
|
21
|
Reyer H, Oster M, Wittenburg D, Murani E, Ponsuksili S, Wimmers K. Genetic Contribution to Variation in Blood Calcium, Phosphorus, and Alkaline Phosphatase Activity in Pigs. Front Genet 2019; 10:590. [PMID: 31316547 PMCID: PMC6610066 DOI: 10.3389/fgene.2019.00590] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 06/04/2019] [Indexed: 12/18/2022] Open
Abstract
Blood values of calcium (Ca), inorganic phosphorus (IP), and alkaline phosphatase activity (ALP) are valuable indicators for mineral status and bone mineralization. The mineral homeostasis is maintained by absorption, retention, and excretion processes employing a number of known and unknown sensing and regulating factors with implications on immunity. Due to the high inter-individual variation of Ca and P levels in the blood of pigs and to clarify molecular contributions to this variation, the genetics of hematological traits related to the Ca and P balance were investigated in a German Landrace population, integrating both single-locus and multi-locus genome-wide association study (GWAS) approaches. Genomic heritability estimates suggest a moderate genetic contribution to the variation of hematological Ca (N = 456), IP (N = 1049), ALP (N = 439), and the Ca/P ratio (N = 455), with values ranging from 0.27 to 0.54. The genome-wide analysis of markers adds a number of genomic regions to the list of quantitative trait loci, some of which overlap with previous results. Despite the gaps in knowledge of genes involved in Ca and P metabolism, genes like THBS2, SHH, PTPRT, PTGS1, and FRAS1 with reported connections to bone metabolism were derived from the significantly associated genomic regions. Additionally, genomic regions included TRAFD1 and genes coding for phosphate transporters (SLC17A1-SLC17A4), which are linked to Ca and P homeostasis. The study calls for improved functional annotation of the proposed candidate genes to derive features involved in maintaining Ca and P balance. This gene information can be exploited to diagnose and predict characteristics of micronutrient utilization, bone development, and a well-functioning musculoskeletal system in pig husbandry and breeding.
Collapse
Affiliation(s)
- Henry Reyer
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Michael Oster
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Dörte Wittenburg
- Biomathematics and Bioinformatics Unit, Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Eduard Murani
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Functional Genome Analysis Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Klaus Wimmers
- Genomics Unit, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany.,Department of Animal Breeding and Genetics, Faculty of Agricultural and Environmental Sciences, University of Rostock, Rostock, Germany
| |
Collapse
|
22
|
Padmanabhan P, Goodhill GJ. Axon growth regulation by a bistable molecular switch. Proc Biol Sci 2019; 285:rspb.2017.2618. [PMID: 29669897 DOI: 10.1098/rspb.2017.2618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
For the brain to function properly, its neurons must make the right connections during neural development. A key aspect of this process is the tight regulation of axon growth as axons navigate towards their targets. Neuronal growth cones at the tips of developing axons switch between growth and paused states during axonal pathfinding, and this switching behaviour determines the heterogeneous axon growth rates observed during brain development. The mechanisms controlling this switching behaviour, however, remain largely unknown. Here, using mathematical modelling, we predict that the molecular interaction network involved in axon growth can exhibit bistability, with one state representing a fast-growing growth cone state and the other a paused growth cone state. Owing to stochastic effects, even in an unchanging environment, model growth cones reversibly switch between growth and paused states. Our model further predicts that environmental signals could regulate axon growth rate by controlling the rates of switching between the two states. Our study presents a new conceptual understanding of growth cone switching behaviour, and suggests that axon guidance may be controlled by both cell-extrinsic factors and cell-intrinsic growth regulatory mechanisms.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Geoffrey J Goodhill
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia .,School of Mathematics and Physics, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| |
Collapse
|
23
|
Baranes K, Hibsh D, Cohen S, Yamin T, Efroni S, Sharoni A, Shefi O. Comparing Transcriptome Profiles of Neurons Interfacing Adjacent Cells and Nanopatterned Substrates Reveals Fundamental Neuronal Interactions. NANO LETTERS 2019; 19:1451-1459. [PMID: 30704243 DOI: 10.1021/acs.nanolett.8b03879] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Developing neuronal axons are directed by chemical and physical signals toward a myriad of target cells. According to current dogma, the resulting network architecture is critically shaped by electrical interconnections, the synapses; however, key mechanisms translating neuronal interactions into neuronal growth behavior during network formation are still unresolved. To elucidate these mechanisms, we examined neurons interfacing nanopatterned substrates and compared them to natural interneuron interactions. We grew similar neuronal populations under three connectivity conditions, (1) the neurons are isolated, (2) the neurons are interconnected, and (3) the neurons are connected only to artificial substrates, then quantitatively compared both the cell morphologies and the transcriptome-expression profiles. Our analysis shows that whereas axon-guidance signaling pathways in isolated neurons are predominant, in isolated neurons interfacing nanotopography, these pathways are downregulated, similar to the interconnected neurons. Moreover, in nanotopography, interfacing neuron genes related to synaptogenesis and synaptic regulation are highly expressed, that is, again resembling the behavior of interconnected neurons. These molecular findings demonstrate that interactions with nanotopographies, although not leading to electrical coupling, play a comparable functional role in two major routes, neuronal guidance and network formation, with high relevance to the design of regenerative interfaces.
Collapse
Affiliation(s)
- Koby Baranes
- Faculty of Engineering , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Bar-Ilan Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Dror Hibsh
- Bar-Ilan Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Faculty of Life Sciences , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Sharon Cohen
- Faculty of Engineering , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Bar-Ilan Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Gonda Multidisciplinary Brain Research Center , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Tony Yamin
- Bar-Ilan Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Department of Physics , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Sol Efroni
- Bar-Ilan Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Faculty of Life Sciences , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Amos Sharoni
- Bar-Ilan Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Department of Physics , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| | - Orit Shefi
- Faculty of Engineering , Bar-Ilan University , Ramat-Gan 5290002 , Israel
- Bar-Ilan Institute of Nanotechnology and Advanced Materials , Bar-Ilan University , Ramat-Gan 5290002 , Israel
| |
Collapse
|
24
|
Santoni G, Amantini C. The Transient Receptor Potential Vanilloid Type-2(TRPV2) Ion Channels in Neurogenesis andGliomagenesis: Cross-Talk between TranscriptionFactors and Signaling Molecules. Cancers (Basel) 2019; 11:cancers11030322. [PMID: 30845786 PMCID: PMC6468602 DOI: 10.3390/cancers11030322] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 12/20/2022] Open
Abstract
Recently, the finding of cancer stem cells in brain tumors has increased the possibilities for advancing new therapeutic approaches with the aim to overcome the limits of current available treatments. In addition, a role for ion channels, particularly of TRP channels, in developing neurons as well as in brain cancer development and progression have been demonstrated. Herein, we focus on the latest advancements in understanding the role of TRPV2, a Ca2+ permeable channel belonging to the TRPV subfamily in neurogenesis and gliomagenesis. TRPV2 has been found to be expressed in both neural progenitor cells and glioblastoma stem/progenitor-like cells (GSCs). In developing neurons, post-translational modifications of TRPV2 (e.g., phosphorylation by ERK2) are required to stimulate Ca2+ signaling and nerve growth factor-mediated neurite outgrowth. TRPV2 overexpression also promotes GSC differentiation and reduces gliomagenesis in vitro and in vivo. In glioblastoma, TRPV2 inhibits survival and proliferation, and induces Fas/CD95-dependent apoptosis. Furthermore, by proteomic analysis, the identification of a TRPV2 interactome-based signature and its relation to glioblastoma progression/recurrence, high or low overall survival and drug resistance strongly suggest an important role of the TRPV2 channel as a potential biomarker in glioblastoma prognosis and therapy.
Collapse
Affiliation(s)
- Giorgio Santoni
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy.
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy.
| |
Collapse
|
25
|
Pan Y, Chai X, Gao Q, Zhou L, Zhang S, Li L, Luan S. Dynamic Interactions of Plant CNGC Subunits and Calmodulins Drive Oscillatory Ca 2+ Channel Activities. Dev Cell 2019; 48:710-725.e5. [PMID: 30713075 DOI: 10.1016/j.devcel.2018.12.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/03/2018] [Accepted: 12/28/2018] [Indexed: 12/20/2022]
Abstract
Calcium is a universal signal in all eukaryotes, but the mechanism for encoding calcium signatures remains largely unknown. Calcium oscillations control pollen tube growth and fertilization in flowering plants, serving as a model for dissecting the molecular machines that mediate calcium fluctuations. We report that pollen-tube-specific cyclic nucleotide-gated channels (CNGC18, CNGC8, and CNGC7) together with calmodulin 2 (CaM2) constitute a molecular switch that either opens or closes the calcium channel depending on cellular calcium levels. Under low calcium, calcium-free calmodulin 2 (Apo-CaM2) interacts with CNGC18-CNGC8 complex, leading to activation of the influx channel and consequently increasing cytosolic calcium levels. Calcium-bound CaM2 dissociates from CNGC18/8 heterotetramer, closing the channel and initiating a downturn of cellular calcium levels. We further reconstituted the calcium oscillator in HEK293 cells, supporting the model that Ca2+-CaM-dependent regulation of CNGC channel activity provides an auto-regulatory feedback mechanism for calcium oscillations during pollen tube growth.
Collapse
Affiliation(s)
- Yajun Pan
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Xuyang Chai
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Qifei Gao
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Liming Zhou
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Sisi Zhang
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Legong Li
- College of Life Sciences, Capital Normal University, Beijing 100048, China.
| | - Sheng Luan
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
26
|
Donato A, Kagias K, Zhang Y, Hilliard MA. Neuronal sub-compartmentalization: a strategy to optimize neuronal function. Biol Rev Camb Philos Soc 2019; 94:1023-1037. [PMID: 30609235 PMCID: PMC6617802 DOI: 10.1111/brv.12487] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022]
Abstract
Neurons are highly polarized cells that consist of three main structural and functional domains: a cell body or soma, an axon, and dendrites. These domains contain smaller compartments with essential roles for proper neuronal function, such as the axonal presynaptic boutons and the dendritic postsynaptic spines. The structure and function of these compartments have now been characterized in great detail. Intriguingly, however, in the last decade additional levels of compartmentalization within the axon and the dendrites have been identified, revealing that these structures are much more complex than previously thought. Herein we examine several types of structural and functional sub-compartmentalization found in neurons of both vertebrates and invertebrates. For example, in mammalian neurons the axonal initial segment functions as a sub-compartment to initiate the action potential, to select molecules passing into the axon, and to maintain neuronal polarization. Moreover, work in Drosophila melanogaster has shown that two distinct axonal guidance receptors are precisely clustered in adjacent segments of the commissural axons both in vivo and in vitro, suggesting a cell-intrinsic mechanism underlying the compartmentalized receptor localization. In Caenorhabditis elegans, a subset of interneurons exhibits calcium dynamics that are localized to specific sections of the axon and control the gait of navigation, demonstrating a regulatory role of compartmentalized neuronal activity in behaviour. These findings have led to a number of new questions, which are important for our understanding of neuronal development and function. How are these sub-compartments established and maintained? What molecular machinery and cellular events are involved? What is their functional significance for the neuron? Here, we reflect on these and other key questions that remain to be addressed in this expanding field of biology.
Collapse
Affiliation(s)
- Alessandra Donato
- Clem Jones Centre for Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Konstantinos Kagias
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, U.S.A
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, U.S.A
| | - Massimo A Hilliard
- Clem Jones Centre for Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
27
|
Goodhill GJ. Theoretical Models of Neural Development. iScience 2018; 8:183-199. [PMID: 30321813 PMCID: PMC6197653 DOI: 10.1016/j.isci.2018.09.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/06/2018] [Accepted: 09/19/2018] [Indexed: 12/22/2022] Open
Abstract
Constructing a functioning nervous system requires the precise orchestration of a vast array of mechanical, molecular, and neural-activity-dependent cues. Theoretical models can play a vital role in helping to frame quantitative issues, reveal mathematical commonalities between apparently diverse systems, identify what is and what is not possible in principle, and test the abilities of specific mechanisms to explain the data. This review focuses on the progress that has been made over the last decade in our theoretical understanding of neural development.
Collapse
Affiliation(s)
- Geoffrey J Goodhill
- Queensland Brain Institute and School of Mathematics and Physics, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
28
|
Baba K, Yoshida W, Toriyama M, Shimada T, Manning CF, Saito M, Kohno K, Trimmer JS, Watanabe R, Inagaki N. Gradient-reading and mechano-effector machinery for netrin-1-induced axon guidance. eLife 2018; 7:34593. [PMID: 30082022 PMCID: PMC6080949 DOI: 10.7554/elife.34593] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/05/2018] [Indexed: 12/28/2022] Open
Abstract
Growth cones navigate axonal projection in response to guidance cues. However, it is unclear how they can decide the migratory direction by transducing the local spatial cues into protrusive forces. Here we show that knockout mice of Shootin1 display abnormal projection of the forebrain commissural axons, a phenotype similar to that of the axon guidance molecule netrin-1. Shallow gradients of netrin-1 elicited highly polarized Pak1-mediated phosphorylation of shootin1 within growth cones. We demonstrate that netrin-1–elicited shootin1 phosphorylation increases shootin1 interaction with the cell adhesion molecule L1-CAM; this, in turn, promotes F-actin–adhesion coupling and concomitant generation of forces for growth cone migration. Moreover, the spatially regulated shootin1 phosphorylation within growth cones is required for axon turning induced by netrin-1 gradients. Our study defines a mechano-effector for netrin-1 signaling and demonstrates that shootin1 phosphorylation is a critical readout for netrin-1 gradients that results in a directional mechanoresponse for axon guidance. Neurons communicate with each other by forming intricate webs that link cells together according to a precise pattern. A neuron can connect to another by growing a branch-like structure known as the axon. To contact the correct neuron, the axon must develop and thread its way to exactly the right place in the brain. Scientists know that the tip of the axon is extraordinarily sensitive to gradients of certain molecules in its surroundings, which guide the budding structure towards its final destination. In particular, two molecules seem to play an important part in this process: netrin-1, which is a protein found outside cells that attracts a growing axon, and shootin1a, which is present inside neurons. Previous studies have shown that netrin-1 can trigger a cascade of reactions that activates shootin1a. In turn, activated shootin1a molecules join the internal skeleton of the cell with L1-CAM, a molecule that attaches the neuron to its surroundings. If the internal skeleton is the engine of the axon, L1-CAMs are the wheels, and shootin1a the clutch. However, it is not clear whether shootin1a is involved in guiding growing axons, and how it could help neurons ‘understand’ and react to gradients of netrin-1. Here, Baba et al. discover that when shootin1a is absent in mice, the axons do not develop properly. Further experiments in rat neurons show that if there is a little more netrin-1 on one side of the tip of an axon, this switches on the shootin1a molecules on that edge. Activated shootin1a promote interactions between the internal skeleton and L1-CAM, helping the axon curve towards the area that has more netrin-1. In fact, if the activated shootin1a is present everywhere on the axon, and not just on one side, the structure can develop, but not turn. Taken together, the results suggest that shootin1a can read the gradients of netrin-1 and then coordinate the turning of a growing axon in response. Wound healing, immune responses or formation of organs are just a few examples of processes that rely on cells moving in an orderly manner through the body. Dissecting how axons are guided through their development may shed light on the migration of cells in general. Ultimately, this could help scientists to understand disorders such as birth abnormalities or neurological disabilities, which arise when this process goes awry.
Collapse
Affiliation(s)
- Kentarou Baba
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Wataru Yoshida
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Michinori Toriyama
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Tadayuki Shimada
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Colleen F Manning
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, United States
| | - Michiko Saito
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kenji Kohno
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, United States
| | - Rikiya Watanabe
- Department of Applied Chemistry, Graduate School of Engineering, University of Tokyo, Tokyo, Japan
| | - Naoyuki Inagaki
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
29
|
Zargar Kharazi A, Dini G, Naser R. Fabrication and evaluation of a nerve guidance conduit capable of Ca 2+ ion release to accelerate axon extension in peripheral nerve regeneration. J Biomed Mater Res A 2018; 106:2181-2189. [PMID: 29637737 DOI: 10.1002/jbm.a.36425] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/16/2018] [Accepted: 03/28/2018] [Indexed: 11/11/2022]
Abstract
In this study, biodegradable nanocomposites consisting of poly (glycerol sebacate) (PGS) elastomeric matrix and the reinforcing phase of calcium titanate (CaTiO3 ) nanoparticles were fabricated as a nerve guidance conduit (NGC) for peripheral nerve regeneration. CaTiO3 nanoparticles were synthesized via the sol-gel method and calcined at 800°C for 60 min. PGS elastomer was synthesized via the polycondensation reaction of glycerol and sebacate (1:1) and 2.5 and 5 wt. percentages of the synthesized CaTiO3 nanoparticles were added to the PGS prepolymer solution. The composites obtained were heated in order to make crosslinks in the pre-polymer. CaTiO3 nanoparticles, PGS elastomer, and the composites fabricated were characterized in terms of their structural, chemical, physical, mechanical, and cell response properties to evaluate the feasibility of using the nanocomposite for NGC applications. The results indicated that CaTiO3 nanoparticles were 50 nm in size. When the nanoparticles were added to the PGS, the elastic modulus and tensile strength of the nanocomposite reached values of about 1 and 0.5 MPa, respectively that are near those of natural nerves. The degradation behavior and swelling of the nanocomposites, as compared with those of the PGS elastomer, were controlled by introducing CaTiO3 into the PGS, which swelling limitation could prevent nerve compression. It was observed that Ca2+ ions established chemical bonds with PGS, which led to high crosslink densities that, in turn, contribute to improved mechanical properties of the composite. The Ca2+ ions released from the nanocomposite samples were in the nontoxic range. The PC12 cell line on the surface of the nanocomposite specimens showed good cell adhesion and proliferation with improved axon outgrowth and extension. Based on the results obtained the fabricated PGS/CaTiO3 nanocomposite may be recommended as a suitable NGC with desirable effects on peripheral nerve regeneration. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2181-2189, 2018.
Collapse
Affiliation(s)
- Anousheh Zargar Kharazi
- Biomaterials Nanotechnology and Tissue Engineering faculty, School of Advanced Medical Technology, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| | - Ghasem Dini
- Department of Nanotechnology Engineering, Faculty of Advanced Sciences and Technologies, University of Isfahan, Isfahan, 81746-73441, Iran
| | - Reza Naser
- Biomaterials Nanotechnology and Tissue Engineering faculty, School of Advanced Medical Technology, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| |
Collapse
|
30
|
Alonso‐Jiménez A, Ramón C, Dols‐Icardo O, Roig C, Gallardo E, Clarimón J, Núñez‐Peralta C, Díaz‐Manera J. Corpus callosum agenesis, myopathy and pinpoint pupils: consider Stormorken syndrome. Eur J Neurol 2018; 25:e25-e26. [DOI: 10.1111/ene.13545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Affiliation(s)
- A. Alonso‐Jiménez
- Neuromuscular Disorders Unit Neurology Department Hospital de la Santa Creu I Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - C. Ramón
- Neurology Department Hospital Universitario Central de Asturias Oviedo Spain
| | - O. Dols‐Icardo
- Memory Unit Neurology Department and Sant Pau Biomedical Research Institute Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid Spain
| | - C. Roig
- Neuromuscular Disorders Unit Neurology Department Hospital de la Santa Creu I Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - E. Gallardo
- Neuromuscular Disorders Unit Neurology Department Hospital de la Santa Creu I Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
- Centro de Investigación Básica en Red en Enfermedades Raras (CIBERER) Madrid Spain
| | - J. Clarimón
- Memory Unit Neurology Department and Sant Pau Biomedical Research Institute Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid Spain
| | - C. Núñez‐Peralta
- Radiology Department Hospital de la Santa Creu i Sant Pau Barcelona Spain
| | - J. Díaz‐Manera
- Neuromuscular Disorders Unit Neurology Department Hospital de la Santa Creu I Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
- Centro de Investigación Básica en Red en Enfermedades Raras (CIBERER) Madrid Spain
| |
Collapse
|
31
|
Theisen U, Hennig C, Ring T, Schnabel R, Köster RW. Neurotransmitter-mediated activity spatially controls neuronal migration in the zebrafish cerebellum. PLoS Biol 2018; 16:e2002226. [PMID: 29300740 PMCID: PMC5754045 DOI: 10.1371/journal.pbio.2002226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 11/22/2017] [Indexed: 11/27/2022] Open
Abstract
Neuronal migration during embryonic development contributes to functional brain circuitry. Many neurons migrate in morphologically distinct stages that coincide with differentiation, requiring tight spatial regulation. It had been proposed that neurotransmitter-mediated activity could exert this control. Here, we demonstrate that intracellular calcium transients occur in cerebellar neurons of zebrafish embryos during migration. We show that depolarization increases and hyperpolarization reduces the speed of tegmental hindbrain neurons using optogenetic tools and advanced track analysis optimized for in vivo migration. Finally, we introduce a compound screening assay to identify acetylcholine (ACh), glutamate, and glycine as regulators of migration, which act regionally along the neurons’ route. We summarize our findings in a model describing how different neurotransmitters spatially interact to control neuronal migration. The high evolutionary conservation of the cerebellum and hindbrain makes it likely that polarization state-driven motility constitutes an important principle in building a functional brain. Postmitotic neurons migrate from their site of origin to their final destination in the developing brain to form functional structures. These neurons typically follow defined routes through the tissue. Previous studies investigating progress along such route have identified neurotransmitters—chemicals that transmit the signals between neurons—as important regulators in neuronal migration using mostly rodent brain slice cultures and cultivated neurons. In this study, we use live zebrafish embryos to test the influence of neurotransmitters on migrating hindbrain neurons. First, we demonstrate that calcium transients can be measured in these neurons using genetically encoded reporters. Next, we use optogenetic channels to specifically de- or hyperpolarize the plasma membrane of the neurons to show that the polarization state is linked to migratory speed. Finally, we use a screening method to identify the neurotransmitter systems involved in migration progress control. We summarize these findings in a model that suggests that there are regions of influence for different neurotransmitters that act successively on the neurons to ensure their timely arrival at their destination.
Collapse
Affiliation(s)
- Ulrike Theisen
- Technische Universität Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany
| | - Christian Hennig
- Technische Universität Braunschweig, Institute for Genetics, Braunschweig, Germany
| | - Tobias Ring
- Technische Universität Braunschweig, Institute for Engineering Design, Vibroacoustics, Braunschweig, Germany
| | - Ralf Schnabel
- Technische Universität Braunschweig, Institute for Genetics, Braunschweig, Germany
| | - Reinhard W. Köster
- Technische Universität Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
32
|
Drebrin in Neuronal Migration and Axonal Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:141-155. [PMID: 28865019 DOI: 10.1007/978-4-431-56550-5_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
During development, production of neurons from neural stem cells, migration of neurons from their birthplace to their final location, and extension of neurites, axons, and dendrites are important for the formation of functional neuronal circuits. The actin cytoskeleton has major roles in the morphological development of neurons. In this chapter, we focused on the distribution and function of the actin-binding protein, drebrin, to elucidate the importance of drebrin-bound F-actin in neurons during early developmental stages of neurons in embryonic, postnatal, and adult brains. There are three major isoforms of drebrin in the chicken brain (E1, E2, and A) and two major isoforms in the mammalian brain (E and A). Among these drebrin isoforms, drebrin E1 and E2 in chicken and drebrin E in the mammalian brain are involved in these neuronal stages. In migrating neurons of the developing and adult brain, drebrin is localized at the base of filopodia of leading processes, to regulate neuronal migration. In axonal growth cones, drebrin is localized in the transitional zone to regulate axonal growth by inhibiting actomyosin interactions and mediating the interactions between F-actin and microtubules. For axonal collateral branching, drebrin is localized at axonal actin patches and the base of filopodia, to accelerate the transition from actin patches to filopodia and stabilize the filopodia.
Collapse
|
33
|
Fields C, Levin M. Multiscale memory and bioelectric error correction in the cytoplasm-cytoskeleton-membrane system. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2017; 10. [DOI: 10.1002/wsbm.1410] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/19/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Chris Fields
- 21 Rue des Lavandiéres, 11160 Caunes Minervois; France
| | - Michael Levin
- Allen Discovery Center at Tufts University; Medford MA USA
| |
Collapse
|
34
|
Abu-Omar N, Das J, Szeto V, Feng ZP. Neuronal Ryanodine Receptors in Development and Aging. Mol Neurobiol 2017; 55:1183-1192. [DOI: 10.1007/s12035-016-0375-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 12/28/2016] [Indexed: 01/09/2023]
|
35
|
Ly N, Cyert MS. Calcineurin, the Ca 2+-dependent phosphatase, regulates Rga2, a Cdc42 GTPase-activating protein, to modulate pheromone signaling. Mol Biol Cell 2017; 28:576-586. [PMID: 28077617 PMCID: PMC5328617 DOI: 10.1091/mbc.e16-06-0432] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 01/04/2017] [Accepted: 01/04/2017] [Indexed: 11/11/2022] Open
Abstract
Calcineurin, the conserved Ca2+/calmodulin-activated phosphatase, is required for viability during prolonged exposure to pheromone and acts through multiple substrates to down-regulate yeast pheromone signaling. Calcineurin regulates Dig2 and Rod1/Art4 to inhibit mating-induced gene expression and activate receptor internalization, respectively. Recent systematic approaches identified Rga2, a GTPase-activating protein (GAP) for the Cdc42 Rho-type GTPase, as a calcineurin substrate. Here we establish a physiological context for this regulation and show that calcineurin dephosphorylates and positively regulates Rga2 during pheromone signaling. Mating factor activates the Fus3/MAPK kinase, whose substrates induce gene expression, cell cycle arrest, and formation of the mating projection. Our studies demonstrate that Fus3 also phosphorylates Rga2 at inhibitory S/TP sites, which are targeted by Cdks during the cell cycle, and that calcineurin opposes Fus3 to activate Rga2 and decrease Cdc42 signaling. Yeast expressing an Rga2 mutant that is defective for regulation by calcineurin display increased gene expression in response to pheromone. This work is the first to identify cross-talk between Ca2+/calcineurin and Cdc42 signaling and to demonstrate modulation of Cdc42 activity through a GAP during mating.
Collapse
Affiliation(s)
- Nina Ly
- Department of Biology, Stanford University, Stanford, CA 94305
| | - Martha S Cyert
- Department of Biology, Stanford University, Stanford, CA 94305
| |
Collapse
|
36
|
Amare AT, Schubert KO, Klingler-Hoffmann M, Cohen-Woods S, Baune BT. The genetic overlap between mood disorders and cardiometabolic diseases: a systematic review of genome wide and candidate gene studies. Transl Psychiatry 2017; 7:e1007. [PMID: 28117839 PMCID: PMC5545727 DOI: 10.1038/tp.2016.261] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/21/2016] [Accepted: 10/31/2016] [Indexed: 12/11/2022] Open
Abstract
Meta-analyses of genome-wide association studies (meta-GWASs) and candidate gene studies have identified genetic variants associated with cardiovascular diseases, metabolic diseases and mood disorders. Although previous efforts were successful for individual disease conditions (single disease), limited information exists on shared genetic risk between these disorders. This article presents a detailed review and analysis of cardiometabolic diseases risk (CMD-R) genes that are also associated with mood disorders. First, we reviewed meta-GWASs published until January 2016, for the diseases 'type 2 diabetes, coronary artery disease, hypertension' and/or for the risk factors 'blood pressure, obesity, plasma lipid levels, insulin and glucose related traits'. We then searched the literature for published associations of these CMD-R genes with mood disorders. We considered studies that reported a significant association of at least one of the CMD-R genes and 'depression' or 'depressive disorder' or 'depressive symptoms' or 'bipolar disorder' or 'lithium treatment response in bipolar disorder', or 'serotonin reuptake inhibitors treatment response in major depression'. Our review revealed 24 potential pleiotropic genes that are likely to be shared between mood disorders and CMD-Rs. These genes include MTHFR, CACNA1D, CACNB2, GNAS, ADRB1, NCAN, REST, FTO, POMC, BDNF, CREB, ITIH4, LEP, GSK3B, SLC18A1, TLR4, PPP1R1B, APOE, CRY2, HTR1A, ADRA2A, TCF7L2, MTNR1B and IGF1. A pathway analysis of these genes revealed significant pathways: corticotrophin-releasing hormone signaling, AMPK signaling, cAMP-mediated or G-protein coupled receptor signaling, axonal guidance signaling, serotonin or dopamine receptors signaling, dopamine-DARPP32 feedback in cAMP signaling, circadian rhythm signaling and leptin signaling. Our review provides insights into the shared biological mechanisms of mood disorders and cardiometabolic diseases.
Collapse
Affiliation(s)
- A T Amare
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - K O Schubert
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Northern Adelaide Local Health Network, Mental Health Services, Adelaide, SA, Australia
| | - M Klingler-Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - S Cohen-Woods
- School of Psychology, Faculty of Social and Behavioural Sciences, Flinders University, Adelaide, SA, Australia
| | - B T Baune
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Discipline of Psychiatry, School of Medicine, The University of Adelaide, North Terrace, Adelaide, SA 5005, Australia. E-mail:
| |
Collapse
|
37
|
Sergi PN, Cavalcanti-Adam EA. Biomaterials and computation: a strategic alliance to investigate emergent responses of neural cells. Biomater Sci 2017; 5:648-657. [DOI: 10.1039/c6bm00871b] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Synergistic use of biomaterials and computation allows to identify and unravel neural cell responses.
Collapse
Affiliation(s)
- Pier Nicola Sergi
- The Biorobotics Institute
- Sant’ Anna Scuola Universitaria Superiore
- Pontedera
- 56025 Italy
| | - Elisabetta Ada Cavalcanti-Adam
- Max Planck Institute for Medical Research
- Dept Cellular Biophysics and Heidelberg University
- Dept Biophysical Chemistry
- Heidelberg
- Germany
| |
Collapse
|
38
|
Wieraszko A, Ahmed Z. Direct Current-Induced Calcium Trafficking in Different Neuronal Preparations. Neural Plast 2016; 2016:2823735. [PMID: 28074161 PMCID: PMC5198193 DOI: 10.1155/2016/2823735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 10/03/2016] [Accepted: 10/23/2016] [Indexed: 11/30/2022] Open
Abstract
The influence of direct current (DC) stimulation on radioactive calcium trafficking in sciatic nerve in vivo and in vitro, spinal cord, and synaptosomes was investigated. The exposure to DC enhanced calcium redistribution in all of these preparations. The effect was dependent on the strength of the stimulation and extended beyond the phase of exposure to DC. The DC-induced increase in calcium sequestration by synaptosomes was significantly reduced by cobalt and rupture of synaptosomes by osmotic shock. Although both anodal and cathodal currents were effective, the experiments with two electrodes of different areas revealed that cathodal stimulation exerted stronger effect. The exposure to DC induced not only relocation but also redistribution of calcium within segments of the sciatic nerve. Enzymatic removal of sialic acid by preincubation of synaptosomes with neuroaminidase, or carrying out the experiments in sodium-free environment, amplified DC-induced calcium accumulation.
Collapse
Affiliation(s)
- Andrzej Wieraszko
- Department of Biology, The College of Staten Island/City University of New York, 2800 Victory Boulevard, Staten Island, NY 10314, USA
| | - Zaghloul Ahmed
- Department of Physical Therapy, The College of Staten Island/City University of New York, 2800 Victory Boulevard, Staten Island, NY 10314, USA
| |
Collapse
|
39
|
Watanabe K, Bizen N, Sato N, Takebayashi H. Endoplasmic Reticulum-Localized Transmembrane Protein Dpy19L1 Is Required for Neurite Outgrowth. PLoS One 2016; 11:e0167985. [PMID: 27959946 PMCID: PMC5154530 DOI: 10.1371/journal.pone.0167985] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/28/2016] [Indexed: 11/18/2022] Open
Abstract
The endoplasmic reticulum (ER), including the nuclear envelope, is a continuous and intricate membrane-bound organelle responsible for various cellular functions. In neurons, the ER network is found in cell bodies, axons, and dendrites. Recent studies indicate the involvement of the ER network in neuronal development, such as neuronal migration and axonal outgrowth. However, the regulation of neural development by ER-localized proteins is not fully understood. We previously reported that the multi-transmembrane protein Dpy19L1 is required for neuronal migration in the developing mouse cerebral cortex. A Dpy19L family member, Dpy19L2, which is a causative gene for human Globozoospermia, is suggested to act as an anchor of the acrosome to the nuclear envelope. In this study, we found that the patterns of exogenous Dpy19L1 were partially coincident with the ER, including the nuclear envelope in COS-7 cells at the level of the light microscope. The reticular distribution of Dpy19L1 was disrupted by microtubule depolymerization that induces retraction of the ER. Furthermore, Dpy19L1 showed a similar distribution pattern with a ER marker protein in embryonic mouse cortical neurons. Finally, we showed that Dpy19L1 knockdown mediated by siRNA resulted in decreased neurite outgrowth in cultured neurons. These results indicate that transmembrane protein Dpy19L1 is localized to the ER membrane and regulates neurite extension during development.
Collapse
Affiliation(s)
- Keisuke Watanabe
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Gross Anatomy and Morphogenesis, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- * E-mail:
| | - Norihisa Bizen
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Noboru Sato
- Division of Gross Anatomy and Morphogenesis, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
40
|
Xu B, Bressloff PC. Model of Growth Cone Membrane Polarization via Microtubule Length Regulation. Biophys J 2016; 109:2203-14. [PMID: 26588578 DOI: 10.1016/j.bpj.2015.09.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/04/2015] [Accepted: 09/21/2015] [Indexed: 01/18/2023] Open
Abstract
We present a mathematical model of membrane polarization in growth cones. We proceed by coupling an active transport model of cytosolic proteins along a two-dimensional microtubule (MT) network with a modified Dogterom-Leibler model of MT growth. In particular, we consider a Rac1-stathmin-MT pathway in which the growth and catastrophe rates of MTs are regulated by cytosolic stathmin, while the stathmin is regulated by Rac1 at the membrane. We use regular perturbation theory and numerical simulations to determine the steady-state stathmin concentration, the mean MT length distribution, and the resulting distribution of membrane-bound proteins. We thus show how a nonuniform Rac1 distribution on the membrane generates a polarized distribution of membrane proteins. The mean MT length distribution and hence the degree of membrane polarization are sensitive to the precise form of the Rac1 distribution and parameters such as the catastrophe-promoting constant and tubulin association rate. This is a consequence of the fact that the lateral diffusion of stathmin tends to weaken the effects of Rac1 on the distribution of mean MT lengths.
Collapse
Affiliation(s)
- Bin Xu
- Mathematics, University of Utah, Salt Lake City, Utah
| | | |
Collapse
|
41
|
Nesler KR, Starke EL, Boin NG, Ritz M, Barbee SA. Presynaptic CamKII regulates activity-dependent axon terminal growth. Mol Cell Neurosci 2016; 76:33-41. [PMID: 27567686 DOI: 10.1016/j.mcn.2016.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 08/12/2016] [Accepted: 08/22/2016] [Indexed: 12/27/2022] Open
Abstract
Spaced synaptic depolarization induces rapid axon terminal growth and the formation of new synaptic boutons at the Drosophila larval neuromuscular junction (NMJ). Here, we identify a novel presynaptic function for the Calcium/Calmodulin-dependent Kinase II (CamKII) protein in the control of activity-dependent synaptic growth. Consistent with this function, we find that both total and phosphorylated CamKII (p-CamKII) are enriched in axon terminals. Interestingly, p-CamKII appears to be enriched at the presynaptic axon terminal membrane. Moreover, levels of total CamKII protein within presynaptic boutons globally increase within one hour following stimulation. These effects correlate with the activity-dependent formation of new presynaptic boutons. The increase in presynaptic CamKII levels is inhibited by treatment with cyclohexamide suggesting a protein-synthesis dependent mechanism. We have previously found that acute spaced stimulation rapidly downregulates levels of neuronal microRNAs (miRNAs) that are required for the control of activity-dependent axon terminal growth at this synapse. The rapid activity-dependent accumulation of CamKII protein within axon terminals is inhibited by overexpression of activity-regulated miR-289 in motor neurons. Experiments in vitro using a CamKII translational reporter show that miR-289 can directly repress the translation of CamKII via a sequence motif found within the CamKII 3' untranslated region (UTR). Collectively, our studies support the idea that presynaptic CamKII acts downstream of synaptic stimulation and the miRNA pathway to control rapid activity-dependent changes in synapse structure.
Collapse
Affiliation(s)
- Katherine R Nesler
- Department of Biological Sciences, Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA
| | - Emily L Starke
- Department of Biological Sciences, Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA
| | - Nathan G Boin
- Department of Biological Sciences, Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA
| | - Matthew Ritz
- Department of Biological Sciences, Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA
| | - Scott A Barbee
- Department of Biological Sciences, Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA; Molecular and Cellular Biophysics Program, University of Denver, Denver, CO 80210, USA.
| |
Collapse
|
42
|
Emergence of ion channel modal gating from independent subunit kinetics. Proc Natl Acad Sci U S A 2016; 113:E5288-97. [PMID: 27551100 DOI: 10.1073/pnas.1604090113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many ion channels exhibit a slow stochastic switching between distinct modes of gating activity. This feature of channel behavior has pronounced implications for the dynamics of ionic currents and the signaling pathways that they regulate. A canonical example is the inositol 1,4,5-trisphosphate receptor (IP3R) channel, whose regulation of intracellular Ca(2+) concentration is essential for numerous cellular processes. However, the underlying biophysical mechanisms that give rise to modal gating in this and most other channels remain unknown. Although ion channels are composed of protein subunits, previous mathematical models of modal gating are coarse grained at the level of whole-channel states, limiting further dialogue between theory and experiment. Here we propose an origin for modal gating, by modeling the kinetics of ligand binding and conformational change in the IP3R at the subunit level. We find good agreement with experimental data over a wide range of ligand concentrations, accounting for equilibrium channel properties, transient responses to changing ligand conditions, and modal gating statistics. We show how this can be understood within a simple analytical framework and confirm our results with stochastic simulations. The model assumes that channel subunits are independent, demonstrating that cooperative binding or concerted conformational changes are not required for modal gating. Moreover, the model embodies a generally applicable principle: If a timescale separation exists in the kinetics of individual subunits, then modal gating can arise as an emergent property of channel behavior.
Collapse
|
43
|
Nawrotek K, Tylman M, Rudnicka K, Gatkowska J, Balcerzak J. Tubular electrodeposition of chitosan–carbon nanotube implants enriched with calcium ions. J Mech Behav Biomed Mater 2016; 60:256-266. [DOI: 10.1016/j.jmbbm.2016.02.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/30/2016] [Accepted: 02/05/2016] [Indexed: 01/20/2023]
|
44
|
Nawrotek K, Tylman M, Rudnicka K, Balcerzak J, Kamiński K. Chitosan-based hydrogel implants enriched with calcium ions intended for peripheral nervous tissue regeneration. Carbohydr Polym 2016; 136:764-71. [DOI: 10.1016/j.carbpol.2015.09.105] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/27/2015] [Accepted: 09/28/2015] [Indexed: 01/30/2023]
|
45
|
Dynamic Trk and G Protein Signalings Regulate Dopaminergic Neurodifferentiation in Human Trophoblast Stem Cells. PLoS One 2015; 10:e0143852. [PMID: 26606046 PMCID: PMC4659658 DOI: 10.1371/journal.pone.0143852] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022] Open
Abstract
Understanding the mechanisms in the generation of neural stem cells from pluripotent stem cells is a fundamental step towards successful management of neurodegenerative diseases in translational medicine. Albeit all-trans retinoic acid (RA) has been associated with axon outgrowth and nerve regeneration, the maintenance of differentiated neurons, the association with degenerative disease like Parkinson's disease, and its regulatory molecular mechanism from pluripotent stem cells to neural stem cells remain fragmented. We have previously reported that RA is capable of differentiation of human trophoblast stem cells to dopamine (DA) committed progenitor cells. Intracranial implantation of such neural progenitor cells into the 6-OHDA-lesioned substantia nigra pars compacta successfully regenerates dopaminergic neurons and integrity of the nigrostriatal pathway, ameliorating the behavioral deficits in the Parkinson’s disease rat model. Here, we demonstrated a dynamic molecular network in systematic analysis by addressing spatiotemporal molecular expression, intracellular protein-protein interaction and inhibition, imaging study, and genetic expression to explore the regulatory mechanisms of RA induction in the differentiation of human trophoblast stem cells to DA committed progenitor cells. We focused on the tyrosine receptor kinase (Trk), G proteins, canonical Wnt2B/β-catenin, genomic and non-genomic RA signaling transductions with Tyrosine hydroxylase (TH) gene expression as the differentiation endpoint. We found that at the early stage, integration of TrkA and G protein signalings aims for axonogenesis and morphogenesis, involving the novel RXRα/Gαq/11 and RARβ/Gβ signaling pathways. While at the later stage, five distinct signaling pathways together with epigenetic histone modifications emerged to regulate expression of TH, a precursor of dopamine. RA induction generated DA committed progenitor cells in one day. Our results provided substantial mechanistic evidence that human trophoblast stem cell-derived neural stem cells can potentially be used for neurobiological study, drug discovery, and as an alternative source of cell-based therapy in neurodegenerative diseases like Parkinson’s disease.
Collapse
|
46
|
Nerve Growth Factor Regulates Transient Receptor Potential Vanilloid 2 via Extracellular Signal-Regulated Kinase Signaling To Enhance Neurite Outgrowth in Developing Neurons. Mol Cell Biol 2015; 35:4238-52. [PMID: 26416880 DOI: 10.1128/mcb.00549-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/21/2015] [Indexed: 12/13/2022] Open
Abstract
Neurite outgrowth is key to the formation of functional circuits during neuronal development. Neurotrophins, including nerve growth factor (NGF), increase neurite outgrowth in part by altering the function and expression of Ca(2+)-permeable cation channels. Here we report that transient receptor potential vanilloid 2 (TRPV2) is an intracellular Ca(2+)-permeable TRPV channel upregulated by NGF via the mitogen-activated protein kinase (MAPK) signaling pathway to augment neurite outgrowth. TRPV2 colocalized with Rab7, a late endosome protein, in addition to TrkA and activated extracellular signal-regulated kinase (ERK) in neurites, indicating that the channel is closely associated with signaling endosomes. In line with these results, we showed that TRPV2 acts as an ERK substrate and identified the motifs necessary for phosphorylation of TRPV2 by ERK. Furthermore, neurite length, TRPV2 expression, and TRPV2-mediated Ca(2+) signals were reduced by mutagenesis of these key ERK phosphorylation sites. Based on these findings, we identified a previously uncharacterized mechanism by which ERK controls TRPV2-mediated Ca(2+) signals in developing neurons and further establish TRPV2 as a critical intracellular ion channel in neuronal function.
Collapse
|
47
|
Majewski L, Kuznicki J. SOCE in neurons: Signaling or just refilling? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1940-52. [DOI: 10.1016/j.bbamcr.2015.01.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 01/14/2023]
|
48
|
He Y, Ren Y, Wu B, Decourt B, Lee AC, Taylor A, Suter DM. Src and cortactin promote lamellipodia protrusion and filopodia formation and stability in growth cones. Mol Biol Cell 2015. [PMID: 26224308 PMCID: PMC4569314 DOI: 10.1091/mbc.e15-03-0142] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
How Src tyrosine kinase and cortactin control actin organization and dynamics in neuronal growth cones is not well understood. Using multiple high-resolution imaging techniques, this study shows that Src and cortactin control the persistence of lamellipodial protrusion as well as the formation, stability, and elongation of filopodia in growth cones. Src tyrosine kinases have been implicated in axonal growth and guidance; however, the underlying cellular mechanisms are not well understood. Specifically, it is unclear which aspects of actin organization and dynamics are regulated by Src in neuronal growth cones. Here, we investigated the function of Src2 and one of its substrates, cortactin, in lamellipodia and filopodia of Aplysia growth cones. We found that up-regulation of Src2 activation state or cortactin increased lamellipodial length, protrusion time, and actin network density, whereas down-regulation had opposite effects. Furthermore, Src2 or cortactin up-regulation increased filopodial density, length, and protrusion time, whereas down-regulation promoted lateral movements of filopodia. Fluorescent speckle microscopy revealed that rates of actin assembly and retrograde flow were not affected in either case. In summary, our results support a model in which Src and cortactin regulate growth cone motility by increasing actin network density and protrusion persistence of lamellipodia by controlling the state of actin-driven protrusion versus retraction. In addition, both proteins promote the formation and stability of actin bundles in filopodia.
Collapse
Affiliation(s)
- Yingpei He
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Yuan Ren
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Bingbing Wu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Boris Decourt
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Aih Cheun Lee
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Aaron Taylor
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907 )
| |
Collapse
|
49
|
Roccasalvo IM, Micera S, Sergi PN. A hybrid computational model to predict chemotactic guidance of growth cones. Sci Rep 2015; 5:11340. [PMID: 26086936 PMCID: PMC4471899 DOI: 10.1038/srep11340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 05/15/2015] [Indexed: 11/08/2022] Open
Abstract
The overall strategy used by growing axons to find their correct paths during the nervous system development is not yet completely understood. Indeed, some emergent and counterintuitive phenomena were recently described during axon pathfinding in presence of chemical gradients. Here, a novel computational model is presented together with its ability to reproduce both regular and counterintuitive axonal behaviours. In this model, the key role of intracellular calcium was phenomenologically modelled through a non standard Gierer-Meinhardt system, as a crucial factor influencing the growth cone behaviour both in regular and complex conditions. This model was able to explicitly reproduce neuritic paths accounting for the complex interplay between extracellular and intracellular environments, through the sensing capability of the growth cone. The reliability of this approach was proven by using quantitative metrics, numerically supporting the similarity between in silico and biological results in regular conditions (control and attraction). Finally, the model was able to qualitatively predict emergent and counterintuitive phenomena resulting from complex boundary conditions.
Collapse
Affiliation(s)
| | - Silvestro Micera
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Pisa, Italy
- Bertarelli Foundation Chair in Translational NeuroEngineering Laboratory, Institute of Bioengineering, School of Engineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
- Center for Neuroprosthetics, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
50
|
Tojima T, Kamiguchi H. Exocytic and endocytic membrane trafficking in axon development. Dev Growth Differ 2015; 57:291-304. [DOI: 10.1111/dgd.12218] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 04/09/2015] [Accepted: 04/09/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Takuro Tojima
- Laboratory for Neuronal Growth Mechanisms; RIKEN Brain Science Institute; 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Hiroyuki Kamiguchi
- Laboratory for Neuronal Growth Mechanisms; RIKEN Brain Science Institute; 2-1 Hirosawa Wako Saitama 351-0198 Japan
| |
Collapse
|