Open access peer-reviewed chapter - ONLINE FIRST

Neurostimulation in Neuro-Ophthalmology: Mechanisms and Therapeutic Potential

Written By

Nour Shaheen, Mohamed Khaled, Serah Seo, Yarema Bezchlibnyk, Oliver Flouty and Vishal Bharmauria

Submitted: 10 May 2024 Reviewed: 20 May 2024 Published: 01 July 2024

DOI: 10.5772/intechopen.115105

Current Concepts in Neuro-Ophthalmology IntechOpen
Current Concepts in Neuro-Ophthalmology Edited by Kemal Örnek

From the Edited Volume

Current Concepts in Neuro-Ophthalmology [Working Title]

Prof. Kemal Örnek

Chapter metrics overview

View Full Metrics

Abstract

Visual processing constitutes a substantial portion of cognitive, executive, and sensorimotor functions of the brain. Understandably, damage to visual areas and pathways results in various impairments. Neuro-ophthalmology addresses these complexities, yet traditional management approaches often have limited efficacy and undesirable side effects. In recent years, neurostimulation has emerged as a promising alternative, offering strong therapeutic benefits with minimal adverse effects. While extensively explored in neurological and psychiatric disorders, its application in ophthalmology remains relatively underexplored. This chapter navigates recent advancements in neurostimulation techniques, focusing on their potential in treating neuro-ophthalmic illnesses. We begin with an introduction to the visual system and then cover major neuro-ophthalmologic illnesses and related stimulation principles while also describing associated neurochemical and neuroplastic changes. Two major types of neurostimulation modalities in ophthalmology are discussed—invasive and non-invasive—highlighting their mechanisms and therapeutic potentials. Finally, we address current challenges, gaps, and prospects in neurostimulation research in ophthalmology in managing neuro-ophthalmic disorders.

Keywords

  • neurostimulation
  • neuro-ophthalmology
  • visual function
  • therapeutic applications
  • visual prosthetics

1. Introduction

We, like many animals, are primarily visual creatures, and processing visual information for cognition and behavior occupies a sizable section of the cerebral cortex [1]. About 30% of the cerebral cortex is dedicated to visual processing in conjunction with numerous additional non-visual areas dispersed all over the brain [2, 3, 4]. Therefore, the loss/damage of visual areas and pathways may lead to several perceptive, cognitive, spatial, memory-related, sensorimotor, and other declines in behavior [5, 6].

Neuro-ophthalmology is a special branch of medicine that deals with a variety of complex visual issues caused by neurological conditions, thus posing significant challenges to the affected individual [7]. Despite several developments in traditional management strategies (pharmacological or surgical) for various neurological disorders to control such conditions, the undesirable adverse effects and low treatment response rates have paved the way for alternative modern techniques [8, 9, 10, 11]. Neurostimulation, the process of modifying or modulating nerve activity by administering electrical stimulants directly to a specific brain region, is a rapidly growing technique with few adverse effects and strong therapeutic benefits [12]. Notably, these techniques are adaptable and customizable contingent upon the subject [13] and can be applied as a single treatment, by constantly stimulating the region (on a duty cycle with specific parameters), or in response to physiological changes [14], with applications extending to several diseases/impairments [15]. Additionally, it can also be directed at a malfunctioning brain area or network in neurological and psychiatric disorders, such as dystonia [16], obsessive-compulsive disorder [17], focal epilepsy [18], bipolar disorder [19], schizophrenia [20], chronic pain [21, 22], and depression [23].

Neurostimulation in ophthalmology is rather underexplored for its therapeutic potential. In this chapter, centered around the advances, mostly over the past few years, we navigate it as a promising prospect in neuro-ophthalmic illnesses. We first introduce different types of stimulation and the principles of stimulation (with focus on ophthalmology). We then overview the general visual perception pathway, followed by a description of some major neuro-ophthalmologic illnesses. We then discuss neurochemical changes and mechanisms along with neuroplastic changes, followed by two major types (invasive and non-invasive) of neurostimulation modulations in ophthalmology. Finally, we discuss challenges and gaps with future forecasts in ophthalmological research and applications.

Advertisement

2. Types of neurostimulation: invasive and non-invasive

Neurostimulation is classified into two types: invasive and non-invasive techniques. While the former requires the insertion of an electrode or stimulation tool inside the specific area of interest, the latter is carried out externally, making it safer and easier for applicability [24, 25], but with inadequate spatial resolution and shallow penetration [26]. The invasive techniques primarily include visual implants/prostheses and deep brain stimulation (DBS). On the other hand, the non-invasive techniques fall into several categories, such as transcorneal electrical stimulation (TES), transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), transcranial alternating current stimulation (tACS), random noise stimulation (RNS), transcranial ultrasound stimulation (TUS), and vagus nerve stimulation (VNS) [27, 28, 29].

In neuro-ophthalmology, depending on the stimulation site, stimulation falls into two categories: pre-chiasmatic and post-chiasmatic [30]. The pre-chiasmatic stimulation includes corneal (transcorneal alternating current stimulation, tcES), eyelid (transpalpebral alternating current stimulation, tpES), and peri-orbital zone (repetitive transorbital alternating current stimulation, rtACS) stimulations. These are performed to treat optic neuropathy, retinitis pigmentosa, glaucoma, retinal artery occlusion, macular degeneration, and others [31]. The post-chiasmatic stimulation comprises tDCS and high-frequency random noise stimulation (hf-RNS) to treat brain disorders, such as hemianopsia and brain damage. Notably, these are also delivered to treat pre-chiasmatic diseases, such as amblyopia and myopia [30, 32, 33].

Advertisement

3. The visual pathway: overview and organization

The visual cortex is one of the most studied areas of the brain, and we have a relatively good understanding of the architecture, morphology, physiology, and functional organization of the visual pathways across many species [34]—a fundamental premise to comprehend the outcomes of electrical stimulation of the visual cortex in relation to different eye diseases. The visual pathway (and perception) begins with the globes (eyeballs), extending all the way to the cortex.

3.1 The optic pathway

The optic pathway begins in the retina, a complex structure composed of ten distinct layers, each with its own function. The photoreceptor layers, which include rods and cones, generate action potentials through photosensitive cycles involving rhodopsin.

The ganglion cell layer and the nerve fiber layer form the basis of the optic nerve; the ganglion cell layer contains cell bodies, while the nerve fiber layer contains axons that extend across the retina [35, 36]. These axons consist of two types of fibers: temporal fibers, which control the nasal visual field, and nasal fibers, which control the temporal visual field. These fibers converge at the optic disc and exit the eye posteriorly to form the orbital portion of the optic nerve. The nerve is encased in the dura mater, continuous with that of the brain, allowing cerebrospinal fluid to move freely between the eye and the intracranial space. The axons leave the orbit through the orbital foramen along with the ophthalmic artery and sympathetic fibers [35, 36]. The retina of the eye has two types of photoreceptor cells: rods and cones that are specialized neurons with photosensitive proteins that are uniquely activated by the different wavelengths of light [37, 38]. The fovea is the part of the retina where the acuity of the image is the sharpest and only contains cones (responsible for vision in high-intensity light during the day), whereas the peripheral retina has both rods (responsible for vision in low-intensity light during the night) and cones [39]. The information is then relayed to the retinal ganglion cells (RGCs) through bipolar cells and finally reaches the optic nerve (the second cranial nerve; CN II) [36].

Figure 1A displays the parts of an eye. As the light from an object hits the cornea, it then is focused by the pupil and the lens to relay visual data from the surrounding environment onto the innermost nervous layer of the eyes, the retina — the eye’s neurosensory structure, where the image of the object is first represented in an inverted fashion [40]. Through the optic canal, the optic nerve exits the orbit and travels to the optic chiasm, where it converges with the contralateral nerve in the suprasellar cistern [41]. Figure 1B illustrates the pathway from the eye to the retina and to the cortex. Some medial axons from each optic nerve decussate (cross over) joining the lateral axons at the contralateral side [42]. In this way, the right and the left visual fields are represented in the brain; that is, the information coming from the left side of the visual scene mostly enters the right part of the brain and vice versa [43].

Figure 1.

From the eyes to the visual cortex. (A) Illustration of the anatomical structure of the eye, spanning from the cornea to the optic nerve. The purple triangle represents an object (external light source) that is first represented as an inverted image on the retina (fovea, the region of highest acuity on the retina). (B) Shows the right (red) and left (blue) visual hemifields. The decussation of the optic nerves at the optic chiasm results in the processing of the right visual field by the left hemisphere. Similarly, the left visual field is processed and represented by the right hemisphere. The visual cortex is organized into retinotopic maps, i.e., sensory space is systematically organized in the cortex. Note: This figure is an original figure created by the authors.

3.2 Retinogeniculate and retinotectal pathways

From the retina, the information is relayed to the lateral geniculate nucleus (LGN) in the thalamus (retinogeniculate pathway) or directly to the superior colliculus in the midbrain (retinotectal pathway) [44]. The optic radiations, composed of second-order neural cells, originates from the thalamus and relays the information to the visual cortex [45]. Thus, the visual information reaches the extrastriate cortex via these two separate pathways [46].

The retinogeniculate pathway (RGVP) travels through the optic nerve, crosses at the optic chiasm, continues along the optic tract, and ultimately reaches the LGN [41, 47]. Around 53–58% of RGVP fibers cross over at the optic chiasm [48, 49, 50]. This results in the RGVP being categorized into four anatomical segments: two that cross (medial) and two that do not cross (lateral), with each segment responsible for transmitting visual information from visual hemi-fields [48, 51, 52]. Studying the RGVP is crucial for various research and clinical purposes. Many diseases can impact the RGVP, such as pituitary tumors [53], glioma [54], optic neuritis [55, 56, 57], ischemic optic neuropathy [58, 59], and optic nerve sheath meningioma. Specifically, visualizing the RGVP is beneficial for planning surgical approaches to both intrinsic and extrinsic lesions of the pathway [60].

On the other hand, the retinotectal pathway through the superior colliculus processes this information to coordinate eye movements and direct attention to visual stimuli, thus playing a crucial role in visual orientation and reflexive responses [6162]. Neurostimulation of this pathway can potentially enhance or restore visual function, particularly in individuals with visual impairments. Techniques such as transcranial magnetic stimulation (TMS) or optogenetics can modulate the activity of neurons in the superior colliculus, influencing visual processing and eye movement control [24, 63, 64, 65, 66].

3.3 Visual streams: dorsal vs. ventral

Different regions of the visual cortex in the brain have specialized functions related to processing visual information [67]. The primary visual cortex (V1) in the occipital lobe is the first part of the cortex where the information arrives for primary interpretation [68]. V1 plays a crucial role in identifying spatial frequencies, orientation, and color [69]. V2, another area of the visual cortex, supplements V1’s functions by assisting in the perception of contours [70]. Area V1 receives input primarily from the lateral geniculate nucleus (LGN) and contains ocular dominance columns (ODCs) which facilitate spatial frequency recognition and contribute to binocular vision [71, 72]. The V1 comprises functional modules where neurons with similar receptive fields align into columns [73]. From V1 onwards the visual information travels parallelly in two separate [68] yet interconnected [73] pathways: the dorsal and ventral pathways (Figure 2).

Figure 2.

Dorsal and visual pathways. The information from the retina to the V1 takes two separate pathways: through LGN (retinogeniculate, orange) and through SC (retinotectal, yellow). From V1, the visual information flow is divided into the dorsal (where pathway, blue) and ventral (what pathway, red) pathways, with cross-interactions between them (magenta arrow). These pathways also interact with the frontal cortex (green arrow). This network suggests that loss/damage to any area(s) along these pathways may cause loss of multiplicity of functions: visual, cognitive, motor, and others [68, 73]. Note: This figure is an original figure created by the authors.

The ventral pathway (what) begins with V1 relaying information in a hierarchical manner to the secondary (V2) and other associative areas (V3, V4), terminating in the inferior temporal (IT) cortex [74, 75, 76]. Notably, the receptive fields (area of space that modulates neural activity) project specifically from one area to another forming contralateral retinotopic maps, thereby integrating simple information into a complex percept [74]. Conversely, the dorsal pathway (where pathway) branches into the dorsal part of the brain from V3 onwards to the medial temporal (MT, also called V5), terminating in the superior parietal lobule (SPL) [77, 78]. Collectively, as the names suggest, the ventral (what pathway) is responsible for detecting and combining different characteristics (orientation, color, spatial frequency, temporal frequency, direction, etc.) [79, 80, 81], whereas the dorsal pathway (where) encodes the spatial location and action [82, 83, 84].

Advertisement

4. Disorders of the eye

Eye disorders are classified according to the part of the eye affected. Below, we cover some major orders across both types.

4.1 Retinal disorders

Photoreceptor degeneration, which includes both structural and functional deterioration of the cones and the rods [85], leads to a remarkable reduction in the ability of the retina to perceive light, ultimately resulting in subsequent vision loss. This may end up with retinal remodeling, an event cascade that permanently changes the neural retinal structure due to an altered retinal environment [86].

Retinal remodeling is often described in three phases in which multiple cellular and molecular events occur in an attempt for injury adaptation [85, 87, 88]. The early phase of retinal remodeling begins with photoreceptor stress. The stress on these cells leads to the dislocation of photoreceptor opsins, which are light-sensitive pigments found within rods and cones (types of photoreceptor cells). This dislocation activates cellular reactions that cause the photoreceptor cells to hyperpolarize (change their electrical charge), and their outer segments move into inner adjacent retinal layers, disrupting the normal organization of the retina [85]. This disruption results in impaired transmission of visual signals, affecting vision. Photoreceptors’ death with subsequent activation of microglial cells [89] results in an even greater loss of photoreceptors [90] and bipolar deafferentation. Müller cell hypertrophy [89, 91], an enlargement or swelling of Müller cells, a type of glial cell found in the retina leads to the separation of the neuronal retinal layer from the retinal pigmented epithelium (RPE) and choroid [92]. The RPE and choroid are layers of tissue beneath the retina that provide essential support functions, including blood supply, nourishment, and immune regulation, which are crucial for the health and function of the retina. Briefly, Müller cell hypertrophy disrupts the normal relationship between the neuronal retinal layer and the supporting layers beneath it, potentially impacting the retina’s blood supply, nourishment, and immune regulation, which are essential for its proper functioning [92]. In the chronic phase, persistent remodeling with gross rewiring, de-novo axonal formation, sprouts from all the remaining neurons [93]. Combined with neuronal migration, it leads to restructuring of the retina, irreversibly altering retinal architecture and function, thus causing long-term progression of visual loss [93].

Two conditions mostly contribute to the photoreceptor degeneration: age-related macular degeneration (AMD) and retinitis pigmentosa (RP). AMD is typically acquired later in life and is characterized by the accumulation of yellowish deposits called drusen under the retina [94]. These deposits accumulate in the atrophic (degenerated) areas of the RPE, especially at the macula, the central region of the retina responsible for detailed central vision [94]. The presence of drusen and the atrophy of the RPE can lead to a progressive loss of central vision over time, as the function of the photoreceptors becomes compromised. AMD is a leading cause of vision loss in older adults and can significantly impact the quality of life. There are two main forms of AMD: dry (non-neovascular) AMD, characterized by the presence of drusen and RPE atrophy, and wet (neovascular) AMD, characterized by the growth of abnormal blood vessels beneath the retina, which can lead to sudden and severe vision loss if left untreated [95]. RP is a group of rare genetic disorders that affect the retina. These disorders typically have an early onset and may initially affect the peripheral vision before progressing toward the center of the retina [85, 96, 97]. It is associated with defects in various components of the retina, including opsins (the light-sensitive proteins found in photoreceptor cells), the RPE, or other molecules involved in retinal function [98]. Despite the specific molecular defects involved, the common outcome in RP is the degeneration of photoreceptor cells. As photoreceptor cells degenerate, individuals with RP often experience progressive vision loss, starting with night blindness and peripheral vision loss and eventually leading to central vision impairment or blindness in severe cases. The specific genetic mutations and the rate of disease progression can vary among individuals affected by RP [99].

Diabetic retinopathy is another common retinal disorder and a leading cause of blindness in diabetics [100]. Elevated sugar levels detrimentally affect the neurovascular system in the eye. This condition is categorized based on its severity. In less severe cases, known as non-proliferative diabetic retinopathy, there are observable microvascular indicators such as microaneurysms, macular edema, and hemorrhages [101]. In more severe cases, termed proliferative diabetic retinopathy, there is the growth of abnormal and fragile blood vessels, leading to significant vision impairment due to bleeding in the vitreous cavity and detachment of the vitreous [101, 102].

A class of neurodegenerative illnesses known as glaucoma is marked by damage to the optic nerve and the gradual degeneration of RGCs. The pathophysiology of all types of glaucoma involves the cupping of the optic nerve head and the death of RGCs and their axons [103]. The elevation of intraocular pressure is thought to be the primary cause of the increased apoptosis of RGCs in glaucoma, as it is caused by either increased production or decreased outflow of aqueous fluid [104].

4.2 Optic nerve disorders

Optic neuropathy is a condition caused by any damage to the optic nerve that can lead to a visual impairment(s). It encompasses a wide range of disorders with different reasons (e.g., compressive, ischemic, inflammatory, diabetic, and toxic) [105]. Ischemic optic neuropathy (ION) is a condition caused by a diminished blood supply to the optic nerve resulting in nerve injury. The ischemic insult can be anterior (at the optic disk, AION) [106] or posterior (to the optic disk, PION) [107]. ION can be further classified according to the cause into arteritic (inflammation of blood vessels like giant cell arteritis) [108] or non-arteritic (other causes) [109]. Optic neuritis (ON) is an inflammatory demyelination of the optic nerve. It is associated with autoimmune reactions resulting from either an underlying autoimmune condition (e.g., multiple sclerosis) or other factors [110, 111, 112]. The inflammation may extend to the optic chiasma (chiasmal optic neuritis) that can cause bitemporal hemianopia and impaired vision of the outer halves of both eyes [113].

Advertisement

5. The principle of neurostimulation in ophthalmology

The earliest thorough and extensive studies on electrical stimulation (ES) of the human visual cortex were pioneered by neurosurgeon Wilder Penfield [114, 115]. After individuals had direct cortical stimulation (DCS), Penfield recorded their visual perceptions or phosphenes, and he demonstrated that areas close to the occipital pole were more likely to generate visual perceptions [115].

The majority of neuromodulatory stimulation techniques rely on electricity, but ultrasound and magnetic field stimulation-based techniques also exist [116, 117]. ES is used in various neurological disorders to modulate the neuronal activity and restore the normal physiology of the dysfunctional neural networks for therapeutic purposes [118]. In ophthalmology, three main types of ES are done depending on the mechanism of action. The first category involves the replacement of a damaged area by mimicking its function allowing for downstream stimulation of the intact visual pathway (i.e., visual prostheses such as retinal implants) [119]. The second category involves the alternation of network activity restoring the normal circuit needed for proper visual function (e.g., tDCS, tACS, DBS, and rTMS) [24]. The third category involves activating neural survival pathways in a damaged part of the neurovisual system to improve visual outcomes [e.g., transcorneal alternating current stimulation (TcACS)] [120].

Numerous clinical trials have been conducted investigating the different neurostimulation devices and protocols to optimize the current intensity, electrode positioning, current frequency, and duration. This, in turn, improves the electrode design and surgical techniques for better outcomes; however, there is still no consensus regarding the standardization and optimization of ES protocols [121]. ES can be delivered to many sites along the visuo-cortical pathway. However, the retinal ES implant technique has attracted the attention of many researchers because of lower surgical complications and better accessibility [122]. There are two forms of electrical neurostimulation in use: prosthetics and non-invasive electric stimulation. While using prosthetics, the downstream visual pathways are stimulated by bypassing the less conductive dysfunctional areas [123]. In non-invasive, low-level electrical stimulation (100–1000 μA), one or more electrodes are positioned close to the eye [124, 125]. The invasive approach, however, being precise in the delivery of ES also carries the risks of infection and surgical complications [126]. The non-invasive approach has a safer profile with only mild adverse effects (e.g., itching and tingling) [127], but it has less spatial resolution [128, 129]. Thus, a combination of devices and approaches allows a trade-off giving the patient and the doctor a range of treatment options.

Advertisement

6. Electrical stimulation in degenerative ophthalmic disorders: neurochemical, immunomodulatory, neuroplastic, and other changes in

Multiple basic science studies have explored the cellular and molecular mechanisms of ES (especially the minimally invasive approaches), explaining the improvement of visual outcomes in a variety of eye diseases [31]. Ample evidence from molecular and electrophysiological studies advocates for this technique for several visual disorders. We highlight specific neurostimulation mechanisms in some inherited and acquired eyes diseases:

6.1 Neurochemistry and immunomodulation: neuroprotective roles in different retinal diseases

The widely accepted explanation for the neuroprotective benefits is that ES may both increase the intrinsic sensitivity of neurons to endogenous neurotrophic factors and up-regulate their expression levels. For example, ES-induced Müller cells’ mRNA levels and proteins for vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and insulin-like growth factor-1 (IGF-1) increase noticeably [130, 131, 132, 133, 134, 135]. Hanif et al. [136] found an upregulation of gene expression of BDNF, caspase 3, (FGF2), and glutamine synthetase (GS) after a 30-min low-level (4 μA) electrical stimulation to a rodent model with RP. These are important factors for triggering the intrinsic survival system [137, 138, 139, 140, 141], thus preventing the systems against damage [142, 143]. Specifically, Kanamoto et al. [144] identified an upregulation of 25 proteins mediating retinal function through signaling, immunomodulatory effects, and axonal and dendritic regenerative processes [135].

Similarly other apoptotic genes are downregulated [143] when applied direct ES to Müller cell culture [132]. Conclusively, several studies point to the fact that BDNF plays a central role in neuroprotection and is upregulated during ES [133, 143, 145]. Moreover, authors [132, 146, 147] have also shown that ES on axotomized RGCs of the optic nerve (invasive/non-invasive) led to an enhanced survival through activation of neuroprotection mechanisms including IGF-1. ES has also shown a rise in GTPase levels modulating calcium signaling and enhancing directed RGC axon growth toward the cathode electrode in a rodent model [148, 149].

Furthermore, it has been shown that ES plays a role in limiting the inflammatory response in some eye diseases, thus playing a significant role in immunomodulation [150]. It interferes with the activation of inflammatory cells and reduces the production of pro-inflammatory cytokines [151, 152]. Microglial cells, the most abundant CNS immune cells, responsible for the innate immunity of the CNS and injury repair are involved in inflammatory response modulation [153, 154]. Notably, depending on the condition, microglial activation may be beneficial or damaging in a variety of brain and retinal disorders [155]. Retinal microglial cells get activated after ocular insults mediating local inflammatory response and triggering pro-inflammatory signaling. Tumor necrosis factor-alpha (TNF-α) is the most prominent cytokine expressed by the activated microglial cells that is responsible for harmful neuroinflammatory and neurodegenerative processes by acting on its receptor (TNFR1) [156, 157, 158]. Additionally, it mediates the activation of nuclear factor kappa B (NF-κB) and c-Jun N-terminal kinase (JNK) pathway, further reactivating the glial cells and production of neurotoxic pro-inflammatory cytokines, eventually causing the loss of RGCs [159, 160, 161, 162]. After TES treatment in rodent models, a significantly lower number of activated microglial cells were reported [120, 145]. This was associated with a decline in pro-inflammatory cytokines expression (IL-6, TNF-α, and COX-2) along with suppression of the NF-κB pathway and upregulation of anti-inflammatory cytokines, IL-10. Similarly, the application of transpalpebral electrical stimulation (TpES) has shown a significant decrease in activated microglial cells with prolonged photoreceptor survival in inherited photoreceptor degeneration mice [163].

6.2 Promotion of neuroplasticity

A fundamental property of neurons is their capacity to reorganize the synaptic network and the strength and efficacy of synaptic transmission through a diverse number of activity-dependent (e.g., when learning a new skill, practice, or forming memory) [164, 165, 166] or training-induced (adaptation, deprivation, and environmental enrichment) [165, 167] mechanisms. These mechanisms are characterized by synaptic redistributions and synaptic scaling, typically referred to as synaptic plasticity [120, 158, 168]. Interestingly, recent research has shown evidence of improvement in visual function after damage to the visual system in response to electrical stimulation, either structurally through neuronal regeneration or functionally via modulating network connectivity and electrophysiological parameters [169, 170, 171, 172, 173]. ES techniques, such as retinal prostheses or TcACS, aim to bypass damaged photoreceptors and directly stimulate surviving retinal cells or higher visual processing centers. By delivering electrical impulses to the retina or the visual cortex, these approaches can induce neuroplastic changes, including synaptic remodeling and functional reorganization, to enhance visual perception and restore vision [174, 175]. Additionally, ES may facilitate the integration of artificial visual inputs with residual visual pathways, promoting adaptation and improving visual outcomes in individuals with various eye diseases [176]. In summary, by enhancing synaptic efficacy and facilitating the formation of new connections, ES can aid in restoring function after neurological damage or promoting skill acquisition and cognitive enhancement in healthy individuals.

6.3 Enhancement of retinal blood flow

The retina is one of the most oxygen-requiring tissues in the human body and maintaining efficient oxygen circulation to the retina is imperative for optimal visual function [177, 178, 179]. Studies have shown that ES can induce blood flow into the retina. Evidence comes from different kinds of electric treatment, such as transcorneal alternating current stimulation (tcACS) and TES, revealing their positive effect on chorioretinal blood flow in healthy individuals [180] and people with eye conditions such as RP [181, 182]. This increased blood flow comes (without any effects on the systemic blood flow) through adjustments in the vascular tone, thus influencing the neurovascular system, but the exact mechanism is not yet well-known [183].

Some studies suggest an upregulation of some growth factors, such as vascular endothelial growth factors (VEGF), which are responsible for the increased chorioretinal blood flow [184, 185]. Other studies point toward ES-induced upregulation of arachidonic acid metabolites and neurotrophic factors released by Müller cells [132149, 186, 187]. Additionally, a study has reported the role of nitric oxide (induced by IGF-1 released by activated Müller cells) in mediating vasodilatory effects [188]. In summary, ES leads to increases in blood flow to the eye without the need for surgeries.

Advertisement

7. Electrical stimulation in neuro-ophthalmology: invasive and non-invasive

Several neurostimulatory approaches have evolved over the last few decades. Initially, invasive techniques were developed during the second half of the last century as promising devices for visual field defects [115, 119]. However, in the early 2000s, non-invasive ES offered an easier method with fewer complications when applied with low current tDCS to the visual cortex, thus inducing phosphenes [189].

7.1 Invasive visual stimulation (visual prostheses/bionic eye)

Invasive stimulation approaches are silicon-based bioelectronic devices that help restore sight only in conditions with limited damage along a specific point of the visual pathway while the rest of the visual system is intact. Figure 3 highlights these visual areas. These are composed of an electrode array (a group of electrodes in a grid) producing a sequence of tiny electrical pulses from images taken using an external tiny video camera worn like glasses. Anywhere along the visual pathway, this device can be implanted to provide neural ES, including the retina, the optic nerve, the visual cortex, and the thalamus. Visual restoration using prosthetics is often referred to as “artificial vision”.

Figure 3.

Visual pathway and different visual areas that can be electrically stimulated for prosthetic applications, thus restoring vision. Note: This is a tractography image generated by us; however, the display of the retina (brown) is an artistic rendering of the eyeball.

The underlying principle of artificial vision devices mainly stems from neurophysiologic principles of the visual pathway. One principle is that the electric stimulus can serve as a substitute for light leading to visual perception [115]. Another principle underlines that prosthetic replacement of the impaired hub/point in the visual pathway fills the functional/structural hole left by impaired areas, thus subserving the remaining intact pathway [190]. Additionally, the retinotopic organization of the visual cortex allows for better localization of points for electrical stimulations to produce rational visual percepts [190].

7.1.1 Retinal prostheses

Retinal prostheses are designed for the damaged photoreceptors of retina and mimic the activity of photoreceptors by converting light into electrical impulses, thus activating ablated/silent downstream neural pathways in degenerative diseases such as RP or AMD [191]. Patients can then see their environment (almost like a grid) when phosphenes of different strengths are saliently triggered in different areas of their retinotopic map [192]. In retinal prostheses, triggering (direct or indirect) the ganglion cells is the key objective of ES [193, 194]. In direct stimulation, the ganglion cells are directly activated without any presynaptic impulses from other retinal cells, such as bipolar cells, that send synaptic projections to the RGCs [195, 196]. Somatic stimulation can be achieved by triggering the cell bodies [197, 198], whereas axonal stimulation activates the axons of the ganglion cells that are right under the electrode [199]. On the other hand, the indirect stimulation method entails the activation of other cells in the retina, namely the bipolar cells, amacrine cells, or horizontal cells [200, 201]. These cells then excite the ganglion cells, thus ameliorating the visual condition by bridging the broken pathway. Table 1 summarizes the main eye diseases and their ES techniques.

TechniqueFunctionalityImplantation SitesComponentsBenefitsLimitations
I. Invasive
Retinal prostheses for RP, AMD [202, 203, 204, 205]Mimic photoreceptors’ activity by converting light into electrical impulses, activating downstream neural pathways, and replacing damaged photoreceptors in degenerative diseases, such as RP or AMD.Epiretinal, subretinal, suprachoroidal locationsElectrode arrayRestoration of visual perception in degenerative diseases. Higher resolution and specificity with direct stimulation methods.Delicacy of retinal structure. Ongoing degeneration in visual pathway. Engineering challenges for high-resolution vision.
Optic nerve prosthesis for RP [206, 207]Stimulating optic nerve to bypass damaged retina.
Penetrating electrodes provide more focused stimulation.
Extracranial part of optic nerveSpiral cuff electrodes, penetrating electrodesPerception of phosphenes across broad visual field.Less likelihood to impair retinal synaptic processes.
Surgical challenges.
Lateral geniculate nucleus prosthesis for optic neuropathies, retinal degenerative diseases, and eye trauma [208, 209]Stimulate LGN to restore vision, exploiting its spatial distribution of receptive fields and structural organization.Lateral geniculate nucleusMicroelectrode arrayLess-density electrode array with wider receptive fields. Potential for selective layer stimulation.Complex surgical procedure.
Proper synchronization between LGNs required.
Visual cortex prostheses for various visual impairments [210, 211, 212]Stimulate primary visual cortex with penetrating electrode arrays for high-resolution artificial vision.Primary visual cortexPenetrating electrode arraysHigher resolution due to the large surface area of visual cortex. Alternative option for other ineffective prostheses.Challenges in biocompatibility. Ongoing trials for enhanced encoding. Tolerance by host tissue.
II. Noninvasive
TES for optic and retinal disorders [213, 214, 215, 216, 217]Electrical stimulation via silver thread DTL electrodes or contact lens on the cornea. Reference electrode placed under the skin.CorneaSilver-thread DTL electrodes or contact lens, reference electrodeImproved visual function outcomes in clinical trials. Neuroprotective and therapeutic potential on retinal disorders.DTL electrodes may cause corneal complications and require careful placement.
rtACS for optic nerve injuries [169, 218, 219, 220, 221]Delivery of alternating currents through electrodes near supraorbital and infraorbital ridges. Entrainment of brain oscillations to external force.Supraorbital and infraorbital ridgesMultichannel device delivering sinusoidal pulses through four separate periorbital electrodes.Enhanced visual outcomes observed in optic nerve injuries. Modulation of oscillatory brain activity, synchronization, and synaptic plasticity.May cause less corneal complications compared with TcES.
TpES for AMD (TpES) [222, 223, 224].Application of electrodes on the eyelid, minimizing direct contact with the cornea.EyelidElectrodes on the eyelidEffective in treating AMD. Minimizes the risk of local eye complications. Helps lower IOP in primary open-angle glaucoma (POAG)Limited to treating certain eye conditions.
LIFUS for various eye disorders
[225, 226, 227].
Noninvasive stimulation of neurons using low-intensity focused ultrasound.Retina, visual cortexLow-intensity focused ultrasoundHigher spatial resolution and deeper penetration. Stimulation of RGCs or visual cortex with potential for vision restoration.Not yet approved for clinical use. Limited studies on safety and efficacy in humans. Requires development and validation.
tRNS on the visual cortex
[228, 229, 230]
A biphasic sinusoidal alternating current with weak intensity (in mA) in unpredictable random frequencies is applied transcranially targeting the visual cortexPrimary visual cortexTwo electrodes, the active is placed over the visual cortexPromotes visual perceptual learning in patients with central visual lossMechanism of action is not known, thus limiting the ability to improve the protocol and design

Table 1.

Summary of the techniques, principles, mechanisms, targeted disorders, implantation sites, components, benefits, limitations, and devices associated with various methods of sight restoration, including invasive (retinal prostheses, optic nerve prosthesis, lateral geniculate nucleus prosthesis, and visual cortex prostheses) and non-invasive [transcorneal electrical stimulation (TES), repetitive transorbital alternating current stimulation (rtACS), transpalpebar current stimulation (TpES), low-intensity focused ultrasound (LIFUS), and transcranial random noise stimulation (tRNS)].

Abbreviations: VP or BE: Visual Prostheses or Bionic Eye, RP: Retinitis Pigmentosa, AMD: Age-Related Macular Degeneration, RGC: Retinal Ganglion Cells, DTL: Dacryolingostatoplasty, RPE: Retinal Pigment Epithelium, LGN: Lateral Geniculate Nucleus, TES: Transcorneal Electrical Stimulation, rtACS: Repetitive Transorbital Alternating Current Stimulation, TpES: Transpalpebar Current Stimulation, LIFUS: Low-Intensity Focused Ultrasound, tRNS: Transcranial Random Noise Stimulation.

The placement of a retinal prosthetic may be done is a subject-specific fashion. The electrode array can be placed in epiretinal, subretinal, or suprachoroidal locations. The epiretinal prosthesis is placed directly above the ganglion cells on the inner surface of the retina [e.g., Argus-II Retinal Prosthesis [202] and IMI retinal implant [231]]. It requires an intact inner retinal layer and thus comes with a limitation for extensive retinal degeneration [232, 233, 234]. Notably, surgical complications like retinal damage may occur. The subretinal prosthesis [e.g., Alpha IMS [203] and Prima System implant [204]] is implanted between the retina and RPE. It has a lower risk of retinal damage but a higher risk of retinal detachment [235, 236]. Notably, both epiretinal and subretinal devices are closer to RGCs, thereby providing them direct stimulation that results in higher resolution and specificity [237]. In contrast, the suprachoroidal (STS) implant is placed between the sclera and the choroid, preserving the structure of the retina with lower propensity for surgical complications, such as retinal detachment. However, the indirect stimulation of RGCs leads to a coarser resolution [238]. Moreover, suprachoroidal prostheses have greater stimulation thresholds, raising the possibility of retinal injury [239].

Retinal implants have multiple limitations, mainly causing damage to the retinal structure while planting the electrodes. Additionally, despite technological advancements in retinal implants, the ongoing degeneration throughout the visual pathway may limit the potential of visual prostheses to restore visual perception, because retinal implants are only replacement options [240, 241]. In other words, they do not halt the progression of the ongoing degeneration that may extend from the photoreceptors to the RGCs [94, 242, 243]. A substantial portion of the pathway should still be intact for retinal devices to send decipherable and meaningful electrical impulses. Various engineering challenges in obtaining a high-resolution artificial vision may further limit retinal prosthetics [244, 245, 246].

7.1.2 Optic nerve prosthesis

Optic nerve stimulation is an intriguing method for visual restoration. It is less likely to impair the intricate synaptic processes of the retina, as the electrodes (spiral cuff and penetrating) are implanted far from the retina at the extracranial part of the optic nerve [247]. The spiral cuff electrodes wrap around the optic nerve, thus providing a greater surface area of stimulation, albeit less selectivity, thus requiring a higher number of electrodes for stimulations [248, 249, 250, 251]. The penetrating electrodes, on the other hand, can deliver more focused stimulation [252]. Multiple studies have investigated the direct stimulation of the optic nerve in patients with RP bypassing the damaged retina. Whether using a spiral cuff or penetrating electrodes, the optic nerve stimulation results in the perception of phosphenes across a broad area of the visual field [206, 253, 254, 255, 256, 257, 258].

While optic nerve prostheses hold promise for restoring vision in individuals with optic nerve damage, the technology is still in the experimental stages and faces several challenges, including achieving sufficient visual acuity, ensuring biocompatibility and long-term stability of the device, and developing effective methods for integrating the prosthesis with the brain’s neural circuits. Research in this area is ongoing, with the goal of improving the efficacy and accessibility of optic nerve prostheses for individuals with vision loss.

7.1.3 Lateral geniculate nucleus prosthesis

The LGN is a thalamic structure that receives input from the retina and delivers the visual data to the V1 [259]. It is an interesting site for visual restoration and artificial vision in irreversibly blind patients due to optic neuropathies or retinal degeneration diseases or even in blindness caused by eye trauma [260, 261]. However, it is a more complex surgical procedure but has advantages over other methods. The LGN is a compact structure with a distinct spatial distribution of receptive fields [262], allowing for a less-density electrode array, thus enabling the capture of a broader spectrum of receptive fields. Moreover, anatomically, the LGN is structurally divided into several layers. Magnocellular layers make up the inner two layers (1 and 2), whilst parvocellular layers make up the outer four layers (3, 4, 5, and 6). Additionally, koniocellular layers are filled between them. Since these layers are sensitive to specific features of the incoming visual data (e.g., color or motion perception), they allow more specific stimulation of the downstream layers. Importantly, two separate electrodes in each LGN are needed with precise synchronization for coherent information transfer. Currently, there is an ongoing effort to develop an implant for the LGN, with preliminary results that are very promising [208]. Indeed, results of recent research have confirmed that LGN microstimulation can cause phosphenes and predictable visual percepts [208, 263, 264, 265, 266].

7.1.4 Visual cortex prostheses

As mentioned above, the V1 is the entry point of visual information in the visual cortex and the final option for vision restoration using stimulation [267]. Unlike the LGN, the visual cortex has a large surface area allowing for a higher number of electrodes, thereby enhancing resolution [268, 269, 270]. Visual cortex prostheses bypass damaged areas along most of the visual system pathway, thus providing a potential treatment option for the blind individuals [271, 272] when the retinal/optic nerve/LGN prostheses prove ineffective. Initially, research efforts in the latter half of the twentieth century were aimed at creating artificial vision in blind individuals using surface electrodes, but without any effectiveness for the completely blind individuals [273, 274, 275]. With major advancements in technologies since then, the shift to penetrating electrode arrays has reignited the hope for profoundly blind [276]. Penetrating electrode arrays offer a closer positioning to target neurons with many small electrodes introducing very low currents. This allows offering more precise and discrete phosphene production, but with challenges of biocompatibility and tolerance by the host tissue [277]. Currently, there are active ongoing trials on developing cortical prostheses with enhanced encoding and integration systems [210, 278, 279, 280].

7.2 Non-invasive neurovisual microstimulation techniques

Unlike invasive techniques, non-invasive stimulation not only offers fewer surgical complications but has also been proven to play a neuroprotective role in multiple retinal disorders [281]. These involve introducing an active electrode on the eyelids (transpalpebral or transdermal) or cornea (transcorneal) or around the orbit (periorbital) to deliver electrical stimulation with adjustments for tailoring the parameters (wavelength, frequency, and intensity), contingent upon the individual and their respective phosphene thresholds [282, 283].

7.2.1 Transcorneal electrical stimulation (TES)

It is currently the most frequent type used in retinal diseases [281], involving sterile silver-thread Dawson, Trick, and Litzkow (DTL) electrodes or contact lens as an active electrode placed on the cornea. Although, these electrodes cause fewer corneal complications like abrasion, but require more care for maintaining a proper placement on the cornea [284, 285, 286]. A reference electrode is placed underneath the skin near the active electrode. Multiple clinical trials have shown improved visual function outcomes in patients with different optic and retinal disorders after TcES application [213, 214, 215, 216, 217]. Also, TcES has been reported effective in cases of RP improving visual acuity and field in basic [287, 288] and clinical studies [289]. Additionally, retinal artery occlusions have shown improvement in multifocal electroretinograms (mfERGs) restoring the amplitude of the N1–P1 waves in the ischemic loci [290, 291]. The biological mechanisms by which TcES acts on the neural tissue helping survival and regeneration were mentioned in Section 6.

Interestingly, the effect of TcES is not confined to the retina or optic nerve as it has shown a neuromodulatory effect on the brain by shifting the altered (due to injury/damage) electrophysiological activity in a more normal range for visual [172173292] function. It is suggested that the visual impairment in blind individuals due to a prechiasmatic lesion is not only related to the primary tissue injury but also to the impaired synchrony and loss/changes of functional connectivity in certain brainwaves (associated with visual perception) [169, 173, 293]. Moreover, prolonged TcES has shown activation of network connectivity and persistent enhancement in the theta, alpha, and beta brainwave synchronization in a retinal degeneration model in both visual and non-visual areas of the brain (V1 and prefrontal cortex) [293, 294, 295]. Xie et al. [296] have found an activation of primary and secondary visual cortices measured by positron emission tomography (PET) and 18 F-fluorodeoxyglucose (FDG) using either DTL or ERG-Jet (contact lens) electrodes. The DTL electrodes resulted in the activation of the V2, while ERG-Jet activated both the V1 and V2, engendering brighter phosphene production.

7.2.2 Repetitive transorbital alternating current stimulation

Repetitive transorbital alternating current stimulation (rtACS) involves the delivery of alternating electrical currents through electrodes placed near supraorbital and infraorbital ridges [297]. This involves a multi-channel device that delivers sinusoidal pulses of oscillatory current through four separate periorbital electrodes bilaterally: two supraorbital and two infraorbital. This method differs from the former in that it makes less contact with the cornea, further lowering the risk of complications like abrasions, dryness, and discomfort [289, 298].

The nature of repetitive stimulation helps in the entrainment of desynchronized brain oscillations, with a periodic application of the externally forced stimulation [299]. Again, the adjustment of the stimulation parameters protocol can be done according to different pathological conditions. It has shown enhanced visual outcomes (visual acuity, visual field, and detection) in optic nerve injury patients [174218, 300, 301]. The suggested mechanism of action involves the modulation of oscillatory brain activity, as evidenced by observed alterations in alpha-band brainwave activity, resulting in synchronization [219]. By targeting the visual system through the orbit, rtACS has the potential to modulate neuronal activity and synaptic plasticity in visual pathways. Enhanced neuroplasticity and network connectivity have been shown on EEG along with resynchronization of the alpha bands reported in recent trials [169, 218, 219, 220, 221]. Moreover, Granata and Falsini [302] have observed subjective clinical improvements in a heterogeneous group of various chronic low vision pathologies, associated with an increase in the amplitude of visually evoked potentials (VEP) after the application of rtACS.

7.2.3 Transpalpebral current stimulation

Transpalpebral current stimulation (TpES) involves the application of electrodes on the eyelid, limiting direct contact with the cornea, thereby minimizing the risk of local eye complications like eye discomfort and mucin homeostasis disruption, a lubricating agent on the corneal surface that prevents eye dryness [298]. TpES has the added benefit of lowered IOP, in addition to the similar mechanisms in other methods. In primary open-angle glaucoma (POAG) patients, it acted akin to tyrosine kinase inhibitors and activated maxi-K+ channel in the trabecular meshwork (TM) leading to relaxation of TM lowering the resistance to aqueous humor outflow, which in turn lowered the high intraocular pressure [303]. TpES has been proven to be most effective in treating individuals with AMD [222, 223, 224].

7.2.4 Low-intensity focused ultrasound

Low-intensity focused ultrasound (LIFUS) is an emerging neuromodulatory non-invasive method for sight restoration [304, 305, 306, 307]. Numerous studies have consistently shown that FUS effectively stimulates neurons both in laboratory settings, including in-vitro [225, 308], ex-vivo [309, 310], and in-vivo [226, 311] methods. It offers an advantage over other non-invasive techniques like tDCS and TMS by providing a higher spatial resolution (at μm levels) and deeper penetration (at cm levels). It is delivered in low intensity to the nervous system inducing excitatory actions while not causing a neuronal injury [312, 313, 314]. The mechanical energy from the ultrasound waves induces various biological and cellular effects such as cavitation and mechanosensitive ion channel activation [315]. For vision, the LIFUS can help in vision restoration in two ways, either by retinal stimulation or visual cortex stimulation. Recently, retinal non-invasive ultrasound prostheses have been developed for retinal stimulation. However, it has not been approved in clinical settings yet, the basic studies have shown the ability to safely stimulate retinal ganglion cells with different patterns in retinal degeneration models producing phosphenes [225, 226, 227].

An alternative method for using ultrasound in visual restoration involves non-invasively stimulating the visual cortex through transcranial-focused ultrasound. This stimulation leads to temporary modulation of the visual cortex [316, 317, 318, 319]. The parameters of the ultrasound waves have different types of cortical responses. The pulsed waves had a prolonged effect than the continuous waves.

7.2.5 Transcranial random noise stimulation (tRNS)

Another recently emerging non-invasive electrical stimulation technique is transcranial random noise stimulation (tRNS) [320]. A biphasic sinusoidal alternating current with weak intensity (in mA) in unpredictable random frequencies is applied transcranially using two electrodes, either in low-frequency spectrum (<100 Hz) (lf-RNS) or high-frequency spectrum (>100 Hz) (hf-tRNS) [321]. The latter has proven superiority in its neuromodulatory potential [228, 322, 323]. tRNS has the same effect as other non-invasive techniques concerning the modulation of cortical oscillatory activity; however, the exact mechanism is still not well-known [320, 324, 325]. Few studies have shown that the repetitive modulation of voltage-gated sodium channels may explain the results of enhanced neuroplasticity [326, 327, 328]. However, it delivers an alternating electrical current like other techniques such as tACS, and it has proven to be more effective compared with them [329, 330]. This can be explained by the stochastic resonance phenomenon which suggests that delivering noisy stimulation (random frequency current) can amplify the neuron’s sensitivity to weak stimuli [331]. The application of tRNS at the visual cortex promotes visual learning, a rehabilitative approach for functional recovery of central vision loss [332, 333], in both healthy [334, 335, 336] and visually impaired individuals with diseases such as myopia [229], amblyopia [230], and macular degeneration [337].

Advertisement

8. Conclusion

Preclinical research and clinical trials collectively demonstrate that neurostimulation holds promise in enhancing retinal sensitivity, improving visual function, and potentially restoring lost vision. By targeting specific regions of the visual pathway, neurostimulation approaches seek to promote neuronal regeneration and plasticity while avoiding dysfunctional or damaged areas. Despite its potential, neurostimulation faces several challenges and limitations. These include the need for further optimization of stimulation parameters, standardization of protocols, long-term safety, and efficacy assessments, and ensuring broad accessibility of the technology for clinical use. Moreover, the heterogeneity of visual disorders and varied responses to neurostimulation underscore the importance of personalized treatment plans and ongoing research efforts.

Furthermore, researchers are exploring innovative stimulation techniques beyond traditional electrical stimulation, such as optogenetics, magnetic stimulation, and ultrasound stimulation, aiming for more precise and targeted modulation of neural activity within the visual system [338, 339, 340, 341]. Advancements in imaging technologies, such as functional magnetic resonance imaging (fMRI) and optical coherence tomography (OCT), can improve the precise targeting and localization of neurostimulation within the visual pathways, leading to more effective and personalized treatment approaches [342, 343]. Development of closed-loop neurostimulation systems for neuromodulations, which dynamically adjust stimulation parameters based on real-time feedback, may optimize treatment outcomes. Integration of neurostimulation with other therapeutic modalities, such as pharmacotherapy or gene therapy, may synergistically enhance treatment efficacy and promote neuroprotection or neuroregeneration. Continued refinement of non-invasive neurostimulation techniques offers safer and more accessible alternatives for modulating visual cortical excitability. Bridging the gap between preclinical research findings and clinical implementation, along with prioritizing patient-centered outcomes research, is crucial for the widespread adoption of neurostimulation therapies in neuro-ophthalmology and improving the lives of affected individuals.

Advertisement

Contribution and acknowledgements

NS contributed to conceptualization and drafted the original version of manuscript. MK also contributed to drafting the original version of manuscript. SS finalized the figures and contributed to editing. YB, OF, and VB contributed to writing and editing of the manuscript. This research was supported by the University of South Florida (USF) seed grant and the support of the Department of Neurosurgery and Brain Repair, USF.

References

  1. 1. Grossman ED, Blake R. Brain areas active during visual perception of biological motion. Neuron. 2002;35(6):1167-1175
  2. 2. Sells SB, Fixott S. Evaluation of research on effects of visual training on visual functions. American Journal of Ophthalmology. 1957;44(2):230-236
  3. 3. Werner JS, Chalupa L. The Visual Neurosciences. Cambridge, MA: MIT Press; 2004
  4. 4. Erichsen JT, Woodhouse JM. Human and animal vision. In: Batchelor BG, editor. Machine Vision Handbook. London: Springer London; 2012. pp. 89-115
  5. 5. Varadaraj V, Munoz B, Deal JA, An Y, Albert MS, Resnick SM, et al. Association of vision impairment with cognitive decline across multiple domains in older adults. JAMA Network Open. 2021;4(7):e2117416
  6. 6. Nagarajan N, Assi L, Varadaraj V, Motaghi M, Sun Y, Couser E, et al. Vision impairment and cognitive decline among older adults: A systematic review. BMJ Open. 2022;12(1):e047929
  7. 7. Lee AG, Sinclair A, Sadaka A, Berry S, Mollan S. Neuro-Ophthalmology: Global Trends in Diagnosis, Treatment and Management. Cham: Springer International Publishing; 2019
  8. 8. Rissardo JP, Vora NM, Tariq I, Mujtaba A, Caprara ALF. Deep brain stimulation for the management of refractory neurological disorders: A comprehensive review. Medicina (Kaunas). 2023;59(11):1991
  9. 9. Giacobbe P, Burhan AM, Waxman R, Ng E. Interventional psychiatry and neurotechnologies: Education and ethics training. The Canadian Journal of Neurological Sciences. 2023;50(s1):s10-s16
  10. 10. Jiang W-L, Cai D-B, Sun C-H, Yin F, Goerigk S, Brunoni AR, et al. Adjunctive tDCS for treatment-refractory auditory hallucinations in schizophrenia: A meta-analysis of randomized, double-blinded, sham-controlled studies. Asian Journal of Psychiatry. 2022;73:103100
  11. 11. Steuber ER, McGuire JF. A meta-analysis of transcranial magnetic stimulation in obsessive-compulsive disorder. Biological Psychiatry. Cognitive Neuroscience and Neuroimaging. 2023;8(11):1145-1155
  12. 12. Zohuri B, McDaniel P. Electrical brain stimulation to treat neurological disorders. In: Transcranial Magnetic and Electrical Brain Stimulation for Neurological Disorders. New York, NY, USA: Academic Press; 2022. pp. 267-302
  13. 13. Medaglia JD, Erickson B, Zimmerman J, Kelkar A. Personalizing neuromodulation. International Journal of Psychophysiology. 2020;154:101-110
  14. 14. Doucet BM, Lam A, Griffin L. Neuromuscular electrical stimulation for skeletal muscle function. The Yale Journal of Biology and Medicine. 2012;85(2):201-215
  15. 15. Amengual JL, Vernet M, Adam C, Valero-Cabré A. Local entrainment of oscillatory activity induced by direct brain stimulation in humans. Scientific Reports. 2017;7:41908
  16. 16. Bari AA, Thum J, Babayan D, Lozano AM. Current and expected advances in deep brain stimulation for movement disorders. Progress in Neurological Surgery. 2018;33:222-229
  17. 17. Bergfeld IO, Dijkstra E, Graat I, de Koning P, van den Boom BJG, Arbab T, et al. Invasive and non-invasive neurostimulation for OCD. Current Topics in Behavioral Neurosciences. 2021;49:399-436
  18. 18. Li MCH, Cook MJ. Deep brain stimulation for drug-resistant epilepsy. Epilepsia. 2018;59(2):273-290
  19. 19. Hsu C-W, Chou P-H, Brunoni AR, Hung K-C, Tseng P-T, Liang C-S, et al. Comparing different non-invasive brain stimulation interventions for bipolar depression treatment: A network meta-analysis of randomized controlled trials. Neuroscience and Biobehavioral Reviews. 2024;156:105483
  20. 20. Corripio I, Roldán A, Sarró S, McKenna PJ, Alonso-Solís A, Rabella M, et al. Deep brain stimulation in treatment resistant schizophrenia: A pilot randomized cross-over clinical trial. eBioMedicine. 2020;51:102568
  21. 21. Szymoniuk M, Chin J-H, Domagalski Ł, Biszewski M, Jóźwik K, Kamieniak P. Brain stimulation for chronic pain management: A narrative review of analgesic mechanisms and clinical evidence. Neurosurgical Review. 2023;46(1):127
  22. 22. Shaheen N, Shaheen A, Elgendy A, Bezchlibnyk YB, Zesiewicz T, Dalm B, et al. Deep brain stimulation for chronic pain: A systematic review and meta-analysis. Frontiers in Human Neuroscience. 2023;17:1297894
  23. 23. Delaloye S, Holtzheimer PE. Deep brain stimulation in the treatment of depression. Dialogues in Clinical Neuroscience. 2014;16(1):83-91
  24. 24. Bhattacharya A, Mrudula K, Sreepada SS, Sathyaprabha TN, Pal PK, Chen R, et al. An overview of noninvasive brain stimulation: Basic principles and clinical applications. The Canadian Journal of Neurological Sciences. 2022;49(4):479-492
  25. 25. Xu S, Wang W, Chen S, Wu Q , Li C, Ma X, et al. Deep brain stimulation complications in patients with Parkinson’s disease and surgical modifications: A single-center retrospective analysis. Frontiers in Human Neuroscience. 2021;15:684895
  26. 26. Liu X, Qiu F, Hou L, Wang X. Review of noninvasive or minimally invasive deep brain stimulation. Frontiers in Behavioral Neuroscience. 2021;15:820017
  27. 27. Zimerman M, Hummel FC. Brain stimulation and its role in neurological diseases. In: Cohen Kadosh R, editor. The Stimulated Brain: Cognitive Enhancement Using Non-Invasive Brain Stimulation. London: Academic Press; 2014. pp. 333-369
  28. 28. Davidson B, Bhattacharya A, Sarica C, Darmani G, Raies N, Chen R, et al. Neuromodulation techniques – From non-invasive brain stimulation to deep brain stimulation. Neurotherapeutics. 2024;21(3):e00330
  29. 29. Aderinto N, Olatunji G, Muili A, Kokori E, Edun M, Akinmoju O, et al. A narrative review of non-invasive brain stimulation techniques in neuropsychiatric disorders: Current applications and future directions. The Egyptian Journal of Neurology, Psychiatry and Neurosurgery. 2024;60(1):50
  30. 30. Perin C, Viganò B, Piscitelli D, Matteo BM, Meroni R, Cerri CG. Non-invasive current stimulation in vision recovery: A review of the literature. Restorative Neurology and Neuroscience. 2020;38(3):239-250
  31. 31. Liu J, Ma AKH, So KF, Lee VWH, Chiu K. Mechanisms of electrical stimulation in eye diseases: A narrative review. Advances in Ophthalmology Practice and Research. 2022;2(2):100060
  32. 32. Alej P, González N. Effectiveness and Safety of Vision Modulation and Restoration Using Noninvasive Electrical Stimulation: A Systematic Review and Meta-Analysis. Universidad de los Andes; 2022. 13 p
  33. 33. Muñoz Negrete FJ, Rebolleda G. Automated perimetry and neuro-ophthalmology. Topographic correlation. Archivos de la Sociedad Española de Oftalmología. 2002;77(8):413-428
  34. 34. Forrester JV, Dick AD, McMenamin PG, Roberts F, Pearlman E. The Eye: Basic Sciences in Practice. 4th ed. Edinburgh, UK: Elsevier; 2016. 568 p
  35. 35. Kelts EA. The basic anatomy of the optic nerve and visual system (or, why Thoreau was wrong). NeuroRehabilitation. 2010;27(3):217-222
  36. 36. De Moraes CG. Anatomy of the visual pathways. Journal of Glaucoma. 2013;22(Suppl. 5):S2-S7
  37. 37. Kaufman PL, Alm A, Adler FH. Adler’s Physiology of the Eye: Clinical Application. 10th ed. St. Louis: Mosby; 2003
  38. 38. Roof DJ, Makino CL. The structure and function of retinal photoreceptors. In: Albert DA, Jakobiec FA, editors. Principles and Practice of Ophthalmology. 2nd ed. Philadelphia: Saunders; 2000. pp. 1624-1673
  39. 39. Wells-Gray EM, Choi SS, Bries A, Doble N. Variation in rod and cone density from the fovea to the mid-periphery in healthy human retinas using adaptive optics scanning laser ophthalmoscopy. Eye. 2016;30(8):1135-1143
  40. 40. Cattell JM. The inverted image on the retina. Science. 1895;2(41):487-487
  41. 41. Salazar JJ, Ramírez AI, De Hoz R, Salobrar-Garcia E, Rojas P, Fernández-Albarral AJ, et al. Anatomy of the human optic nerve: Structure and function. In: Ferreri MF, editor. Optic Nerve. London, UK: IntechOpen; 2019
  42. 42. Ichinose T, Habib S. ON and OFF signaling pathways in the retina and the visual system. Frontiers in Ophthalmology. 2022;2:989002
  43. 43. Trattler WB, Kaiser PK, Friedman NJ. Review of Ophthalmology. 2nd ed. Philadelphia, U.S.: Elsevier Saunders; 2012. pp. 102-103
  44. 44. Callaway EM. Structure and function of parallel pathways in the primate early visual system. Journal of Physiology (London). 2005;566(Pt. 1):13-19
  45. 45. Smith AM, Czyz CN. Neuroanatomy, cranial nerve 2 (optic). In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2024
  46. 46. Ireland AC, Carter IB. Neuroanatomy, optic chiasm. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2024
  47. 47. Purves D, Augustine GJ, Fitzpatrick D, et al., editors. Central projections of retinal ganglion cells. In: Neuroscience. 2nd ed. Sunderland (MA): Sinauer Associates; 2001
  48. 48. Sántha K. Über das Verhalten der primären optischen Zentren bei einseitiger peripherer Blindheit. Albrecht von Graefes Archiv für Ophthalmologie. 1932;129(2):224-237
  49. 49. Kupfer C, Chumbley L, Downer JC. Quantitative histology of optic nerve, optic tract and lateral geniculate nucleus of man. Journal of Anatomy. 1967;101(Pt. 3):393-401
  50. 50. Chacko LW. The laminar pattern of the lateral geniculate body in the primates. Journal of Neurology, Neurosurgery, and Psychiatry. 1948;11(3):211-224
  51. 51. Purves D. Neuroscience. Scholarpedia. 2009;4(8):7204
  52. 52. Quigley HA, Addicks EM, Green WR. Optic nerve damage in human glaucoma. III. Quantitative correlation of nerve fiber loss and visual field defect in glaucoma, ischemic neuropathy, papilledema, and toxic neuropathy. Archives of Ophthalmology. 1982;100(1):135-146
  53. 53. Laws ER, Trautmann JC, Hollenhorst RW. Transsphenoidal decompression of the optic nerve and chiasm. Visual results in 62 patients. Journal of Neurosurgery. 1977;46(6):717-722
  54. 54. Hales PW, Smith V, Dhanoa-Hayre D, O’Hare P, Mankad K, d’Arco F, et al. Delineation of the visual pathway in paediatric optic pathway glioma patients using probabilistic tractography, and correlations with visual acuity. NeuroImage. Clinical. 2018;17:541-548
  55. 55. Optic Neuritis Study Group. The 5-year risk of MS after optic neuritis. Experience of the optic neuritis treatment trial. Neurology. 1997;49(5):1404-1413
  56. 56. Brex PA, Ciccarelli O, O’Riordan JI, Sailer M, Thompson AJ, Miller DH. A longitudinal study of abnormalities on MRI and disability from multiple sclerosis. The New England Journal of Medicine. 2002;346(3):158-164
  57. 57. Beck RW, Trobe JD, Moke PS, Gal RL, Xing D, Bhatti MT, et al. High- and low-risk profiles for the development of multiple sclerosis within 10 years after optic neuritis: Experience of the optic neuritis treatment trial. Archives of Ophthalmology. 2003;121(7):944-949
  58. 58. Cho KH, Ahn SJ, Jung C, Han MK, Park KH, Woo SJ. Ischemic injury of the papillomacular bundle is a predictive marker of poor vision in eyes with branch retinal artery occlusion. American Journal of Ophthalmology. 2016;162:107-120.e2
  59. 59. Attyé A, Jean C, Remond P, Peyrin C, Lecler A, Boudiaf N, et al. Track-weighted imaging for neuroretina: Evaluations in healthy volunteers and ischemic optic neuropathy. Journal of Magnetic Resonance Imaging. 2018;48:737-747
  60. 60. Ma J, Su S, Yue S, Zhao Y, Li Y, Chen X, et al. Preoperative visualization of cranial nerves in skull base tumor surgery using diffusion tensor imaging technology. Turkish Neurosurgery. 2016;26(6):805-812
  61. 61. Steward O. Visual system IV. In: Functional Neuroscience. New York, NY: Springer New York; 2000. pp. 349-361
  62. 62. Johnson KG, Harris WA. Connecting the eye with the brain: The formation of the retinotectal pathway. Results and Problems in Cell Differentiation. 2000;31:157-177
  63. 63. Lockhofen DEL, Mulert C. Neurochemistry of visual attention. Frontiers in Neuroscience. 2021;15:643597
  64. 64. Maunsell JHR. Neuronal mechanisms of visual attention. Annual Review of Vision Science. 2015;1:373-391
  65. 65. Sehic A, Guo S, Cho K-S, Corraya RM, Chen DF, Utheim TP. Electrical stimulation as a means for improving vision. The American Journal of Pathology. 2016;186(11):2783-2797
  66. 66. Bisley JW. The neural basis of visual attention. Journal of Physiology (London). 2011;589(Pt. 1):49-57
  67. 67. Breedlove SM, Watson NV. Behavioral Neuroscience. Eighth ed. Sunderland, Massachusetts: Sinauer Associates, Inc; 2017
  68. 68. Sheth B, Young R. Ventral and dorsal streams in cortex: Focal vs. ambient processing/exploitation vs. exploration. Journal of Vision. 2014;14(10):51-51
  69. 69. Hubel DH, Wiesel TN. Receptive fields, binocular interaction and functional architecture in the cat’s visual cortex. Journal of Physiology (London). 1962;160:106-154
  70. 70. von der Heydt R, Peterhans E, Baumgartner G. Illusory contours and cortical neuron responses. Science. 1984;224(4654):1260-1262
  71. 71. Horton JC, Adams DL. The cortical column: A structure without a function. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 2005;360(1456):837-862
  72. 72. Shmuel A, Chaimow D, Raddatz G, Ugurbil K, Yacoub E. Mechanisms underlying decoding at 7 T: Ocular dominance columns, broad structures, and macroscopic blood vessels in V1 convey information on the stimulated eye. NeuroImage. 2010;49(3):1957-1964
  73. 73. Milner AD. How do the two visual streams interact with each other? Experimental Brain Research. 2017;235(5):1297-1308
  74. 74. Wandell BA, Dumoulin SO, Brewer AA. Visual field maps in human cortex. Neuron. 2007;56(2):366-383
  75. 75. Bachatene L, Bharmauria V, Cattan S, Rouat J, Molotchnikoff S. Reprogramming of orientation columns in visual cortex: A domino effect. Scientific Reports. 2015;5:9436
  76. 76. Bachatene L, Bharmauria V, Molotchnikoff S. Adaptation and neuronal network in visual cortex. In: Molotchnikoff S, editor. Visual Cortex - Current Status and Perspectives. London, UK: InTechOpen; 2012
  77. 77. Goodale MA, Milner AD. Separate visual pathways for perception and action. Trends in Neurosciences. 1992;15(1):20-25
  78. 78. Choi S-H, Jeong G, Kim Y-B, Cho Z-H. Proposal for human visual pathway in the extrastriate cortex by fiber tracking method using diffusion-weighted MRI. NeuroImage. 2020;220:117145
  79. 79. Ponce CR, Hartmann TS, Livingstone MS. End-stopping predicts curvature tuning along the ventral stream. The Journal of Neuroscience. 2017;37(3):648-659
  80. 80. Salzman CD, Britten KH, Newsome WT. Cortical microstimulation influences perceptual judgements of motion direction. Nature. 1990;346(6280):174-177
  81. 81. Afraz S-R, Kiani R, Esteky H. Microstimulation of inferotemporal cortex influences face categorization. Nature. 2006;442(7103):692-695
  82. 82. Priebe NJ, Cassanello CR, Lisberger SG. The neural representation of speed in macaque area MT/V5. The Journal of Neuroscience. 2003;23(13):5650-5661
  83. 83. Born RT, Tootell RB. Segregation of global and local motion processing in primate middle temporal visual area. Nature. 1992;357(6378):497-499
  84. 84. Mineault PJ, Khawaja FA, Butts DA, Pack CC. Hierarchical processing of complex motion along the primate dorsal visual pathway. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(16):E972-E980
  85. 85. Marc RE, Jones BW. Retinal remodeling in inherited photoreceptor degenerations. Investigative Ophthalmology & Visual Science . 2003;28(2):139-147
  86. 86. Jones BW, Kondo M, Terasaki H, Lin Y, McCall M, Marc RE. Retinal remodeling. Japanese Journal of Ophthalmology. 2012;56(4):289-306
  87. 87. Pfeiffer RL, Jones BW. Current perspective on retinal remodeling: Implications for therapeutics. Frontiers in Neuroanatomy. 2022;16:1099348
  88. 88. García-Ayuso D, Esquiva G, Hombrebueno JR. Editorial: Understanding retinal remodeling: Retinal alterations and therapeutic implications. Frontiers in Neuroanatomy. 2023;17:1191906
  89. 89. Zhao TT, Tian CY, Yin ZQ. Activation of Müller cells occurs during retinal degeneration in RCS rats. Advances in Experimental Medicine and Biology. 2010;664:575-583
  90. 90. Roque RS, Rosales AA, Jingjing L, Agarwal N, Al-Ubaidi MR. Retina-derived microglial cells induce photoreceptor cell death in vitro. Brain Research. 1999;836(1-2):110-119
  91. 91. Bringmann A, Wiedemann P. Müller glial cells in retinal disease. Ophthalmologica. 2012;227(1):1-19
  92. 92. Strauss O. The retinal pigment epithelium in visual function. Physiological Reviews. 2005 Jul;85(3):845-881
  93. 93. Gupta N, Brown KE, Milam AH. Activated microglia in human retinitis pigmentosa, late-onset retinal degeneration, and age-related macular degeneration. Experimental Eye Research. 2003;76(4):463-471
  94. 94. Jones BW, Pfeiffer RL, Ferrell WD, Watt CB, Tucker J, Marc RE. Retinal remodeling and metabolic alterations in human AMD. Frontiers in Cellular Neuroscience. 2016;10:103
  95. 95. Waugh N, Loveman E, Colquitt J, Royle P, Yeong JL, Hoad G, et al. Treatments for dry age-related macular degeneration and Stargardt disease: A systematic review. In: Introduction to Age-Related Macular Degeneration. Winchester, England: Health Technology Assessment; 2018
  96. 96. Phelan JK, Bok D. A brief review of retinitis pigmentosa and the identified retinitis pigmentosa genes. Molecular Vision. 2000;6:116-124
  97. 97. Weleber RG, Gregory-Evance K. Retinitis pigmentosa and allied disorders. In: Hinton DR, editor. Basic Science and Inherited Retinal Disease. St. Louis: Mosby; 2006. pp. 395-498
  98. 98. Desai S, Alibhai AY. Retinitis pigmentosa. In: Atlas of Retinal OCT: Optical Coherence Tomography. Amsterdam, The Netherlands: Elsevier; 2018. p. 148
  99. 99. Patil SB, Hurd TW, Ghosh AK, Murga-Zamalloa CA, Khanna H. Variable effect of human disease causing mutations in X-linked RP-associated ciliary protein RP2 on its function; insights into associated clinical heterogeneity. Investigative Ophthalmology & Visual Science. 2011
  100. 100. Kropp M, Golubnitschaja O, Mazurakova A, Koklesova L, Sargheini N, Vo T-TKS, et al. Diabetic retinopathy as the leading cause of blindness and early predictor of cascading complications-risks and mitigation. The EPMA Journal. 2023;14(1):21-42
  101. 101. Shukla UV, Tripathy K. Diabetic retinopathy. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2024
  102. 102. Kollias AN, Ulbig MW. Diabetic retinopathy: Early diagnosis and effective treatment. Deutsches Ärzteblatt International. 2010;107(5):75-83; quiz 84
  103. 103. Waisberg E, Micieli JA. Neuro-ophthalmological optic nerve cupping: An overview. Eye Brain. 2021;13:255-268
  104. 104. Guo L, Moss SE, Alexander RA, Ali RR, Fitzke FW, Cordeiro MF. Retinal ganglion cell apoptosis in glaucoma is related to intraocular pressure and IOP-induced effects on extracellular matrix. Investigative Ophthalmology & Visual Science. 2005;46(1):175-182
  105. 105. Osborne B. Optic neuropathies [Internet]. UpToDate. 2024. Available from: https://www.uptodate.com/contents/optic-neuropathies [Accessed: May 10, 2024]
  106. 106. Hayreh SS. Anterior ischaemic optic neuropathy. I. Terminology and pathogenesis. The British Journal of Ophthalmology. 1974;58(12):955-963
  107. 107. Hayreh SS. Posterior ischemic optic neuropathy. Ophthalmologica. 1981;182(1):29-41
  108. 108. Steigerwalt RD, Cesarone MR, Belcaro G, Pascarella A, De Angelis M, Gattegna R, et al. Arteritic anterior ischemic optic neuropathy treated with intravenous prostaglandin E(1) and steroids. International Journal of Angiology. 2010;19(3):e113-e115
  109. 109. Hayreh SS. Inter-individual variation in blood supply of the optic nerve head. Its importance in various ischemic disorders of the optic nerve head, and glaucoma, low-tension glaucoma and allied disorders. Documenta Ophthalmologica. 1985;59(3):217-246
  110. 110. Fraser C, Plant GT. Optic neuritis: Pathophysiology, clinical features, and management. In: Minagar A, editor. Neuroinflammation. London; Burlington, MA: Elsevier; 2011. pp. 253-276
  111. 111. Guier CP, Stokkermans TJ. Optic neuritis. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2024
  112. 112. Osborne B. Optic neuritis: Pathophysiology, clinical features, and diagnosis - UpToDate [Internet]. 2024. Available from: https://www.uptodate.com/contents/optic-neuritis-pathophysiology-clinical-features-and-diagnosis [Accessed: May 5, 2024]
  113. 113. Newman NJ, Lessell S, Winterkorn JM. Optic chiasmal neuritis. Neurology. 1991;41(8):1203-1210
  114. 114. Penfield W. Some observations on the cerebral cortex of man. Proceedings of the Royal Society of London. Series B, Biological Sciences. 1947;134(876):329-347
  115. 115. Penfield W, Rasmussen T. The cerebral cortex of man: A clinical study of localization of function. Journal of the American Medical Association. 1950;144(16):1412
  116. 116. Pasquinelli C, Hanson LG, Siebner HR, Lee HJ, Thielscher A. Safety of transcranial focused ultrasound stimulation: A systematic review of the state of knowledge from both human and animal studies. Brain Stimulation. 2019;12(6):1367-1380
  117. 117. Peterchev AV, Wagner TA, Miranda PC, Nitsche MA, Paulus W, Lisanby SH, et al. Fundamentals of transcranial electric and magnetic stimulation dose: Definition, selection, and reporting practices. Brain Stimulation. 2012;5(4):435-453
  118. 118. Ye M, Zestos AG, Welle CG, Wu GK, Cui XT. Editorial: Safety and efficacy evaluation of electrical stimulation devices for neural modulation. Frontiers in Neuroscience. 2023;17:1143989
  119. 119. Lewis PM, Rosenfeld JV. Electrical stimulation of the brain and the development of cortical visual prostheses: An historical perspective. Brain Research. 2016;1630:208-224
  120. 120. Fu L, Fung FK, Lo AC-Y, Chan Y-K, So K-F, Wong IY-H, et al. Transcorneal electrical stimulation inhibits retinal microglial activation and enhances retinal ganglion cell survival after acute ocular hypertensive injury. Translational Vision Science & Technology. 2018;7(3):7
  121. 121. Fernandez E. Development of visual neuroprostheses: Trends and challenges. Bioelectronic Medicine. 2018;4:12
  122. 122. Ghezzi D. Retinal prostheses: Progress toward the next generation implants. Frontiers in Neuroscience. 2015;9:290
  123. 123. Cheng DL, Greenberg PB, Borton DA. Advances in retinal prosthetic research: A systematic review of engineering and clinical characteristics of current prosthetic initiatives. Current Eye Research. 2017;42(3):334-347
  124. 124. Pardue MT, Ciavatta VT, Hetling JR. Neuroprotective effects of low level electrical stimulation therapy on retinal degeneration. Advances in Experimental Medicine and Biology. 2014;801:845-851
  125. 125. Rahmani S, Bogdanowicz L, Thomas J, Hetling JR. Chronic delivery of low-level exogenous current preserves retinal function in pigmented P23H rat. Vision Research. 2013;76:105-113
  126. 126. Cabral AM, Pereira AA, Vieira MF, Pessôa BL, de Oliveira AA. Prevalence of distinct types of hardware failures related to deep brain stimulation. Neurosurgical Review. 2022;45(2):1123-1134
  127. 127. Krishnan C, Santos L, Peterson MD, Ehinger M. Safety of noninvasive brain stimulation in children and adolescents. Brain Stimulation. 2015;8(1):76-87
  128. 128. Airan R. Neuromodulation with nanoparticles. Science. 2017;357(6350):465
  129. 129. Woods AJ, Antal A, Bikson M, Boggio PS, Brunoni AR, Celnik P, et al. A technical guide to tDCS, and related non-invasive brain stimulation tools. Clinical Neurophysiology. 2016;127(2):1031-1048
  130. 130. Ciavatta VT, Kim M, Wong P, Nickerson JM, Shuler RK, McLean GY, et al. Retinal expression of Fgf2 in RCS rats with subretinal microphotodiode array. Investigative Ophthalmology & Visual Science. 2009;50(10):4523-4530
  131. 131. Mocko JA, Kim M, Faulkner AE, Cao Y, Ciavatta VT, Pardue MT. Effects of subretinal electrical stimulation in Mer-KO mice. Investigative Ophthalmology & Visual Science. 2011;52(7):4223-4230
  132. 132. Morimoto T, Miyoshi T, Matsuda S, Tano Y, Fujikado T, Fukuda Y. Transcorneal electrical stimulation rescues axotomized retinal ganglion cells by activating endogenous retinal IGF-1 system. Investigative Ophthalmology & Visual Science. 2005;46(6):2147-2155
  133. 133. Ni Y, Gan D, Xu H, Xu G, Da C. Neuroprotective effect of transcorneal electrical stimulation on light-induced photoreceptor degeneration. Experimental Neurology. 2009;219(2):439-452
  134. 134. Tao Y, Chen T, Liu Z-Y, Wang L-Q , Xu W-W, Qin L-M, et al. Topographic quantification of the transcorneal electrical stimulation (TES)-induced protective effects on N-methyl-N-nitrosourea-treated retinas. Investigative Ophthalmology & Visual Science. 2016;57(11):4614-4624
  135. 135. Yu H, Enayati S, Chang K, Cho K, Lee SW, Talib M, et al. Noninvasive electrical stimulation improves photoreceptor survival and retinal function in mice with inherited photoreceptor degeneration. Investigative Ophthalmology & Visual Science. 2020;61(4):5
  136. 136. Hanif AM, Kim MK, Thomas JG, Ciavatta VT, Chrenek M, Hetling JR, et al. Whole-eye electrical stimulation therapy preserves visual function and structure in P23H-1 rats. Experimental Eye Research. 2016;149:75-83
  137. 137. Smith AW, Das A, Guyton MK, Ray SK, Rohrer B, Banik NL. Calpain inhibition attenuates apoptosis of retinal ganglion cells in acute optic neuritis. Investigative Ophthalmology & Visual Science. 2011;52(7):4935-4941
  138. 138. Yildiz-Unal A, Korulu S, Karabay A. Neuroprotective strategies against calpain-mediated neurodegeneration. Neuropsychiatric Disease and Treatment. 2015;11:297-310
  139. 139. Thomas CN, Berry M, Logan A, Blanch RJ, Ahmed Z. Caspases in retinal ganglion cell death and axon regeneration. Cell Death Discovery. 2017;3:17032
  140. 140. Kale J, Osterlund EJ, Andrews DW. BCL-2 family proteins: Changing partners in the dance towards death. Cell Death and Differentiation. 2018;25(1):65-80
  141. 141. Brunelle JK, Letai A. Control of mitochondrial apoptosis by the Bcl-2 family. Journal of Cell Science. 2009;122(Pt. 4):437-441
  142. 142. Willmann G, Schäferhoff K, Fischer MD, Arango-Gonzalez B, Bolz S, Naycheva L, et al. Gene expression profiling of the retina after transcorneal electrical stimulation in wild-type Brown Norway rats. Investigative Ophthalmology & Visual Science. 2011;52(10):7529-7537
  143. 143. Sato T, Fujikado T, Lee T-S, Tano Y. Direct effect of electrical stimulation on induction of brain-derived neurotrophic factor from cultured retinal Müller cells. Investigative Ophthalmology & Visual Science. 2008;49(10):4641-4646
  144. 144. Kanamoto T, Souchelnytskyi N, Kurimoto T, Ikeda Y, Sakaue H, Munemasa Y, et al. Proteomic study of retinal proteins associated with transcorneal electric stimulation in rats. Journal of Ophthalmology. 2015;2015:492050
  145. 145. Jassim AH, Cavanaugh M, Shah JS, Willits R, Inman DM. Transcorneal electrical stimulation reduces neurodegenerative process in a mouse model of glaucoma. Annals of Biomedical Engineering. 2021;49(2):858-870
  146. 146. Tagami Y, Kurimoto T, Miyoshi T, Morimoto T, Sawai H, Mimura O. Axonal regeneration induced by repetitive electrical stimulation of crushed optic nerve in adult rats. Japanese Journal of Ophthalmology. 2009;53(3):257-266
  147. 147. Morimoto T, Miyoshi T, Fujikado T, Tano Y, Fukuda Y. Electrical stimulation enhances the survival of axotomized retinal ganglion cells in vivo. Neuroreport. 2002;13(2):227-230
  148. 148. Gokoffski KK, Jia X, Shvarts D, Xia G, Zhao M. Physiologic electrical fields direct retinal ganglion cell axon growth in vitro. Investigative Ophthalmology & Visual Science. 2019;60(10):3659-3668
  149. 149. Sato T, Fujikado T, Morimoto T, Matsushita K, Harada T, Tano Y. Effect of electrical stimulation on IGF-1 transcription by L-type calcium channels in cultured retinal Müller cells. Japanese Journal of Ophthalmology. 2008;52(3):217-223
  150. 150. Lennikov A, Yang M, Chang K, Pan L, Saddala MS, Lee C, et al. Direct modulation of microglial function by electrical field. Frontiers in Cell and Development Biology. 2022;10:980775
  151. 151. Zhou W, Ni Y, Jin Z, Zhang M, Wu J, Zhu Y, et al. Electrical stimulation ameliorates light-induced photoreceptor degeneration in vitro via suppressing the proinflammatory effect of microglia and enhancing the neurotrophic potential of Müller cells. Experimental Neurology. 2012;238(2):192-208
  152. 152. Enayati S, Chang K, Achour H, Cho KS, Xu F, Guo S, et al. Electrical stimulation induces retinal Müller cell proliferation and their progenitor cell potential. Cells. 2020;9(3):781
  153. 153. Singh D. Astrocytic and microglial cells as the modulators of neuroinflammation in Alzheimer’s disease. Journal of Neuroinflammation. 2022;19(1):206
  154. 154. Ramirez AI, de Hoz R, Salobrar-Garcia E, Salazar JJ, Rojas B, Ajoy D, et al. The role of microglia in retinal neurodegeneration: Alzheimer’s disease, Parkinson, and glaucoma. Frontiers in Aging Neuroscience. 2017;9:214
  155. 155. Vainchtein ID, Molofsky AV. Astrocytes and microglia: In sickness and in health. Trends in Neurosciences. 2020;43(3):144-154
  156. 156. Tezel G. TNF-alpha signaling in glaucomatous neurodegeneration. Progress in Brain Research. 2008;173:409-421
  157. 157. Berger S, Savitz SI, Nijhawan S, Singh M, David J, Rosenbaum PS, et al. Deleterious role of TNF-alpha in retinal ischemia-reperfusion injury. Investigative Ophthalmology & Visual Science. 2008;49(8):3605-3610
  158. 158. Ji M, Sun Q , Zhang G, Huang Z, Zhang Y, Shen Q , et al. Microglia-derived TNF-α mediates Müller cell activation by activating the TNFR1-NF-κB pathway. Experimental Eye Research. 2022;214:108852
  159. 159. Dvoriantchikova G, Barakat D, Brambilla R, Agudelo C, Hernandez E, Bethea JR, et al. Inactivation of astroglial NF-kappa B promotes survival of retinal neurons following ischemic injury. The European Journal of Neuroscience. 2009;30(2):175-185
  160. 160. Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005;50(4):427-434
  161. 161. Papa S, Bubici C, Zazzeroni F, Pham CG, Kuntzen C, Knabb JR, et al. The NF-kappaB-mediated control of the JNK cascade in the antagonism of programmed cell death in health and disease. Cell Death and Differentiation. 2006;13(5):712-729
  162. 162. Kamata H, Honda S-I, Maeda S, Chang L, Hirata H, Karin M. Reactive oxygen species promote TNF alpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell. 2005;120(5):649-661
  163. 163. Gunes K, Chang K, Tai WL, Cho K-S, Utheim TP, Chen D. Non-invasive transpalpebral electrical stimulation reduces microglia activation in mice with inherited photoreceptor degeneration. Investigative Ophthalmology & Visual Science. 2023;64(8):2310
  164. 164. Xu T, Yu X, Perlik AJ, Tobin WF, Zweig JA, Tennant K, et al. Rapid formation and selective stabilization of synapses for enduring motor memories. Nature. 2009;462(7275):915-919
  165. 165. Draganski B, Gaser C, Busch V, Schuierer G, Bogdahn U, May A. Neuroplasticity: Changes in grey matter induced by training. Nature. 2004;427(6972):311-312
  166. 166. Buonomano DV, Merzenich MM. Cortical plasticity: From synapses to maps. Annual Review of Neuroscience. 1998;21:149-186
  167. 167. Bharmauria V, Ouelhazi A, Lussiez R, Molotchnikoff S. Adaptation-induced plasticity in the sensory cortex. Journal of Neurophysiology. 2022;128(4):946-962
  168. 168. Castaldi E, Lunghi C, Morrone MC. Neuroplasticity in adult human visual cortex. Neuroscience and Biobehavioral Reviews. 2020;112:542-552
  169. 169. Bola M, Gall C, Moewes C, Fedorov A, Hinrichs H, Sabel BA. Brain functional connectivity network breakdown and restoration in blindness. Neurology. 2014;83(6):542-551
  170. 170. Kasten FH, Dowsett J, Herrmann CS. Sustained after effect of α-tACS lasts up to 70 min after stimulation. Frontiers in Human Neuroscience. 2016;10:245
  171. 171. Ma Z, Cao P, Sun P, Li L, Lu Y, Yan Y, et al. Optical imaging of visual cortical responses evoked by transcorneal electrical stimulation with different parameters. Investigative Ophthalmology & Visual Science. 2014;55(8):5320-5331
  172. 172. Sergeeva EG, Bola M, Wagner S, Lazik S, Voigt N, Mawrin C, et al. Repetitive transcorneal alternating current stimulation reduces brain idling state after long-term vision loss. Brain Stimulation. 2015;8(6):1065-1073
  173. 173. Sergeeva EG, Fedorov AB, Henrich-Noack P, Sabel BA. Transcorneal alternating current stimulation induces EEG “aftereffects” only in rats with an intact visual system but not after severe optic nerve damage. Journal of Neurophysiology. 2012;108(9):2494-2500
  174. 174. Sabel BA, Fedorov AB, Naue N, Borrmann A, Herrmann C, Gall C. Non-invasive alternating current stimulation improves vision in optic neuropathy. Restorative Neurology and Neuroscience. 2011;29(6):493-505
  175. 175. Sabel BA, Hamid AIA, Borrmann C, Speck O, Antal A. Transorbital alternating current stimulation modifies BOLD activity in healthy subjects and in a stroke patient with hemianopia: A 7 Tesla fMRI feasibility study. International Journal of Psychophysiology. 2020;154:80-92
  176. 176. Zaehle T, Rach S, Herrmann CS. Transcranial alternating current stimulation enhances individual alpha activity in human EEG. PLoS One. 2010;5(11):e13766
  177. 177. Sun Y, Smith LEH. Retinal vasculature in development and diseases. Annual Review of Vision Science. 2018;4:101-122
  178. 178. Schmidt M, Giessl A, Laufs T, Hankeln T, Wolfrum U, Burmester T. How does the eye breathe? Evidence for neuroglobin-mediated oxygen supply in the mammalian retina. The Journal of Biological Chemistry. 2003;278(3):1932-1935
  179. 179. Eshaq RS, Wright WS, Harris NR. Oxygen delivery, consumption, and conversion to reactive oxygen species in experimental models of diabetic retinopathy. Redox Biology. 2014;2:661-666
  180. 180. Kurimoto T, Oono S, Oku H, Tagami Y, Kashimoto R, Takata M, et al. Transcorneal electrical stimulation increases chorioretinal blood flow in normal human subjects. Clinical Ophthalmology. 2010;4:1441-1446
  181. 181. Della Volpe-Waizel M, Zuche HC, Müller U, Rickmann A, Scholl HPN, Todorova MG. Metabolic monitoring of transcorneal electrical stimulation in retinitis pigmentosa. Graefe's Archive for Clinical and Experimental Ophthalmology. 2020;258(1):79-87
  182. 182. Bittner AK, Seger K, Salveson R, Kayser S, Morrison N, Vargas P, et al. Randomized controlled trial of electro-stimulation therapies to modulate retinal blood flow and visual function in retinitis pigmentosa. Acta Ophthalmologica. 2018;96(3):e366-e376
  183. 183. Freitag S, Hunold A, Klemm M, Klee S, Link D, Nagel E, et al. Pulsed electrical stimulation of the human eye enhances retinal vessel reaction to flickering light. Frontiers in Human Neuroscience. 2019;13:371
  184. 184. Nagasaka M, Kohzuki M, Fujii T, Kanno S, Kawamura T, Onodera H, et al. Effect of low-voltage electrical stimulation on angiogenic growth factors in ischaemic rat skeletal muscle. Clinical and Experimental Pharmacology & Physiology. 2006;33(7):623-627
  185. 185. Kanno S, Oda N, Abe M, Saito S, Hori K, Handa Y, et al. Establishment of a simple and practical procedure applicable to therapeutic angiogenesis. Circulation. 1999;99(20):2682-2687
  186. 186. Newman EA. Glial cell regulation of neuronal activity and blood flow in the retina by release of gliotransmitters. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 2015;370(1672):20140195
  187. 187. Metea MR, Newman EA. Glial cells dilate and constrict blood vessels: A mechanism of neurovascular coupling. The Journal of Neuroscience. 2006;26(11):2862-2870
  188. 188. Cheah LS, Gwee M, Das R, Ballard H, Yang YF, Daniel EE, et al. Evidence for the existence of a constitutive nitric oxide synthase in vascular smooth muscle. Clinical and Experimental Pharmacology & Physiology. 2002;29(8):725-727
  189. 189. Antal A, Kincses TZ, Nitsche MA, Paulus W. Manipulation of phosphene thresholds by transcranial direct current stimulation in man. Experimental Brain Research. 2003;150(3):375-378
  190. 190. Maynard EM. Visual prostheses. Annual Review of Biomedical Engineering. 2001;3:145-168
  191. 191. Lin T-C, Chang H-M, Hsu C-C, Hung K-H, Chen Y-T, Chen S-Y, et al. Retinal prostheses in degenerative retinal diseases. Journal of the Chinese Medical Association. 2015;78(9):501-505
  192. 192. Caspi A, Roy A, Dorn JD, Greenberg RJ. Retinotopic to spatiotopic mapping in blind patients implanted with the Argus II retinal prosthesis. Investigative Ophthalmology & Visual Science. 2017;58(1):119-127
  193. 193. Freeman DK, Rizzo JF, Fried SI. Encoding visual information in retinal ganglion cells with prosthetic stimulation. Journal of Neural Engineering. 2011;8(3):035005
  194. 194. Chang Y-C, Haji Ghaffari D, Chow RH, Weiland JD. Stimulation strategies for selective activation of retinal ganglion cell soma and threshold reduction. Journal of Neural Engineering. 2019;16(2):026017
  195. 195. Sekirnjak C, Hottowy P, Sher A, Dabrowski W, Litke AM, Chichilnisky EJ. Electrical stimulation of mammalian retinal ganglion cells with multielectrode arrays. Journal of Neurophysiology. 2006;95(6):3311-3327
  196. 196. Jensen RJ, Ziv OR, Rizzo JF. Thresholds for activation of rabbit retinal ganglion cells with relatively large, extracellular microelectrodes. Investigative Ophthalmology & Visual Science. 2005;46(4):1486-1496
  197. 197. Grosberg LE, Hottowy P, Jepson LH, Ito S, Kellison-Linn F, Sher A, et al. Axon activation with focal epiretinal stimulation in primate retina. Investigative Ophthalmology & Visual Science. 2015;56(7):780
  198. 198. Grosberg LE, Ganesan K, Goetz GA, Madugula SS, Bhaskhar N, Fan V, et al. Activation of ganglion cells and axon bundles using epiretinal electrical stimulation. Journal of Neurophysiology. 2017;118(3):1457-1471
  199. 199. Fried SI, Lasker ACW, Desai NJ, Eddington DK, Rizzo JF. Axonal sodium-channel bands shape the response to electric stimulation in retinal ganglion cells. Journal of Neurophysiology. 2009;101(4):1972-1987
  200. 200. Margalit E, Thoreson WB. Inner retinal mechanisms engaged by retinal electrical stimulation. Investigative Ophthalmology & Visual Science. 2006;47(6):2606-2612
  201. 201. Freeman DK, Eddington DK, Rizzo JF, Fried SI. Selective activation of neuronal targets with sinusoidal electric stimulation. Journal of Neurophysiology. 2010;104(5):2778-2791
  202. 202. Ahuja AK, Dorn JD, Caspi A, McMahon MJ, Dagnelie G, Dacruz L, et al. Blind subjects implanted with the Argus II retinal prosthesis are able to improve performance in a spatial-motor task. The British Journal of Ophthalmology. 2011;95(4):539-543
  203. 203. Stingl K, Bartz-Schmidt KU, Besch D, Braun A, Bruckmann A, Gekeler F, et al. Artificial vision with wirelessly powered subretinal electronic implant alpha-IMS. Proceedings of the Biological Sciences. 2013;280(1757):20130077
  204. 204. Muqit MMK, Hubschman JP, Picaud S, McCreery DB, van Meurs JC, Hornig R, et al. PRIMA subretinal wireless photovoltaic microchip implantation in non-human primate and feline models. PLoS One. 2020;15(4):e0230713
  205. 205. Richard G, Feucht M, Bornfeld N, Laube T, Rössler G, Velikay-Parel M, et al. Multicenter study on acute electrical stimulation of the human retina with an epiretinal implant: clinical results in 20 patients. Investigative Ophthalmology & Visual Science. 2005;46(13):1143
  206. 206. Veraart C, Raftopoulos C, Mortimer JT, Delbeke J, Pins D, Michaux G, et al. Visual sensations produced by optic nerve stimulation using an implanted self-sizing spiral cuff electrode. Brain Research. 1998;813(1):181-186
  207. 207. Nishida K, Sakaguchi H, Kamei M, Cecilia-Gonzalez C, Terasawa Y, Velez-Montoya R, et al. Visual sensation by electrical stimulation using a new direct optic nerve electrode device. Brain Stimulation. 2015;8(3):678-681
  208. 208. Panetsos F, Sanchez-Jimenez A, Cerio ED, Diaz-Guemes I, Sanchez FM. Consistent phosphenes generated by electrical microstimulation of the visual thalamus. An experimental approach for thalamic visual neuroprostheses. Frontiers in Neuroscience. 2011;5:84
  209. 209. Pezaris JS, Reid RC. Simulations of electrode placement for a thalamic visual prosthesis. IEEE Transactions on Biomedical Engineering. 2009;56(1):172-178
  210. 210. Allen PJ, Ayton LN. Development and experimental basis for the future of prosthetic vision. In: Chang A, Mieler WF, Ohji M, editors. Macular Surgery: Current Practice and Trends. Singapore: Springer Singapore; 2020. pp. 449-462
  211. 211. Fernández E, Normann RA. CORTIVIS approach for an intracortical visual prostheses. In: Gabel VP, editor. Artificial Vision. Cham: Springer International Publishing; 2017. pp. 191-201
  212. 212. Barry MP, Sadeghi R, Towle VL, Stipp K, Puhov H, Diaz W, et al. Preliminary visual function for the first human with the intracortical visual prosthesis (ICVP). Investigative Ophthalmology & Visual Science. 2023;64(8):2842
  213. 213. Robles-Camarillo D, Niño-de-Rivera L, López-Miranda J, Gil-Carrasco F, Quiroz-Mercado H. The effect of transcorneal electrical stimulation in visual acuity: Retinitis pigmentosa. JBiSE. 2013;06(10):1-7
  214. 214. Bittner AK, Seger K. Longevity of visual improvements following transcorneal electrical stimulation and efficacy of retreatment in three individuals with retinitis pigmentosa. Graefe's Archive for Clinical and Experimental Ophthalmology. 2018;256(2):299-306
  215. 215. Schatz A, Röck T, Naycheva L, Willmann G, Wilhelm B, Peters T, et al. Transcorneal electrical stimulation for patients with retinitis pigmentosa: A prospective, randomized, sham-controlled exploratory study. Investigative Ophthalmology & Visual Science. 2011;52(7):4485-4496
  216. 216. Jolly JK, Wagner SK, Martus P, MacLaren RE, Wilhelm B, Webster AR, et al. Transcorneal electrical stimulation for the treatment of retinitis Pigmentosa: A Multicenter safety study of the OkuStim® system (TESOLA-study). Ophthalmic Research. 2020;63(3):234-243
  217. 217. Dizdar Yigit D, Sevik MO, Şahin Ö. Transcorneal electrical stimulation therapy may have a stabilization effect on multifocal electroretinography for patients with retinitis pigmentosa. Retina (Philadelphia, PA). 2022;42(5):923-933
  218. 218. Gall C, Sgorzaly S, Schmidt S, Brandt S, Fedorov A, Sabel BA. Noninvasive transorbital alternating current stimulation improves subjective visual functioning and vision-related quality of life in optic neuropathy. Brain Stimulation. 2011;4(4):175-188
  219. 219. Schmidt S, Mante A, Rönnefarth M, Fleischmann R, Gall C, Brandt SA. Progressive enhancement of alpha activity and visual function in patients with optic neuropathy: A two-week repeated session alternating current stimulation study. Brain Stimulation. 2013;6(1):87-93
  220. 220. Gall C, Schmidt S, Schittkowski MP, Antal A, Ambrus GG, Paulus W, et al. Alternating current stimulation for vision restoration after optic nerve damage: A randomized clinical trial. PLoS One. 2016;11(6):e0156134
  221. 221. Räty S, Ruuth R, Silvennoinen K, Sabel BA, Tatlisumak T, Vanni S. Resting-state functional connectivity after occipital stroke. Neurorehabilitation and Neural Repair. 2022;36(2):151-163
  222. 222. Shinoda K, Imamura Y, Matsuda S, Seki M, Uchida A, Grossman T, et al. Transcutaneous electrical retinal stimulation therapy for age-related macular degeneration. The Open Ophthalmology Journal. 2008;2:132-136
  223. 223. Anastassiou G, Schneegans A-L, Selbach M, Kremmer S. Transpalpebral electrotherapy for dry age-related macular degeneration (AMD): An exploratory trial. Restorative Neurology and Neuroscience. 2013;31(5):571-578
  224. 224. Chaikin L, Kashiwa K, Bennet M, Papastergiou G, Gregory W. Microcurrent stimulation in the treatment of dry and wet macular degeneration. Clinical Ophthalmology. 2015;9:2345-2353
  225. 225. Menz MD, Ye P, Firouzi K, Nikoozadeh A, Pauly KB, Khuri-Yakub P, et al. Radiation force as a physical mechanism for ultrasonic neurostimulation of the ex vivo retina. The Journal of Neuroscience. 2019;39(32):6251-6264
  226. 226. Naor O, Hertzberg Y, Zemel E, Kimmel E, Shoham S. Towards multifocal ultrasonic neural stimulation II: Design considerations for an acoustic retinal prosthesis. Journal of Neural Engineering. 2012;9(2):026006
  227. 227. Menz MD, Oralkan O, Khuri-Yakub PT, Baccus SA. Precise neural stimulation in the retina using focused ultrasound. The Journal of Neuroscience. 2013;33(10):4550-4560
  228. 228. Terney D, Chaieb L, Moliadze V, Antal A, Paulus W. Increasing human brain excitability by transcranial high-frequency random noise stimulation. The Journal of Neuroscience. 2008;28(52):14147-14155
  229. 229. Camilleri R, Pavan A, Campana G. The application of online transcranial random noise stimulation and perceptual learning in the improvement of visual functions in mild myopia. Neuropsychologia. 2016;89:225-231
  230. 230. Moret B, Camilleri R, Pavan A, Lo Giudice G, Veronese A, Rizzo R, et al. Differential effects of high-frequency transcranial random noise stimulation (hf-tRNS) on contrast sensitivity and visual acuity when combined with a short perceptual training in adults with amblyopia. Neuropsychologia. 2018;114:125-133
  231. 231. Hornig R, Zehnder T, Velikay-Parel M, Laube T, Feucht M, Richard G. The IMI retinal implant system. In: Humayun MS, Weiland JD, Chader G, Greenbaum E, editors. Artificial Sight. New York, NY: Springer New York; 2007. pp. 111-128
  232. 232. Santos A, Humayun MS, de Juan E, Greenburg RJ, Marsh MJ, Klock IB, et al. Preservation of the inner retina in retinitis pigmentosa. A morphometric analysis. Archives of Ophthalmology. 1997;115(4):511-515
  233. 233. Humayun MS, Weiland JD, Fujii GY, Greenberg R, Williamson R, Little J, et al. Visual perception in a blind subject with a chronic microelectronic retinal prosthesis. Vision Research. 2003;43(24):2573-2581
  234. 234. Rao C, Yuan X-H, Zhang SJ, Wang QL, Huang YS. Epiretinal prosthesis forouter retinal degenerative diseases. International Journal of Ophthalmology. 2008;1:273-276
  235. 235. Vu QA, Seo HW, Choi K-E, Kim N, Kang YN, Lee J, et al. Structural changes in the retina after implantation of subretinal three-dimensional implants in mini pigs. Frontiers in Neuroscience. 2022;16:1010445
  236. 236. Adekunle AN, Adkins A, Wang W, Kaplan HJ, de Castro JF, Lee SJ, et al. Integration of perforated subretinal prostheses with retinal tissue. Translational Vision Science & Technology. 2015;4(4):5
  237. 237. Chenais NAL, Airaghi Leccardi MJI, Ghezzi D. Photovoltaic retinal prosthesis restores high-resolution responses to single-pixel stimulation in blind retinas. Communications Materials. 2021;2(1):28
  238. 238. Miyoshi T, Morimoto T, Sawai H, Fujikado T. Spatial resolution of suprachoroidal-transretinal stimulation estimated by recording single-unit activity from the cat lateral geniculate nucleus. Frontiers in Neuroscience. 2021;15:717429
  239. 239. Kanda H, Morimoto T, Fujikado T, Tano Y, Fukuda Y, Sawai H. Electrophysiological studies of the feasibility of suprachoroidal-transretinal stimulation for artificial vision in normal and RCS rats. Investigative Ophthalmology & Visual Science. 2004;45(2):560-566
  240. 240. Zrenner E, Miliczek KD, Gabel VP, Graf HG, Guenther E, Haemmerle H, et al. The development of subretinal microphotodiodes for replacement of degenerated photoreceptors. Ophthalmic Research. 1997;29(5):269-280
  241. 241. Zrenner E, Stett A, Weiss S, Aramant RB, Guenther E, Kohler K, et al. Can subretinal microphotodiodes successfully replace degenerated photoreceptors? Vision Research. 1999;39(15):2555-2567
  242. 242. Jones BW, Pfeiffer RL, Ferrell WD, Watt CB, Marmor M, Marc RE. Retinal remodeling in human retinitis pigmentosa. Experimental Eye Research. 2016;150:149-165
  243. 243. Jones BW, Marc RE, Pfeiffer RL. Retinal degeneration, remodeling and plasticity. In: Kolb H, Fernandez E, Nelson R, editors. Webvision: The Organization of the Retina and Visual System [Internet]. Salt Lake City (UT): University of Utah Health Sciences Center; 1995
  244. 244. Tong W, Meffin H, Garrett DJ, Ibbotson MR. Stimulation strategies for improving the resolution of retinal prostheses. Frontiers in Neuroscience. 2020;14:262
  245. 245. Flores T, Lei X, Huang T, Lorach H, Dalal R, Galambos L, et al. Optimization of pillar electrodes in subretinal prosthesis for enhanced proximity to target neurons. Journal of Neural Engineering. 2018;15(3):036011
  246. 246. Chen ZC, Wang BY, Kochnev Goldstein A, Butt E, Mathieson K, Palanker D. Photovoltaic implant simulator reveals resolution limits in subretinal prosthesis. Journal of Neural Engineering. 2022;19:055008
  247. 247. Pezaris JS, Eskandar EN. Getting signals into the brain: Visual prosthetics through thalamic microstimulation. Neurosurgical Focus. 2009;27(1):E6
  248. 248. Sweeney JD, Ksienski DA, Mortimer JT. A nerve cuff technique for selective excitation of peripheral nerve trunk regions. IEEE Transactions on Biomedical Engineering. 1990;37(7):706-715
  249. 249. Rozman J, Trlep M. Multielectrode spiral cuff for selective stimulation of nerve fibres. Journal of Medical Engineering & Technology. 1992;16(5):194-203
  250. 250. Hoekema R, Venner K, Struijk JJ, Holsheimer J. Multigrid solution of the potential field in modeling electrical nerve stimulation. Computers and Biomedical Research. 1998;31(5):348-362
  251. 251. Rozman J, Sovinec B, Trlep M, Zorko B. Multielectrode spiral cuff for ordered and reversed activation of nerve fibres. Journal of Biomedical Engineering. 1993;15(2):113-120
  252. 252. Branner A, Normann RA. A multielectrode array for intrafascicular recording and stimulation in sciatic nerve of cats. Brain Research Bulletin. 2000;51(4):293-306
  253. 253. Sakaguchi H, Kamei M, Nishida K, Terasawa Y, Fujikado T, Ozawa M, et al. Implantation of a newly developed direct optic nerve electrode device for artificial vision in rabbits. Journal of Artificial Organs. 2012;15(3):295-300
  254. 254. Sakaguchi H, Fujikado T, Kanda H, Osanai M, Fang X, Nakauchi K, et al. Electrical stimulation with a needle-type electrode inserted into the optic nerve in rabbit eyes. Japanese Journal of Ophthalmology. 2004;48(6):552-557
  255. 255. Brelén ME, De Potter P, Gersdorff M, Cosnard G, Veraart C, Delbeke J. Intraorbital implantation of a stimulating electrode for an optic nerve visual prosthesis. Case report. Journal of Neurosurgery. 2006;104(4):593-597
  256. 256. Veraart C, Wanet-Defalque M-C, Gérard B, Vanlierde A, Delbeke J. Pattern recognition with the optic nerve visual prosthesis. Artificial Organs. 2003;27(11):996-1004
  257. 257. Sakaguchi H, Kamei M, Fujikado T, Yonezawa E, Ozawa M, Cecilia-Gonzalez C, et al. Artificial vision by direct optic nerve electrode (AV-DONE) implantation in a blind patient with retinitis pigmentosa. Journal of Artificial Organs. 2009;12(3):206-209
  258. 258. Brelén ME, Vince V, Gérard B, Veraart C, Delbeke J. Measurement of evoked potentials after electrical stimulation of the human optic nerve. Investigative Ophthalmology & Visual Science. 2010;51(10):5351-5355
  259. 259. Covington BP, Al Khalili Y. Neuroanatomy, nucleus lateral geniculate. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2024
  260. 260. Nguyen HT, Tangutooru SM, Rountree CM, Kantzos AJK, Tarlochan F, Yoon WJ, et al. Thalamic visual prosthesis. IEEE Transactions on Biomedical Engineering. 2016;63(8):1573-1580
  261. 261. Kyada MJ, Killian NJ, Pezaris JS. Thalamic visual prosthesis project. In: Gabel VP, editor. Artificial Vision. Cham: Springer International Publishing; 2017. pp. 177-189
  262. 262. Wiesel TN, Hubel DH. Spatial and chromatic interactions in the lateral geniculate body of the rhesus monkey. Journal of Neurophysiology. 1966;29(6):1115-1156
  263. 263. Pezaris JS, Reid RC. Demonstration of artificial visual percepts generated through thalamic microstimulation. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(18):7670-7675
  264. 264. Vurro M, Crowell AM, Pezaris JS. Simulation of thalamic prosthetic vision: Reading accuracy, speed, and acuity in sighted humans. Frontiers in Human Neuroscience. 2014;8:816
  265. 265. Rassia KEK, Moutoussis K, Pezaris JS. Reading text works better than watching videos to improve acuity in a simulation of artificial vision. Scientific Reports. 2022;12(1):12953
  266. 266. Troy JB, Nguyen HT, Meng C, Rountree CM. Electrode and stimulation parameters for a thalamic visual prosthesis. In: Qatar Foundation Annual Research Conference Proceedings; 2016 Mar; Doha, Qatar. Doha, Qatar: Hamad Bin Khalifa University Press (HBKU Press); 2016. Abstract HBPP1881
  267. 267. Pio-Lopez L, Poulkouras R, Depannemaecker D. Visual cortical prosthesis: An electrical perspective. Journal of Medical Engineering & Technology. 2021;45(5):394-407
  268. 268. Daniel PM, Whitteridge D. The representation of the visual field on the cerebral cortex in monkeys. Journal of Physiology (London). 1961;159:203-221
  269. 269. Tchoe Y, Bourhis AM, Cleary DR, Stedelin B, Lee J, Tonsfeldt KJ, et al. Human brain mapping with multithousand-channel PtNRGrids resolves spatiotemporal dynamics. Science Translational Medicine. 2022;14(628):eabj1441
  270. 270. Christie BP, Ashmont KR, House PA, Greger B. Approaches to a cortical vision prosthesis: Implications of electrode size and placement. Journal of Neural Engineering. 2016;13(2):025003
  271. 271. Beauchamp MS, Oswalt D, Sun P, Foster BL, Magnotti JF, Niketeghad S, et al. Dynamic stimulation of visual cortex produces form vision in sighted and blind humans. Cell. 2020;181(4):774-783.e5
  272. 272. Najarpour Foroushani A, Pack CC, Sawan M. Cortical visual prostheses: From microstimulation to functional percept. Journal of Neural Engineering. 2018;15(2):021005
  273. 273. Pollen DA. Some perceptual effects of electrical stimulation of the visual cortex in man. In: Tower DB, editor. The Nervous System, Vol. 2: The Clinical Neurosciences. New York: Raven Press; 1975. pp. 519-528
  274. 274. Dobelle WH, Mladejovsky MG. Phosphenes produced by electrical stimulation of human occipital cortex, and their application to the development of a prosthesis for the blind. Journal of Physiology (London). 1974;243(2):553-576
  275. 275. Brindley GS, Lewin WS. The sensations produced by electrical stimulation of the visual cortex. Journal of Physiology (London). 1968;196(2):479-493
  276. 276. Schmidt EM, Bak MJ, Hambrecht FT, Kufta CV, O’Rourke DK, Vallabhanath P. Feasibility of a visual prosthesis for the blind based on intracortical microstimulation of the visual cortex. Brain. 1996;119(Pt. 2):507-522
  277. 277. Fernández E, Alfaro A, González-López P. Toward long-term communication with the brain in the blind by Intracortical stimulation: Challenges and future prospects. Frontiers in Neuroscience. 2020;14:681
  278. 278. Troyk PR. The intracortical visual prosthesis project. In: Gabel VP, editor. Artificial Vision. Cham: Springer International Publishing; 2017. pp. 203-214
  279. 279. Coulombe J, Sawan M, Gervais JF. A highly flexible system for microstimulation of the visual cortex: Design and implementation. IEEE Transactions on Biomedical Circuits and Systems. 2007;1(4):258-269
  280. 280. Pelayo FJ, Martinez A, Romero S, Morillas CA, Ros E, Fernández E. Cortical visual neuro-prosthesis for the blind: Retina-like software/hardware preprocessor. In: IEEE-EMBS International Conference on Neural Engineering. Capri, Italy; Mar 2003
  281. 281. Li J, Zhou W, Liang L, Li Y, Xu K, Li X, et al. Noninvasive electrical stimulation as a neuroprotective strategy in retinal diseases: A systematic review of preclinical studies. Journal of Translational Medicine. 2024;22(1):28
  282. 282. Morimoto T, Miyoshi T, Sawai H, Fujikado T. Optimal parameters of transcorneal electrical stimulation (TES) to be neuroprotective of axotomized RGCs in adult rats. Experimental Eye Research. 2010;90(2):285-291
  283. 283. Kanai R, Chaieb L, Antal A, Walsh V, Paulus W. Frequency-dependent electrical stimulation of the visual cortex. Current Biology. 2008;18(23):1839-1843
  284. 284. Hébert M, Vaegan LP. Reproducibility of ERG responses obtained with the DTL electrode. Vision Research. 1999;39(6):1069-1070
  285. 285. Brouwer AH, de Wit GC, de Boer JH, van Genderen MM. Effects of DTL electrode position on the amplitude and implicit time of the electroretinogram. Documenta Ophthalmologica. 2020;140(3):201-209
  286. 286. Woo J, Jung S, Gauvin M, Lachapelle P. The DTL ERG electrode comes in different shapes and sizes: Are they all good? Documenta Ophthalmologica. 2017;135(2):155-164
  287. 287. Morimoto T, Kanda H, Kondo M, Terasaki H, Nishida K, Fujikado T. Transcorneal electrical stimulation promotes survival of photoreceptors and improves retinal function in rhodopsin P347L transgenic rabbits. Investigative Ophthalmology & Visual Science. 2012;53(7):4254-4261
  288. 288. Morimoto T, Fujikado T, Choi J-S, Kanda H, Miyoshi T, Fukuda Y, et al. Transcorneal electrical stimulation promotes the survival of photoreceptors and preserves retinal function in royal college of surgeons rats. Investigative Ophthalmology & Visual Science. 2007;48(10):4725-4732
  289. 289. Schatz A, Pach J, Gosheva M, Naycheva L, Willmann G, Wilhelm B, et al. Transcorneal electrical stimulation for patients with retinitis pigmentosa: A prospective, randomized, sham-controlled follow-up study over 1 year. Investigative Ophthalmology & Visual Science. 2017;58(1):257-269
  290. 290. Inomata K, Shinoda K, Ohde H, Tsunoda K, Hanazono G, Kimura I, et al. Transcorneal electrical stimulation of retina to treat longstanding retinal artery occlusion. Graefe's Archive for Clinical and Experimental Ophthalmology. 2007;245(12):1773-1780
  291. 291. Oono S, Kurimoto T, Kashimoto R, Tagami Y, Okamoto N, Mimura O. Transcorneal electrical stimulation improves visual function in eyes with branch retinal artery occlusion. Clinical Ophthalmology. 2011;5:397-402
  292. 292. Su X, Zhou M, Di L, Chen J, Zhai Z, Liang J, et al. The visual cortical responses to sinusoidal transcorneal electrical stimulation. Brain Research. 2022;1785:147875
  293. 293. Agadagba SK, Eldaly ABM, Chan LLH. Transcorneal electrical stimulation induces long-lasting enhancement of brain functional and directional connectivity in retinal degeneration mice. Frontiers in Cellular Neuroscience. 2022;16:785199
  294. 294. Agadagba SK, Eldaly ABM, Chan LLH. ECoG power alterations across stages of prolonged transcorneal electrical stimulation in the blind mice. Annual International Conference of the IEEE Engineering in Medicine and Biology Society. 2021;2021:5784-5787
  295. 295. Agadagba SK, Li X, Chan LLH. Excitation of the pre-frontal and primary visual cortex in response to transcorneal electrical stimulation in retinal degeneration mice. Frontiers in Neuroscience. 2020;14:572299
  296. 296. Xie J, Wang G-J, Yow L, Cela JC, Humayun MS, Weiland JD, et al. Modeling and percept of transcorneal electrical stimulation in humans. IEEE Transactions on Biomedical Engineering. 2011;58(7):1932-1939
  297. 297. Lee S, Park J, Kwon J, Kim DH, Im C-H. Multi-channel transorbital electrical stimulation for effective stimulation of posterior retina. Scientific Reports. 2021;11(1):9745
  298. 298. Yang M, Lennikov A, Chang K, Ashok A, Lee C, Cho K-S, et al. Transcorneal but not transpalpebral electrical stimulation disrupts mucin homeostasis of the ocular surface. BMC Ophthalmology. 2022;22(1):490
  299. 299. Thut G, Schyns PG, Gross J. Entrainment of perceptually relevant brain oscillations by non-invasive rhythmic stimulation of the human brain. Frontiers in Psychology. 2011;2:170
  300. 300. Gall C, Fedorov AB, Ernst L, Borrmann A, Sabel BA. Repetitive transorbital alternating current stimulation in optic neuropathy. NeuroRehabilitation. 2010;27(4):335-341
  301. 301. Fedorov A, Jobke S, Bersnev V, Chibisova A, Chibisova Y, Gall C, et al. Restoration of vision after optic nerve lesions with noninvasive transorbital alternating current stimulation: A clinical observational study. Brain Stimulation. 2011;4(4):189-201
  302. 302. Granata G, Falsini B. Preliminary results of transorbital alternating current stimulation in chronic low vision: Correlation of clinical and neurophysiological results. Neuromodulation. 2023;26(4):892-894
  303. 303. Gil-Carrasco F, Ochoa-Contreras D, Torres MA, Santiago-Amaya J, Pérez-Tovar FW, Gonzalez-Salinas R, et al. Transpalpebral electrical stimulation as a novel therapeutic approach to decrease intraocular pressure for open-angle glaucoma: A pilot study. Journal of Ophthalmology. 2018;2018:2930519
  304. 304. Yuan Y, Wang Z, Wang X, Yan J, Liu M, Li X. Low-intensity pulsed ultrasound stimulation induces coupling between ripple neural activity and hemodynamics in the mouse visual cortex. Cerebral Cortex. 2019;29(7):3220-3223
  305. 305. Jiang Q , Li G, Zhao H, Sheng W, Yue L, Su M, et al. Temporal neuromodulation of retinal ganglion cells by low-frequency focused ultrasound stimulation. IEEE Transactions on Neural Systems and Rehabilitation Engineering. 2018;26(5):969-976
  306. 306. Fomenko A, Neudorfer C, Dallapiazza RF, Kalia SK, Lozano AM. Low-intensity ultrasound neuromodulation: An overview of mechanisms and emerging human applications. Brain Stimulation. 2018;11(6):1209-1217
  307. 307. Baek H, Pahk KJ, Kim H. A review of low-intensity focused ultrasound for neuromodulation. Biomedical Engineering Letters. 2017;7(2):135-142
  308. 308. Cafarelli A, Marino A, Vannozzi L, Puigmartí-Luis J, Pané S, Ciofani G, et al. Piezoelectric nanomaterials activated by ultrasound: The pathway from discovery to future clinical adoption. ACS Nano. 2021;15(7):11066-11086
  309. 309. Sarica C, Fomenko A, Nankoo J-F, Darmani G, Vetkas A, Yamamoto K, et al. Toward focused ultrasound neuromodulation in deep brain stimulator implanted patients: Ex-vivo thermal, kinetic and targeting feasibility assessment. Brain Stimulation. 2022;15(2):376-379
  310. 310. Blackmore J, Shrivastava S, Sallet J, Butler CR, Cleveland RO. Ultrasound neuromodulation: A review of results, mechanisms and safety. Ultrasound in Medicine & Biology. 2019;45(7):1509-1536
  311. 311. Lee W, Kim H, Jung Y, Song I-U, Chung YA, Yoo S-S. Image-guided transcranial focused ultrasound stimulates human primary somatosensory cortex. Scientific Reports. 2015;5:8743
  312. 312. Lee W, Kim H-C, Jung Y, Chung YA, Song I-U, Lee J-H, et al. Transcranial focused ultrasound stimulation of human primary visual cortex. Scientific Reports. 2016;6:34026
  313. 313. Dalecki D. Mechanical bioeffects of ultrasound. Annual Review of Biomedical Engineering. 2004;6:229-248
  314. 314. O’Brien WD. Ultrasound-biophysics mechanisms. Progress in Biophysics and Molecular Biology. 2007;93(1-3):212-255
  315. 315. Kamimura HAS, Conti A, Toschi N, Konofagou EE. Ultrasound neuromodulation: Mechanisms and the potential of multimodal stimulation for neuronal function assessment. Frontiers of Physics. 2020;8:150
  316. 316. Lu G, Qian X, Castillo J, Li R, Jiang L, Lu H, et al. Transcranial focused ultrasound for noninvasive neuromodulation of the visual cortex. IEEE Transactions on Ultrasonics, Ferroelectrics, and Frequency Control. 2021;68(1):21-28
  317. 317. Naor O, Krupa S, Shoham S. Ultrasonic neuromodulation. Journal of Neural Engineering. 2016;13(3):031003
  318. 318. Kim H, Park MY, Lee SD, Lee W, Chiu A, Yoo S-S. Suppression of EEG visual-evoked potentials in rats through neuromodulatory focused ultrasound. Neuroreport. 2015;26(4):211-215
  319. 319. Legon W, Bansal P, Tyshynsky R, Ai L, Mueller JK. Transcranial focused ultrasound neuromodulation of the human primary motor cortex. Scientific Reports. 2018;8(1):10007
  320. 320. Potok W, van der Groen O, Bachinger M, Edwards D, Wenderoth N. Transcranial random noise stimulation modulates neural processing of sensory and motor circuits, from potential cellular mechanisms to behavior: A scoping review. eNeuro. 2022;9:5
  321. 321. Moret B, Donato R, Nucci M, Cona G, Campana G. Transcranial random noise stimulation (tRNS): A wide range of frequencies is needed for increasing cortical excitability. Scientific Reports. 2019;9(1):15150
  322. 322. Schoisswohl S, Langguth B, Gebel N, Poeppl TB, Kreuzer PM, Schecklmann M. Electrophysiological evaluation of high and low-frequency transcranial random noise stimulation over the auditory cortex. Progress in Brain Research. 2021;263:95-108
  323. 323. Joos K, De Ridder D, Vanneste S. The differential effect of low- versus high-frequency random noise stimulation in the treatment of tinnitus. Experimental Brain Research. 2015;233(5):1433-1440
  324. 324. Yamashiro K, Ikarashi K, Makibuchi T, Anazawa S, Baba Y, Fujimoto T, et al. Transcranial high-frequency random noise stimulation does not modulate Nogo N2 and Go/Nogo reaction times in somatosensory and auditory modalities. Scientific Reports. 2023;13(1):3014
  325. 325. Sánchez-León CA, Sánchez-López Á, Gómez-Climent MA, Cordones I, Cohen Kadosh R, Márquez-Ruiz J. Impact of chronic transcranial random noise stimulation (tRNS) on GABAergic and glutamatergic activity markers in the prefrontal cortex of juvenile mice. Progress in Brain Research. 2021;264:323-341
  326. 326. Remedios L, Mabil P, Flores-Hernández J, Torres-Ramírez O, Huidobro N, Castro G, et al. Effects of short-term random noise electrical stimulation on dissociated pyramidal neurons from the cerebral cortex. Neuroscience. 2019;404:371-386
  327. 327. Chaieb L, Antal A, Paulus W. Transcranial random noise stimulation-induced plasticity is NMDA-receptor independent but sodium-channel blocker and benzodiazepines sensitive. Frontiers in Neuroscience. 2015;9:125
  328. 328. Onorato I, D’Alessandro G, Di Castro MA, Renzi M, Dobrowolny G, Musarò A, et al. Noise enhances action potential generation in mouse sensory neurons via stochastic resonance. PLoS One. 2016;11(8):e0160950
  329. 329. Vanneste S, Fregni F, De Ridder D. Head-to-head comparison of transcranial random noise stimulation, transcranial AC stimulation, and transcranial DC stimulation for tinnitus. Frontiers in Psychiatry. 2013;4:158
  330. 330. Murphy OW, Hoy KE, Wong D, Bailey NW, Fitzgerald PB, Segrave RA. Transcranial random noise stimulation is more effective than transcranial direct current stimulation for enhancing working memory in healthy individuals: Behavioural and electrophysiological evidence. Brain Stimulation. 2020;13(5):1370-1380
  331. 331. van der Groen O, Wenderoth N. Transcranial random noise stimulation of visual cortex: Stochastic resonance enhances central mechanisms of perception. The Journal of Neuroscience. 2016;36(19):5289-5298
  332. 332. Maniglia M, Soler V, Cottereau B, Trotter Y. Spontaneous and training-induced cortical plasticity in MD patients: Hints from lateral masking. Scientific Reports. 2018;8(1):90
  333. 333. Maniglia M, Soler V, Trotter Y. Combining fixation and lateral masking training enhances perceptual learning effects in patients with macular degeneration. Journal of Vision. 2020;20(10):19
  334. 334. Herpich F, Melnick MD, Agosta S, Huxlin KR, Tadin D, Battelli L. Boosting learning efficacy with noninvasive brain stimulation in intact and brain-damaged humans. The Journal of Neuroscience. 2019;39(28):5551-5561
  335. 335. Contemori G, Trotter Y, Cottereau BR, Maniglia M. tRNS boosts perceptual learning in peripheral vision. Neuropsychologia. 2019;125:129-136
  336. 336. Pirulli C, Fertonani A, Miniussi C. The role of timing in the induction of neuromodulation in perceptual learning by transcranial electric stimulation. Brain Stimulation. 2013;6(4):683-689
  337. 337. Contemori G, Maniglia M, Guénot J, Soler V, Cherubini M, Cottereau BR, et al. tRNS boosts visual perceptual learning in participants with bilateral macular degeneration. Frontiers in Aging Neuroscience. 2024;16:1326435
  338. 338. Parnami K, Bhattacharyya A. Current approaches to vision restoration using optogenetic therapy. Frontiers in Cellular Neuroscience. 2023;17:1236826
  339. 339. Provansal M, Marazova K, Sahel JA, Picaud S. Vision restoration by optogenetic therapy and developments toward sonogenetic therapy. Translational Vision Science & Technology. 2022;11(1):18
  340. 340. Zheng J, Zhang W, Liu L, Hung Yap MK. Low frequency repetitive transcranial magnetic stimulation promotes plasticity of the visual cortex in adult amblyopic rats. Frontiers in Neuroscience. 2023;17:1109735
  341. 341. Qian X, Lu G, Thomas BB, Li R, Chen X, Shung KK, et al. Noninvasive ultrasound retinal stimulation for vision restoration at high spatiotemporal resolution. BME Frontiers. 2022;2022:9829316
  342. 342. Benson NC, Butt OH, Brainard DH, Aguirre GK. Correction of distortion in flattened representations of the cortical surface allows prediction of V1-V3 functional organization from anatomy. PLoS Computational Biology. 2014;10(3):e1003538
  343. 343. Chen Y, Aguirre AD, Ruvinskaya L, Devor A, Boas DA, Fujimoto JG. Optical coherence tomography (OCT) reveals depth-resolved dynamics during functional brain activation. Journal of Neuroscience Methods. 2009;178(1):162-173

Written By

Nour Shaheen, Mohamed Khaled, Serah Seo, Yarema Bezchlibnyk, Oliver Flouty and Vishal Bharmauria

Submitted: 10 May 2024 Reviewed: 20 May 2024 Published: 01 July 2024