1
|
Lu CH, Lee CE, Nakamoto ML, Cui B. Cellular Signaling at the Nano-Bio Interface: Spotlighting Membrane Curvature. Annu Rev Phys Chem 2025; 76:251-277. [PMID: 40258240 PMCID: PMC12043246 DOI: 10.1146/annurev-physchem-090722-021151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
No longer viewed as a passive consequence of cellular activities, membrane curvature-the physical shape of the cell membrane-is now recognized as an active constituent of biological processes. Nanoscale topographies on extracellular matrices or substrate surfaces impart well-defined membrane curvatures on the plasma membrane. This review examines biological events occurring at the nano-bio interface, the physical interface between the cell membrane and surface nanotopography, which activates intracellular signaling by recruiting curvature-sensing proteins. We encompass a wide range of biological processes at the nano-bio interface, including cell adhesion, endocytosis, glycocalyx redistribution, regulation of mechanosensitive ion channels, cell migration, and differentiation. Despite the diversity of processes, we call attention to the critical role of membrane curvature in each process. We particularly highlight studies that elucidate molecular mechanisms involving curvature-sensing proteins with the hope of providing comprehensive insights into this rapidly advancing area of research.
Collapse
Affiliation(s)
- Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, California, USA;
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
| | - Christina E Lee
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
- Biophysics Program, Stanford University School of Medicine, Stanford, California, USA
| | - Melissa L Nakamoto
- Department of Chemistry, Stanford University, Stanford, California, USA;
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California, USA;
- Wu-Tsai Neuroscience Institute and Sarafan ChEM-H Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
2
|
Che H, Du Y, Jiang Y, Zhu Z, Bai M, Zheng J, Yang M, Xiang L, Gong P. Neuronal TRPV1-CGRP axis regulates peripheral nerve regeneration through ERK/HIF-1 signaling pathway. J Neurochem 2025; 169:e16281. [PMID: 39792906 DOI: 10.1111/jnc.16281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025]
Abstract
Severe trauma frequently leads to nerve damage. Peripheral nerves possess a degree of regenerative ability, and actively promoting their recovery can help restore the sensory and functional capacities of tissues. The neuropeptide calcitonin gene-related peptide (CGRP) is believed to regulate the repair of injured peripheral nerves, with neuronal transient receptor potential vanilloid type 1 (TRPV1) potentially serving as a crucial upstream factor. In this study, we established a mouse model of sciatic nerve (SN) crush injury and found that intrathecal injection of capsaicin (Cap) activated the neuronal TRPV1-CGRP axis, thereby promoting SN repair. Conversely, the application of capsazepine (Cpz), which inhibits the neuronal TRPV1-CGRP axis, delayed SN repair. Local restoration of CGRP expression at the injury site enhanced the repair process. In vitro experiments, we employed the rat Schwann cell (SC) line RSC96 to establish an indirect co-culture model of neurons and SCs. We observed that the proliferation, migration, expression of myelination-associated proteins, and neurotrophic secretion functions of RSC96 cells are positively correlated with the degree of activation of neuronal TRPV1. Inhibition of neuronal TRPV1, followed by the restoration of CGRP levels, improved these functions in RSC96 cells. Furthermore, activation of the neuronal TRPV1-CGRP axis resulted in an upregulation of extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation levels and an increase in hypoxia-inducible factor 1α (HIF-1α) accumulation in RSC96 cells, thereby promoting their proliferation and migration. In summary, this study demonstrates that neuronal TRPV1-CGRP axis can regulate biological behavior of SCs and axon regeneration by activating the ERK/HIF-1 signaling pathway following peripheral nerve injury. This finding clarifies the role of CGRP in neuroregulatory networks and provides a novel reference point for the development of drugs and biomaterials for treating nerve damage.
Collapse
Affiliation(s)
- Huiling Che
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Du
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixuan Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhanfeng Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mingxuan Bai
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianan Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mao Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Vecchi JT, Rhomberg M, Allan Guymon C, Hansen MR. Inositol trisphosphate and ryanodine receptor signaling distinctly regulate neurite pathfinding in response to engineered micropatterned surfaces. PLoS One 2024; 19:e0308389. [PMID: 39236043 PMCID: PMC11376539 DOI: 10.1371/journal.pone.0308389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/26/2024] [Indexed: 09/07/2024] Open
Abstract
Micro and nanoscale patterning of surface features and biochemical cues have emerged as tools to precisely direct neurite growth into close proximity with next generation neural prosthesis electrodes. Biophysical cues can exert greater influence on neurite pathfinding compared to the more well studied biochemical cues; yet the signaling events underlying the ability of growth cones to respond to these microfeatures remain obscure. Intracellular Ca2+ signaling plays a critical role in how a growth cone senses and grows in response to various cues (biophysical features, repulsive peptides, chemo-attractive gradients). Here, we investigate the role of inositol triphosphate (IP3) and ryanodine-sensitive receptor (RyR) signaling as sensory neurons (spiral ganglion neurons, SGNs, and dorsal root ganglion neurons, DRGNs) pathfind in response to micropatterned substrates of varied geometries. We find that IP3 and RyR signaling act in the growth cone as they navigate biophysical cues and enable proper guidance to biophysical, chemo-permissive, and chemo-repulsive micropatterns. In response to complex micropatterned geometries, RyR signaling appears to halt growth in response to both topographical features and chemo-repulsive cues. IP3 signaling appears to play a more complex role, as growth cones appear to sense the microfeatures in the presence of xestospongin C but are unable to coordinate turning in response to them. Overall, key Ca2+ signaling elements, IP3 and RyR, are found to be essential for SGNs to pathfind in response to engineered biophysical and biochemical cues. These findings inform efforts to precisely guide neurite regeneration for improved neural prosthesis function, including cochlear implants.
Collapse
Affiliation(s)
- Joseph T Vecchi
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, United States of America
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States of America
| | - Madeline Rhomberg
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States of America
| | - C Allan Guymon
- Department of Chemical Engineering, University of Iowa, Iowa City, IA, United States of America
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, United States of America
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States of America
| |
Collapse
|
4
|
Vecchi JT, Rhomberg M, Guymon CA, Hansen MR. The geometry of photopolymerized topography influences neurite pathfinding by directing growth cone morphology and migration. J Neural Eng 2024; 21:026027. [PMID: 38547528 PMCID: PMC10993768 DOI: 10.1088/1741-2552/ad38dc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Objective. Cochlear implants provide auditory perception to those with severe to profound sensorineural hearing loss: however, the quality of sound perceived by users does not approximate natural hearing. This limitation is due in part to the large physical gap between the stimulating electrodes and their target neurons. Therefore, directing the controlled outgrowth of processes from spiral ganglion neurons (SGNs) into close proximity to the electrode array could provide significantly increased hearing function.Approach.For this objective to be properly designed and implemented, the ability and limits of SGN neurites to be guided must first be determined. In this work, we engineer precise topographical microfeatures with angle turn challenges of various geometries to study SGN pathfinding and use live imaging to better understand how neurite growth is guided by these cues.Main Results.We find that the geometry of the angled microfeatures determines the ability of neurites to navigate the angled microfeature turns. SGN neurite pathfinding fidelity is increased by 20%-70% through minor increases in microfeature amplitude (depth) and by 25% if the angle of the patterned turn is made obtuse. Further, we see that dorsal root ganglion neuron growth cones change their morphology and migration to become more elongated within microfeatures. Our observations also indicate complexities in studying neurite turning. First, as the growth cone pathfinds in response to the various cues, the associated neurite often reorients across the angle topographical microfeatures. Additionally, neurite branching is observed in response to topographical guidance cues, most frequently when turning decisions are most uncertain.Significance.Overall, the multi-angle channel micropatterned substrate is a versatile and efficient system to assess neurite turning and pathfinding in response to topographical cues. These findings represent fundamental principles of neurite pathfinding that will be essential to consider for the design of 3D systems aiming to guide neurite growthin vivo.
Collapse
Affiliation(s)
- Joseph T Vecchi
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA, United States of America
| | - Madeline Rhomberg
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA, United States of America
| | - C Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, United States of America
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States of America
- Department of Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
5
|
Rezzani R, Favero G, Gianò M, Pinto D, Labanca M, van Noorden CJ, Rinaldi F. Transient Receptor Potential Channels in the Healthy and Diseased Blood-Brain Barrier. J Histochem Cytochem 2024; 72:199-231. [PMID: 38590114 PMCID: PMC11020746 DOI: 10.1369/00221554241246032] [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: 09/19/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
The large family of transient receptor potential (TRP) channels are integral membrane proteins that function as environmental sensors and act as ion channels after activation by mechanical (touch), physical (heat, pain), and chemical stimuli (pungent compounds such as capsaicin). Most TRP channels are localized in the plasma membrane of cells but some of them are localized in membranes of organelles and function as intracellular Ca2+-ion channels. TRP channels are involved in neurological disorders but their precise role(s) and relevance in these disorders are not clear. Endothelial cells of the blood-brain barrier (BBB) express TRP channels such as TRP vanilloid 1-4 and are involved in thermal detection by regulating BBB permeability. In neurological disorders, TRP channels in the BBB are responsible for edema formation in the brain. Therefore, drug design to modulate locally activity of TRP channels in the BBB is a hot topic. Today, the application of TRP channel antagonists against neurological disorders is still limited.
Collapse
Affiliation(s)
- Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
| | - Marzia Gianò
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, Milan, Italy
| | - Mauro Labanca
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, Milan, Italy
| |
Collapse
|
6
|
Vecchi JT, Rhomberg M, Guymon CA, Hansen MR. The geometry of photopolymerized topography influences neurite pathfinding by directing growth cone morphology and migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555111. [PMID: 37693432 PMCID: PMC10491164 DOI: 10.1101/2023.08.28.555111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Cochlear implants (CIs) provide auditory perception to those with profound sensorineural hearing loss: however, the quality of sound perceived by a CI user does not approximate natural hearing. This limitation is due in part to the large physical gap between the stimulating electrodes and their target neurons. Therefore, directing the controlled outgrowth of processes from spiral ganglion neurons (SGNs) into close proximity to the electrode array could provide significantly increased hearing function. For this objective to be properly designed and implemented, the ability and limits of SGN neurites to be guided must first be determined. In this work, we engineered precise topographical microfeatures with angle turn challenges of various geometries to study SGN pathfinding. Additionally, we analyze sensory neurite growth in response to topographically patterned substrates and use live imaging to better understand how neurite growth is guided by these cues. In assessing the ability of neurites to sense and turn in response to topographical cues, we find that the geometry of the angled microfeatures determines the ability of neurites to navigate the angled microfeature turns. SGN neurite pathfinding fidelity can be increased by 20-70% through minor increases in microfeature amplitude (depth) and by 25% if the angle of the patterned turn is made more obtuse. Further, by using engineered topographies and live imaging of dorsal root ganglion neurons (DRGNs), we see that DRGN growth cones change their morphology and migration to become more elongated within microfeatures. However, our observations also indicate complexities in studying neurite turning. First, as the growth cone pathfinds in response to the various cues, the associated neurite often reorients across the angle topographical microfeatures. This reorientation is likely related to the tension the neurite shaft experiences when the growth cone elongates in the microfeature around a turn. Additionally, neurite branching is observed in response to topographical guidance cues, most frequently when turning decisions are most uncertain. Overall, the multi-angle channel micropatterned substrate is a versatile and efficient system to assess SGN neurite turning and pathfinding in response to topographical cues. These findings represent fundamental principles of neurite pathfinding that will be essential to consider for the design of 3D systems aiming to guide neurite growth in vivo.
Collapse
Affiliation(s)
- Joseph T. Vecchi
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, USA
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, USA
| | - Madeline Rhomberg
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, USA
| | - C. Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Marlan R. Hansen
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, USA
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
7
|
Zu L, Shi H, Yang J, Zhang C, Fu Y, Xi N, Liu L, Wang W. Unidirectional diphenylalanine nanotubes for dynamically guiding neurite outgrowth. Biomed Mater 2022; 18. [PMID: 36541466 DOI: 10.1088/1748-605x/aca737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022]
Abstract
Neural networks have been culturedin vitroto investigate brain functions and diseases, clinical treatments for brain damage, and device development. However, it remains challenging to form complex neural network structures with desired orientations and connectionsin vitro. Here, we introduce a dynamic strategy by using diphenylalanine (FF) nanotubes for controlling physical patterns on a substrate to regulate neurite-growth orientation in cultivating neural networks. Parallel FF nanotube patterns guide neurons to develop neurites through the unidirectional FF nanotubes while restricting their polarization direction. Subsequently, the FF nanotubes disassemble and the restriction of neurites disappear, and secondary neurite development of the neural network occurs in other direction. Experiments were conducted that use the hippocampal neurons, and the results demonstrated that the cultured neural networks by using the proposed dynamic approach can form a significant cross-connected structure with substantially more lateral neural connections than static substrates. The proposed dynamic approach for neurite outgrowing enables the construction of oriented innervation and cross-connected neural networksin vitroand may explore the way for the bio-fabrication of highly complex structures in tissue engineering.
Collapse
Affiliation(s)
- Lipeng Zu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huiyao Shi
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jia Yang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China.,University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Chuang Zhang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
| | - Yuanyuan Fu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang 110122, People's Republic of China
| | - Ning Xi
- Department of Industrial and Manufacturing Systems Engineering, University of Hong Kong, Pokfulam Road, Hong Kong, People's Republic of China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
| | - Wenxue Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.,Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
| |
Collapse
|
8
|
Interaction of micropatterned topographical and biochemical cues to direct neurite growth from spiral ganglion neurons. Hear Res 2021; 409:108315. [PMID: 34343850 DOI: 10.1016/j.heares.2021.108315] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/07/2021] [Accepted: 07/12/2021] [Indexed: 01/01/2023]
Abstract
Functional outcomes with neural prosthetic devices, such as cochlear implants, are limited in part due to physical separation between the stimulating elements and the neurons they stimulate. One strategy to close this gap aims to precisely guide neurite regeneration to position the neurites in closer proximity to electrode arrays. Here, we explore the ability of micropatterned biochemical and topographic guidance cues, singly and in combination, to direct the growth of spiral ganglion neuron (SGN) neurites, the neurons targeted by cochlear implants. Photopolymerization of methacrylate monomers was used to form unidirectional topographical features of ridges and grooves in addition to multidirectional patterns with 90o angle turns. Microcontact printing was also used to create similar uni- and multi-directional patterns of peptides on polymer surfaces. Biochemical cues included peptides that facilitate (laminin, LN) or repel (EphA4-Fc) neurite growth. On flat surfaces, SGN neurites preferentially grew on LN-coated stripes and avoided EphA4-Fc-coated stripes. LN or EphA4-Fc was selectively adsorbed onto the ridges or grooves to test the neurite response to a combination of topographical and biochemical cues. Coating the ridges with EphA4-Fc and grooves with LN lead to enhanced SGN alignment to topographical patterns. Conversely, EphA4-Fc coating on the grooves or LN coating on the ridges tended to disrupt alignment to topographical patterns. SGN neurites respond to combinations of topographical and biochemical cues and surface patterning that leverages both cues enhance guided neurite growth.
Collapse
|
9
|
Lavanderos B, Silva I, Cruz P, Orellana-Serradell O, Saldías MP, Cerda O. TRP Channels Regulation of Rho GTPases in Brain Context and Diseases. Front Cell Dev Biol 2020; 8:582975. [PMID: 33240883 PMCID: PMC7683514 DOI: 10.3389/fcell.2020.582975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological and neuropsychiatric disorders are mediated by several pathophysiological mechanisms, including developmental and degenerative abnormalities caused primarily by disturbances in cell migration, structural plasticity of the synapse, and blood-vessel barrier function. In this context, critical pathways involved in the pathogenesis of these diseases are related to structural, scaffolding, and enzymatic activity-bearing proteins, which participate in Ca2+- and Ras Homologs (Rho) GTPases-mediated signaling. Rho GTPases are GDP/GTP binding proteins that regulate the cytoskeletal structure, cellular protrusion, and migration. These proteins cycle between GTP-bound (active) and GDP-bound (inactive) states due to their intrinsic GTPase activity and their dynamic regulation by GEFs, GAPs, and GDIs. One of the most important upstream inputs that modulate Rho GTPases activity is Ca2+ signaling, positioning ion channels as pivotal molecular entities for Rho GTPases regulation. Multiple non-selective cationic channels belonging to the Transient Receptor Potential (TRP) family participate in cytoskeletal-dependent processes through Ca2+-mediated modulation of Rho GTPases. Moreover, these ion channels have a role in several neuropathological events such as neuronal cell death, brain tumor progression and strokes. Although Rho GTPases-dependent pathways have been extensively studied, how they converge with TRP channels in the development or progression of neuropathologies is poorly understood. Herein, we review recent evidence and insights that link TRP channels activity to downstream Rho GTPase signaling or modulation. Moreover, using the TRIP database, we establish associations between possible mediators of Rho GTPase signaling with TRP ion channels. As such, we propose mechanisms that might explain the TRP-dependent modulation of Rho GTPases as possible pathways participating in the emergence or maintenance of neuropathological conditions.
Collapse
Affiliation(s)
- Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Octavio Orellana-Serradell
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
10
|
Huo Z, Su Y, Dong Y, Zheng Y, Zhang Q, Duan Y, Wang G. Rho-kinase inhibition by Fasudil promotes tumor maturation and apoptosis in small-cell lung cancer. Am J Transl Res 2020; 12:4354-4370. [PMID: 32913511 PMCID: PMC7476155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/17/2020] [Indexed: 06/11/2023]
Abstract
Small-cell lung cancer (SCLC) is a poorly differentiated neuroendocrine neoplasm with inadequate therapeutic options. Fasudil is a Rho-associated protein kinase 1 and 2 (ROCK1/2) inhibitor whose clinical indications remain limited in cardiocerebrovascular diseases. This study aimed to report a possible implication of Fasudil for SCLC. The expression and prognostic value of ROCK1/2 were investigated immunohistochemically in surgical specimens. The positive rates of ROCK1 (77/113, 68.1%) and ROCK2 (94/113, 83.2%) were distinctly higher in SCLC than in other lung neuroendocrine tumors. The high expression level of ROCK1 was related to the poor long-term survival of patients, especially in the classic SCLC subtype. In vitro, SCLC cell line treated with Fasudil exhibited synapse-like morphologic change, accompanied by a reduction in the expression levels of c-myc and cyclin D1. Cell cycle arrest was further demonstrated, accompanied by sensitivity to starvation-induced apoptosis, indicating tumor maturation. In addition, RNA-seq identified hundreds of differentially expressed genes involved in the positive regulation of neuron differentiation, stem cell differentiation, cell development, and nervous system development. Finally, Fasudil inhibited SCLC growth, promoted structural maturity, and induced apoptosis in BALB/c nude mice xenograft model. In conclusion, these results indicated a potential and novel application of Fasudil for SCLC treatment.
Collapse
Affiliation(s)
- Zitian Huo
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yinxia Su
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yuting Dong
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yiyun Zheng
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Qian Zhang
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yaqi Duan
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Guoping Wang
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| |
Collapse
|
11
|
Surface morphology and property of UV-cured film containing photopolymerizable polysiloxane-based nanogels with initiating capability. INTERNATIONAL JOURNAL OF INDUSTRIAL CHEMISTRY 2019. [DOI: 10.1007/s40090-019-00193-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
12
|
Teotia M, Mittal A, Soni RK. Light-mediated thermoset polymers. MATERIALS FOR BIOMEDICAL ENGINEERING 2019:57-103. [DOI: 10.1016/b978-0-12-816874-5.00003-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Impaired healing of cornea incision injury in a TRPV1-deficient mouse. Cell Tissue Res 2018; 374:329-338. [PMID: 29971480 PMCID: PMC6209059 DOI: 10.1007/s00441-018-2878-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/21/2018] [Indexed: 12/19/2022]
Abstract
The present study attempts to elucidate the role of TRPV1 cation channel receptor on primary repair in an incision-wounded mouse cornea in vivo. Previous study revealed that blocking TRPV1 suppressed myofibroblast formation and expression of transforming growth factor β1 (TGFβ1) in cultured keratocytes or ocular fibroblasts. Male C57BL/6 (wild-type; WT) mice and male C57BL/6 Trpv1-null (KO) mice incurred a full-thickness incision injury (1.8 mm in length, limbus to limbus) in the central cornea of one eye with a surgical blade under general and topical anesthesia. The injury was not sutured. On days 0, 5, and 10, the eyes were enucleated, processed for histology, immunohistochemistry, and real-time RT-PCR gene expression analysis to evaluate the effects of the loss of TRPV1 on primary healing. Electron microscopy observation was also performed to know the effect of the loss of TRPV1 on ultrastructure of keratocytes. The results showed that the loss of Trpv1 gene delayed closure of corneal stromal incision with hindered myofibroblast transdifferentiation along with declines in the expression of collagen Ia1 and TGFβ1. Inflammatory cell infiltration was not affected by the loss of TRPV1. Ultrastructurally endoplasmic reticulum of TRPV1-null keratocytes was more extensively dilated as compared with WT keratocytes, suggesting an impairment of protein secretion by TRPV1-gene knockout. These results indicate that injury-related TRPV1 signal is involved in healing of stromal incision injury in a mouse cornea by selectively stimulating TGFβ-induced granulation tissue formation.
Collapse
|
14
|
Photopolymerized Microfeatures Guide Adult Spiral Ganglion and Dorsal Root Ganglion Neurite Growth. Otol Neurotol 2018; 39:119-126. [PMID: 29227456 DOI: 10.1097/mao.0000000000001622] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
HYPOTHESIS Microtopographical patterns generated by photopolymerization of methacrylate polymer systems will direct growth of neurites from adult neurons, including spiral ganglion neurons (SGNs). BACKGROUND Cochlear implants (CIs) provide hearing perception to patients with severe to profound hearing loss. However, their ability to encode complex auditory stimuli is limited due, in part, to poor spatial resolution caused by spread of the electrical currents in the inner ear. Directing the regrowth of SGN peripheral processes towards stimulating electrodes could help reduce current spread and improve spatial resolution provided by the CI. Previous work has demonstrated that micro- and nano-scale patterned surfaces precisely guide the growth of neurites from a variety of neonatal neurons including SGNs. Here, we sought to determine the extent to which adult neurons likewise respond to these topographical surface features. METHODS Photopolymerization was used to fabricate methacrylate polymer substrates with micropatterned surfaces of varying amplitudes and periodicities. Dissociated adult dorsal root ganglion neurons (DRGNs) and SGNs were cultured on these surfaces and the alignment of the neurite processes to the micropatterns was determined. RESULTS Neurites from both adult DRGNs and SGNs significantly aligned to the patterned surfaces similar to their neonatal counterparts. Further DRGN and SGN neurite alignment increased as the amplitude of the microfeatures increased. Decreased pattern periodicity also improved neurite alignment. CONCLUSION Microscale surface topographic features direct the growth of adult SGN neurites. Topographical features could prove useful for guiding growth of SGN peripheral axons towards a CI electrode array.
Collapse
|
15
|
Bessonov IV, Kotliarova MS, Kopitsyna MN, Fedulov AV, Moysenovich AM, Arkhipova AY, Bogush VG, Bagrov DV, Ramonova AA, Mashkov AE, Shaitan KV, Moisenovich MM. Photocurable Hydrogels Containing Spidroin or Fibroin. ACTA ACUST UNITED AC 2017. [DOI: 10.3103/s0096392518010030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Leigh BL, Truong K, Bartholomew R, Ramirez M, Hansen MR, Allan Guymon C. Tuning Surface and Topographical Features to Investigate Competitive Guidance of Spiral Ganglion Neurons. ACS APPLIED MATERIALS & INTERFACES 2017; 9:31488-31496. [PMID: 28841276 PMCID: PMC6341486 DOI: 10.1021/acsami.7b09258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cochlear Implants (CIs) suffer from limited tonal resolution due, in large part, to spatial separation between stimulating electrode arrays and primary neural receptors. In this work, a combination of physical and chemical micropatterns, formed on acrylate polymers, are used to direct the growth of primary spiral ganglion neurons (SGNs), the inner ear neurons. Utilizing the inherent temporal and spatial control of photopolymerization, physical microgrooves are fabricated using a photomask in a single step process. Biochemical patterns are generated by adsorbing laminin, a cell adhesion protein, to acrylate polymer surfaces followed by irradiation through a photomask with UV light to deactivate protein in exposed areas and generate parallel biochemical patterns. Laminin deactivation was shown increase as a function of UV light exposure while remaining adsorbed to the polymer surface. SGN neurites show alignment to both biochemical and physical patterns when evaluated individually. Competing biochemical and physical patterns were also examined. The relative guiding strength of physical cues was varied by independently changing both the amplitude and the band spacing of the microgrooves, with higher amplitudes and shorter band spacing providing cues that more effective guide neurite growth. SGN neurites aligned to laminin patterns with lower physical pattern amplitude and thus weaker physical cues. Alignment of SGNs shifted toward the physical pattern with higher amplitude and lower periodicity patterns which represent stronger cues. These results demonstrate the ability of photopolymerized microfeatures to modulate alignment of inner ear neurites even in the presence of conflicting physical and biochemical cues laying the groundwork for next generation cochlear implants and neural prosthetic devices.
Collapse
Affiliation(s)
- Braden L. Leigh
- Departments of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Kristy Truong
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Reid Bartholomew
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Mark Ramirez
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan R. Hansen
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
- Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - C. Allan Guymon
- Departments of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
17
|
Thomson SE, Charalambous C, Smith CA, Tsimbouri PM, Déjardin T, Kingham PJ, Hart AM, Riehle MO. Microtopographical cues promote peripheral nerve regeneration via transient mTORC2 activation. Acta Biomater 2017; 60:220-231. [PMID: 28754648 PMCID: PMC5593812 DOI: 10.1016/j.actbio.2017.07.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/04/2017] [Accepted: 07/20/2017] [Indexed: 12/16/2022]
Abstract
Despite microsurgical repair, recovery of function following peripheral nerve injury is slow and often incomplete. Outcomes could be improved by an increased understanding of the molecular biology of regeneration and by translation of experimental bioengineering strategies. Topographical cues have been shown to be powerful regulators of the rate and directionality of neurite regeneration, and in this study we investigated the downstream molecular effects of linear micropatterned structures in an organotypic explant model. Linear topographical cues enhanced neurite outgrowth and our results demonstrated that the mTOR pathway is important in regulating these responses. mTOR gene expression peaked between 48 and 72 h, coincident with the onset of rapid neurite outgrowth and glial migration, and correlated with neurite length at 48 h. mTOR protein was located to glia and in a punctate distribution along neurites. mTOR levels peaked at 72 h and were significantly increased by patterned topography (p < 0.05). Furthermore, the topographical cues could override pharmacological inhibition. Downstream phosphorylation assays and inhibition of mTORC1 using rapamycin highlighted mTORC2 as an important mediator, and more specific therapeutic target. Quantitative immunohistochemistry confirmed the presence of the mTORC2 component rictor at the regenerating front where it co-localised with F-actin and vinculin. Collectively, these results provide a deeper understanding of the mechanism of action of topography on neural regeneration, and support the incorporation of topographical patterning in combination with pharmacological mTORC2 potentiation within biomaterial constructs used to repair peripheral nerves. Statement of Significance Peripheral nerve injury is common and functionally devastating. Despite microsurgical repair, healing is slow and incomplete, with lasting functional deficit. There is a clear need to translate bioengineering approaches and increase our knowledge of the molecular processes controlling nerve regeneration to improve the rate and success of healing. Topographical cues are powerful determinants of neurite outgrowth and represent a highly translatable engineering strategy. Here we demonstrate, for the first time, that microtopography potentiates neurite outgrowth via the mTOR pathway, with the mTORC2 subtype being of particular importance. These results give further evidence for the incorporation of microtopographical cues into peripheral nerve regeneration conduits and indicate that mTORC2 may be a suitable therapeutic target to potentiate nerve regeneration.
Collapse
|
18
|
Irving M, Murphy MF, Lilley F, French PW, Burton DR, Dixon S, Sharp MC. The use of abrasive polishing and laser processing for developing polyurethane surfaces for controlling fibroblast cell behaviour. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:690-697. [PMID: 27987762 DOI: 10.1016/j.msec.2016.10.067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/03/2016] [Accepted: 10/24/2016] [Indexed: 01/02/2023]
Affiliation(s)
- Michael Irving
- General Engineering Research Institute, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Mark F Murphy
- General Engineering Research Institute, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Francis Lilley
- General Engineering Research Institute, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Paul W French
- General Engineering Research Institute, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - David R Burton
- General Engineering Research Institute, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Simon Dixon
- Biomer Technology LTD, 10 Seymour Court, Tudor Road, Manor Park, Runcorn, Cheshire, WA7 1SY, UK
| | - Martin C Sharp
- General Engineering Research Institute, Liverpool John Moores University, Liverpool, L3 3AF, UK
| |
Collapse
|
19
|
A high throughput approach for analysis of cell nuclear deformability at single cell level. Sci Rep 2016; 6:36917. [PMID: 27841297 PMCID: PMC5107983 DOI: 10.1038/srep36917] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 10/12/2016] [Indexed: 01/14/2023] Open
Abstract
Various physiological and pathological processes, such as cell differentiation, migration, attachment, and metastasis are highly dependent on nuclear elasticity. Nuclear morphology directly reflects the elasticity of the nucleus. We propose that quantification of changes in nuclear morphology on surfaces with defined topography will enable us to assess nuclear elasticity and deformability. Here, we used soft lithography techniques to produce 3 dimensional (3-D) cell culture substrates decorated with micron sized pillar structures of variable aspect ratios and dimensions to induce changes in cellular and nuclear morphology. We developed a high content image analysis algorithm to quantify changes in nuclear morphology at the single-cell level in response to physical cues from the 3-D culture substrate. We present that nuclear stiffness can be used as a physical parameter to evaluate cancer cells based on their lineage and in comparison to non-cancerous cells originating from the same tissue type. This methodology can be exploited for systematic study of mechanical characteristics of large cell populations complementing conventional tools such as atomic force microscopy and nanoindentation.
Collapse
|
20
|
Charge-balanced biphasic electrical stimulation inhibits neurite extension of spiral ganglion neurons. Neurosci Lett 2016; 624:92-9. [DOI: 10.1016/j.neulet.2016.04.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/17/2016] [Accepted: 04/28/2016] [Indexed: 11/23/2022]
|
21
|
Li S, Tuft B, Xu L, Polacco M, Clarke JC, Guymon CA, Hansen MR. Intracellular calcium and cyclic nucleotide levels modulate neurite guidance by microtopographical substrate features. J Biomed Mater Res A 2016; 104:2037-48. [PMID: 27062708 DOI: 10.1002/jbm.a.35738] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/29/2016] [Accepted: 04/05/2016] [Indexed: 02/06/2023]
Abstract
Micro- and nanoscale surface features have emerged as potential tools to direct neurite growth into close proximity with next generation neural prosthesis electrodes. However, the signaling events underlying the ability of growth cones to respond to topographical features remain largely unknown. Accordingly, this study probes the influence of [Ca(2+) ]i and cyclic nucleotide levels on the ability of neurites from spiral ganglion neurons (SGNs) to precisely track topographical micropatterns. Photopolymerization and photomasking were used to generate micropatterned methacrylate polymer substrates. Dissociated SGN cultures were plated on the micropatterned surfaces. Calcium influx and release from internal stores were manipulated by elevating extracellular K(+) , maintenance in calcium-free media, or bath application of various calcium channel blockers. Cyclic nucleotide activity was increased by application of cpt-cAMP or 8-Br-cGMP. Elevation of [Ca(2+) ]i by treatment of cultures with elevated potassium reduced neurite alignment to physical microfeatures. Maintenance of cultures in Ca(2+) -free medium or treatment with the non-selective voltage-gated calcium channel blocker cadmium or L-type Ca(2+) channel blocker nifedipine did not signficantly alter SGN neurite alignment. By contrast, ryanodine or xestospongin C, which block release of internal calcium stores via ryanodine-sensitive channels or inositol-1,4,5-trisphosphate receptors respectively, each significantly decreased neurite alignment. Cpt-cAMP significantly reduced neurite alignment while 8-Br-cGMP significantly enhanced neurite alignment. Manipulation of [Ca(2+) ]i or cAMP levels significantly disrupts neurite guidance while elevation of cGMP levels increases neurite alignment. The results suggest intracellular signaling pathways similar to those recruited by chemotactic cues are involved in neurite guidance by topographical features. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2037-2048, 2016.
Collapse
Affiliation(s)
- Shufeng Li
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa, 52242.,Department of Otolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China
| | - Bradley Tuft
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, Iowa, 52242
| | - Linjing Xu
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa, 52242
| | - Marc Polacco
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa, 52242
| | - Joseph C Clarke
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa, 52242
| | - C Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, Iowa, 52242
| | - Marlan R Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa, 52242.,Department of Neurosurgery, University of Iowa, Iowa City, Iowa, 52242
| |
Collapse
|
22
|
Hoon JL, Tan MH, Koh CG. The Regulation of Cellular Responses to Mechanical Cues by Rho GTPases. Cells 2016; 5:cells5020017. [PMID: 27058559 PMCID: PMC4931666 DOI: 10.3390/cells5020017] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The Rho GTPases regulate many cellular signaling cascades that modulate cell motility, migration, morphology and cell division. A large body of work has now delineated the biochemical cues and pathways, which stimulate the GTPases and their downstream effectors. However, cells also respond exquisitely to biophysical and mechanical cues such as stiffness and topography of the extracellular matrix that profoundly influence cell migration, proliferation and differentiation. As these cellular responses are mediated by the actin cytoskeleton, an involvement of Rho GTPases in the transduction of such cues is not unexpected. In this review, we discuss an emerging role of Rho GTPase proteins in the regulation of the responses elicited by biophysical and mechanical stimuli.
Collapse
Affiliation(s)
- Jing Ling Hoon
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Mei Hua Tan
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
- Mechanobiology Institute, Singapore 117411, Singapore.
| |
Collapse
|
23
|
Park M, Oh E, Seo J, Kim MH, Cho H, Choi JY, Lee H, Choi IS. Control over Neurite Directionality and Neurite Elongation on Anisotropic Micropillar Arrays. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1148-52. [PMID: 26395860 DOI: 10.1002/smll.201501896] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/10/2015] [Indexed: 05/20/2023]
Abstract
Control over neurite orientation in primary hippocampal neurons is achieved by using interrupted, anisotropic micropillar arrays as a cell culture platform. Both neurite orientation and neurite length are controlled by a function of interpillar distance.
Collapse
Affiliation(s)
- Matthew Park
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 305-701, South Korea
| | - Eunkyul Oh
- Department of Chemistry, College of Natural Science, Hanyang University, Seoul, 133-791, South Korea
| | - Jeongyeon Seo
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 305-701, South Korea
| | - Mi-Hee Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 305-701, South Korea
| | - Hyeoncheol Cho
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 305-701, South Korea
| | - Ji Yu Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 305-701, South Korea
| | - Haiwon Lee
- Department of Chemistry, College of Natural Science, Hanyang University, Seoul, 133-791, South Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul, 133-791, South Korea
| | - Insung S Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 305-701, South Korea
| |
Collapse
|
24
|
Cheng EL, Leigh B, Guymon CA, Hansen MR. Quantifying Spiral Ganglion Neurite and Schwann Behavior on Micropatterned Polymer Substrates. Methods Mol Biol 2016; 1427:305-18. [PMID: 27259935 DOI: 10.1007/978-1-4939-3615-1_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The first successful in vitro experiments on the cochlea were conducted in 1928 by Honor Fell (Fell, Arch Exp Zellforsch 7(1):69-81, 1928). Since then, techniques for culture of this tissue have been refined, and dissociated primary culture of the spiral ganglion has become a widely accepted in vitro model for studying nerve damage and regeneration in the cochlea. Additionally, patterned substrates have been developed that facilitate and direct neural outgrowth. A number of automated and semi-automated methods for quantifying this neurite outgrowth have been utilized in recent years (Zhang et al., J Neurosci Methods 160(1):149-162, 2007; Tapias et al., Neurobiol Dis 54:158-168, 2013). Here, we describe a method to study the effect of topographical cues on spiral ganglion neurite and Schwann cell alignment. We discuss our microfabrication process, characterization of pattern features, cell culture techniques for both spiral ganglion neurons and spiral ganglion Schwann cells. In addition, we describe protocols for reducing fibroblast count, immunocytochemistry, and methods for quantifying neurite and Schwann cell alignment.
Collapse
Affiliation(s)
- Elise L Cheng
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Braden Leigh
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - C Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Marlan R Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA. .,Department of Neurosurgery, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, 52242, USA.
| |
Collapse
|
25
|
Ren F, Zhang H, Qi C, Gao ML, Wang H, Li XQ. Blockade of transient receptor potential cation channel subfamily V member 1 promotes regeneration after sciatic nerve injury. Neural Regen Res 2015; 10:1324-31. [PMID: 26487864 PMCID: PMC4590249 DOI: 10.4103/1673-5374.162770] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The transient receptor potential cation channel subfamily V member 1 (TRPV1) provides the sensation of pain (nociception). However, it remains unknown whether TRPV1 is activated after peripheral nerve injury, or whether activation of TRPV1 affects neural regeneration. In the present study, we established rat models of unilateral sciatic nerve crush injury, with or without pretreatment with AMG517 (300 mg/kg), a TRPV1 antagonist, injected subcutaneously into the ipsilateral paw 60 minutes before injury. At 1 and 2 weeks after injury, we performed immunofluorescence staining of the sciatic nerve at the center of injury, at 0.3 cm proximal and distal to the injury site, and in the dorsal root ganglia. Our results showed that Wallerian degeneration occurred distal to the injury site, and neurite outgrowth and Schwann cell regeneration occurred proximal to the injury. The number of regenerating myelinated and unmyelinated nerve clusters was greater in the AMG517-pretreated rats than in the vehicle-treated group, most notably 2 weeks after injury. TRPV1 expression in the injured sciatic nerve and ipsilateral dorsal root ganglia was markedly greater than on the contralateral side. Pretreatment with AMG517 blocked this effect. These data indicate that TRPV1 is activated or overexpressed after sciatic nerve crush injury, and that blockade of TRPV1 may accelerate regeneration of the injured sciatic nerve.
Collapse
Affiliation(s)
- Fei Ren
- Department of Pathophysiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Chao Qi
- Department of Pathophysiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mei-Ling Gao
- Department of Pathophysiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xia-Qing Li
- Department of Pathophysiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
26
|
Liu Y, Li S. A Cell Culture Model of Latent and Lytic Herpes Simplex Virus Type 1 Infection in Spiral Ganglion. ORL J Otorhinolaryngol Relat Spec 2015; 77:141-9. [PMID: 26022499 DOI: 10.1159/000381679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/16/2015] [Indexed: 11/19/2022]
Abstract
PURPOSE OF THE STUDY Reactivation of latent herpes simplex virus type 1 (HSV-1) in spiral ganglion neurons (SGNs) is supposed to be one of the causes of idiopathic sudden sensorineural hearing loss. This study aims to establish a cell culture model of latent and lytic HSV-1 infection in spiral ganglia. PROCEDURES In the presence of acyclovir, primary cultures of SGNs were latently infected with HSV-1 expressing green fluorescent protein. Four days later, these cells were treated with trichostatin A (TSA), a known chemical reactivator of HSV-1. TCID50 was used to measure the titers of virus in cultures on Vero cells. RNA from cultures was detected for the presence of transcripts of ICP27 and latency-associated transcript (LAT) using reverse transcription polymerase chain reaction. RESULTS There is no detectable infectious HSV-1 in latently infected cultures, whereas they could be observed in both lytically infected and latently infected/TSA-treated cultures. LAT was the only detectable transcript during latent infection, whereas lytic ICP27 transcript was detected in lytically infected and latently infected/TSA-treated cultures. CONCLUSION Cultured SGNs can be both latently and lytically infected with HSV-1. Furthermore, latently infected SGNs can be reactivated using TSA, yielding infectious virus.
Collapse
Affiliation(s)
- Yuehong Liu
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, PR China
| | | |
Collapse
|