Optogenetics and its Potential Role in the Treatment of Epilepsy

Editorial

Austin J Pharmacol Ther. 2014; 2 (4). 1023

Optogenetics and its Potential Role in the Treatment of Epilepsy

Yi Luo1*and Man-Qi Li2

1Department of Pharmacology, Huazhong University of Science and Technology, China

2Department of Foreign Language, Yangtze University, China

*Corresponding author: : Yi Luo, Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China

Received: June 01, 2014; Accepted: June 02, 2014; Published: June 03, 2014

Editorial

Optogenetics, as the term has been commonly used, refers to the technology to use light to control neuron or neuronal network that has been specially sensitized to light [1]. The peak activation wavelength of light is mostly between 400 to 650 nm [2]. It has been firstly introduced into neuroscience since 2005 [3]. It is an integration of opics and genetics to control the activity of special neurons in living tissue. For example microbial opsin genes can be introduced in to a specific targeted neuron or neuronal population, then the effects of those manipulations can be precisely measured in real–time. The key factor used in optogenetisc is light–sensitive protein, usually referred to opsin. We can achieve gain–or loss–of function of the targeted neuron or neuronal network using different microbial opsins.

According to the function of opsins, there are three major types. A type such as Channelrhodopsin 2 (ChR2) induces the targeted to be depolarization and enables action potential elicited to light pulses. B type such as halorhodopsin (NpHR) and proton pumps enable the targeted to be hyperpolarization to prevent the induction of action potentials. C type such as light–activated membrane–bound G protein–coupled (OptoXR) or soluble (bacterial cyclase) receptors that minic various signaling cascades. The cascade is usually Gq, Gs or Gi signaling.

Apart from A type of opsins, B type is used to light to silence electrical activity in targeted cells. Available B type such as NpHR that directly move ions to achieve silencing is inefficient. Only a single ion per photon across the cell membrane is pumped rather than many ions per photon are allowed to flow through a channel pore. Recently, a class of ChR, which is originally cation–conducting, has been converted into chloride–conducting anion channels [4]. The new class of ChR enables fast optical inhibition of action potentials and displays step–function kinetics for stable inhibition of targeted neuron. Moreover, it is more sensitive to light than NpHR. An approach that is more physiological, efficient, and sensitive optogenetic inhibition has come to us.

Nearly 1% of people worldwide (70 million) have epilepsy [5]. Epilepsy is a common neurological disorders characterized by epileptic seizures. Epilepsy cannot be cured, but seizures can be controlled with medication. The seizures are episodes that vary from brief or almost undetectable to a long period of vigorous shaking. Seizure is the clinical manifestation of a hyperexcitable network, in which the electrical balance underlying normal neuronal activity is pathologically altered.

There are many types of Anti–Epileptic Drugs (AEDs) to different targets. Generally speaking, there are ten classes of targets [6]. A. Voltage–gated ion channels, such as voltage–gated sodium channel, voltage–gated calcium channel, voltage–gated potassium channel, the Hyperpolarization–activated Cyclic Nucleotide–gated cation (HCN) channel, voltage–gated chloride channel. B. Ligandgated ion channels, such as GABAA receptor, nicotinic cholinergic receptor, glycine receptor. C. Ionotropic glutamate receptors, such as NMDA receptor, AMPA receptor. D. Acid–sensing ion channels. E. G–Protein–Coupled Receptors (GPCRs). F. The metabotropic glutamate metabotropic receptors, such as GABAB receptor. G. Neurotransmitter transporters, such as plasma membrane GABA transporter, plasma membrane glutamate transporters, vesicular glutamate transporters. H. Presynaptic proteins which influence synaptic function, such as synaptic vesicle protein, synaptic anchoring proteins. I. Enzymes, such as GABA–transaminase, carbonic anhydrase, protein kinases and phosphatases. J. Gap junctions (connexins). So far the most important classes of targets for currently marketed AEDs are voltage–gated and ligand–gated ion channels. Other targets have been investigated, recently, it is reportedthat hydrogen sulfide aggravates Seizure–Like Events (SLEs) of rats in vivo and in vitro, which may be due to an increase in neuronal excitation [7]. Enzymes that control hydrogen sulfide biosynthesis are potential new targets for the treatment of epilepsy. Although lotsof AEDs are available, effective symptomatic relief is achieved only in about two thirds of the patients [8]. Surgery, neurostimulation or dietary changes may be considered for seizures that do not respond to medication.

As hyperexcitable network is the key factor underline seizure generation, reducing the excitability of network is the goal for treating epilepsy. As mentioned above, the new class of ChR enables fast optical inhibition of action potentials. This new opsin is supposed to directly and completely terminate epileptic discharge by light pulses.

The vast majority of optogenetic study to today has been carried out in mouse or rats. Viral vector delivery, which introduced the opsin to cells, to the primate brain has been well established and used for limited forms of neuromodulation [9] . Two optogenetic works on primate have been performed, demonstrating for the first time that optogenetics can be used in rhesus macaques [10,11] . Given that mammalian optogenetics is just at the beginning, it still has a lot of progress made in non–human primate and toward human use. However, significant technical obstacles are still remained, requiring advances in opsin engineering, vector delivery, and light pulse among other fields. For example, the brain of rhesus is approximately 250 times larger than rat while the brain of human is approximately 10 times larger than rhesus [12]. Thorough safety studies will be required prior to any human use. Moreover, the safety of the expression of opsin and delivery devices of light will need to be tested. Just now, the histological data available from the non–human primate work described above are conducted for several months, in this short time frame, no large–scale neuronal damage is reported [10,11,13].

It is thoughtful that optogenetics with the new class of ChR will be used in the treatment of epilepsy in the foreseeable future.

References

  1. Deisseroth K, Schnitzer MJ. Engineering approaches to illuminating brain structure and dynamics. Neuron. 2013; 80: 568-577.
  2. Tye KM, Deisseroth K. Optogenetic investigation of neural circuits underlying brain disease in animal models. Nat Rev Neurosci. 2012; 13: 251-266.
  3. Boyden ES, Zhang F, Bamberg E, Nagel G, Deisseroth K. Millisecond-timescale, genetically targeted optical control of neural activity. Nat Neurosci. 2005; 8: 1263-1268.
  4. Berndt A, Lee SY, Ramakrishnan C, Deisseroth K. Structure-guided transformation of channelrhodopsin into a light-activated chloride channel. Science. 2014; 344: 420-424.
  5. Thurman DJ, Beghi E, Begley CE, Berg AT, Buchhalter JR, Ding D, et al. Standards for epidemiologic studies and surveillance of epilepsy. Epilepsia. 2011; 52 Suppl 7: 2-26.
  6. Meldrum BS, Rogawski MA. Molecular targets for antiepileptic drug development. Neurotherapeutics. 2007; 4: 18-61.
  7. Luo Y, Wu PF, Zhou J, Xiao W, He JG, Guan XL, et al. Aggravation of seizure-like events by hydrogen sulfide: involvement of multiple targets that control neuronal excitability. CNS Neurosci Ther. 2014; 20: 411-419.
  8. Eadie MJ. Shortcomings in the current treatment of epilepsy. Expert Rev Neurother. 2012; 12: 1419-1427.
  9. Tan EM, Yamaguchi Y, Horwitz GD, Gosgnach S, Lein ES, Goulding M, et al. Selective and quickly reversible inactivation of mammalian neurons in vivo using the Drosophila allatostatin receptor. Neuron. 2006; 51: 157-170.
  10. Han X, Qian X, Bernstein JG, Zhou HH, Franzesi GT, Stern P, et al. Millisecond-timescale optical control of neural dynamics in the nonhuman primate brain. Neuron. 2009; 62: 191-198.
  11. Diester I, Kaufman MT, Mogri M, Pashaie R, Goo W, Yizhar O, et al. An optogenetic toolbox designed for primates. Nat Neurosci. 2011; 14: 387-397.
  12. Kalanithi PS, Henderson JM. Optogenetic neuromodulation. Int Rev Neurobiol. 2012; 107: 185-205.
  13. Han X, Chow BY, Zhou H, Klapoetke NC, Chuong A, Rajimehr R, et al. A high-light sensitivity optical neural silencer: development and application to optogenetic control of non-human primate cortex. Front Syst Neurosci. 2011; 5: 18.

Download PDF

Citation: Yi Luo and Man-Qi Li. Optogenetics and its Potential Role in the Treatment of Epilepsy. Austin J Pharmacol Ther. 2014; 2 (4). 1023. ISSN: 2373-6208.

Home
Journal Scope
Online First
Current Issue
Editorial Board
Instruction for Authors
Submit Your Article
Contact Us