And Then There Was Light: Perspectives of Optogenetics for Deep Brain Stimulation and Neuromodulation

Deep Brain Stimulation (DBS) has evolved into a well-accepted add-on treatment for patients with severe Parkinsons disease as well as for other chronic neurological conditions. The focal action of electrical stimulation can yield better responses and it exposes the patient to fewer side effects compared to pharmaceuticals distributed throughout the body toward the brain. On the other hand, the current practice of DBS is hampered by the relatively coarse level of neuromodulation achieved. Optogenetics, in contrast, offers the perspective of much more selective actions on the various physiological structures, provided that the stimulated cells are rendered sensitive to the action of light. Optogenetics has experienced tremendous progress since its first in vivo applications about 10 years ago. Recent advancements of viral vector technology for gene transfer substantially reduce vector-associated cytotoxicity and immune responses. This brings about the possibility to transfer this technology into the clinic as a possible alternative to DBS and neuromodulation. New paths could be opened toward a rich panel of clinical applications. Some technical issues still limit the long term use in humans but realistic perspectives quickly emerge. Despite a rapid accumulation of observations about patho-physiological mechanisms, it is still mostly serendipity and empiric adjustments that dictate clinical practice while more efficient logically designed interventions remain rather exceptional. Interestingly, it is also very much the neuro technology developed around optogenetics that offers the most promising tools to fill in the existing knowledge gaps about brain function in health and disease. The present review examines Parkinson's disease and refractory epilepsy as use cases for possible optogenetic stimulation therapies.

[1]  W. Carlezon,et al.  Use of Adeno‐Associated and Herpes Simplex Viral Vectors for In Vivo Neuronal Expression in Mice , 2015, Current protocols in neuroscience.

[2]  S. Gray,et al.  Methods for gene transfer to the central nervous system. , 2014, Advances in genetics.

[3]  J. Bennett,et al.  A deviant immune response to viral proteins and transgene product is generated on subretinal administration of adenovirus and adeno-associated virus. , 2002, Molecular therapy : the journal of the American Society of Gene Therapy.

[4]  Hyung Ho Yoon,et al.  Optogenetic inactivation of the subthalamic nucleus improves forelimb akinesia in a rat model of Parkinson disease. , 2014, Neurosurgery.

[5]  Patrick Degenaar,et al.  A Processing Platform for Optoelectronic/Optogenetic Retinal Prosthesis , 2013, IEEE Transactions on Biomedical Engineering.

[6]  I. Soltesz,et al.  On-demand optogenetic control of spontaneous seizures in temporal lobe epilepsy , 2013, Nature Communications.

[7]  Christofer Toumazou,et al.  Modeling Study of the Light Stimulation of a Neuron Cell With Channelrhodopsin-2 Mutants , 2011, IEEE Transactions on Biomedical Engineering.

[8]  Mark A. Rossi,et al.  A wirelessly controlled implantable LED system for deep brain optogenetic stimulation , 2015, Front. Integr. Neurosci..

[9]  K. Thompson,et al.  Overview on Research and Clinical Applications of Optogenetics , 2016, Current protocols in pharmacology.

[10]  Ling Wang,et al.  Light distribution and thermal effects in the rat brain under optogenetic stimulation , 2016, Journal of biophotonics.

[11]  R. Ransohoff,et al.  Development, maintenance and disruption of the blood-brain barrier , 2013, Nature Medicine.

[12]  Dimiter Prodanov,et al.  Mechanical and Biological Interactions of Implants with the Brain and Their Impact on Implant Design , 2016, Front. Neurosci..

[13]  A. Zorzos,et al.  Multiwaveguide implantable probe for light delivery to sets of distributed brain targets. , 2010, Optics letters.

[14]  W. C. Hall,et al.  High-speed mapping of synaptic connectivity using photostimulation in Channelrhodopsin-2 transgenic mice , 2007, Proceedings of the National Academy of Sciences.

[15]  Xindong Liu,et al.  Microelectrode array for chronic deep-brain microstimulation and recording , 2006, IEEE Transactions on Biomedical Engineering.

[16]  R. Ramakrishnan,et al.  Deletion of multiple immediate–early genes from herpes simplex virus reduces cytotoxicity and permits long-term gene expression in neurons , 1998, Gene Therapy.

[17]  J. Uney,et al.  Using viral vectors as gene transfer tools (Cell Biology and Toxicology Special Issue: ETCS-UK 1 day meeting on genetic manipulation of cells) , 2009, Cell Biology and Toxicology.

[18]  A. Benabid,et al.  Chronic electrical stimulation of the ventralis intermedius nucleus of the thalamus as a treatment of movement disorders. , 1996, Journal of neurosurgery.

[19]  Robert Puers,et al.  Low Loss CMOS-Compatible PECVD Silicon Nitride Waveguides and Grating Couplers for Blue Light Optogenetic Applications , 2016, IEEE Photonics Journal.

[20]  Richard O. Snyder,et al.  Adeno-Associated Virus , 2011, Methods in Molecular Biology.

[21]  Eran Stark,et al.  Diode probes for spatiotemporal optical control of multiple neurons in freely moving animals. , 2012, Journal of neurophysiology.

[22]  Olivier Marre,et al.  Red‐shifted channelrhodopsin stimulation restores light responses in blind mice, macaque retina, and human retina , 2016, EMBO molecular medicine.

[23]  Donna Webb,et al.  Coupling optogenetic stimulation with NanoLuc-based luminescence (BRET) Ca++ sensing , 2016, Nature Communications.

[24]  J. Volkmann,et al.  Introduction to the programming of deep brain stimulators , 2002, Movement disorders : official journal of the Movement Disorder Society.

[25]  Jack A. Wells,et al.  fMRI response to blue light delivery in the naïve brain: Implications for combined optogenetic fMRI studies , 2013, NeuroImage.

[26]  Steve S. Chung,et al.  Long-term efficacy and safety of thalamic stimulation for drug-resistant partial epilepsy , 2015, Neurology.

[27]  Xue Han,et al.  In vivo application of optogenetics for neural circuit analysis. , 2012, ACS chemical neuroscience.

[28]  James M. Wilson,et al.  Gene Therapy Vectors Based on Adeno-Associated Virus Type 1 , 1999, Journal of Virology.

[29]  Shriya Srinivasan,et al.  Transdermal optogenetic peripheral nerve stimulation , 2017, Journal of neural engineering.

[30]  F. Lim,et al.  General Considerations on the Biosafety of Virus-derived Vectors Used in Gene Therapy and Vaccination , 2013, Current gene therapy.

[31]  Dinh Tuan Vo Biomedical photonics handbook , 2003 .

[32]  E. Barcelon,et al.  Optogenetic Glia Manipulation: Possibilities and Future Prospects , 2016, Experimental neurobiology.

[33]  Weien Yuan,et al.  Current Experimental Studies of Gene Therapy in Parkinson's Disease , 2017, Front. Aging Neurosci..

[34]  Michael Z. Lin,et al.  Characterization of engineered channelrhodopsin variants with improved properties and kinetics. , 2009, Biophysical journal.

[35]  S. Duan,et al.  Entorhinal Principal Neurons Mediate Brain-stimulation Treatments for Epilepsy , 2016, EBioMedicine.

[36]  Takeharu Nagai,et al.  Genetically encoded bioluminescent voltage indicator for multi-purpose use in wide range of bioimaging , 2017, Scientific Reports.

[37]  Denise M. Piscopo,et al.  Rhythmic brain stimulation reduces anxiety-related behavior in a mouse model based on meditation training , 2017, Proceedings of the National Academy of Sciences.

[38]  Jason Chung,et al.  Long-term channelrhodopsin-2 (ChR2) expression can induce abnormal axonal morphology and targeting in cerebral cortex , 2013, Front. Neural Circuits.

[39]  Christian Hauptmann,et al.  Coordinated reset has sustained aftereffects in Parkinsonian monkeys , 2012, Annals of neurology.

[40]  M. Häusser,et al.  All-Optical Interrogation of Neural Circuits , 2015, The Journal of Neuroscience.

[41]  T. Eggerman,et al.  Rapid Kupffer cell death after intravenous injection of adenovirus vectors. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[42]  V. Haurigot,et al.  Safety of AAV Factor IX Peripheral Transvenular Gene Delivery to Muscle in Hemophilia B Dogs , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[43]  J. R. Augustine,et al.  Evolution and rebirth of functional stereotaxy in the subthalamus. , 2013, World neurosurgery.

[44]  Chethan Pandarinath,et al.  Retinal prosthetic strategy with the capacity to restore normal vision , 2012, Proceedings of the National Academy of Sciences.

[45]  O. Yoshida,et al.  Herpes simplex viral-vector design for efficient transduction of nonneuronal cells without cytotoxicity , 2015, Proceedings of the National Academy of Sciences.

[46]  Alex J Walsh,et al.  Action potential block in neurons by infrared light. , 2016, Neurophotonics.

[47]  J. Wickens,et al.  Interfacing with Neural Activity via Femtosecond Laser Stimulation of Drug-Encapsulating Liposomal Nanostructures , 2016, eNeuro.

[48]  M. Hayden,et al.  Correction of feline lipoprotein lipase deficiency with adeno-associated virus serotype 1-mediated gene transfer of the lipoprotein lipase S447X beneficial mutation. , 2006, Human gene therapy.

[49]  S. Kleinlogel Optogenetic user's guide to Opto-GPCRs. , 2016, Frontiers in bioscience.

[50]  R. Herzog,et al.  Progress and prospects: immune responses to viral vectors , 2010, Gene Therapy.

[51]  J. Rasko,et al.  Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response , 2006, Nature Medicine.

[52]  Leonardo Sileo,et al.  Micro- and Nanotechnologies for Optical Neural Interfaces , 2016, Front. Neurosci..

[53]  Hyung Ho Yoon,et al.  Optogenetic Inhibition of the Subthalamic Nucleus Reduces Levodopa-Induced Dyskinesias in a Rat Model of Parkinson's Disease , 2016, Stereotactic and Functional Neurosurgery.

[54]  Jing Wang,et al.  Integrated device for combined optical neuromodulation and electrical recording for chronic in vivo applications , 2012, Journal of neural engineering.

[55]  Mohamed S. Emara,et al.  Dynamic illumination of spatially restricted or large brain volumes via a single tapered optical fiber , 2017, Nature Neuroscience.

[56]  Andrei Faraon,et al.  Patterned photostimulation via visible-wavelength photonic probes for deep brain optogenetics , 2016, Neurophotonics.

[57]  B. Roska,et al.  Emerging therapies for inherited retinal degeneration , 2016, Science Translational Medicine.

[58]  R. Herzog,et al.  Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. , 2009, Blood.

[59]  Katja M Arndt,et al.  Strategies for the photo-control of endogenous protein activity. , 2017, Current opinion in structural biology.

[60]  P. O'Suilleabhain,et al.  Tremor response to polarity, voltage, pulsewidth and frequency of thalamic stimulation , 2003, Neurology.

[61]  Niklas Smedemark-Margulies,et al.  Tools, methods, and applications for optophysiology in neuroscience , 2013, Front. Mol. Neurosci..

[62]  Vinay Goyal,et al.  Parkinson's disease: A review , 2018, Neurology India.

[63]  Warren M Grill,et al.  Implanted neural interfaces: biochallenges and engineered solutions. , 2009, Annual review of biomedical engineering.

[64]  Edward B. Miller,et al.  Alpha2,3 and alpha2,6 N-linked sialic acids facilitate efficient binding and transduction by adeno-associated virus types 1 and 6. , 2006, Journal of virology.

[65]  Gil Bub,et al.  All‐optical control of cardiac excitation: combined high‐resolution optogenetic actuation and optical mapping , 2016, The Journal of physiology.

[66]  Pui-In Mak,et al.  An Integrated Circuit for Simultaneous Extracellular Electrophysiology Recording and Optogenetic Neural Manipulation , 2017, IEEE Transactions on Biomedical Engineering.

[67]  B. Pál,et al.  Astrocyte-Dependent Slow Inward Currents (SICs) Participate in Neuromodulatory Mechanisms in the Pedunculopontine Nucleus (PPN) , 2017, Front. Cell. Neurosci..

[68]  M. Clyde,et al.  Clinical response to varying the stimulus parameters in deep brain stimulation for essential tremor , 2006, Movement disorders : official journal of the Movement Disorder Society.

[69]  Steffen B. E. Wolff,et al.  A polymer-based neural microimplant for optogenetic applications: design and first in vivo study. , 2013, Lab on a chip.

[70]  Thomas J. Davidson,et al.  Bidirectional Control of Generalized Epilepsy Networks via Rapid Real-Time Switching of Firing Mode , 2017, Neuron.

[71]  S. Randell,et al.  Terminal N-Linked Galactose Is the Primary Receptor for Adeno-associated Virus 9* , 2011, The Journal of Biological Chemistry.

[72]  K. Eliceiri,et al.  Blue Light Modulates Murine Microglial Gene Expression in the Absence of Optogenetic Protein Expression , 2016, Scientific Reports.

[73]  A. Rezai,et al.  AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomised trial , 2011, The Lancet Neurology.

[74]  M. Fee,et al.  Using temperature to analyze temporal dynamics in the songbird motor pathway , 2008, Nature.

[75]  Christoph Zrenner,et al.  Closed-Loop Neuroscience and Non-Invasive Brain Stimulation: A Tale of Two Loops , 2016, Front. Cell. Neurosci..

[76]  Kishan Dholakia,et al.  Fast targeted gene transfection and optogenetic modification of single neurons using femtosecond laser irradiation , 2013, Scientific Reports.

[77]  Murtaza Z Mogri,et al.  Optical Deconstruction of Parkinsonian Neural Circuitry , 2009, Science.

[78]  R. H. Smith Adeno-associated virus integration: virus versus vector , 2008, Gene Therapy.

[79]  Keyun Qing,et al.  Adeno-associated virus 2 co-receptors?-first reply , 1999, Nature Medicine.

[80]  Karl Deisseroth,et al.  Closed-Loop and Activity-Guided Optogenetic Control , 2015, Neuron.

[81]  N. Bessis,et al.  Immune responses to gene therapy vectors: influence on vector function and effector mechanisms , 2004, Gene therapy.

[82]  E. Bamberg,et al.  Channelrhodopsin-1: A Light-Gated Proton Channel in Green Algae , 2002, Science.

[83]  K. Suk,et al.  Reversible Induction of Pain Hypersensitivity following Optogenetic Stimulation of Spinal Astrocytes. , 2016, Cell reports.

[84]  Charles B. Mikell,et al.  Deep brain stimulation: a mechanistic and clinical update. , 2013, Neurosurgical focus.

[85]  J. French,et al.  Refractory Epilepsy: One Size Does Not Fit All , 2006, Epilepsy currents.

[86]  E. S. Gibson,et al.  Optogenetic Control of Synaptic Composition and Function , 2017, Neuron.

[87]  Edward B. Miller,et al.  α2,3 and α2,6 N-Linked Sialic Acids Facilitate Efficient Binding and Transduction by Adeno-Associated Virus Types 1 and 6 , 2006, Journal of Virology.

[88]  Simon C Watkins,et al.  Prolonged gene expression and cell survival after infection by a herpes simplex virus mutant defective in the immediate-early genes encoding ICP4, ICP27, and ICP22 , 1996, Journal of virology.

[89]  B. Bacskai,et al.  Optogenetic Restoration of Disrupted Slow Oscillations Halts Amyloid Deposition and Restores Calcium Homeostasis in an Animal Model of Alzheimer’s Disease , 2017, PloS one.

[90]  Mufti Mahmud,et al.  Differential Modulation of Excitatory and Inhibitory Neurons during Periodic Stimulation , 2016, Front. Neurosci..

[91]  Sadegh Nabavi,et al.  Engineering a memory with LTD and LTP , 2014, Nature.

[92]  Simon R. Schultz,et al.  Prospects for Optogenetic Augmentation of Brain Function , 2015, Front. Syst. Neurosci..

[93]  G. Buzsáki,et al.  Tools for probing local circuits: high-density silicon probes combined with optogenetics , 2015, Neuron.

[94]  F. Bezanilla,et al.  Photosensitivity of Neurons Enabled by Cell-Targeted Gold Nanoparticles , 2015, Neuron.

[95]  J. Mittelstrass,et al.  What is transdisciplinarity ? , 2003 .

[96]  D.B. McCreery,et al.  A characterization of the effects on neuronal excitability due to prolonged microstimulation with chronically implanted microelectrodes , 1997, IEEE Transactions on Biomedical Engineering.

[97]  Bard Ermentrout,et al.  Optogenetic Stimulation Shifts the Excitability of Cerebral Cortex from Type I to Type II: Oscillation Onset and Wave Propagation , 2017, PLoS Comput. Biol..

[98]  K. Dikranian,et al.  Surgical extraction of human dorsal root ganglia from organ donors and preparation of primary sensory neuron cultures , 2016, Nature Protocols.

[99]  Kevin T. Beier,et al.  A Brainstem-Spinal Cord Inhibitory Circuit for Mechanical Pain Modulation by GABA and Enkephalins , 2017, Neuron.

[100]  J. Sommer,et al.  Effects of transient immunosuppression on adenoassociated, virus-mediated, liver-directed gene transfer in rhesus macaques and implications for human gene therapy. , 2006, Blood.

[101]  Giovanni Di Pasquale,et al.  Identification of PDGFR as a receptor for AAV-5 transduction , 2003, Nature Medicine.

[102]  F. B. Johnson,et al.  Attachment of adeno-associated virus type 3H to fibroblast growth factor receptor 1 , 2006, Archives of Virology.

[103]  Karl Deisseroth,et al.  Optogenetics in Neural Systems , 2011, Neuron.

[104]  W. Jagust,et al.  Safety and tolerability of putaminal AADC gene therapy for Parkinson disease , 2009, Neurology.

[105]  Ahmed Soltan,et al.  High density, high radiance μLED matrix for optogenetic retinal prostheses and planar neural stimulation , 2017, IEEE Transactions on Biomedical Circuits and Systems.

[106]  V. Haurigot,et al.  Peripheral transvenular delivery of adeno-associated viral vectors to skeletal muscle as a novel therapy for hemophilia B. , 2010, Blood.

[107]  Patrick Ruther,et al.  Short and long term biocompatibility of NeuroProbes silicon probes , 2010, Journal of Neuroscience Methods.

[108]  R. Samulski,et al.  Viral vectors for gene delivery to the central nervous system , 2012, Neurobiology of Disease.

[109]  Kristin N. Parent,et al.  Structural Characterization of the Dual Glycan Binding Adeno-Associated Virus Serotype 6 , 2010, Journal of Virology.

[110]  Jie Tian,et al.  Adenovirus Activates Complement by Distinctly Different Mechanisms In Vitro and In Vivo: Indirect Complement Activation by Virions In Vivo , 2009, Journal of Virology.

[111]  D. Goldstein,et al.  Correction of a rat model of Parkinson's disease by coexpression of tyrosine hydroxylase and aromatic amino acid decarboxylase from a helper virus-free herpes simplex virus type 1 vector. , 2003, Human gene therapy.

[112]  Jie Tian,et al.  Induction of shock after intravenous injection of adenovirus vectors: a critical role for platelet-activating factor. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[113]  J. Brotchie,et al.  High frequency stimulation or elevated K+ depresses neuronal activity in the rat entopeduncular nucleus , 2007, Neuroscience.

[114]  A. Michael,et al.  Brain Tissue Responses to Neural Implants Impact Signal Sensitivity and Intervention Strategies , 2014, ACS chemical neuroscience.

[115]  Jing Wang,et al.  A coaxial optrode as multifunction write-read probe for optogenetic studies in non-human primates , 2013, Journal of Neuroscience Methods.

[116]  Robert Puers,et al.  High-density optrode-electrode neural probe using SixNy photonics for in vivo optogenetics , 2015, 2015 IEEE International Electron Devices Meeting (IEDM).

[117]  D. Shera,et al.  Long-Term Follow-Up After Gene Therapy for Canavan Disease , 2012, Science Translational Medicine.

[118]  P. Golshani,et al.  WONOEP appraisal: Optogenetic tools to suppress seizures and explore the mechanisms of epileptogenesis , 2014, Epilepsia.

[119]  T. Schlaepfer,et al.  Integrative Neuroscience Mini Review Article Origin and Evolution of Deep Brain Stimulation , 2022 .

[120]  Li Zhong,et al.  Human hepatocyte growth factor receptor is a cellular coreceptor for adeno-associated virus serotype 3. , 2010, Human gene therapy.

[121]  N. D. Di Paolo,et al.  Recognition of virus infection and innate host responses to viral gene therapy vectors. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[122]  M. Grubb,et al.  Activity-dependent relocation of the axon initial segment fine-tunes neuronal excitability , 2010, Nature.

[123]  J. Gordon,et al.  Modeling the Spatiotemporal Dynamics of Light and Heat Propagation for In Vivo Optogenetics. , 2015, Cell reports.

[124]  Rats can acquire conditional fear of faint light leaking through the acrylic resin used to mount fiber optic cannulas. , 2016, Learning & memory.

[125]  E. Bamberg,et al.  Optogenetic Control of Ca2+ and Voltage-Dependent Large Conductance (BK) Potassium Channels. , 2017, Journal of molecular biology.

[126]  Susana Q. Lima,et al.  Remote Control of Behavior through Genetically Targeted Photostimulation of Neurons , 2005, Cell.

[127]  Ronald L Klein,et al.  Efficient neuronal gene transfer with AAV8 leads to neurotoxic levels of tau or green fluorescent proteins. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[128]  Lauren E. Mueller,et al.  Elevated potassium provides an ionic mechanism for deep brain stimulation in the hemiparkinsonian rat , 2013, The European journal of neuroscience.

[129]  D. Kleinfeld,et al.  ReaChR: A red-shifted variant of channelrhodopsin enables deep transcranial optogenetic excitation , 2013, Nature Neuroscience.

[130]  Xue Han,et al.  Prosthetic systems for therapeutic optical activation and silencing of genetically targeted neurons , 2008, SPIE BiOS.

[131]  K. Deisseroth Optogenetics: 10 years of microbial opsins in neuroscience , 2015, Nature Neuroscience.

[132]  A. Benabid,et al.  The impact on Parkinson’s disease of electrical parameter settings in STN stimulation , 2002, Neurology.

[133]  M. Dours-Zimmermann,et al.  Extracellular matrix of the central nervous system: from neglect to challenge , 2008, Histochemistry and Cell Biology.

[134]  Application of optogenetics-mediated motor cortex stimulation in the treatment of chronic neuropathic pain. , 2016, Journal of translational science.

[135]  E. Moser,et al.  Relationship between neuronal activity and brain temperature in rats. , 1996, Neuroreport.

[136]  A. Kleinschmidt,et al.  Noninvasive Functional Imaging of Human Brain Using Light , 2000, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[137]  S. Taverna,et al.  Optogenetic activation of VGLUT2-expressing excitatory neurons blocks epileptic seizure-like activity in the mouse entorhinal cortex , 2017, Scientific Reports.

[138]  S. Kochanek,et al.  Frequency and Stability of Chromosomal Integration of Adenovirus Vectors , 1999, Journal of Virology.

[139]  Nicholas B Langhals,et al.  Providing a Sense of Touch to Prosthetic Hands , 2015, Plastic and reconstructive surgery.

[140]  M. Welsh,et al.  Binding of Adeno-associated Virus Type 5 to 2,3-Linked Sialic Acid Is Required for Gene Transfer* , 2001, The Journal of Biological Chemistry.

[141]  J. Chiorini,et al.  Epidermal growth factor receptor is a co-receptor for adeno-associated virus serotype 6 , 2010, Nature Medicine.

[142]  Fred Wolf,et al.  Controlling the oscillation phase through precisely timed closed-loop optogenetic stimulation: a computational study , 2013, Front. Neural Circuits.

[143]  L. Rozamus,et al.  Long-term pharmacologically regulated expression of erythropoietin in primates following AAV-mediated gene transfer. , 2005, Blood.

[144]  H. Lüders,et al.  Deep Brain Stimulation in Epilepsy , 2001, Journal of clinical neurophysiology : official publication of the American Electroencephalographic Society.

[145]  P. Osborne Problematizing Disciplinarity, Transdisciplinary Problematics , 2015, Theory, culture & society.

[146]  J. Obeso,et al.  Striatal activation by optogenetics induces dyskinesias in the 6‐hydroxydopamine rat model of Parkinson disease , 2017, Movement disorders : official journal of the Movement Disorder Society.

[147]  E. Dietrichs,et al.  Long-term follow-up of thalamic deep brain stimulation for essential tremor – patient satisfaction and mortality , 2014, BMC Neurology.

[148]  K. Deisseroth,et al.  eNpHR: a Natronomonas halorhodopsin enhanced for optogenetic applications , 2008, Brain cell biology.

[149]  R. Nicoll,et al.  Single-Cell Optogenetic Excitation Drives Homeostatic Synaptic Depression , 2010, Neuron.

[150]  S. Kleinlogel,et al.  Chronic activation of the D156A point mutant of Channelrhodopsin-2 signals apoptotic cell death: the good and the bad , 2016, Cell Death & Disease.

[151]  Mikhail G. Shapiro,et al.  Infrared light excites cells by changing their electrical capacitance , 2012, Nature Communications.

[152]  Keyun Qing,et al.  Human fibroblast growth factor receptor 1 is a co-receptor for infection by adeno-associated virus 2 , 1999, Nature Medicine.

[153]  Jaimie M. Henderson,et al.  Patient-specific analysis of the volume of tissue activated during deep brain stimulation , 2007, NeuroImage.

[154]  S S Stensaas,et al.  The reaction of the cerebral cortex to chronically implanted plastic needles. , 1976, Acta neuropathologica.

[155]  G. Friehs,et al.  Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience , 2010, Molecular Psychiatry.

[156]  Robert E. Gross,et al.  Deep Brain Stimulation for the Treatment of Epilepsy: Circuits, Targets, and Trials , 2014, Neurotherapeutics.

[157]  R. Samulski,et al.  Membrane-Associated Heparan Sulfate Proteoglycan Is a Receptor for Adeno-Associated Virus Type 2 Virions , 1998, Journal of Virology.

[158]  Hisao Tsukamoto,et al.  Homologs of vertebrate Opn3 potentially serve as a light sensor in nonphotoreceptive tissue , 2013, Proceedings of the National Academy of Sciences.

[159]  Karen L. Smith,et al.  Effects of insertion conditions on tissue strain and vascular damage during neuroprosthetic device insertion , 2006, Journal of neural engineering.

[160]  A. Cavanna,et al.  Clinical utility of implantable neurostimulation devices as adjunctive treatment of uncontrolled seizures , 2014, Neuropsychiatric disease and treatment.

[161]  Jae-Woong Jeong,et al.  Preparation and implementation of optofluidic neural probes for in vivo wireless pharmacology and optogenetics , 2017, Nature Protocols.

[162]  Matthew D. Johnson,et al.  Mechanisms and targets of deep brain stimulation in movement disorders , 2008, Neurotherapeutics.

[163]  Elisa E. Konofagou,et al.  Non-invasive, Focused Ultrasound-Facilitated Gene Delivery for Optogenetics , 2017, Scientific Reports.

[164]  T. Oertner,et al.  Optical induction of synaptic plasticity using a light-sensitive channel , 2007, Nature Methods.

[165]  R A Bakay,et al.  Ablative surgery and deep brain stimulation for Parkinson's disease. , 1999, Neurosurgery.

[166]  S. Kochanek,et al.  High-capacity 'gutless' adenoviral vectors. , 2001, Current opinion in molecular therapeutics.

[167]  E. Agosin,et al.  Optogenetic switches for light-controlled gene expression in yeast , 2017, Applied Microbiology and Biotechnology.

[168]  The Activation Function of TMS on a Finite Element Model of a Cortical Sulcus , 2007, 2007 29th Annual International Conference of the IEEE Engineering in Medicine and Biology Society.

[169]  E. Ben-Menachem,et al.  VNS Therapy versus the latest antiepileptic drug. , 2005, Epileptic disorders : international epilepsy journal with videotape.

[170]  R. Mandel,et al.  Immune responses to adenovirus and adeno-associated vectors used for gene therapy of brain diseases: the role of immunological synapses in understanding the cell biology of neuroimmune interactions. , 2007, Current gene therapy.

[171]  Fabio Benfenati,et al.  Long-term optical stimulation of channelrhodopsin-expressing neurons to study network plasticity , 2013, Front. Mol. Neurosci..

[172]  Patrícia A. Correia,et al.  Transient inhibition and long-term facilitation of locomotion by phasic optogenetic activation of serotonin neurons , 2017, eLife.

[173]  P H Peckham,et al.  Peripheral nerve stimulation for restoration of motor function. , 1997, Journal of clinical neurophysiology : official publication of the American Electroencephalographic Society.

[174]  H Bostock,et al.  The strength‐duration relationship for excitation of myelinated nerve: computed dependence on membrane parameters. , 1983, The Journal of physiology.

[175]  M. Kay,et al.  The 37/67-Kilodalton Laminin Receptor Is a Receptor for Adeno-Associated Virus Serotypes 8, 2, 3, and 9 , 2006, Journal of Virology.

[176]  M. Avoli,et al.  Optogenetic Low-Frequency Stimulation of Specific Neuronal Populations Abates Ictogenesis , 2017, The Journal of Neuroscience.

[177]  M. Rice,et al.  Characterization of Optically and Electrically Evoked Dopamine Release in Striatal Slices from Digenic Knock-in Mice with DAT-Driven Expression of Channelrhodopsin , 2017, ACS chemical neuroscience.

[178]  C B Maks,et al.  Deep brain stimulation activation volumes and their association with neurophysiological mapping and therapeutic outcomes , 2008, Journal of Neurology, Neurosurgery, and Psychiatry.

[179]  Philip J. Hahn,et al.  Network perspectives on the mechanisms of deep brain stimulation , 2010, Neurobiology of Disease.

[180]  R. Samulski,et al.  Cross-Packaging of a Single Adeno-Associated Virus (AAV) Type 2 Vector Genome into Multiple AAV Serotypes Enables Transduction with Broad Specificity , 2002, Journal of Virology.

[181]  Karl Deisseroth,et al.  Genetic Reactivation of Cone Photoreceptors Restores Visual Responses in Retinitis Pigmentosa , 2010, Science.

[182]  W Zhang,et al.  A tyrosine hydroxylase-neurofilament chimeric promoter enhances long-term expression in rat forebrain neurons from helper virus-free HSV-1 vectors. , 2000, Brain research. Molecular brain research.

[183]  F. Bushman,et al.  Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. , 2008, The Journal of clinical investigation.

[184]  R. Campbell,et al.  Emerging fluorescent protein technologies. , 2015, Current opinion in chemical biology.

[185]  R. Herzog,et al.  Hepatic gene transfer as a means of tolerance induction to transgene products. , 2009, Current gene therapy.

[186]  K. Mathieson,et al.  Optogenetic activation of neocortical neurons in vivo with a sapphire-based micro-scale LED probe , 2015, Front. Neural Circuits.

[187]  Keyoumars Ashkan,et al.  Accuracy of stimulating electrode placement in paediatric pallidal deep brain stimulation for primary and secondary dystonia , 2013, Acta Neurochirurgica.

[188]  D. Szarowski,et al.  Cerebral Astrocyte Response to Micromachined Silicon Implants , 1999, Experimental Neurology.

[189]  M. Weitzman,et al.  Adeno-associated virus biology. , 2011, Methods in molecular biology.

[190]  Gonzalo Arias-Gil,et al.  Measurement, modeling, and prediction of temperature rise due to optogenetic brain stimulation , 2016, Neurophotonics.

[191]  G. Charras,et al.  Optogenetic control of cellular forces and mechanotransduction , 2017, Nature Communications.

[192]  J. Chiorini,et al.  Adeno-Associated Virus Serotype 4 (AAV4) and AAV5 Both Require Sialic Acid Binding for Hemagglutination and Efficient Transduction but Differ in Sialic Acid Linkage Specificity , 2001, Journal of Virology.

[193]  Seok Hyun Yun,et al.  Light in diagnosis, therapy and surgery , 2016, Nature Biomedical Engineering.

[194]  J. B. Ranck,et al.  Which elements are excited in electrical stimulation of mammalian central nervous system: A review , 1975, Brain Research.

[195]  K. Deisseroth,et al.  Millisecond-timescale, genetically targeted optical control of neural activity , 2005, Nature Neuroscience.

[196]  V. Dhawan,et al.  Modulation of metabolic brain networks after subthalamic gene therapy for Parkinson's disease , 2007, Proceedings of the National Academy of Sciences.

[197]  Michael A. Henninger,et al.  High-Performance Genetically Targetable Optical Neural Silencing via Light-Driven Proton Pumps , 2010 .

[198]  T. Larcher,et al.  Lack of immunotoxicity after regional intravenous (RI) delivery of rAAV to nonhuman primate skeletal muscle. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[199]  Edward S Boyden,et al.  Principles of designing interpretable optogenetic behavior experiments , 2015, Learning & memory.

[200]  R. Kotin,et al.  Immunological Aspects of Recombinant Adeno-Associated Virus Delivery to the Mammalian Brain , 2002, Journal of Virology.

[201]  Volker Busskamp,et al.  Biophysical Properties of Optogenetic Tools and Their Application for Vision Restoration Approaches , 2016, Front. Syst. Neurosci..

[202]  K. Vonck,et al.  Clinical experience with vagus nerve stimulation and deep brain stimulation in epilepsy. , 2007, Acta neurochirurgica. Supplement.

[203]  I. Melnick,et al.  Astrocytes and presynaptic plasticity in the striatum: Evidence and unanswered questions , 2018, Brain Research Bulletin.

[204]  R. Samulski,et al.  Viral vectors and delivery strategies for CNS gene therapy. , 2010, Therapeutic delivery.

[205]  Svjetlana Miocinovic,et al.  Experimental and theoretical characterization of the voltage distribution generated by deep brain stimulation , 2009, Experimental Neurology.

[206]  J. Sommer,et al.  Human immunoglobulin inhibits liver transduction by AAV vectors at low AAV2 neutralizing titers in SCID mice. , 2006, Blood.

[207]  K. Ono,et al.  Rapid and efficient gene delivery into the adult mouse brain via focal electroporation , 2016, Scientific Reports.

[208]  D. I. Stephanova,et al.  A distributed-parameter model of the myelinated human motor nerve fibre: temporal and spatial distributions of action potentials and ionic currents , 1995, Biological Cybernetics.

[209]  Matthias Prigge,et al.  Enhancing Channelrhodopsins: An Overview. , 2016, Methods in molecular biology.

[210]  K. High,et al.  Immune responses to AAV vectors: overcoming barriers to successful gene therapy. , 2013, Blood.

[211]  Alan McClelland,et al.  Evidence for gene transfer and expression of factor IX in haemophilia B patients treated with an AAV vector , 2000, Nature Genetics.

[212]  C. Sladek,et al.  Comparison of the efficacy of four viral vectors for transducing hypothalamic magnocellular neurosecretory neurons in the rat supraoptic nucleus , 2011, Journal of Neuroscience Methods.

[213]  Matthew T. Kaufman,et al.  An optogenetic toolbox designed for primates , 2011, Nature Neuroscience.

[214]  Y. Agid,et al.  Disappearance of parkinsonian signs after spontaneous vascular 'thalamotomy'. , 1986, Archives of neurology.

[215]  R. Chen,et al.  Adeno-associated virus-mediated gene transfer to the brain: duration and modulation of expression. , 1999, Human gene therapy.

[216]  J. A. Varela,et al.  Shielded Coaxial Optrode Arrays for Neurophysiology , 2016, Front. Neurosci..

[217]  Jacob G. Bernstein,et al.  Millisecond-Timescale Optical Control of Neural Dynamics in the Nonhuman Primate Brain , 2009, Neuron.

[218]  Alexander Gottschalk,et al.  Optogenetic Long-Term Manipulation of Behavior and Animal Development , 2011, PloS one.

[219]  A. Wise,et al.  Challenges for the application of optical stimulation in the cochlea for the study and treatment of hearing loss , 2017, Expert opinion on biological therapy.

[220]  Dimitri M. Kullmann,et al.  Optogenetic approaches to treat epilepsy , 2016, Journal of Neuroscience Methods.

[221]  B. Carter,et al.  Adeno-associated virus vectors in clinical trials. , 2005, Human gene therapy.

[222]  C. McIntyre,et al.  Deep brain stimulation mechanisms: the control of network activity via neurochemistry modulation , 2016, Journal of neurochemistry.

[223]  James L Olds,et al.  Positive reinforcement produced by electrical stimulation of septal area and other regions of rat brain. , 1954, Journal of comparative and physiological psychology.

[224]  G. Nagel,et al.  Channelrhodopsin-2–XXL, a powerful optogenetic tool for low-light applications , 2014, Proceedings of the National Academy of Sciences.

[225]  Claus-Peter Richter,et al.  Optical cochlear implants: Evaluation of surgical approach and laser parameters in cats , 2010, Hearing Research.

[226]  Dimiter Prodanov,et al.  A model of space-fractional-order diffusion in the glial scar. , 2016, Journal of theoretical biology.

[227]  Jason T. Ritt,et al.  Control strategies for underactuated neural ensembles driven by optogenetic stimulation , 2013, Front. Neural Circuits.

[228]  Ege Iseri,et al.  Implantable optoelectronic probes for in vivo optogenetics , 2017, Journal of neural engineering.

[229]  B. Davidson,et al.  Transduction of nonhuman primate brain with adeno-associated virus serotype 1: vector trafficking and immune response. , 2008, Human gene therapy.

[230]  Valentina Emiliani,et al.  Computer Generated Holography with Intensity-Graded Patterns , 2016, Front. Cell. Neurosci..

[231]  R. Samulski,et al.  Directed Evolution of a Novel Adeno-associated Virus (AAV) Vector That Crosses the Seizure-compromised Blood-Brain Barrier (BBB). , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[232]  Nader Pouratian,et al.  Proceedings of the Third Annual Deep Brain Stimulation Think Tank: A Review of Emerging Issues and Technologies , 2016, Front. Neurosci..

[233]  B. Connors,et al.  Semiconductor ultra-violet light-emitting diodes for flash photolysis , 2007, Journal of Neuroscience Methods.

[234]  Mary Collins,et al.  One Contribution to the Special Feature 'evolution and Genetics in Medicine' Guest Edited Gene Therapy: Progress and Predictions , 2022 .

[235]  Sohee Kim,et al.  Characterization of fiber-optic light delivery and light-induced temperature changes in a rodent brain for precise optogenetic neuromodulation. , 2016, Biomedical optics express.

[236]  David Eidelberg,et al.  Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial , 2007, The Lancet.

[237]  A. Malyshev,et al.  Chloride conducting light activated channel GtACR2 can produce both cessation of firing and generation of action potentials in cortical neurons in response to light , 2017, Neuroscience Letters.