scholarly journals Interaction of micropatterned topographical and biochemical cues to direct neurite growth from spiral ganglion neurons

2021 ◽  
Author(s):  
Kristy Truong ◽  
Braden Leigh ◽  
Joseph T. Vecchi ◽  
Reid Bartholomew ◽  
Linjing Xu ◽  
...  

AbstractFunctional 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 90° 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.

2020 ◽  
Vol 2020 ◽  
pp. 1-8
Author(s):  
Na Shen ◽  
Lei Zhou ◽  
Bin Lai ◽  
Shufeng Li

Background. Cochlear implant-based electrical stimulation may be an important reason to induce the residual hearing loss after cochlear implantation. In our previous study, we found that charge-balanced biphasic electrical stimulation inhibited the neurite growth of spiral ganglion neurons (SGNs) and decreased Schwann cell density in vitro. In this study, we want to know whether cochlear implant-based electrical stimulation can induce the change of electrical activity in cultured SGNs. Methods. Spiral ganglion neuron electrical stimulation in vitro model is established using the devices delivering cochlear implant-based electrical stimulation. After 48 h treatment by 50 μA or 100 μA electrical stimulation, the action potential (AP) and voltage depended calcium current (ICa) of SGNs are recorded using whole-cell electrophysiological method. Results. The results show that the ICa of SGNs is decreased significantly in 50 μA and 100 μA electrical stimulation groups. The reversal potential of ICa is nearly +80 mV in control SGN, but the reversal potential decreases to +50 mV in 50 μA and 100 μA electrical stimulation groups. Interestingly, the AP amplitude, the AP latency, and the AP duration of SGNs have no statistically significant differences in all three groups. Conclusion. Our study suggests cochlear implant-based electrical stimulation only significantly inhibit the ICa of cultured SGNs but has no effect on the firing of AP, and the relation of ICa inhibition and SGN damage induced by electrical stimulation and its mechanism needs to be further studied.


2020 ◽  
Vol 21 (22) ◽  
pp. 8511
Author(s):  
Kiran Kumar Sriperumbudur ◽  
Revathi Appali ◽  
Anthony W. Gummer ◽  
Ursula van Rienen

Sensorineural deafness is caused by the loss of peripheral neural input to the auditory nerve, which may result from peripheral neural degeneration and/or a loss of inner hair cells. Provided spiral ganglion cells and their central processes are patent, cochlear implants can be used to electrically stimulate the auditory nerve to facilitate hearing in the deaf or severely hard-of-hearing. Neural degeneration is a crucial impediment to the functional success of a cochlear implant. The present, first-of-its-kind two-dimensional finite-element model investigates how the depletion of neural tissues might alter the electrically induced transmembrane potential of spiral ganglion neurons. The study suggests that even as little as 10% of neural tissue degeneration could lead to a disproportionate change in the stimulation profile of the auditory nerve. This result implies that apart from encapsulation layer formation around the cochlear implant electrode, tissue degeneration could also be an essential reason for the apparent inconsistencies in the functionality of cochlear implants.


2020 ◽  
Author(s):  
Chen Liu ◽  
Shu Fang ◽  
Da-xiong Ding ◽  
Han-dai Qin ◽  
Shuo-long Yuan ◽  
...  

AbstractCochlear implants (CIs) are by far the optimal option to partially restore hearing for the patients of sensorineural hearing impairment (HI) by electrically stimulating spiral ganglion neurons (SGNs). However, wide current spread from each electrode constitute an interface which restricts precision and quality of the electrical CIs. Recently, optogenetic stimulation of the cochlea has been proved as a more optimized approach via adeno-associated virus (AAV) carrying the gene encoding the light-sensitive channelrhodopsin-2. Here, we focus on summarizing recent work on stable and accurate ChR2 expression and compare the electrophysiological recording of optogenetic and acoustic stimulation in adult guinea pigs. Light stimulation generated auditory responses that was similar to that of acoustic stimulation. Moreover, normal hearing adult guinea pigs responded with a rise in amplitudes with increasing light intensity. In conclusion, optogenetic cochlear stimulation achieved good spectral selectivity of artificial sound encoding in a new adult rodent model, suggesting that the capabilities of optogenetics might be applied to improve cochlear implants in the future.


2022 ◽  
Vol 15 ◽  
Author(s):  
Li Zhang ◽  
Sen Chen ◽  
Yu Sun

Sensorineural hearing loss (SNHL) is one of the most prevalent sensory deficits in humans, and approximately 360 million people worldwide are affected. The current treatment option for severe to profound hearing loss is cochlear implantation (CI), but its treatment efficacy is related to the survival of spiral ganglion neurons (SGNs). SGNs are the primary sensory neurons, transmitting complex acoustic information from hair cells to second-order sensory neurons in the cochlear nucleus. In mammals, SGNs have very limited regeneration ability, and SGN loss causes irreversible hearing loss. In most cases of SNHL, SGN damage is the dominant pathogenesis, and it could be caused by noise exposure, ototoxic drugs, hereditary defects, presbycusis, etc. Tremendous efforts have been made to identify novel treatments to prevent or reverse the damage to SGNs, including gene therapy and stem cell therapy. This review summarizes the major causes and the corresponding mechanisms of SGN loss and the current protection strategies, especially gene therapy and stem cell therapy, to promote the development of new therapeutic methods.


2019 ◽  
Vol 2019 ◽  
pp. 1-12
Author(s):  
Xue Wang ◽  
Yuechen Han ◽  
Man Wang ◽  
Chuan Bo ◽  
Zhenbiao Zhang ◽  
...  

It has been reported that paclitaxel administration could cause sensorineural hearing loss, and Wnt activation is important for the development and cell protection of mouse cochlea. However, the effect of Wnt signaling in spiral ganglion neurons (SGNs) damage induced by paclitaxel has not yet been elucidated. In this study, we explored the effect of paclitaxel on SGNs in the mouse cochlea and the neuroprotective effects of Wnt signaling pathway against paclitaxel-induced SGN damage by using Wnt agonist/antagonists in vitro. We first found that paclitaxel treatment resulted in a degenerative change and reduction of cell numbers in SGNs and induced caspase-mediated apoptosis in SGNs. The expression levels of β-catenin and C-myc were increased, thus indicating Wnt signaling was activated in SGNs after paclitaxel treatment. The activation of Wnt signaling pathway protected against SGN loss after exposure to paclitaxel, whereas the suppression of Wnt signaling in SGNs made them more vulnerable to paclitaxel treatment. We also showed that activation of Wnt signaling in SGNs inhibited caspase-mediated apoptosis. Our findings demonstrated that Wnt signaling had an important role in protecting SGNs against paclitaxel-induced damage and thus might be an effective therapeutic target for the prevention of paclitaxel-induced SGN death.


2021 ◽  
pp. 108315
Author(s):  
Kristy Truong ◽  
Braden Leigh ◽  
Joseph T. Vecchi ◽  
Reid Bartholomew ◽  
Linjing Xu ◽  
...  

2011 ◽  
Vol 278 (1-2) ◽  
pp. 77-85 ◽  
Author(s):  
Patrick J. Atkinson ◽  
Chang-Hyun Cho ◽  
Marlan R. Hansen ◽  
Steven H. Green

Sign in / Sign up

Export Citation Format

Share Document