Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR CONTROLLING PAIN
Document Type and Number:
WIPO Patent Application WO/2015/023782
Kind Code:
A1
Abstract:
The present disclosure provides compositions and methods for controlling pain. The present disclosure provides methods for identifying agents that control pain.

Inventors:
IYER SHRIVATS MOHAN (US)
DELP SCOTT L (US)
MONTGOMERY KATHRYN L (US)
DEISSEROTH KARL A (US)
TOWNE CHRISTOPHER (US)
Application Number:
PCT/US2014/050938
Publication Date:
February 19, 2015
Filing Date:
August 13, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV LELAND STANFORD JUNIOR (US)
CIRCUIT THERAPEUTICS INC (US)
International Classes:
C12N15/864
Domestic Patent References:
WO2011127088A22011-10-13
WO2013090356A22013-06-20
WO1999011764A21999-03-11
WO2008024998A22008-02-28
WO1991018088A11991-11-28
WO1993009239A11993-05-13
WO2010056970A22010-05-20
Foreign References:
US20120165904A12012-06-28
US20120093772A12012-04-19
US20130225664A12013-08-29
US20110028893W2011-03-17
US20090093403A12009-04-09
US20100145418A12010-06-10
US6649811B22003-11-18
US5387742A1995-02-07
US7632679B22009-12-15
US6566118B12003-05-20
US6989264B22006-01-24
US6995006B22006-02-07
US20050027091W2005-07-29
US4797368A1989-01-10
US6596535B12003-07-22
US5139941A1992-08-18
EP0488528A11992-06-03
Other References:
LISKE ET AL., MUSCLE & NERVE, vol. 47, 2013, pages 916
LLEWELLYN ET AL., NATURE MED., vol. 16, 2010, pages 1161
JI ET AL., PLOS ONE, vol. 7, 2012, pages e32699
WANGZYLKA, . NEUROSCI., vol. 29, 2009, pages 13020
DAOU ET AL., SOC. NEUROSCI. CONF., 2012
DAOU ET AL., J. NEUROSCI., vol. 33, 2013, pages 47
MATTIS ET AL., NAT. METHODS, vol. 9, 2012, pages 159
KOKEL ET AL., NAT. CHEM. BIOL., vol. 9, 2013, pages 257
WILLIAMSDENISON, SCI. TRANSL. MED., vol. 5, 2013, pages 177ps5
TOWNE ET AL., GENE THER., vol. 17, 2010, pages 141
TOWNE ET AL., MOL. PAIN, vol. 5, 2009, pages 52
IYER ET AL., NAT. BIOTECH., vol. 32, 2014, pages 3
MEYER ET AL., TEXTBOOK OF PAIN, 1994, pages 13 - 44
KATZMELZACK, SURG. CLIN. NORTH AM., vol. 79, 1999, pages 231
HICKS ET AL., PAIN, vol. 93, 2001, pages 173
SCHMADER, CLIN. J. PAIN, vol. 18, 2002, pages 350
STEWART ET AL., ARCH. DIS. CHILD., vol. 89, 2004, pages 625
PAYEN ET AL., CRITICAL CARE MEDICINE, vol. 29, 2001, pages 2258
CLEELANDRYAN, ANN. ACAD. MED. SINGAPORE, vol. 23, 1994, pages 129
FELDT, PAIN MANAG. NURS., vol. 1, 2000, pages 13
GELINAS ET AL., AM. J. CRIT. CARE, vol. 15, 2006, pages 420
AMBUEL ET AL., J. PEDIATRIC PSYCHOL., vol. 17, 1992, pages 95
OZGULER ET AL., SPINE, vol. 27, 2002, pages 1783
HARDY ET AL.: "Pain Sensations and Reactions Baltimore", 1952, WILLIAMS & WILKINS CO.
MELZACK, PAIN, vol. 1, 1975, pages 277
GRACELYKWILOSZ, PAIN, vol. 35, 1988, pages 279
JENSEN ET AL., CLIN. J. PAIN, vol. 5, 1989, pages 153
HARTRICK ET AL., PAIN PRACT., vol. 3, 2003, pages 310
HUSKISSON, J. RHEUMATOL., vol. 9, 1982, pages 768
STEIN ET AL., J. VIROL, vol. 73, 1999, pages 34243429
DAVIDSON ET AL., PNAS, vol. 97, 2000, pages 3428 - 3432
DAVIDSON ET AL., NAT. GENET., vol. 3, 1993, pages 219 - 223
ALISKYDAVIDSON, HUM. GENE THER., vol. 11, 2000, pages 2315 - 2329
LLEWELLYN ET AL., NAT. MED., vol. 16, no. 10, 2010, pages 1161 - 1166
"GenBank", Database accession no. S62283
CHEN ET AL., CELL, vol. 51, 1987, pages 7 - 19
NUCL. ACIDS. RES., vol. 15, 1987, pages 2363 - 2384
RADOVICK ET AL., PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 3402 - 3406
OBERDICK ET AL., SCIENCE, vol. 248, 1990, pages 223 - 226
BARTGE ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 3648 - 3652
COMB ET AL., EMBO J., vol. 17, 1988, pages 3793 - 3805
LIU ET AL., GENE THERAPY, vol. 11, 2004, pages 52 - 60
LEE ET AL., DEVELOPMENT, vol. 131, 2004, pages 3295 - 3306
MAYFORD ET AL., PROC. NATL. ACAD. SCI. USA, vol. 93, 1996, pages 13250
BLACKLOW, PARVOVIRUSES AND HUMAN DISEASE, 1988, pages 165 - 174
ROSE, COMPREHENSIVE VIROLOGY, vol. 3, 1974, pages 1
P. TATTERSALLHUDDER ARNOLD: "Parvoviruses", 2006, article "The Evolution of Parvovirus Taxonomy", pages: 5 - 14
DE BOWLESJE RABINOWITZRJ SAMULSKI: "The Genus Dependovirus", 2006, pages: 15 - 23
KIOURTI: "Biomedical Telemetry: Communication between Implanted Devices and the External World", OPTICON, vol. l826, no. 8, 2010
SOHAL ET AL., NATURE, vol. 459, 2009, pages 698
RAO ET AL., NEUROPHARMACOL., vol. 35, 1996, pages 393
MOGI ET AL., METHODS IN PAIN RESEARCH, FRONTIERS IN NEUROSCIENCE, 2001
CARTERSHIEH: "Nociception: Guide to Research Techniques in Neuroscience", 2010, ACADEMIC PRESS, pages: 51 - 52
BANNONMALMBERG: "Current Protocols in Neuroscience", 2007, WILEY
MOGIL ET AL.: "Methods in Pain Research", FRONTIERS IN NEUROSCIENCE, 2001
See also references of EP 3033427A4
Attorney, Agent or Firm:
BORDEN, Paula, A. (Field & Francis LLP1900 University Avenue, Suite 20, East Palo Alto California, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for controlling pain in an individual, the method comprising introducing into a primary afferent neuron of the individual a nucleic acid comprising a nucleotide sequence encoding an opsin polypeptide that provides for hyperpolanzation of the primary afferent neuron in response to light of a wavelength that activates the opsin.

2. The method of claim 1, wherein the primary afferent neuron is a nociceptor.

3. The method of claim 1, wherein the light is delivered transdermally.

4. The method of claim 1, wherein the opsin comprises an amino acid sequence having at least about 75% amino acid sequence identity to one of SEQ ID NOs:l, 3, 4, 6, 15, and 16.

5. The method of claim 1, wherein the pain is neuropathic pain.

6. The method of claim 1, wherein said introducing is via injection into a nerve or via intramuscular injection.

7. The method of claim 1, wherein said introducing is via topical, intradermal, intravenous, intrathecal, or intrapleural administration.

8. The method of claim 2, wherein the nociceptor is one that is normally activated by a thermal, mechanical, or chemical stimulus.

9. The method of claim 1, wherein the nucleic acid is a recombinant expression vector.

10. The method of claim 7, wherein the recombinant expression vector is a viral vector.

11. The method of claim 10, wherein the viral vector is a lentivirus vector or an adeno- associated virus (AAV) vector.

12. The method of claim 11, wherein the AAV vector is an AAV6 vector or an AAV8 vector.

13. The method of claim 1, wherein the nucleotide sequence is operably linked to a promoter that provides for selective expression in a neuron.

14. The method of claim 13, wherein the promoter is a synapsin-I promoter, a human synuclein 1 promoter, a human Thyl promoter, or a calcium/calmodulin-dependent kinase II alpha (CAMKIIa) promoter.

15. The method of claim 1, wherein the individual is a mammal.

16. The method of claim 15, wherein the individual is a human, a rat, or a mouse.

17. The method of claim 1, wherein activation of the opsin provides for an at least 10% reduction in pain.

18. A non-human animal model of pain, wherein the non-human animal expresses in a primary afferent neuron of the animal a nucleic acid comprising a nucleotide sequence encoding an opsin polypeptide that provides for depolarization of the nociceptor in response to light of a wavelength that activates the opsin.

19. The non-human animal model of claim 18, wherein the primary afferent neuron is a nociceptor.

20. The non-human animal model of claim 18, wherein the opsin comprises an amino acid sequence having at least about 75% amino acid sequence identity to one of SEQ ID NOs:8-14 and 19-21.

21. The non-human animal model of claim 18, wherein the nucleic acid is a recombinant expression vector.

22. The non-human animal model of claim 18, wherein the recombinant expression vector is a viral vector.

23. The non-human animal model of claim 22, wherein the viral vector is a lentivirus vector or an adeno-associated virus (AAV) vector.

24. The non-human animal model of claim 23, wherein the AAV vector is an AAV6 vector or an AAV8 vector.

25. The non-human animal model of claim 18, wherein the nucleotide sequence is operably linked to a promoter that provides for selective expression in a neuron.

26. The non-human animal model of claim 25, wherein the promoter is a synapsin-I promoter, a human synuclein 1 promoter, a human Thyl promoter, or a calcium/calmodulin- dependent kinase II alpha (CAMKIIa) promoter.

27. The non-human animal model of claim 18, wherein the animal is a rat or a mouse.

28. A method of identifying an agent that reduces pain, the method comprising:

a) administering a test agent to the non-human animal of claim 18; and

b) determining the effect, if any, of the test agent on pain when the depolarizing light- activated polypeptide is activated with light, wherein a test agent that reduces pain in the non-human animal, compared to the level of pain induced by light activation of the depolarizing light-activated polypeptide in the absence of the test agent, indicates that the test agent is a candidate agent for reducing pain.

29. A method of identifying an agent that reduces pain, the method comprising:

a) administering a test agent to the non-human animal of claim 18; and

b) determining the effect, if any, of the test agent on the amount of light required to induce pain following administration of the test agent,

wherein a test agent that increases the amount of light required to induce pain is a candidate agent for reducing pain.

Description:
COMPOSITIONS AND METHODS FOR CONTROLLING PAIN

CROSS-REFERENCE

[0001] This application claims the benefit of U.S. Provisional Patent Application No. 61/865,962, filed

August 14, 2013, which application is incorporated herein by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

[0002] This invention was made with government support under Grant No. NS080954 awarded by the

National Institutes of Health. The government has certain rights in the invention.

INTRODUCTION

[0003] Primary nociceptors are the first neurons in a complex pain-processing system that regulates normal and pathological pain. The ability to excite and inhibit these neurons for the purposes of research and therapy has been limited by pharmacological and electrical stimulation constraints; thus non-invasive, spatially localized control of nociceptors in freely moving animals has not been possible. Literature

[0004] Liske et al. (2013) Muscle & Nerve 47:916; Llewellyn et al. (2010) Nature Med. 16: 1 161; Ji et al. (2012) PLoS One 7:e32699; Wang and Zylka (2009) J. Neurosci. 29:13020; Daou et al. (2012) Soc. Neurosci. Conf. 575.06/1111; Daou et al. (2013) J. Neurosci. 33:47; Mourot et al. (2012) Nat. Methods 9:396; Kokel et al. (2013) Nat. Chem. Biol. 9:257; Mattis et al. (2012) Nat. Methods 9: 159; Williams and Denison (2013) Sci. Transl. Med. 5: 177ps6; Chow and Boyden (2013) Sci. Transl. Med. 5:177ps5; Towne et al. (2010) Gene Ther. 17: 141 ; Towne et al. (2009) Mol. Pain 5:52; Iyer et al. (2014) Nat. Biotech. 32:3.

SUMMARY

[0005] The present disclosure provides compositions and methods for controlling pain. The present disclosure provides methods for identifying agents that control pain.

FEATURES

[0006] The present disclosure features a method for controlling pain in an individual, the method comprising introducing into a nociceptor of the individual a nucleic acid comprising a nucleotide sequence encoding an opsin polypeptide that provides for hyperpolarization of the nociceptor in response to light of a wavelength that activates the opsin. In some cases, the light is delivered transdermally. In some cases, the opsin comprises an amino acid sequence having at least about 75% amino acid sequence identity to one of SEQ ID NOs:l, 3, 4, 6, 15, and 16. In some cases, the pain is neuropathic pain. In some cases, the nucleic acid comprising a nucleotide encoding the opsin is administered to the individual via injection into a nerve, via intramuscular injection, or via intravenous injection. In some cases, the nucleic acid is administered to the individual at or near a treatment site (e.g., a site of pain). In some cases, the nucleic acid is a recombinant expression vector, e.g., the recombinant expression vector is a viral vector. In some instances, where the recombinant expression vector is a viral vector, the viral vector is a lentivirus vector or an adeno-associated virus (AAV) vector. In some instances, the AAV vector is an AAV6 vector or an AAV8 vector. The nucleotide sequence can be operably linked to a promoter that provides for selective expression in a neuron. For example, the promoter can be a synapsin-I promoter, a human synuclein 1 promoter, a human Thyl promoter, or a calcium calmodulin-dependent kinase II alpha (CAMKIIa) promoter. The individual can be a mammal; e.g., a human, a rat, or a mouse. In some case, activation of the opsin provides for an at least 10%, at least 20%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%, reduction in pain. In some case, activation of the opsin provides a 100% reduction in pain, i.e., the individual experiences substantially no pain.

[0007] The present disclosure features a non-human animal model of neuropathic pain, where the non- human animal expresses in a nociceptor of the animal a nucleic acid comprising a nucleotide sequence encoding an opsin polypeptide that provides for depolarization of the nociceptor in response to light of a wavelength that activates the opsin. In some cases, the opsin comprises an amino acid sequence having at least about 75% amino acid sequence identity to one of SEQ ID NOs:8-14 and 19-21. In some cases, the nucleic acid is a recombinant expression vector, e.g., a viral vector. For example, in some cases, the viral vector is a lentivirus vector or an adeno-associated virus (AAV) vector, e.g., an AAV6 vector or an AAV8 vector. In some instances, the nucleotide sequence encoding the opsin is operably linked to a promoter that provides for selective expression in a neuron; e.g., the promoter can be a synapsin-I promoter, a human synuclein 1 promoter, a human Thyl promoter, or a calcium/calmodulin- dependent kinase II alpha (CAMKIIa) promoter. In some instances, the animal is a rat. In some instances, the animal is a mouse.

[0008] The present disclosure features a method of identifying an agent that reduces pain, the method comprising: a) administering a test agent to a subject non-human animal; and b) determining the effect, if any, of the test agent on pain when the depolarizing light-activated polypeptide is activated with light, where a test agent that reduces pain in the non-human animal, compared to the level of pain induced by light activation of the depolarizing light-activated polypeptide in the absence of the test agent, indicates that the test agent is a candidate agent for reducing pain.

[0009] The present disclosure features a method of identifying an agent that reduces pain, the method comprising: a) administering a test agent to a subject non-human animal; and b) determining the effect, if any, of the test agent on the amount of light required to induce pain through the activation of a depolarizing light-activated polypeptide following administration of the test agent, wherein a test agent that increases the amount of light required to produce a sign of pain, compared with the amount of light required to produce a sign of pain in the absence of the test agent indicates that the test agent is a candidate agent for reducing pain. BRIEF DESCRIPTION OF THE DRAWINGS

[0010] Figures 1 A-C depict data showing that intra-sciatic injection of rAAV2/6-hSyn-ChR2(Hl 34R)- eYFP transduced unmyelinated nociceptors that projected to spinal cord lamina.

[0011] Figures 2A-F depict data showing that transdermal illumination of ChR2 + mice resulted in tunable pain-like behavior.

[0012] Figures 3A-G depict responses in blue light-sensitized ChR2-expressing mice and yellow light- desensitized NpHR-expressing mice to mechanical and thermal stimuli.

[0013] Figures 4A and 4B depict data showing that yellow light stimulation of NpHR + mice reversed mechanical allodynia and thermal hyperalgesia caused by a chronic constriction injury.

[0014] Figures 5A-G depict amino acid sequences of various light-activated polypeptides.

[0015] Figures 6A and 6B depict size distribution of opsin transduction.

[0016] Figures 7A-D depict representative images of NpHR transduction observed after intra-sciatic injection of AAV2/6-hSyn-eNpHR3.0-eYFP.

[0017] Figures 8A-F depict electrophysiological recording from ChR2+ and NpHR+ DRG neurons.

[0018] Figure 9 provides Table 1.

[0019] Figures 10A and 10B depict AAV8 transduction in the lumbar and thoracic spinal cord.

[0020] Figure 11 depicts expression of eNpHR3.0 in trigeminal ganglion sensory neurons following direct injection into ganglion.

[0021] Figure 12 depicts mechanical thresholds for mice expressing GFP or eNpHR3.0 in nociceptive fibers using AAV6.

DEFINITIONS

[0022] As used herein, an "individual," "subject," or "patient" is an animal, e.g., a mammal, including a human. Mammals include, but are not limited to, ungulates, canines, felines, bovines, ovines, non- human primates, lagomorphs, and rodents (e.g., mice and rats). In one aspect, an individual is a human. In another aspect, an individual is a non-human mammal.

[0023] Amino acid substitutions in a native protein sequence may be "conservative" or "non-conservative" and such substituted amino acid residues may or may not be one encoded by the genetic code. A

"conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a chemically similar side chain (i.e., replacing an amino acid possessing a basic side chain with another amino acid with a basic side chain). A "non-conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a chemically different side chain (i.e., replacing an amino acid having a basic side chain with an amino acid having an aromatic side chain). The standard twenty amino acid "alphabet" is divided into chemical families based on chemical properties of their side chains. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g. , glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and side chains having aromatic groups (e.g., tyrosine, phenylalanine, tryptophan, histidine).

[0024] As used herein, an "effective dosage" or "effective amount" of a recombinant expression vector, or a pharmaceutical composition comprising a recombinant expression vector, is an amount sufficient to effect beneficial or desired results. For prophylactic use, beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results include clinical results such as decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival. An effective dosage can be administered in one or more administrations. For purposes of this disclosure, an effective dosage of a recombinant expression vector, or a pharmaceutical composition comprising a recombinant expression vector, is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. For example, an effective dosage of a recombinant expression vector, or a pharmaceutical composition comprising a recombinant expression vector, can be an amount sufficient to reduce pain (e.g., neuropathic pain). As is understood in the clinical context, an effective dosage of a recombinant expression vector, or a pharmaceutical composition comprising a recombinant expression vector, may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an "effective dosage" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.

[0025] As used herein, "treatment" or "treating" is an approach for obtaining beneficial or desired results including clinical results. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals. For example, "treatment" or "treating" can refer to reduction in pain.

[0026] Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims. [0027] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.

[0028] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.

[0029] It must be noted that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an opsin" includes a plurality of such opsin and reference to "the nociceptor" includes reference to one or more nociceptors and equivalents thereof known to those skilled in the art, and so forth. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as "solely," "only" and the like in connection with the recitation of claim elements, or use of a "negative" limitation.

[0030] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment.

Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. All combinations of the embodiments pertaining to the invention are specifically embraced by the present invention and are disclosed herein just as if each and every combination was individually and explicitly disclosed. In addition, all sub-combinations of the various embodiments and elements thereof are also specifically embraced by the present invention and are disclosed herein just as if each and every such subcombination was individually and explicitly disclosed herein.

[0031] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed. DETAILED DESCRIPTION

[0032] The present disclosure provides compositions and methods for controlling pain in an individual.

The present disclosure provides methods for identifying agents that control pain.

METHODS OF REDUCING PAIN

[0033] The present disclosure provides compositions and methods for controlling pain in an individual.

In some cases, methods for controlling pain according to the present disclosure generally involve introducing into a neuron of an individual a nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the cell in response to light of a wavelength that activates the opsin; such a method provides for reduction of pain. The nucleic acid enters the neuron (e.g., a primary afferent neuron, such as a small- or a large-diameter primary afferent neuron; e.g., a nociceptor), the opsin is produced in the neuron, and the opsin is inserted into the cell membrane. The terms "opsin," "light-responsive protein," "light-responsive polypeptide," "light-activated protein," and "light-activated polypeptide," are used interchangeably herein.

[0034] In some cases, a nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the cell in response to light of a wavelength that activates the opsin provides for expression of the opsin in a neuron (e.g., primary afferent neuron; e.g., a nociceptor). In some cases, a nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the cell in response to light of a wavelength that activates the opsin provides for expression of the opsin in a sub-population of nociceptors. Targeting expression of a light-activated polypeptide to a sub- population of nociceptors can be achieved by one or more of: selection of the vector (e.g., AAV6; AAV1; AAV8; etc.); selection of a promoter; and delivery means. For example, injection into the sciatic nerve can provide for production of a light-activated polypeptide in unmyelinated nociceptors (putative C-fibers).

[0035] The present disclosure provides methods for reducing pain, e.g., pain such as acute pain,

chronic pain, neuropathic pain, nociceptive pain, allodynia, inflammatory pain, inflammatory hyperalgesia, neuropathies, neuralgia, diabetic neuropathy, human immunodeficiency virus-related neuropathy, nerve injury, rheumatoid arthritic pain, osteoarthritic pain, burns, back pain, eye pain, visceral pain, cancer pain (e.g. bone cancer pain), dental pain, headache, migraine, carpal tunnel syndrome, fibromyalgia, neuritis, sciatica, pelvic hypersensitivity, pelvic pain, post herpetic neuralgia, post-operative pain, post stroke pain, and menstrual pain.

[0036] Pain can be classified as acute or chronic. Acute pain begins suddenly and is short-lived

(usually in twelve weeks or less). It is usually associated with a specific cause such as a specific injury and is often sharp and severe. It is the kind of pain that can occur after specific injuries resulting from surgery, dental work, a strain, or a sprain. Acute pain does not generally result in any persistent psychological response. In contrast, chronic pain is long-term pain, typically persisting for more than three months and leading to significant psychological and emotional problems. Common examples of chronic pain are neuropathic pain (e.g. painful diabetic neuropathy, postherpetic neuralgia), carpal tunnel syndrome, back pain, headache, cancer pain, arthritic pain and chronic post-surgical pain. In some cases, a method of the present disclosure is effective in reducing acute pain. In some cases, a method of the present disclosure is effective in reducing chronic pain.

[0037] Clinical pain is present when discomfort and abnormal sensitivity feature among the patient's symptoms. Individuals can present with various pain symptoms. Such symptoms include: 1) spontaneous pain which may be dull, burning, or stabbing; 2) exaggerated pain responses to noxious stimuli (hyperalgesia); and 3) pain produced by normally innocuous stimuli (allodynia-Meyer et al., 1994, Textbook of Pain, 13-44). Although patients suffering from various forms of acute and chronic pain may have similar symptoms, the underlying mechanisms may be different and may, therefore, require different treatment strategies. Pain can also therefore be divided into a number of different subtypes according to differing pathophysiology, including nociceptive pain, inflammatory pain, and neuropathic pain. In some cases, a method of the present disclosure is effective in reducing nociceptive pain. In some cases, a method of the present disclosure is effective in reducing inflammatory pain. In some cases, a method of the present disclosure is effective in reducing neuropathic pain.

[0038] Nociceptive pain is induced by tissue injury or by intense stimuli with the potential to cause injury. Moderate to severe acute nociceptive pain is a prominent feature of pain from central nervous system trauma, strains/sprains, burns, myocardial infarction and acute pancreatitis, post-operative pain (pain following any type of surgical procedure), posttraumatic pain, renal colic, cancer pain and back pain. Cancer pain may be chronic pain such as tumor related pain (e.g. bone pain, headache, facial pain or visceral pain) or pain associated with cancer therapy (e.g. postchemotherapy syndrome, chronic postsurgical pain syndrome or post radiation syndrome). Cancer pain may also occur in response to chemotherapy, immunotherapy, hormonal therapy or radiotherapy. Back pain may be due to herniated or ruptured intervertebral discs or abnormalities of the lumber facet joints, sacroiliac joints, paraspinal muscles or the posterior longitudinal ligament. Back pain may resolve naturally but in some patients, where it lasts over 12 weeks, it becomes a chronic condition which can be particularly debilitating.

[0039] Neuropathic pain can be defined as pain initiated or caused by a primary lesion or dysfunction in the nervous system. Etiologies of neuropathic pain include, e.g., peripheral neuropathy, diabetic neuropathy, post herpetic neuralgia, trigeminal neuralgia, back pain, cancer neuropathy, HIV

neuropathy, phantom limb pain, carpal tunnel syndrome, central post-stroke pain and pain associated with chronic alcoholism, hypothyroidism, uremia, multiple sclerosis, spinal cord injury, Parkinson's disease, epilepsy, and vitamin deficiency.

[0040] The inflammatory process is a complex series of biochemical and cellular events, activated in response to tissue injury or the presence of foreign substances, which results in swelling and pain. Arthritic pain is a common inflammatory pain.

[00 1] Other types of pain include: pain resulting from musculoskeletal disorders, including myalgia, fibromyalgia, spondylitis, sero-negative (non-rheumatoid) arthropathies, non-articular rheumatism, dystrophinopathy, glycogenolysis, polymyositis and pyomyositis; heart and vascular pain, including pain caused by angina, myocardical infarction, mitral stenosis, pericarditis, Raynaud's phenomenon, scleredoma and skeletal muscle ischemia; head pain, such as migraine (including migraine with aura and migraine without aura), cluster headache, tension-type headache mixed headache and headache associated with vascular disorders; and orofacial pain, including dental pain, otic pain, burning mouth syndrome, and temporomandibular myofascial pain.

[0042] In some cases, a subject method of reducing pain involves introducing into a nociceptor (a sensory neuron that responds to potentially damaging stimuli by sending nerve signals to the spinal cord and brain) a nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the nociceptor in response to light of a wavelength that activates the opsin, thereby reducing pain. The nociceptor can be a thermal nociceptor, a mechanical nociceptor, a chemical nociceptor, or other type of nociceptor. Nociceptive markers include, but are not limited to, IB4, Substance P, TRPVl, and somatostatin.

[0043] Whether pain is reduced can be determined in a human subject using a variety of pain scales.

Patient self-reporting can be used to assess whether pain is reduced; see, e.g., Katz and Melzack (1999) Surg. Clin. North Am. 79:231. Alternatively, an observational pain scale can be used. The LANSS Pain Scale can be used to assess whether pain is reduced; see, e.g., Bennett (2001) Pain 92:147. A visual analog pain scale can be used; see, e.g., Schmader (2002) Clin. J. Pain 18:350. The Likert pain scale can be used; e.g., where 0 is no pain, 5 is moderate pain, and 10 is the worst pain possible. Self -report pain scales for children include, e.g., Faces Pain Scale; Wong-Baker FACES Pain Rating Scale; and Colored Analog Scale. Self-report pain scales for adults include, e.g., Visual Analog Scale; Verbal Numerical Rating Scale; Verbal Descriptor Scale; and Brief Pain Inventory. Pain measurement scales include, e.g., Alder Hey Triage Pain Score (Stewart et al. (2004) Arch. Dis. Child. 89:625); Behavioral Pain Scale (Payen et al. (2001) Critical Care Medicine 29:2258); Brief Pain Inventory (Cleeland and Ryan (1994) Ann. Acad. Med. Singapore 23: 129); Checklist of Nonverbal Pain Indicators (Feldt (2000) Pain Manag. Nurs. 1 : 13); Critical-Care Pain Observation Tool (Gelinas et al. (2006) Am. J. Crit. Care 15:420); COMFORT scale (Ambuel et al. (1992) J. Pediatric Psychol. 17:95); Dallas Pain

Questionnaire (Ozguler et al. (2002) Spine 27:1783); Dolorimeter Pain Index (Hardy et al. (1952) Pain Sensations and Reactions Baltimore: The Williams & Wilkins Co.); Faces Pain Scale - Revised (Hicks et al. (2001) Pain 93:173); Face Legs Activity Cry Consolability Scale; McGill Pain Questionnaire (Melzack (1975) Pain 1 :277); Descriptor Differential Scale (Gracely and Kwilosz (1988) Pain 35:279); Numerical 1 1 point Box (Jensen et al. (1989) Clin. J. Pain 5: 153); Numeric Rating Scale (Hartrick et al. (2003) Pain Pract. 3:310); Wong-Baker FACES Pain Rating Scale; and Visual Analog Scale

(Huskisson (1982) J. Rheumatol. 9:768).

[0044] In some cases, the light used to activate an opsin expressed in a neuron (e.g., a nociceptor) has an intensity of from about 0.05 mW/mm 2 to about 0.1 mW/mm 2 , from about 0.1 mW/mm 2 to about 0.2 mW/mrn 2 , from about 0.2 mW/mm 2 to about 0.3 rnW/mm 2 , from about 0.3 mW/mm 2 to about 0.4 mW/mrn 2 , from about 0.4 rnW/mm 2 to about 0.5 rnW/mm 2 , from about 0.5 mW/mm 2 to about 0.6 mW/mm 2 , from about 0.6 mW/mm 2 to about 0.7 mW/mm 2 , from about about 0.7 mW/mm 2 to about 0.8 mW/mm 2 , from about 0.8 mW/mm 2 to about 0.9 mW/mm 2 , or from about about 0.9 mW/mm 2 to about 1.0 mW/mm 2 . In some cases, the light used to activate an opsin expressed in a neuron (e.g., a nociceptor) has an intensity of from about 1.0 mW/mm 2 to about 1.1 mW/mm 2 , from about 1.1 mW/mm 2 to about 1.2 mW/mm 2 , from about 1.2 mW/mm 2 to about 1.3 mW/mm 2 , from 1.3 mW/mm 2 to about 1.4 mW/mm 2 , from about 1.4 mW/mm 2 to about 1.5 mW/mm 2 , from about 1.5 mW/mm 2 to about 1.6 mW/mm 2 , from about 1.6 mW/mm 2 to about 1.7 mW/mm 2 , from about 1.7 mW/mm 2 to about 1.8 mW/mm 2 , from about 1.8 mW/mm 2 to about 1.9 mW/mm 2 , from about 1.9 mW/mm 2 to about 2.0 mW/mm 2 , from about 2.0 mW/mm 2 to about 2.5 mW/mm 2 , from about 2.5 mW/mm 2 to about 3 mW/mm 2 , from about 3 mW/mm 2 to about 3.5 mW/mm 2 , from about 3.5 mW/mm 2 to about 4 mW/mm 2 , from about 4 mW/mm 2 to about 4.5 mW/mm 2 , from about 4.5 mW/mm 2 to about 5 mW/mm 2 , from about 5 mW/mm 2 to about 5.5 mW/mm 2 , from about 5.5 mW/mm 2 to about 6 mW/mm 2 , from about 6 mW/mm 2 to about 7 mW/mm 2 , or from about 7 mW/mm 2 to about 10 mW/mm 2 . In some cases, the light used to activate an opsin expressed in a neuron (e.g., a nociceptor) has an intensity of from about 0.05 mW/mm 2 to about 0.1 mW/mm 2 . In some cases, the light used to activate an opsin expressed in a neuron (e.g., a nociceptor) has an intensity of about 0.25 mW/mm 2 . In some cases, the light used to activate an opsin expressed in a neuron (e.g., a nociceptor) has an intensity of about 1 mW/mm 2 .

[0045] In some cases, the light is delivered transdermally or transcutaneously. In some cases, an

implantable light source is used; and the light is delivered to a site within the body. In some cases, the light is delivered to a treatment site within the body. In some cases, the light is delivered intracranially.

[0046] A nucleic acid comprising a nucleotide sequence encoding an opsin that provides for

hyperpolarizafion of the cell in response to light of a wavelength that activates the opsin can be introduced into a neuron (e.g., a nociceptor) by any convenient means. For example, a nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the cell in response to light of a wavelength that activates the opsin can be introduced (e.g., injected) into a nerve bundle or nerve fiber, such that the nucleic acid enters a neuron (e.g., a nociceptor), where the opsin is produced in the neuron and is inserted into the cell membrane. A nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the cell in response to light of a wavelength that activates the opsin can be introduced (e.g., injected) proximal to a nerve. Stereotactic injection can be used; see, e.g., Stein et al., J. Virol, 73 :34243429, 1999; Davidson et al, PNAS, 97:3428-3432, 2000; Davidson et al, Nat. Genet. 3 :219-223, 1993; and Alisky & Davidson, Hum. Gene Ther. 11 :2315-2329, 2000, the contents of each of which are hereby incorporated by reference herein in their entireties.

[0047] A nucleic acid comprising a nucleotide sequence encoding an opsin that provides for

hyperpolarizafion of the cell in response to light of a wavelength that activates the opsin can be introduced (e.g., injected) intramuscularly. A nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the cell in response to light of a wavelength that activates the opsin can be administered via any means, including, e.g., intravenous, intramuscular, intracranial, into a nerve, at or near a treatment site, and the like. Administration of an opsin-encoding nucleic acid can be carried out via injection; via implantation at or near a treatment site of a composition comprising a nucleic acid encoding a light-activated polypeptide; via a catheter; or via any other means of delivery. Administration of an opsin-encoding nucleic acid can be carried out via topical, intradermal, intravenous, intrathecal, or intrapleural administration. Administration of an opsin- encoding nucleic acid can be carried out via intradermal administration.

[0048] A light-activated protein (opsin) can be implanted into or proximal to a nerve using a number of different methods. Example methods include, but are not limited to, the use of various delivery devices, such as gelatin capsules, liquid injections and the like. Such methods also include the use of stereotactic surgery techniques such as frames or computerized surgical navigation systems to implant or otherwise access areas of the body.

[0049] In some cases, a nucleic acid comprising a nucleotide sequence encoding a light-responsive opsin protein can be delivered directly to the neurons responsible for pain, where the delivery can be accomplished with a needle, catheter, or related device, using neurosurgical techniques known in the art, such as by stereotactic injection or fluoroscopy. Other methods to deliver a nucleic acid comprising a nucleotide sequence encoding a light-responsive opsin protein to the nerves of interest can also be used, such as, but not limited to, transfection with ionic lipids or polymers, electroporation, optical transfection, impalefection, or via gene gun.

Hyperpolarizing light-responsive polypeptides

[0050] As discussed above, methods for controlling pain according to the present disclosure generally involve introducing into a neuron of an individual a nucleic acid comprising a nucleotide sequence encoding a light-activated polypeptide (an opsin) that provides for hyperpolarization of the cell in response to light of a wavelength that activates the light-activated polypeptide. A light-activated polypeptide can be a polypeptide that allows one or more ions to pass through the plasma membrane of a target cell when the protein is illuminated with light of an activating wavelength. Light-activated proteins may be characterized as ion pump proteins, which facilitate the passage of a small number of ions through the plasma membrane per photon of light, or as ion channel proteins, which allow a stream of ions to freely flow through the plasma membrane when the channel is open. Suitable light-activated proteins for use in a subject method of reducing pain include hyperpolarizing light-activated polypeptides.

[0051] Examples of suitable light-responsive polypeptides include, e.g., the Halorhodopsin family of light- responsive chloride pumps (e.g., NpHR, NpHR2.0, NpHR3.0, NpHR3.1). As another example, the GtR3 proton pump can be used to promote neural cell membrane hyperpolarization in response to light. As another example, eArch (a proton pump) can be used to promote neural cell membrane hyperpolarization in response to light. As another example, an ArchT opsin protein or a Mac opsin protein can be used to promote neural cell membrane hyperpolarization in response to light. Enhanced intracellular transport amino acid motifs

[0052] In some embodiments, the light -responsive opsin proteins expressed in a cell can be fused to one or more amino acid sequence motifs selected from the group consisting of a signal peptide, an ER export signal, a membrane trafficking signal, and/or an N-terminal golgi export signal. The one or more amino acid sequence motifs which enhance light-responsive protein transport to the plasma membranes of mammalian cells can be fused to the N-terminus, the C-terminus, or to both the N- and C-terminal ends of the light-responsive protein. In some cases, the one or more amino acid sequence motifs which enhance light-responsive protein transport to the plasma membranes of mammalian cells is fused internally within a light-activated polypeptide. Optionally, the light-responsive protein and the one or more amino acid sequence motifs may be separated by a linker. In some embodiments, the light- responsive protein can be modified by the addition of a trafficking signal (ts) which enhances transport of the protein to the cell plasma membrane. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence

KSRITSEGEYIPLDQIDINV (SEQ ID NO:22).

[0053] Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least

85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)).

[0054] A trafficking sequence can have a length of from about 10 amino acids to about 50 amino acids, e.g., from about 10 amino acids to about 20 amino acids, from about 20 amino acids to about 30 amino acids, from about 30 amino acids to about 40 amino acids, or from about 40 amino acids to about 50 amino acids.

[0055] Signal sequences that are suitable for use can comprise an amino acid sequence having at least 85%,

90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such as one of the following:

[0056] 1) the signal peptide of hChR2 (e.g., MDYGGALSAVGRELLFVTNPVWNGS (SEQ ID NO:23))

[0057] 2) the β2 subunit signal peptide of the neuronal nicotinic acetylcholine receptor (e.g.,

MAGHSNSMALFSFSLLWLCSGVLGTEF (SEQ ID NO:24));

[0058] 3) a nicotinic acetylcholine receptor signal sequence (e.g., MGLRALMLWLLAAAGLVRESLQG

(SEQ ID NO:25)); and

[0059] 4) a nicotinic acetylcholine receptor signal sequence (e.g., MRGTPLLLWSLFSLLQD (SEQ ID

NO:26)).

[0060] A signal sequence can have a length of from about 10 amino acids to about 50 amino acids, e.g., from about 10 amino acids to about 20 amino acids, from about 20 amino acids to about 30 amino acids, from about 30 amino acids to about 40 amino acids, or from about 40 amino acids to about 50 amino acids. [0061] ER export sequences that are suitable for use in a modified opsin include, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.);

NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like. An ER export sequence can have a length of from about 5 amino acids to about 25 amino acids, e.g., from about 5 amino acids to about 10 amino acids, from about 10 amino acids to about 15 amino acids, from about 15 amino acids to about 20 amino acids, or from about 20 amino acids to about 25 amino acids.

[0062] In some embodiments, the signal peptide sequence in the protein can be deleted or substituted with a signal peptide sequence from a different protein.

Arch

[0063] In some embodiments, a suitable light-activated protein is an Archaerhodopsin (Arch) proton pump (e.g., a proton pump derived from Halorubrum sodomense) that can transport one or more protons across the plasma membrane of a cell when the cell is illuminated with light. The light can have a wavelength between about 530 and about 595 nm or can have a wavelength of about 560 nm. In some embodiments, the Arch protein can comprise an amino acid sequence that is at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:l (Arch). The Arch protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the Arch protein to transport ions across the plasma membrane of a target cell. Additionally, the Arch protein can contain one or more conservative amino acid substitutions and/or one or more non- conservative amino acid substitutions. The Arch protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to transport ions across the plasma membrane of a target cell in response to light.

[0064] In some embodiments, an Arch protein comprises at least one (such as one, two, three, or more) amino acid sequence motifs that enhance transport to the plasma membranes of target cells selected from the group consisting of a signal peptide, an ER export signal, and a membrane trafficking signal. In some embodiments, the Arch protein comprises an N-terminal signal peptide and a C-terminal ER export signal. In some embodiments, the Arch protein comprises an N-terminal signal peptide and a C- terminal trafficking signal. In some embodiments, the Arch protein comprises an N-terminal signal peptide, a C-terminal ER export signal, and a C-terminal trafficking signal. In some embodiments, the Arch protein comprises a C-terminal ER export signal and a C-terminal trafficking signal. In some embodiments, the C-terminal ER export signal and the C-terminal trafficking signal are linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER export signal is more C-terminally located than the trafficking signal. In some embodiments the trafficking signal is more C- terminally located than the ER Export signal.

[0065] In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%o, 99%, or 100%>, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g.,

KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.);

NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

ArchT

[0066] In some embodiments, a suitable light-activated protein is an Archaerhodopsin (ArchT) proton pump (e.g., a proton pump derived from Halorubrum sp. TP009) that can transport one or more protons across the plasma membrane of a cell when the cell is illuminated with light. The light can have a wavelength between about 530 and about 595 nm or can have a wavelength of about 560 nm. In some embodiments, the Arch protein can comprise an amino acid sequence that is at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:3 (ArchT). The ArchT protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the ArchT protein to transport ions across the plasma membrane of a target cell. Additionally, the ArchT protein can contain one or more conservative amino acid substitutions and/or one or more non- conservative amino acid substitutions. The ArchT protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to transport ions across the plasma membrane of a target cell in response to light.

[0067] In some cases, the ArchT polypeptide comprises a membrane trafficking signal and/or an ER export signal. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%., 95%, 96%, 97%, 98%, 99%, or 100%., amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYrPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.); NANSFCYENEVALTS (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID N0:31); and the like.

GtR3

[0068] In some embodiments, the light -responsive proton pump protein can be responsive to blue light and can be derived from Guillardia theta, wherein the proton pump protein can be capable of mediating a hyp er ol arizing current in the cell when the cell is illuminated with blue light. The light can have a wavelength between about 450 and about 495 nm or can have a wavelength of about 490 nm. In another embodiment, the light-responsive proton pump protein can comprise an amino acid sequence at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:4 (GtR3). The light-responsive proton pump protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the light-responsive proton pump protein to regulate the polarization state of the plasma membrane of the cell. Additionally, the light-responsive proton pump protein can contain one or more conservative amino acid substitutions and/or one or more non-conservative amino acid substitutions. The light-responsive proton pump protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to hyperpolarize the plasma membrane of a neuronal cell in response to light.

[0069] In other aspects of the methods disclosed herein, the light-responsive proton pump protein can comprise a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:4 and at least one (such as one, two, three, or more) amino acid sequence motifs which enhance transport to the plasma membranes of mammalian cells selected from the group consisting of a signal peptide, an ER export signal, and a membrane trafficking signal. In some embodiments, the light-responsive proton pump protein comprises an N-terminal signal peptide and a C-terminal ER export signal. In some embodiments, the light-responsive proton pump protein comprises an N-terminal signal peptide and a C-terminal trafficking signal. In some embodiments, the light-responsive proton pump protein comprises an N- terminal signal peptide, a C-terminal ER Export signal, and a C-terminal trafficking signal. In some embodiments, the light-responsive proton pump protein comprises a C-terminal ER Export signal and a C-terminal trafficking signal. In some embodiments, the C-terminal ER Export signal and the C- terminal trafficking signal are linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER Export signal is more C-terminally located than the trafficking signal. In some embodiments the trafficking signal is more C-terminally located than the ER Export signal. [0070] In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%o, 99%, or 100%>, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g.,

KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.);

NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

[0071] Also provided herein are isolated polynucleotides encoding any of the light-responsive proton pump proteins described herein, such as a light-responsive proton pump protein comprising a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:4. Also provided herein are expression vectors (such as a viral vector described herein) comprising a polynucleotide encoding the proteins described herein, such as a light-responsive proton pump protein comprising a core amino acid sequence at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:4.

Oxy

[0072] In some embodiments, a light-activated protein is an Oxyrrhis marina (Oxy) proton pump that can transport one or more protons across the plasma membrane of a cell when the cell is illuminated with light. The light can have a wavelength between about 500 and about 560 nm or can have a wavelength of about 530 nm. In some embodiments, the Oxy protein can comprise an amino acid sequence that is at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:5. The Oxy protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the Oxy protein to transport ions across the plasma membrane of a target cell. Additionally, the Oxy protein can contain one or more conservative amino acid substitutions and or one or more non- conservative amino acid substitutions. The Oxy protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to transport ions across the plasma membrane of a target cell in response to light.

[0073] In some embodiments, an Oxy protein comprises at least one (such as one, two, three, or more) amino acid sequence motifs that enhance transport to the plasma membranes of target cells selected from the group consisting of a signal peptide, an ER export signal, and a membrane trafficking signal. In some embodiments, the Oxy protein comprises an N-terminal signal peptide and a C-terminal ER export signal. In some embodiments, the Oxy protein comprises an N-terminal signal peptide and a C-terminal trafficking signal. In some embodiments, the Oxy protein comprises an N-terminal signal peptide, a C- terminal ER export signal, and a C-terminal trafficking signal. In some embodiments, the Oxy protein comprises a C-terminal ER export signal and a C-terminal trafficking signal. In some embodiments, the C-terminal ER export signal and the C-terminal trafficking signal are linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER export signal is more C-terminally located than the trafficking signal. In some embodiments the trafficking signal is more C-terminally located than the ER Export signal.

Mac

[0074] In some embodiments, the light-responsive proton pump protein can be responsive to light and can be derived from Leptosphaeria maculans, wherein the proton pump protein can be capable of pumping protons across the membrane of a cell when the cell is illuminated with 520 nm to 560 nm light. The light can have a wavelength between about 520 nm to about 560 nm. In another embodiment, the light-responsive proton pump protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 6 or SEQ ID NO: 7 (Mac; Mac 3.0). The light-responsive proton pump protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the light-responsive proton pump protein to regulate the polarization state of the plasma membrane of the cell. Additionally, the light-responsive proton pump protein can contain one or more conservative amino acid substitutions and/or one or more non-conservative amino acid substitutions. The light-responsive proton pump protein comprising substitutions, deletions, and/ or insertions introduced into the native amino acid sequence suitably retains the ability to pump protons across the plasma membrane of a neuronal cell in response to light.

[0075] In other aspects of the methods disclosed herein, the light-responsive proton pump protein can comprise a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%o, or 100%) identical to the sequence shown in SEQ ID NO:6 and at least one (such as one, two, three, or more) amino acid sequence motifs which enhance transport to the plasma membranes of mammalian cells selected from the group consisting of a signal peptide, an ER export signal, and a membrane trafficking signal. In some embodiments, the light-responsive proton pump protein comprises an N-terminal signal peptide and a C-terminal ER export signal. In some embodiments, the light-responsive proton pump protein comprises an N-terminal signal peptide and a C-terminal trafficking signal. In some embodiments, the light-responsive proton pump protein comprises an N- terminal signal peptide, a C-terminal ER Export signal, and a C-terminal trafficking signal. In some embodiments, the light-responsive proton pump protein comprises a C-terminal ER Export signal and a C-terminal trafficking signal. In some embodiments, the C-terminal ER Export signal and the C- terminal trafficking signal are linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER Export signal is more C-terminally located than the trafficking signal. In some embodiments the trafficking signal is more C-terminally located than the ER Export signal.

[0076] In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDI V (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%o, 99%, or 100%>, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g.,

KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.);

NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

[0077] Also provided herein are isolated polynucleotides encoding any of the light-responsive proton pump proteins described herein, such as a light-responsive proton pump protein comprising a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:6. Also provided herein are expression vectors (such as a viral vector described herein) comprising a polynucleotide encoding the proteins described herein, such as a light-responsive proton pump protein comprising a core amino acid sequence at least 85%, 90%, 91%, 92%, 93%o, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:6.

[0078] Further disclosure related to light-activated proton pump proteins can be found in International Patent Application No. PCT/US2011/028893, the disclosure of which is hereby incorporated by reference in its entirety.

NpHR

[0079] In some cases, a suitable light -responsive chloride pump proteins expressed on the plasma membranes of the neurons described above can be derived from Natronomonas pharaonis. In some embodiments, the / g ¾/-responsive chloride pump proteins can be responsive to amber light as well as red light and can mediate a hyperpolarizing current in the neuron when the light-responsive chloride pump proteins are illuminated with amber or red light. The wavelength of light which can activate the light-responsive chloride pumps can be between about 580 and 630 nm. In some embodiments, the light can be at a wavelength of about 589 nm or the light can have a wavelength greater than about 630 nm (e.g. less than about 740 nm). In another embodiment, the light has a wavelength of around 630 nm. In some embodiments, the light-responsive chloride pump protein can hyperpolarize a neural membrane for at least about 90 minutes when exposed to a continuous pulse of light. In some embodiments, the light- responsive chloride pump protein can comprise an amino acid sequence at least about 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:16. Additionally, the light-responsive chloride pump protein can comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the light-responsive protein to regulate the polarization state of the plasma membrane of the cell. In some embodiments, the light-responsive chloride pump protein contains one or more conservative amino acid substitutions. In some embodiments, the light-responsive protein contains one or more non-conservative amino acid substitutions. The light-responsive protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to hyperpolarize the plasma membrane of a neuronal cell in response to light.

[0080] Additionally, in other aspects, the light-responsive chloride pump protein can comprise a core amino acid sequence at least about 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 16 and an endoplasmic reticulum (ER) export signal. This ER export signal can be fused to the C-terminus of the core amino acid sequence or can be fused to the N-terminus of the core amino acid sequence. In some embodiments, the ER export signal is linked to the core amino acid sequence by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments, the ER export signal can comprise the amino acid sequence FXYENE (SEQ ID NO:30), where X can be any amino acid. In another embodiment, the ER export signal can comprise the amino acid sequence VXXSL, where X can be any amino acid. In some embodiments, the ER export signal can comprise the amino acid sequence FCYENEV (SEQ ID NO:31).

[0081] Endoplasmic reticulum (ER) export sequences that are suitable for use in a modified opsin of the

present disclosure include, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.); NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (where X is any amino acid) (SEQ ID NO:30), e.g., FCYENEV (SEQ ID NO:31); and the like. An ER export sequence can have a length of from about 5 amino acids to about 25 amino acids, e.g., from about 5 amino acids to about 10 amino acids, from about 10 amino acids to about 15 amino acids, from about 1 amino acids to about 20 amino acids, or from about 20 amino acids to about 25 amino acids.

[0082] In other aspects, the light-responsive chloride pump proteins described herein can comprise a light- responsive protein expressed on the cell membrane, wherein the protein comprises a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 16 and a trafficking signal (e.g., which can enhance transport of the light-responsive chloride pump protein to the plasma membrane). The trafficking signal may be fused to the C-terminus of the core amino acid sequence or may be fused to the N- terminus of the core amino acid sequence. In some embodiments, the trafficking signal can be linked to the core amino acid sequence by a linker which can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22).

[0083] In some aspects, the light-responsive chloride pump protein can comprise a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 16 and at least one (such as one, two, three, or more) amino acid sequence motifs which enhance transport to the plasma membranes of mammalian cells selected from the group consisting of an ER export signal, a signal peptide, and a membrane trafficking signal. In some embodiments, the light-responsive chloride pump protein comprises an N-terminal signal peptide, a C-tenninal ER Export signal, and a C-terminal trafficking signal. In some embodiments, the C- terminal ER Export signal and the C-terminal trafficking signal can be linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker can also further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER Export signal can be more C-terminally located than the trafficking signal. In other embodiments the trafficking signal is more C-terminally located than the ER Export signal. In some embodiments, the signal peptide comprises the amino acid sequence MTETLPPVTESAVALQAE (SEQ ID NO:32). In another embodiment, the light-responsive chloride pump protein comprises an amino acid sequence at least 95% identical to SEQ ID NO: 17.

[0084] Moreover, in other aspects, the light-responsive chloride pump proteins can comprise a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 16, wherein the N-terminal signal peptide of SEQ ID NO: 16 is deleted or substituted. In some embodiments, other signal peptides (such as signal peptides from other opsins) can be used. The light-responsive protein can further comprise an ER transport signal and/or a membrane trafficking signal described herein. In some embodiments, the light- responsive chloride pump protein comprises an amino acid sequence at least 95% identical to SEQ ID NO: 18.

[0085] In some embodiments, the light -responsive opsin protein is a NpHR opsin protein comprising an amino acid sequence at least 75%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical to the sequence shown in SEQ ID NO:16. In some embodiments, the NpHR opsin protein further comprises an endoplasmic reticulum (ER) export signal and/or a membrane trafficking signal. For example, the NpHR opsin protein comprises an amino acid sequence at least 95% identical to the sequence shown in SEQ ID NO: 16 and an endoplasmic reticulum (ER) export signal. In some embodiments, the amino acid sequence at least 95% identical to the sequence shown in SEQ ID NO: 16 is linked to the ER export signal through a linker. In some embodiments, the ER export signal comprises the amino acid sequence FXYENE (SEQ ID NO:30), where X can be any amino acid. In another embodiment, the ER export signal comprises the amino acid sequence VXXSL, where X can be any amino acid. In some embodiments, the ER export signal comprises the amino acid sequence FCYENEV (SEQ ID NO:31). In some embodiments, the NpHR opsin protein comprises an amino acid sequence at least 95% identical to the sequence shown in SEQ ID NO: 16, an ER export signal, and a membrane trafficking signal. In other embodiments, the NpHR opsin protein comprises, from the N-terminus to the C-terminus, the amino acid sequence at least 95% identical to the sequence shown in SEQ ID NO: 16, the ER export signal, and the membrane trafficking signal. In other embodiments, the NpHR opsin protein comprises, from the N-terminus to the C-terminus, the amino acid sequence at least 95% identical to the sequence shown in SEQ ID NO: 16, the membrane trafficking signal, and the ER export signal. In some embodiments, the membrane trafficking signal is derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In some embodiments, the membrane trafficking signal comprises the amino acid sequence

KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). In some embodiments, the membrane trafficking signal is linked to the amino acid sequence at least 95% identical to the sequence shown in SEQ ID NO: 16 by a linker. In some embodiments, the membrane trafficking signal is linked to the ER export signal through a linker. The linker may comprise any of 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments, the light- responsive opsin protein further comprises an N-terminal signal peptide. In some embodiments, the light-responsive opsin protein comprises the amino acid sequence of SEQ ID NO: 17. In some embodiments, the light-responsive opsin protein comprises the amino acid sequence of SEQ ID NO: 18. Also provided herein are polynucleotides encoding any of the light-responsive chloride ion pump proteins described herein, such as a light-responsive protein comprising a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:16, an ER export signal, and a membrane trafficking signal. In another embodiment, the polynucleotides comprise a sequence which encodes an amino acid at least 95% identical to SEQ ID NO:17 and SEQ ID NO: 18. The polynucleotides may be in an expression vector (such as, but not limited to, a viral vector described herein). The polynucleotides may be used for expression of the light-responsive chloride ion pump proteins. [0087] Further disclosure related to light-responsive chloride pump proteins can be found in U.S. Patent Application Publication Nos: 2009/0093403 and 2010/0145418 as well as in International Patent Application No: PCT/US2011/028893, the disclosures of each of which are hereby incorporated by reference in their entireties.

Dunaliella salina light-activated polypeptide

[0088] In some embodiments, a suitable light-responsive ion channel protein can be responsive to 470 nm -

510 nm light and can be derived from Dunaliella salina, wherein the ion channel protein can be capable of mediating a hyperpolarizing current in the cell when the cell is illuminated with light. The light can have a wavelength between about 470 nm and about 510 nm or can have a wavelength of about 490 nm. In some embodiments, the light-responsive ion channel protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 15. The light-responsive ion channel protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the light-responsive ion channel protein to regulate the polarization state of the plasma membrane of the cell. Additionally, the light-responsive ion channel protein can contain one or more conservative amino acid substitutions and/or one or more non-conservative amino acid substitutions. The light-responsive ion channel protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to transport ions across the plasma membrane of a neuronal cell in response to light.

[0089] In other aspects of the methods disclosed herein, the light-responsive ion channel protein can comprise a core amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 15 and at least one (such as one, two, three, or more) amino acid sequence motifs which enhance transport to the plasma membranes of mammalian cells selected from the group consisting of a signal peptide, an ER export signal, and a membrane trafficking signal. In some embodiments, the light-responsive proton ion channel comprises an N-terminal signal peptide and a C-terminal ER export signal. In some embodiments, the light- responsive ion channel protein comprises an N-terminal signal peptide and a C-terminal trafficking signal. In some embodiments, the light-responsive ion channel protein comprises an N-terminal signal peptide, a C-terminal ER Export signal, and a C-terminal trafficking signal. In some embodiments, the light-responsive ion channel protein comprises a C-terminal ER Export signal and a C-terminal trafficking signal. In some embodiments, the C-terminal ER Export signal and the C-terminal trafficking signal are linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER Export signal is more C-terminally located than the trafficking signal. In some embodiments the trafficking signal is more C-terminally located than the ER Export signal.

[0090] Also provided herein are isolated polynucleotides encoding any of the light-responsive channel proteins described herein, such as a light-responsive ion channel protein comprising a core amino acid sequence at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:15. Also provided herein are expression vectors (such as a viral vector described herein) comprising a polynucleotide encoding the proteins described herein, such as a light- responsive channel protein comprising a core amino acid sequence at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 15.

Polynucleotides and Vectors

[0091] As discussed above, methods for controlling pain according to the present disclosure generally involve introducing into a neuron (e.g., a nociceptor) of an individual a nucleic acid comprising a nucleotide sequence encoding an opsin that provides for hyperpolarization of the cell in response to light of a wavelength that activates the opsin. Suitable nucleic acids comprise a nucleotide sequence that encodes one or more of the light-activated polypeptides (opsins) described herein (e.g., one or more light- activated polypeptides as described herein). In some embodiments, a polynucleotide comprises an expression cassette, wherein the expression cassette contains a plurality of components (e.g., coding sequences; transcription control sequences; etc.) that are utilized to express one or more proteins encoded by the polynucleotide in a target cell.

[0092] In some embodiments, a portion of a polynucleotide encoding a light-activated polypeptide is operably linked to a promoter sequence. Any suitable promoter that functions in a target cell can be used for expression of a polynucleotide encoding a light-activated polypeptide. In certain embodiments, a promoter sequence can be a promoter that is specific to a particular target cell type or to a particular tissue type, such as a particular neuron or a pan-neuronal promoter. Initiation control regions of promoters, which are useful to drive expression of polynucleotides in a specific animal cell, are numerous and familiar to those skilled in the art. Virtually any promoter capable of driving expression of the subject polynucleotides can be used. In some embodiments, the promoter used to drive expression of a subject protein can be the Thyl promoter (See, e.g., Llewellyn, et al., 2010, Nat. Med., 16(10): 1161 -1166). In some embodiments, the promoter used to drive expression of a subject protein can be a human synapsin (hSyn) promoter, a human elongation factor 1- (EFl ) promoter, a cytomegalovirus (CMV) promoter, a CMV early enhancer/chicken β actin (CAG) promoter, a synapsin-I promoter (e.g., a human synapsin-I promoter), a human synuclein 1 promoter, a human Thyl promoter, a calcium/calmodulin-dependent kinase II alpha (CAMKIIa) promoter, or any other promoter capable of driving expression of the a subject nucleic acid sequence in a target cell.

[0093] Neuron-specific promoters and other control elements (e.g., enhancers) are known in the art, and can be operably linked to an opsin-encoding nucleotide sequence. Suitable neuron-specific control sequences include, but are not limited to, a neuron-specific enolase (NSE) promoter (see, e.g., EMBL HSEN02, X51956; see also, e.g., U.S. Pat. No. 6,649,811 , U.S. Pat. No. 5,387,742); an aromatic amino acid decarboxylase (AADC) promoter; a neurofilament promoter (see, e.g., GenBank HUMNFL, L04147); a synapsin promoter (see, e.g., GenBank HUMSYNIB, M55301); a thy-1 promoter (see, e.g., Chen et al. (1987) Cell 51 :7-19); a serotonin receptor promoter (see, e.g., GenBank S62283); a tyrosine hydroxylase promoter (TH) (see, e.g., Nucl. Acids. Res. 15:2363-2384 (1 87) and Neuron 6:583-594 (1991)); a GnRH promoter (see, e.g., Radovick et al, Proc. Natl. Acad. Sci. USA 88:3402-3406 (1991)); an L7 promoter (see, e.g., Oberdick et al., Science 248:223-226 (1990)); a DNMT promoter (see, e.g., Bartge et al., Proc. Natl. Acad. Sci. USA 85:3648-3652 (1988)); an enkephalin promoter (see, e.g., Comb et al, EMBO J. 17:3793-3805 (1988)); a myelin basic protein (MBP) promoter; a CMV enhancer/platelet-derived growth factor-β promoter (see, e.g., Liu et al. (2004) Gene Therapy 11 :52- 60); a motor neuron-specific gene Hb9 promoter (see, e.g., U.S. Pat. No. 7, 632,679; and Lee et al. (2004) Development 131 :3295-3306); and an alpha subunit of Ca( 2+ )-calmodulin-dependent protein kinase II (CaMKIIa) promoter (see, e.g., Mayford et al. (1996) Proc. Natl. Acad. Sci. USA 93:13250).

[0094] In some embodiments, a promoter may be an inducible promoter. For example, the promoter may be induced by a trans-acting factor that responds to an exogenously administered drug. Examples of inducible promoters include, but are not limited to, tetracycline-on or tetracycline-off promoters, or tamoxifen-inducible CreER.

[0095] In some embodiments, a subject polynucleotide may comprise a ribosomal skip sequence that can be used to generate two separate proteins from the same transcript. In such embodiments, a subject polynucleotide will typically include a coding sequence that encodes a light-activated protein as well as a response protein. In these embodiments, a ribosomal skip sequence may be placed between the two coding sequences to produce two distinct proteins (namely, the light-activated protein and the response protein) from the same transcript.

[0096] As noted above, in some cases, the nucleic acid is a recombinant expression vector comprising a

nucleotide sequence encoding a light-activated polypeptide or any variant thereof as described herein. Suitable expression vectors include vectors comprising a nucleotide sequence that encodes an RNA (e.g., an mRNA) that when transcribed from the polynucleotides of the vector will result in the accumulation of a subject protein on the plasma membranes of target cells. Vectors which may be used include, without limitation, lentiviral, herpes simplex virus, adenoviral, and adeno-associated virus (AAV) vectors. Lentiviral vectors include, but are not limited to human immunodeficiency virus (HTV)- based vectors. Lentiviral vectors may be pseudotyped with the envelope proteins of other viruses, including, but not limited to vesicular stomatitis virus (VSV), rabies, Mo-murine leukemia virus (MLV), baculovirus and Ebola. Such vectors may be prepared using standard methods in the art.

[0097] In some embodiments, a vector may be a recombinant AAV vector. AAV vectors are DNA viruses of relatively small size that can integrate, in a stable and site-specific manner, into the genome of the cells that they infect. They are able to infect a wide spectrum of cells without inducing any effects on cellular growth, morphology or differentiation, and they do not appear to be involved in human pathologies. The AAV genome has been cloned, sequenced and characterized. It encompasses approximately 4700 bases and contains an inverted terminal repeat (ITR) region of approximately 145 bases at each end, which serves as an origin of replication for the virus. The remainder of the genome is divided into two essential regions that carry the encapsidation functions: the left-hand part of the genome that contains the rep gene involved in viral replication and expression of the viral genes; and the right-hand part of the genome that contains the cap gene encoding the capsid proteins of the virus.

[0098] AAV vectors may be prepared using standard methods in the art. Adeno-associated viruses of any serotype are suitable (see, e.g., Blacklow, pp. 165-174 of "Parvoviruses and Human Disease" J. R. Pattison, ed. (1988); Rose, Comprehensive Virology 3:1, 1974; P. Tattersall "The Evolution of Parvovirus Taxonomy" In Parvoviruses (JR Kerr, SF Cotmore. ME Bloom, RM Linden, CR Parrish, Eds.) p5-14, Hudder Arnold, London, UK (2006); and DE Bowles, JE Rabinowitz, RJ Samulski "The Genus Dependovirus" (JR Kerr, SF Cotmore. ME Bloom, RM Linden, CR Parrish, Eds.) pl5-23, Hudder Arnold, London, UK (2006), the disclosures of each of which are hereby incorporated by reference herein in their entireties). Methods for purifying for vectors may be found in, for example, U.S. Pat. Nos. 6,566,118, 6,989,264, and 6,995,006 and WO/1999/011764 titled "Methods for Generating High Titer Helper-free Preparation of Recombinant AAV Vectors", the disclosures of which are herein incorporated by reference in their entirety. Methods of preparing AAV vectors in a baculovirus system are described in, e.g., WO 2008/024998. AAV vectors can be self-complementary or single-stranded. Preparation of hybrid vectors is described in, for example, PCT Application No. PCT/US2005/0270 1, the disclosure of which is herein incorporated by reference in its entirety. The use of vectors derived from the AAVs for transferring genes in vitro and in vivo has been described (See e.g., International Patent Application Publication Nos. : 91/18088 and WO 93/09239; U.S. Patent Nos.: 4,797,368, 6,596,535, and 5,139,941 ; and European Patent No.: 0488528, all of which are hereby incorporated by reference herein in their entireties). These publications describe various AAV-derived constructs in which the rep and/or cap genes are deleted and replaced by a gene of interest, and the use of these constructs for transferring the gene of interest in vitro (into cultured cells) or in vivo (directly into an organism). The replication-defective recombinant AAVs according to the present disclosure can be prepared by co-transfecting a plasmid containing the nucleic acid sequence of interest flanked by two AAV inverted terminal repeat (ITR) regions, and a plasmid carrying the AAV encapsidation genes (rep and cap genes), into a cell line that is infected with a human helper virus (for example an adenovirus). The AAV recombinants that are produced are then purified by standard techniques.

[0099] In some embodiments, the vector(s) for use in the methods of the present disclosure are encapsidated into a virus particle (e.g. AAV virus particle including, but not limited to, AAV1 , AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAVl 1, AAV12, AAV13, AAV14, AAV15, and AAV16). Accordingly, the present disclosure includes a recombinant virus particle (recombinant because it contains a recombinant polynucleotide) comprising any of the vectors described herein. Methods of producing such particles are known in the art and are described in U.S. Patent No. 6,596,535, the disclosure of which is hereby incorporated by reference in its entirety. In some cases, AAV6 is used. In some cases, AAV1 is used.

Pharmaceutical compositions

[00100] Aspects of the present disclosure include pharmaceutical compositions that polynucleotides, vectors, or components thereof, described above. The subject pharmaceutical compositions may be administered to a subject for purposes of genetically modifying a target cell so that the target cell expresses one or more light-activated proteins. A subject pharmaceutical composition may, in some embodiments, comprise a pharmaceutically acceptable excipient. In some embodiments, a pharmaceutical composition may comprise components to facilitate delivery of the subject polynucleotides or vectors to a target cell, including but not limited to transfection reagents or components thereof, such as lipids, polymers, and the like.

[00101] In some embodiments, a subject pharmaceutical composition will be suitable for injection into a subject, e.g., will be sterile. For example, in some embodiments, a subject pharmaceutical composition will be suitable for injection into a subject, e.g., where the composition is sterile and is free of detectable pyrogens and or other toxins.

[00102] Pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public as well, and may be incorporated into the pharmaceutical compositions of the present disclosure without limitation.

Delivery Devices

[00103] In some cases, a delivery device is used to deliver a nucleic acid encoding a light- activated polypeptide, or a pharmaceutical composition comprising same, to a target cell. The delivery device may provide regular, irregular, programmed, or clinician- or patient-activated doses of the nucleic acid or pharmaceutical composition to one or more target cells to ensure that the target cells continue to express the encoded light- activated polypeptide.

[00104] Suitable delivery devices may generally include one or more components, such as reservoirs, pumps, actuators, tubing components, needles, catheters, and any other suitable components for delivering the nucleic acid or pharmaceutical composition to a target cell or tissue of an individual. Delivery devices may also include components that facilitate computerized operation, such as a power source, a processor comprising a memory, a user input device, and/or a graphical user interface. In some embodiments, a delivery device may be completely or partially implantable within a patient. In some embodiments, a delivery device may be operated by a caregiver, wherein the device is introduced into a portion of the patient's body, e.g., into the patient's brain, and a subject pharmaceutical composition is delivered to a target tissue, e.g., a portion of the patient's brain. In some embodiments, following delivery of the pharmaceutical composition, the device may be removed. In other embodiments, the device may be kept in place for later delivery of additional pharmaceutical compositions.

Light-generating Devices

[00105] In carrying out a subject method of controlling pain, a light-generating device can be used to deliver light to target cells that express one or more light-activated polypeptides. Light-generating devices suitable for use with a method of the present disclosure can generally produce light of a variety of different wavelengths from one or more light sources on the device. In some embodiments, a light- generating device may include a light cuff or sleeve that can be placed around or near target cells expressing one or more subject proteins. In some embodiments, a portion of the light source or the entire light source may be implantable. The subject light-generating devices may be of any useful configuration for stimulating the light-activated proteins disclosed herein. In some embodiments, for example, a light-generating device may comprise components that facilitate exclusive illumination of a target cell or tissue. For example, in some embodiments, a light-generating device may exclusively direct light to a target cell, a portion of a target cell, e.g., a particular axon of a nerve cell, or a specific anatomical structure, such as, e.g. a bundle of nerve fibers, a target tissue, or a portion of the spinal cord. By "exclusively direct light" is meant that the light-generating device only delivers light to the specific target structure, and does not illuminate other structures. For examples, in some embodiments, a light-generating device may be configured to illuminate an axon of a nerve cell, but not illuminate any other portion of the nerve cell. In this way, the light from the light-generating device only affects light- activated proteins in the specific target structure that is illuminated.

[00106] In some embodiments, a light-generating device may not completely surround the region containing a target cell expressing a light-activated protein, but, rather, can have a U-shape. In some embodiments, a light-generating device can have an attachment arm that can be used to guide the light- generating device to a specific region or target structure, e.g., a specific neuronal region. The attachment arm can be removed following implantation of the light-generating device or can be left in place to fix the position of the light-generating device in proximity to the target cells of interest.

[00107] In some embodiments, the subject light-generating devices may comprise an inner body, the inner body having at least one means for generating light which is connected to a power source. In some embodiments, the power source can be an internal battery for powering the light-generating device. In some embodiments, an implantable light-generating device may comprise an external antenna for receiving wirelessly transmitted electromagnetic energy from an external source for powering device. The wirelessly transmitted electromagnetic energy can be a radio wave, a microwave, or any other electromagnetic energy source that can be transmitted from an external source to power the light-generating device. In some embodiments, the light-generating device is controlled by, e.g., an integrated circuit produced using semiconductor or other processes known in the art.

[00108] In some embodiments, the light-generating device may comprise a light emitting diode (LED).

In some embodiments, the LED can generate blue and/or green light. In other embodiments, the LED can generate amber and/or yellow light. In some embodiments, several micro LEDs are embedded into the inner body of the light-generating device. In other embodiments, the light-generating device is a solid state laser diode or any other means capable of generating light. The light-generating device can generate light having a wavelength and intensity sufficient to activate a subject light-activated protein. In some embodiments, a light-generating device produces light having an intensity of any of about 0.05 mW/mm 2 , 0.1 mW/mm 2 , 0.2 mW/mm 2 , 0.3 mW/mm 2 , 0.4 mW/mm 2 , 0.5 mW/mm 2 , about 0.6 mW/mm , about 0.7 mW/mm , about 0.8 mW/mm , about 0.9 mW/mm , about 1.0 mW/mm , about 1.1 mW/mm , about 1.2 mW/mm 2 , about 1.3 mW/mm 2 , about 1.4 mW/mm , about 1.5 mW/mm 2 , about 1.6 mW/mm 2 , about 1.7 mW/mm 2 , about 1.8 mW/mm 2 , about 1.9 mW/mm 2 , about 2.0 mW/mm 2 , about 2.1 mW/mrn 2 , about 2.2 mW/mm 2 , about 2.3 mW/mm 2 , about 2.4 mW/mm 2 , about 2.5 mW/mm 2 , about 3 mW/mm 2 , about 3.5 mW/mm 2 , about 4 mW/mm 2 , about 4.5 mW/mm 2 , about 5 mW/mm 2 , about 5.5 mW/mm 2 , about 6 mW/mm 2 , about 7 mW/mm 2 , about 8 mW/mm 2 , about 9 mW/mm 2 , or about 10 mW/mm 2 , inclusive, including values in between these numbers. In some embodiments, the light- generating device produces light having an intensity of at least about 10 Hz, such as up to about 25 Hz, such as up to about 50 Hz, such as up to about 75 Hz, such as up to about 100 Hz.

[00109] Suitable light-generating devices are generally capable of generating light having a wavelength ranging from about 350 nm, up to about 360 nm, up to about 370 nm, up to about 380 nm, up to about 390 nm, up to about 400 nm, up to about 410 nm, up to about 420 nm, up to about 430 nm, up to about 440 nm, up to about 450 nm, up to about 460 nm, up to about 470 nm, up to about 480 nm, up to about 490 nm, up to about 500 nm, up to about 510 nm, up to about 520 nm, up to about 530 nm, up to about 540 nm, up to about 550 nm, up to about 560 nm, up to about 570 nm, up to about 580 nm, up to about 590 nm, up to about 600 nm, up to about 610 nm, up to about 620 nm, up to about 630 nm, up to about 640 nm, up to about 650 nm, up to about 660 nm, up to about 670 nm, up to about 680 nm, up to about 690 nm, up to about 700 nm, up to about 710 nm, up to about 720 nm, up to about 730 nm, up to about 740 nm, and/or up to about 750 nm.

[00110] In some embodiments, a suitable light-generating device may include one or more optical fibers that can transmit light from a light source and deliver the light to a target structure. The optical fibers may comprise plastic or glass materials, and in some embodiments may be suitably flexible to facilitate placement of the light-generating device in locations that could not be accommodated by rigid structures. For example, in some embodiments, a light-generating device may comprise a light source that generates light, as well as one or more optical fibers that can be placed in various locations on or in the patient's body. Light from the light source can pass through the optical fiber, passing around corners and bends in the optical fiber, and emerge at the end of the optical fiber to deliver light to a target structure.

[00111] In some embodiments, a suitable light-generating device may comprise a plurality of light sources that can be used to illuminate a target tissue with different wavelengths of light. For example, in some embodiments, a light-generating device may comprise a first light source that generates light of a first wavelength, e.g., red light, and a second light source that generates light of a second wavelength, e.g., green light. Such light-generating devices may be used to simultaneously illuminate the same target tissue with light of both wavelengths, or may alternately illuminate the target tissue with light of the first wavelength and light of the second wavelength. In some embodiments, such light generating devices may be used to deliver light from the same light source different target tissues. For example, in some embodiments a light-generating device may deliver light of a first wavelength to a first target tissue, and may deliver light of a second wavelength to a different target tissue.

Control Devices

[00112] In some cases, a control device that can control, or modulate, the amount of light that is emitted from the light-generating device is used in a subject method. In some embodiments, a control device may be configured to modulate the wavelength and/or the intensity of light that is delivered to a target tissue from a light-generating device. In some embodiments, a control device may be configured to modulate the frequency and/or duration of light that is delivered to a target tissue from a light- generating device. For example, in some embodiments, a control device may be configured to deliver pulses of light from the light-generating device to a target tissue. The control device can modulate the frequency and or duration of the light pulses such that the target tissue is illuminated with light from the light-generating device, e.g., at a regular or irregular rate, according to a user input, etc. In some embodiments, a control device can produce pulses of light from the light-generating device that have a duration ranging from about 1 millisecond or less, up to about 1 second, up to about 10 seconds, up to about 20 seconds, up to about 30 seconds, up to about 40 seconds, up to about 50 seconds, up to about 60 seconds or more. In some embodiments, a control device can produce pulses of light from the light- generating device that have a frequency of 1 pulse per millisecond, up to about 1 pulse per second, up about 1 pulse per minute, up to about 1 pulse per 10 minutes, up to about 1 pulse per 20 minutes, up to about 1 pulse per 30 minutes.

[00113] In some embodiments, a suitable control device may comprise a power source that can be mounted to a wireless transmitter. In some embodiments, a suitable control device may comprise a power source that can be mounted to a transmitting coil. In some embodiments, a battery can be connected to the power source for providing power thereto. A switch can be connected to the power source, allowing an operator (e.g., a patient or caregiver) to manually activate or deactivate the power source. In some embodiments, upon activation of the switch, the power source can provide power to the light-generating device through electromagnetic coupling between the transmitting coil on the control device and an antenna (which may be an external antenna or an internal antenna) of an implantable light-generating device (such as a light cuff or sleeve). The transmitting coil can establish an electromagnetic coupling with the external antenna of the implantable light-generating device when in proximity thereof, for supplying power to the light-generating device and for transmitting one or more control signals to the light-generating device. In some embodiments, the electromagnetic coupling between the transmitting coil of the control device and the external antenna of the implantable light-generating device can be radio-frequency magnetic inductance coupling. When radio-frequency magnetic inductance coupling is used, the operational frequency of the radio wave can be between about 1 and 20 MHz, inclusive, including any values in between these numbers (for example, about 1 MHz, about 2 MHz, about 3 MHz, about 4 MHz, about 5 MHz, about 6 MHz, about 7 MHz, about 8 MHz, about 9 MHz, about 10 MHz, about 11 MHz, about 12 MHz, about 13 MHz, about 14 MHz, about 15 MHz, about 16 MHz, about 17 MHz, about 18 MHz, about 19 MHz, or about 20 MHz). In some cases, the operational frequency of the radio wave can be between about 20 MHz and 5 GHz, e.g., from about 20 MHz to about 50 MHz, from about 50 MHz to about 250 MHz, from about 250 MHz to about 500 MHz, from about 500 MHz to about 750 MHz, from about 750 MHz to about 1 GHz, from about 1 GHz to about 2 GHz, from about 2 GHz to about 3 GHz, from about 3 GHz to about 4 GHz, or from about 4 GHz to about 5 GHz. For example, where midfield radiofrequency coupling is used, the operational frequency can be from 690 MHz to 2.2 GHz. Other coupling techniques may be used, such as an optical receiver, infrared, or a biomedical telemetry system (See, e.g., Kiourti, "Biomedical Telemetry: Communication between Implanted Devices and the External World, Opticonl826, (8): Spring, 2010).

NON-HUMAN ANIMAL MODEL OF PAIN

[00114] The present disclosure provides a non-human animal model of nociceptive pain, where the non- human animal expresses in a neuron (e.g., a primary afferent neuron, such as a small- or a large- diameter primary afferent neuron; e.g., a nociceptor) of the animal a nucleic acid comprising a nucleotide sequence encoding an opsin polypeptide that provides for depolarization of the nociceptor in response to light of a wavelength that activates the opsin. Illumination of the depolarizing opsin, expressed in a membrane of the neuron (e.g., primary afferent neuron, such as a small- or a large- diameter primary afferent neuron; e.g., nociceptor) in the non-human animal, induces pain in the animal.

[00115] A subject non-human animal model is useful for identifying agents that control pain (as

described below). A subject non-human animal model is useful for research applications, e.g., to investigate the role of nociceptor activity in the genesis of pain (e.g., neuropathic pain). In some cases, a subject non-human animal model of pain is a rat. In some cases, a subject non-human animal model of pain is a mouse.

[00116] In some embodiments, the non-human animal model is not a transgenic animal, e.g., the non- human animal model does not include a nucleic acid encoding a light-activated polypeptide integrated into the genome of a germ cell. In some embodiments, the non-human animal model comprises a nucleic acid encoding a light- activated polypeptide in a primary afferent neuron, such as a nociceptor, where the nucleic acid may be integrated into the genome of the primary afferent neuron (e.g., nociceptor). In some embodiments, the non-human animal model comprises a nucleic acid encoding a light-activated polypeptide in a primary afferent neuron (e.g., nociceptor), and not in a non-neuronal cell, where the nucleic acid is integrated into the genome of the nociceptor. In some embodiments, the non-human animal model comprises a nucleic acid encoding a light-activated polypeptide in a primary afferent neuron (e.g., nociceptor), and not in a non-neuronal cell of the animal, where the nucleic acid is not integrated into the genome of the nociceptor.

[00117] In some cases, a Cre-dependent DIO-AAV6 construct (see, e.g., Sohal et al. (2009) Nature

459:698) comprising a nucleotide sequence encoding a depolarizing light-activated polypeptide can be used with a nociceptor-specific Cre mouse line, to achieve opsin expression restricted to sub- populations of nociceptors. For example, a nucleotide sequence encoding a depolarizing light- responsive polypeptide is included within a Cre-dependent DIO-AAV6 construct; and the construct is introduced into nociceptors in a nociceptor-specific Cre mouse.

Depolarizing light-activated Proteins

[00118] As discussed above, a subject non-human animal model of pain expresses in a neuron (e.g., a primary afferent neuron, such as a small- or a large-diameter primary afferent neuron; e.g., a nociceptor) of the animal a nucleic acid comprising a nucleotide sequence encoding an opsin polypeptide that provides for depolarization of the nociceptor in response to light of a wavelength that activates the opsin.

[00119] Examples of suitable light-responsive polypeptides include, e.g., members of the

Channelrhodopsin family of light-responsive cation channel proteins such as Chlamydomonas rheinhardtii channelrhodopsin 2 (ChR2); a step-function opsin (SFO); a stabilized SFO (SSFO); a chimeric opsin such as C1V1; a Volvox carten ' -derived channelrhodopsin (VChRl), etc. Such light- responsive polypeptides can be used to promote neural cell membrane depolarization in response to a light stimulus.

Enhanced intracellular transport amino acid motifs

[00120] Light-responsive opsin proteins having components derived from evolutionarily simpler

organisms may not be expressed or tolerated by mammalian cells or may exhibit impaired subcellular localization when expressed at high levels in mammalian cells. Consequently, in some embodiments, the light-responsive opsin proteins expressed in a cell can be fused to one or more amino acid sequence motifs selected from the group consisting of a signal peptide, an endoplasmic reticulum (ER) export signal, a membrane trafficking signal, and/or an N-terminal golgi export signal. The one or more amino acid sequence motifs which enhance light-responsive protein transport to the plasma membranes of mammalian cells can be fused: a) to the N-terminus of the light-responsive protein; b) to the C-terminus of the light-responsive protein; c) to both the N- and C-terminal ends of the light-responsive protein; or d) internally within the light-responsive protein. Optionally, the light-responsive protein and the one or more amino acid sequence motifs may be separated by a linker.

[00121] In some embodiments, the light-responsive protein can be modified by the addition of a

trafficking signal (ts) which enhances transport of the protein to the cell plasma membrane. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel ir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 1%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g.,

KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)).

[00122] A trafficking sequence can have a length of from about 10 amino acids to about 50 amino acids, e.g., from about 10 amino acids to about 20 amino acids, from about 20 amino acids to about 30 amino acids, from about 30 amino acids to about 40 amino acids, or from about 40 amino acids to about 50 amino acids.

[00123] Signal sequences that are suitable for inclusion in a light-activated polypeptide can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such as one of the following:

[00124] 1 ) the signal peptide of hChR2 (e.g., MDYGGALSAVGRELLFVTNPVWNGS (SEQ ID

NO:23))

[00125] 2) the β2 subunit signal peptide of the neuronal nicotinic acetylcholine receptor (e.g.,

MAGHSNSMALFSFSLLWLCSGVLGTEF (SEQ ID NO:24));

[00126] 3) a nicotinic acetylcholine receptor signal sequence (e.g.,

MGLRALMLWLLAAAGLVRESLQG (SEQ ID NO:25)); and

[00127] 4) a nicotinic acetylcholine receptor signal sequence (e.g., MRGTPLLLWSLFSLLQD (SEQ

ID NO:26)).

[00128] A signal sequence can have a length of from about 10 amino acids to about 50 amino acids, e.g., from about 10 amino acids to about 20 amino acids, from about 20 amino acids to about 30 amino acids, from about 30 amino acids to about 40 amino acids, or from about 40 amino acids to about 50 amino acids.

[00129] Endoplasmic reticulum (ER) export sequences that are suitable for use in a light-responsive polypeptide include, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27);

VLGSL (SEQ ID NO:28); etc.); NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like. An ER export sequence can have a length of from about 5 amino acids to about 25 amino acids, e.g., from about 5 amino acids to about 10 amino acids, from about 10 amino acids to about 15 amino acids, from about 15 amino acids to about 20 amino acids, or from about 20 amino acids to about 25 amino acids.

[00130] In some embodiments, the native signal peptide sequence in the protein can be deleted or substituted with a heterologous signal peptide sequence from a different protein.

ChR

[00131] In some aspects, the light-responsive cation channel protein can be derived from

Chlamydomonas reinhardtii, wherein the cation channel protein can be capable of transporting cations across a cell membrane when the cell is illuminated with light. In another embodiment, the light- responsive cation channel protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:8. The light used to activate the light-responsive cation channel protein derived from

Chlamydomonas reinhardtii can have a wavelength between about 460 and about 495 nm or can have a wavelength of about 480 nm. Additionally, light pulses having a temporal frequency of about 100 Hz can be used to activate the light-responsive protein. In some embodiments, activation of the light- responsive cation channel derived from Chlamydomonas reinhardtii with light pulses having a temporal frequency of about 100 Hz can cause depolarization of the neurons expressing the light-responsive cation channel. The light-responsive cation channel protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the light-responsive cation channel protein to regulate the polarization state of the plasma membrane of the cell. Additionally, the light-responsive cation channel protein can contain one or more conservative amino acid substitutions and/or one or more non-conservative amino acid substitutions. The light-responsive proton pump protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to transport cations across a cell membrane.

[00132] In some embodiments, the light -responsive cation channel comprises a T159C substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises a L132C substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises an E123T substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises an E123A substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises a T159C substitution and an E123T substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light- responsive cation channel comprises a T159C substitution and an E123A substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises a T159C substitution, an L132C substitution, and an E123T substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises a T159C substitution, an L132C substitution, and an El 23 A substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises an L132C substitution and an E123T substitution of the amino acid sequence set forth in SEQ ID NO:8. In some embodiments, the light-responsive cation channel comprises an L132C substitution and an E123A substitution of the amino acid sequence set forth in SEQ ID NO:8.

[00133] In some embodiments, a ChR2 protein comprises at least one (such as one, two, three, or more) amino acid sequence motifs that enhance transport to the plasma membranes of target cells selected from the group consisting of a signal peptide, an ER export signal, and a membrane trafficking signal. In some embodiments, the ChR2 protein comprises an N-terminal signal peptide and a C-terminal ER export signal. In some embodiments, the ChR2 protein comprises an N-terminal signal peptide and a C- terminal trafficking signal. In some embodiments, the ChR2 protein comprises an N-terminal signal peptide, a C-terminal ER export signal, and a C-terminal trafficking signal. In some embodiments, the ChR2 protein comprises a C-terminal ER export signal and a C-terminal trafficking signal. In some embodiments, the C-terminal ER export signal and the C-terminal trafficking signal are linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER export signal is more C-terminally located than the trafficking signal. In some embodiments the trafficking signal is more C- terminally located than the ER Export signal.

[00134] In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)).

[00135] In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL

(SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.); NANSFCYENEVALTSK (SEQ ID NO:29);

FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

Step function opsins and stabilized step function opsins based on ChR2

[00136] In other embodiments, the light-responsive polypeptide is a step function opsin (SFO) protein or a stabilized step function opsin (SSFO) protein that can have specific amino acid substitutions at key positions in the retinal binding pocket of the protein. In some embodiments, the SFO protein can have a mutation at amino acid residue C128 of SEQ ID NO:8. In other embodiments, the SFO protein has a C128A mutation in SEQ ID NO:8. In other embodiments, the SFO protein has a C128S mutation in SEQ ID NO:8. In another embodiment, the SFO protein has a C128T mutation in SEQ ID NO:8. In some embodiments, the SFO protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:9, and comprises an alanine, serine, or threonine at amino acid 128.

[00137] In some embodiments, the SSFO protein can have a mutation at amino acid residue D156 of

SEQ ID NO:8. In other embodiments, the SSFO protein can have a mutation at both amino acid residues CI 28 and D156 of SEQ ID NO:8. In one embodiment, the SSFO protein has an C128S and a D156A mutation in SEQ ID NO:8. In another embodiment, the SSFO protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 10; and comprises an alanine, serine, or threonine at amino acid 128; and comprises a alanine at amino acid 156. In another embodiment, the SSFO protein can comprise a C128T mutation in SEQ ID NO:8. In some embodiments, the SSFO protein comprises C128T and D156A mutations in SEQ ID NO:8.

[00138] In some embodiments the SFO or SSFO proteins provided herein can be capable of mediating a depolarizing current in the cell when the cell is illuminated with blue light. In other embodiments, the light can have a wavelength of about 445 nm. Additionally, in some embodiments the light can be delivered as a single pulse of light or as spaced pulses of light due to the prolonged stability of SFO and SSFO photocurrents. In some embodiments, activation of the SFO or SSFO protein with single pulses or spaced pulses of light can cause depolarization of a neuron expressing the SFO or SSFO protein. In some embodiments, each of the disclosed step function opsin and stabilized step function opsin proteins can have specific properties and characteristics for use in depolarizing the membrane of a neuronal cell in response to light.

[00139] Further disclosure related to SFO or SSFO proteins can be found in International Patent

Application Publication No. WO 2010/056970, the disclosure of which is hereby incorporated by reference in its entirety.

[00140] In some cases, the ChR24jased SFO or SSFO comprises a membrane trafficking signal and/or an ER export signal. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22).

Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.); NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

CI VI chimeric cation channels

[00141] In other embodiments, the light-responsive cation channel protein can be a C1V1 chimeric protein derived from the VChRl protein of Volvox carteri and the C Rl protein from Chlamydomonas reinhardti, wherein the protein comprises the amino acid sequence of VChRl having at least the first and second transmembrane helices replaced by the first and second transmembrane helices of ChRl ; is responsive to light; and is capable of mediating a depolarizing current in the cell when the cell is illuminated with light. In some embodiments, the CI VI protein can further comprise a replacement within the intracellular loop domain located between the second and third transmembrane helices of the chimeric light responsive protein, wherein at least a portion of the intracellular loop domain is replaced by the corresponding portion from ChRl . In another embodiment, the portion of the intracellular loop domain of the CI VI chimeric protein can be replaced with the corresponding portion from ChRl extending to amino acid residue A145 of the ChRl. In other embodiments, the CI VI chimeric protein can further comprise a replacement within the third transmembrane helix of the chimeric light responsive protein, wherein at least a portion of the third transmembrane helix is replaced by the corresponding sequence of ChRl . In yet another embodiment, the portion of the intracellular loop domain of the CI VI chimeric protein can be replaced with the corresponding portion from ChRl extending to amino acid residue W163 of the ChRl . In other embodiments, the CI VI chimeric protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 11.

[00142] In some embodiments, the ClVl protein can mediate a depolarizing current in the cell when the cell is illuminated with green light. In other embodiments, the light can have a wavelength of between about 540 nm to about 560 nm. In some embodiments, the light can have a wavelength of about 542 nm. In some embodiments, the ClVl chimeric protein is not capable of mediating a depolarizing current in the cell when the cell is illuminated with violet light. In some embodiments, the chimeric protein is not capable of mediating a depolarizing current in the cell when the cell is illuminated with light having a wavelength of about 405 nm. Additionally, in some embodiments, light pulses having a temporal frequency of about 100 Hz can be used to activate the ClVl protein.

[00143] In some cases, the ClVl polypeptide comprises a membrane trafficking signal and/or an ER export signal. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.);

NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

CI VI variants

[00144] In some aspects, a suitable light-responsive polypeptide comprises substituted or mutated amino acid sequences, wherein the mutant polypeptide retains the characteristic light-activatable nature of the precursor ClVl chimeric polypeptide but may also possess altered properties in some specific aspects. For example, the mutant light-responsive ClVl chimeric proteins described herein can exhibit an increased level of expression both within an animal cell or on the animal cell plasma membrane; an altered responsiveness when exposed to different wavelengths of light, particularly red light; and/or a combination of traits whereby the chimeric ClVl polypeptide possess the properties of low desensitization, fast deactivation, low violet-light activation for minimal cross-activation with other light-responsive cation channels, and/or strong expression in animal cells.

[00145] Accordingly, provided herein are CI VI chimeric light-responsive opsin proteins that can have specific amino acid substitutions at key positions throughout the retinal binding pocket of the VChRl portion of the chimeric polypeptide. In some embodiments, the CI VI protein can have an amino acid substitution at amino acid residue E122 of SEQ ID NO: l l . In some embodiments, the C1V1 protein can have a substitution at amino acid residue El 62 of SEQ ID NO:l l . In other embodiments, the CI VI protein can have a substitution at both amino acid residues E 162 and E 122 of SEQ ID NO : 11. In other embodiments, the C1V1 protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 12, SEQ ID NO: 13, or SEQ ID NO: 14; and can include one or more of the aforementioned amino acid substitutions.

[00146] In some aspects, the CI V1-E122 mutant chimeric protein is capable of mediating a

depolarizing current in the cell when the cell is illuminated with light. In some embodiments the light can be green light. In other embodiments, the light can have a wavelength of between about 540 nm to about 560 nm. In some embodiments, the light can have a wavelength of about 546 nm. In other embodiments, the C1V1 -E122 mutant chimeric protein can mediate a depolarizing current in the cell when the cell is illuminated with red light. In some embodiments, the red light can have a wavelength of about 630 nm. In some embodiments, the C1V1-E122 mutant chimeric protein does not mediate a depolarizing current in the cell when the cell is illuminated with violet light. In some embodiments, the chimeric protein does not mediate a depolarizing current in the cell when the cell is illuminated with light having a wavelength of about 405 nm. Additionally, in some embodiments, light pulses having a temporal frequency of about 100 Hz can be used to activate the C1V1 -E122 mutant chimeric protein. In some embodiments, activation of the CI V1-E122 mutant chimeric protein with light pulses having a frequency of 100 Hz can cause depolarization of the neurons expressing the C1V1-E122 mutant chimeric protein.

[00147] In other aspects, the C1V1-E162 mutant chimeric protein is capable of mediating a

depolarizing current in the cell when the cell is illuminated with light. In some embodiments the light can be green light. In other embodiments, the light can have a wavelength of between about 535 nm to about 540 nm. In some embodiments, the light can have a wavelength of about 542 nm. In other embodiments, the light can have a wavelength of about 530 nm. In some embodiments, the CI V1-E162 mutant chimeric protein does not mediate a depolarizing current in the cell when the cell is illuminated with violet light. In some embodiments, the chimeric protein does not mediate a depolarizing current in the cell when the cell is illuminated with light having a wavelength of about 405 nm. Additionally, in some embodiments, light pulses having a temporal frequency of about 100 Hz can be used to activate the C1V1-E162 mutant chimeric protein. In some embodiments, activation of the C1V1-E162 mutant chimeric protein with light pulses having a frequency of 100 Hz can cause depolarization-induced synaptic depletion of the neurons expressing the C1V1 -E162 mutant chimeric protein.

[00148] In yet other aspects, the C1V1-E122/E162 mutant chimeric protein is capable of mediating a depolarizing current in the cell when the cell is illuminated with light. In some embodiments the light can be green light. In other embodiments, the light can have a wavelength of between about 540 nm to about 560 nm. In some embodiments, the light can have a wavelength of about 546 nm. In some embodiments, the C1V1 -E122/E162 mutant chimeric protein does not mediate a depolarizing current in the cell when the cell is illuminated with violet light. In some embodiments, the chimeric protein does not mediate a depolarizing current in the cell when the cell is illuminated with light having a wavelength of about 405 nm. In some embodiments, the C1V1-E122/E162 mutant chimeric protein can exhibit less activation when exposed to violet light relative to CI VI chimeric proteins lacking mutations at E122/E162 or relative to other light-responsive cation channel proteins. Additionally, in some embodiments, light pulses having a temporal frequency of about 100 Hz can be used to activate the C1V1-E122/E162 mutant chimeric protein. In some embodiments, activation of the C1V1 - E122/E162 mutant chimeric protein with light pulses having a frequency of 100 Hz can cause depolarization-induced synaptic depletion of the neurons expressing the C1V1 - E122/E162 mutant chimeric protein.

[00149] In some cases, the C1V1 variant polypeptide comprises a membrane trafficking signal and/or an ER export signal. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22).

Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.); NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

Volvox carteri light-activated polypeptide

[00150] In some embodiments, a suitable light-responsive polypeptide is a cation channel derived from

Volvox carteri (VChRl) and is activated by illumination with light of a wavelength of from about 500 nm to about 600 nm, e.g., from about 525 nm to about 550 nm, e.g., 545 nm. In some embodiments, the light-responsive ion channel protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:19. The light-responsive ion channel protein can additionally comprise substitutions, deletions, and/or insertions introduced into a native amino acid sequence to increase or decrease sensitivity to light, increase or decrease sensitivity to particular wavelengths of light, and/or increase or decrease the ability of the light-responsive ion channel protein to regulate the polarization state of the plasma membrane of the cell. Additionally, the light-responsive ion channel protein can contain one or more conservative amino acid substitutions and/or one or more non-conservative amino acid substitutions. The light-responsive ion channel protein comprising substitutions, deletions, and/or insertions introduced into the native amino acid sequence suitably retains the ability to transport ions across the plasma membrane of a neuronal cell in response to light.

[00151] In some cases, a light-responsive cation channel protein can comprise a core amino acid

sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 19 and at least one (such as one, two, three, or more) amino acid sequence motifs which enhance transport to the plasma membranes of mammalian cells selected from the group consisting of a signal peptide, an ER export signal, and a membrane trafficking signal. In some embodiments, the light-responsive proton ion channel comprises an N-terminal signal peptide and a C-terminal ER export signal. In some embodiments, the light-responsive ion channel protein comprises an N-terminal signal peptide and a C-terminal trafficking signal. In some embodiments, the light-responsive ion channel protein comprises an N-terminal signal peptide, a C- terminal ER Export signal, and a C-terminal trafficking signal. In some embodiments, the light- responsive ion channel protein comprises a C-terminal ER Export signal and a C-terminal trafficking signal. In some embodiments, the C-terminal ER Export signal and the C-terminal trafficking signal are linked by a linker. The linker can comprise any of about 5, 10, 20, 30, 40, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 400, or 500 amino acids in length. The linker may further comprise a fluorescent protein, for example, but not limited to, a yellow fluorescent protein, a red fluorescent protein, a green fluorescent protein, or a cyan fluorescent protein. In some embodiments the ER Export signal is more C-terminally located than the trafficking signal. In some embodiments the trafficking signal is more C-terminally located than the ER Export signal.

[00152] In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22). Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.);

NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID NO:31); and the like.

[00153] Also provided herein are isolated polynucleotides encoding any of the light-responsive channel proteins described herein, such as a light-responsive ion channel protem comprising a core amino acid sequence at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 19. Also provided herein are expression vectors (such as a viral vector described herein) comprising a polynucleotide encoding the proteins described herein, such as a light-responsive channel protein comprising a core amino acid sequence at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO: 19. Step function opsins and stabilized step function opsins based on VChRl

[00154] In other embodiments, the light-responsive polypeptide is a SFO or an SSFO based on VChRl .

In some embodiments, the SFO protein can have a mutation at amino acid residue C123 of SEQ ID NO: 19. In other embodiments, the SFO protein has a C123A mutation in SEQ ID NO: 19. In other embodiments, the SFO protein has a C123S mutation in SEQ ID NO: 19. In another embodiment, the SFO protein has a C123T mutation in SEQ ID NO: 19. In some embodiments, the SFO protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:20, and comprises an alanine, serine, or threonine at amino acid 123.

[00155] In some embodiments, the SFO protein can have a mutation at amino acid residue D151 of

SEQ ID NO: 19. In other embodiments, the SFO protein can have a mutation at both amino acid residues CI 23 and D151 of SEQ ID NO: 19. In one embodiment, the SFO protein has an C123S and a D151A mutation in SEQ ID NO: 19. In another embodiment, the SSFO protein can comprise an amino acid sequence at least 75%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the sequence shown in SEQ ID NO:21 ; and comprises an alanine, serine, or threonine at amino acid 122; and comprises a alanine at amino acid 151.

[00156] In some embodiments the SFO or SSFO proteins provided herein can be capable of mediating a depolarizing current in the cell when the cell is illuminated with blue light. In other embodiments, the light can have a wavelength of about 560 nm. Additionally, in some embodiments the light can be delivered as a single pulse of light or as spaced pulses of light due to the prolonged stability of SFO and SSFO photocurrents. In some embodiments, activation of the SFO or SSFO protein with single pulses or spaced pulses of light can cause depolarization of a neuron expressing the SFO or SSFO protein. In some embodiments, each of the disclosed step function opsin and stabilized step function opsin proteins can have specific properties and characteristics for use in depolarizing the membrane of a neuronal cell in response to light.

[00157] In some cases, the VChRl -based SFO or SSFO comprises a membrane trafficking signal and/or an ER export signal. In some embodiments, the trafficking signal can be derived from the amino acid sequence of the human inward rectifier potassium channel Kir2.1. In other embodiments, the trafficking signal can comprise the amino acid sequence KSRITSEGEYIPLDQIDINV (SEQ ID NO:22).

Trafficking sequences that are suitable for use can comprise an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%, amino acid sequence identity to an amino acid sequence such a trafficking sequence of human inward rectifier potassium channel Kir2.1 (e.g., KSRITSEGEYIPLDQIDINV (SEQ ID NO:22)). In some cases, the ER export signal is, e.g., VXXSL (where X is any amino acid) (e.g., VKESL (SEQ ID NO:27); VLGSL (SEQ ID NO:28); etc.); NANSFCYENEVALTSK (SEQ ID NO:29); FXYENE (SEQ ID NO:30) (where X is any amino acid), e.g., FCYENEV (SEQ ID N0:31); and the like.

SCREENING METHODS

[00158] The present disclosure provides methods of identifying an agent that is suitable for use in controlling pain.

Methods of identifying an agent that reduces pain

[00159] In some cases, a subject method involves: a) contacting a non-human animal (e.g., a non- human mammal such as a rat or a mouse) of the present disclosure with a test agent, where the non- human animal expresses a depolarizing light-activated polypeptide in a primary afferent neuron, such as a nociceptor; and b) determining the effect, if any, of the test agent on pain when the depolarizing light- activated polypeptide is illuminated (activated) with light. A test agent that reduces pain in the non- human animal, compared to the level of pain induced by light activation of the depolarizing light- activated polypeptide in the absence of the test agent, indicates that the test agent is a candidate agent for controlling (reducing) pain. In some cases, the non-human animal is a subject non-human animal model, as described above.

[00160] For example, a test agent that reduces pain by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, or more than 25% (e.g., 25% to 50%; 50% to 75%; etc.) is considered a candidate agent for reducing pain.

[00161] As used herein, the term "determining" refers to both quantitative and qualitative

determinations and as such, the term "determining" is used interchangeably herein with "assaying," "measuring," and the like.

[00162] The terms "candidate agent," "test agent," "agent", "substance" and "compound" are used interchangeably herein. Candidate agents encompass numerous chemical classes, typically synthetic, semi-synthetic, or naturally occurring inorganic or organic molecules. Candidate agents include those found in large libraries of synthetic or natural compounds. For example, synthetic compound libraries are commercially available from Maybridge Chemical Co. (Trevillet, Cornwall, UK), ComGenex (South San Francisco, CA), and MicroSource (New Milford, CT). A rare chemical library is available from Aldrich (Milwaukee, Wis.) and can also be used. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available from Pan Labs (Bothell, WA) or are readily producible.

[00163] Candidate agents can be small organic or inorganic compounds having a molecular weight of more than 50 daltons and less than about 2,500 daltons. Candidate agents can comprise functional groups necessary for structural interaction with proteins, e.g., hydrogen bonding, and may include at least an amine, carbonyl, hydroxyl or carboxyl group, and may contain at least two of the functional chemical groups. The candidate agents may comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, and derivatives, structural analogs or combinations thereof.

[00164] Assays of the present disclosure include controls, where suitable controls include a non-human animal model that expresses a depolarizing light-responsive polypeptide in a nociceptor, and that has been exposed to activating light, but has not been administered the test agent.

[00165] Whether a test agent reduces pain in the non-human animal can be determined using any of a variety of assays known in the art. For example, test that measure pain through one or more behaviors such as withdrawal, licking, immobility, and vocalization, can be used. Suitable tests include, e.g.: a) the formalin assay; b) the von Frey test; c) a thermal assay such as tail withdrawal assay, a hot plate assay, a tail flick test (Rao et al. (1996) Neuropharmacol. 35:393); d) the Hargreaves assay; and the like. See, e.g., Mogil, et al. (2001) Methods in Pain Research, Frontiers in Neuroscience; and Carter and Shieh (2010) Nociception: Guide to Research Techniques in Neuroscience, Burlington, MA, Academic Press, pp 1-52; and Bannon and Malmberg (2007) Current Protocols in Neuroscience, Wiley Online Library. Suitable tests include those described in the Examples.

Methods of identifying agents that increase the minimum intensity of light required to produce a pain response

[00166] The present disclosure provides a method of identifying an agent that reduces pain, the method comprising: a) contacting a non-human animal (e.g., a non-human mammal such as a rat or a mouse) of the present disclosure with a test agent, where the non-human animal expresses a depolarizing light- activated polypeptide in a primary afferent neuron, such as a nociceptor; and b) determining the effect, if any, of the test agent on the minimum amount of light required to induce pain following

administration of the test agent.

[00167] A test agent that increases the amount of light required to induce pain is a candidate agent for reducing pain. For example, a test agent that increases the amount of light (as expressed in mW/mm 2 ) required to induce pain by at least 10%, at least 15%, at least 20%, at least 25%, at least 50%, at least 75%>, at least 100%) (or 2-fold), at least 2.5-fold, at least 5-fold, at least 10-fold, or more than 10-fold, is considered a candidate agent for reducing pain.

[00168] For example, if the minimum light intensity of light required to produce a pain response in the non-human animal is 0.5 mW/mm 2 , and the test agent increased the minimum amount of light required to induce pain to 0.75 mW/mm 2 , the test agent would be considered a candidate agent for reducing pain.

[00169] As used herein, the term "determining" refers to both quantitative and qualitative

determinations and as such, the term "determining" is used interchangeably herein with "assaying," "measuring," and the like.

[00170] The terms "candidate agent," "test agent," "agent", "substance" and "compound" are used interchangeably herein. Candidate agents encompass numerous chemical classes, typically synthetic, semi-synthetic, or naturally occurring inorganic or organic molecules. Candidate agents include those found in large libraries of synthetic or natural compounds. For example, synthetic compound libraries are commercially available from Maybridge Chemical Co. (Trevillet, Cornwall, UK), ComGenex (South San Francisco, CA), and MicroSource (New Milford, CT). A rare chemical library is available from Aldrich (Milwaukee, Wis.) and can also be used. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available from Pan Labs (Bothell, WA) or are readily producible.

[00171] Candidate agents can be small organic or inorganic compounds having a molecular weight of more than 50 daltons and less than about 2,500 daltons. Candidate agents can comprise functional groups necessary for structural interaction with proteins, e.g., hydrogen bonding, and may include at least an amine, carbonyl, hydroxyl or carboxyl group, and may contain at least two of the functional chemical groups. The candidate agents may comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, and derivatives, structural analogs or combinations thereof.

[00172] Assays of the present disclosure include controls, where suitable controls include a non-human animal model that expresses a depolarizing light-responsive polypeptide in a nociceptor, and that has been exposed to activating light, but has not been administered the test agent. In some cases, a control is a non-human animal model that has been administered an agent known not to affect pain.

[00173] Whether a test agent increases the minimum amount of light required to induce a pain response in the non-human animal can be determined using any of a variety of assays known in the art. For example, test that measure pain through one or more behaviors such as withdrawal, licking, immobility, and vocalization, can be used. Suitable tests include, e.g.: a) the formalin assay; b) the von Frey test; c) a thermal assay such as tail withdrawal assay, a hot plate assay, a tail flick test (Rao et al. (1996) Neuropharmacol. 35:393); d) the Hargreaves assay; and the like. See, e.g., Mogil, et al. (2001) Methods in Pain Research, Frontiers in Neuroscience; and Carter and Shieh (2010) Nociception: Guide to Research Techniques in Neuroscience, Burlington, MA, Academic Press, pp 51 -52; and Bannon and Malmberg (2007) Current Protocols in Neuroscience, Wiley Online Library. Suitable tests include those described in the Examples.

EXAMPLES

[00174] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius, and pressure is at or near atmospheric. Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pi, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m., intramuscular(ly); i.p., intraperitoneal(ly); s.c, subcutaneous(ly); and the like.

Example 1 : Bidirectional control of pain

[00175] It is demonstrated here that optogenetics can be used to bidirectionally control acute pain in both normal and pathological states. A gene transduction strategy was used that is adaptable and clinically relevant. Adeno-associated virus serotype 6 (AAV6) was used. AAV6 has many attractive features; it has been used for gene delivery in non-human primates, and is a leading candidate for future use in human clinical trials 15 . It is also capable of retrograde transport, and can specifically transduce nociceptors through intraneural delivery, removing the need for risky dorsal root ganglion injections. MATERIALS AND METHODS

Animal test subjects and experiments

[00176] All surgical and behavioral procedures were approved by the Stanford University

Administrative Panel on Lab Animal Care. Female C57BL/6 mice (1 - 4 months old) were housed in groups of 5 under a 12: 12 light:dark cycle. Food and water were available ad libitum.

Intraneural injection of AAV6-hSyn-ChR2(H134R)-eYFP and AAV6-hSyn-eNpHR3.0-eYFP

[00177] Mice were anesthetized with 2-2.5% isoflurane, placed on a heating pad maintained at 37°C, and allowed to reach a stable plane of anesthesia, which was periodically checked through examination of breathing rate and a toe-pinch test. Fur was shaved from the femur, bilaterally or unilaterally, depending on the injection, using an electrical razor. A hair removal cream (Nair) was used to remove any remaining hair from the incision site. The incision site was then sterilized with alternating applications of ethanol and Betadme solution, and the mouse legs taped to the surgical table. 100 μΐ of 1 mg/ml Rimadyl was injected. Sterilized forceps and spring scissors were then used to make a 2 cm incision immediately above the femur. The gluteus superficialis and biceps femoris muscles were identified and the connective tissue between them cut to expose the sciatic nerve cavity. Retractors were used to keep the cavity open and allow for clear access to the nerve. The nerve was carefully freed from the underlying fascia using blunted micromanipulators and spring scissors. 100 μΐ of 0.25%

Bupivacaine was injected into the incision site to simultaneously prevent the nerve from drying and induce local anesthesia. A 35G beveled needle (Nanofil# NF35BV-2, World Precision Instruments) was carefully inserted into the nerve, and 2.5-4 μΐ of virus solution injected at 1 μΐ/min, using a 25 μΐ syringe (Hamilton Company), connected to a Harvard PHD Syringe pump (Harvard Apparatus). When possible, 2 separate injections were made into the common peroneal and tibial branches of the sciatic nerve, to ensure that the nerve was filled uniformly. ChR2 injected mice received 3 X 10 10 vg (from UNC

Vector Core), while NpHR injected mice received either 1 x 10 11 vg or 3 x 10 11 vg (from UNC Vector

Core and Virovek, respectively). Depending on the mouse, this procedure was performed either unilaterally or bilaterally. The incision was then sutured closed using 5-0 suture. Isolation, Culture and Electrophysiology of Opsin-Expressing DRG Neurons

Isolation of DRG

[00178] Dorsal root ganglion (DRG) excision, culture and electrophysiology procedures were largely based on previously reported protocols 23 . Mice, three to four weeks after intraneural injection, were deeply anesthetized with isoflurane 5% and fur was shaved from the back. Mice were then perfused with 4°C sterile phosphate buffered saline. The following isolation steps were rapidly performed, and completed within 5 minutes after perfusion. After removing the skin from the back, using sterile procedure, the muscles along the vertebral column were cut and bone rongeurs used to peel away any muscle or tendon superficial to the vertebrae. The rongeurs were used to break away the vertebral bone directly dorsal to the spinal cord, starting at the base of the spine, and moving rostrally. Muscle lateral to the spinal cord was peeled away until the sciatic nerve branches could be visualized, and bones were broken lateral to the spinal cord to free the path of the nerve. Each nerve branch was cut using small spring scissors, pulled proximally with forceps until the dorsal root ganglion could be visualized, and cut proximal to the DRG. The DRG was then placed in 4°C, sterile MEM-complete solution (minimal essential media, MEM vitamins, antibiotics, and 10% fetal bovine serum). Three DRGs were excised from each expressing side of the mouse.

DRG Culture

[00179] Excised DRGs were desheathed and transferred to MEM-Collagenase solution (minimal essential media, vitamins, antibiotics, no fetal bovine serum, 0.125% collagenase). The tissue was incubated at 37°C for 45 minutes in a water bath and then triturated in 2.5 ml TripleE Express (Invitrogen). The trypsin was quenched with 2.5 ml MEM-complete with 80 ug/ml DNase I, 100 μg/ml trypsin inhibitor from chicken egg white and 2.5mg/ml MgSC>4. Cells were centrifuged, and resuspended in MEM-complete at a cell density of 500,000 cells/ml. 100 ul of the cell suspension was carefully placed as a bubble on matrigel-coated coverslips, and then incubated at 37°C, 3% C0 2 , 90% humidity. Two hours after initial incubation, the cultured neurons were flooded with 1 ml of MEM- complete. Cells were maintained 2-7 days in culture with fresh media changes as needed until electrophysiology was performed.

Electrophysiology

[00180] A Spectra X Light engine (Lumencor) or DG4 xenon lamp (Sutter Instruments) was used to identify fluorescent protein expression, and deliver light pulses for opsin activation. A 475/28 filter was used to apply blue light for ChR2, and a 586/20 filter was used to apply yellow light for NpHR. Light power density through the microscope objective was measured with a power meter (ThorLabs). Whole- cell recordings were obtained with patch pipettes (4-6 ΜΩ) pulled from borosilicate glass capillaries (Sutter Instruments) with a horizontal puller (P-2000, Sutter Instruments). The external recording solution contained (in niM): 125 NaCl, 2 KC1, 2 CaCl 2 , 2MgCl 2 , 30 glucose, 25 HEPES, and 1 μΜ tetrodotoxin when necessary to eliminate escape spikes for peak photocurrent measurements. The internal recording solution contained (in mM): 130 -gluconate, 10 mM KC1, 10 HEPES, 10 EGTA, 2 MgC¾. Recordings were made using a MultiClamp700B amplifier (Molecular Devices), and pClamp 10.3 software (Molecular Devices) was used to record and analyze data. Signals were filtered at 4 kHz using a Bessel filter and digitized at 10 kHz with a Digidata 1440A analog-digital interface (MolecularDevices). Peak and steady-state photocurrents were measured from a 1 s light pulse in voltage-clamp mode, where cells were held at -50 mV. Series resistances were carefully monitored and recordings were not used if the series resistance changed significantly (by >20%) or reached 20 ΜΩ. Latency to Light Measurement

Experimental Protocol:

[00181] Approximately 1 to 5 weeks after intraneural injection, mice were placed in a plastic enclosure with a thin, transparent, plastic floor and allowed to habituate to the test setup for 30 minutes prior to testing. A multimode optical fiber (Thor Labs, #AFS105/125Y ) attached to a laser (OEM Laser Systems, 473 nm, 1 mW/mm 2 ) was directed at the footpad through the floor. To begin the trial, the animal was required to: (1) be awake, (2) have all four paws on the floor, and (3) be at rest, not preparing to walk. Latency was calculated from when the footpad was illuminated to when the paw was withdrawn. To avoid experimenter bias, no subjective criteria were applied to the end-point for latency calculation, i.e. normal ambulation was also considered to end the trial. A maximum latency of 1 minute was set to ensure practicality of data collection. Individual trials were at least 2 minutes apart, and each mouse had five trials, which were averaged together. All trials were video recorded at 30 frames per second and latencies calculated through video analysis post-collection.

Statistics

[00182] A one-way ANOVA was used to analyze changes in ChR2+ mice's latencies in response to blue light in weeks 1 -5 as compared with ChR2+ mice's response to yellow light. Dunnett's post-hoc multiple-comparisons test was used to determine which latencies were significantly different from yellow-light controls. Effect sizes were calculated using g ¥ an extension of Hedges' g for multiple groups 24.

Place aversion

Construction of 2 Chamber Place Aversion Setup

[00183] A 2 chamber place setup was built with an entryway connecting the two 10 cm x 12 cm

chambers. The floor of each chamber, one red, the other blue, was illuminated with a 10 x 12 array of light-emitting diodes (Blue LEDs: 475 nm, Red LEDs: 625 nm, Cree) and directed with mirrors such that the light power density was equivalent in each room (0.15 mW/mm 2 ).

Experimental Protocol

[00184] A single mouse was allowed to explore the 2 chamber set up for 10 minutes prior to testing with the LED array floors turned off. Then, the mouse's location was recorded using a video camera and analyzed using BIOBSERVE Viewer 2 . The mouse position was recorded with the lights off for 10 minutes, and then the lights were switched on, and the position recorded for a further 30 minutes. Statistics

[00185] A two-sided paired Student's f-test was used to examine whether changes in mouse position preference between the 'lights-off and 'lights-on' condition were statistically significant. The percentage change between the two conditions was then calculated for each ChR2+ and NpHR+ mice. These percentages were then compared using a two-sided, unpaired, Student's f-test for heteroscedastic populations). Effect sizes were calculated using Hedges' g.

Measurement of Mechanical Withdrawal Thresholds

[00186] Mechanical allodynia was investigated through von Frey testing. Mice were allowed to

habituate to the test setup for 1 hour prior to testing. Hairs of various forces were applied to the bottom of the foot using the Up-and-down method 25 ' 26 for approximately 2 seconds.

[00187] The appearance of any of the following behaviors was considered as a withdrawal response: (1) rapid flinch or withdrawal of the paw, (2) spreading of the toes, or (3) immediate licking of the paw. If the animal moved the paw for some other reason before the end of the 2 seconds, the test was considered ambiguous and repeated. Depending on the opsin used, we then performed simultaneous illumination of the mouse's paw with blue light (473 nm, 0.15 mW/mm 2 ) or yellow light (593 nm, 0.15 mW/mm 2 ). The von Frey test was conducted by a single examiner for all data collected, who was always blinded to whether the mice being tested had opsin expression or not.

Statistics

[00188] Changes in von Frey threshold were tested for statistical significance using the non-parametric two-sided Wilcoxon signed-rank test. Effect sizes were calculated using Hedges' g.

Measurement of Thermal Withdrawal Latency

[00189] A modified Hargreaves plantar test apparatus was used to measure changes in thermal

sensitivity with different types of illumination. The standard Hargreaves test glass plate was raised slightly to allow placement of an LED ring above the infrared emitter. The LED ring was calibrated to emit 0.15 mW/mm 2 of blue (475 nm, Cree) or yellow (590 nm, OSRAM Opto Semiconductors) light. To control for light-induced confounds, withdrawal latency to infrared heat when mice received on- spectrum illumination (blue light for ChR2 injected mice, and yellow light for NpHR injected mice) was compared with off-spectrum illumination (vice versa). Withdrawal latency was automatically measured between onset of infrared light and the first paw withdrawal. Infrared intensity was kept constant across all trials, and the tester was always blinded as to whether the mice being tested had opsin expression or not.

Statistics

[00190] Changes in thermal withdrawal latency were compared between off-spectrum and on-spectrum illumination conditions using a two-sided, paired, Student's f-test. Effect sizes were calculated using Hedges' g. Chronic constriction injury

[00191] The chronic constriction injury model used here was adapted from an existing protocol 27 .

Animals were anesthetized with isoflurane and the sciatic nerve was exposed unilaterally in a similar fashion to the sciatic nerve exposure used for the intraneural injections. One 7-0 prolene double-knot ligature was tied around the nerve such that the ligature was just able to slide along the nerve, and the free ends of the suture were cut short. Non-absorbable 5-0 suture was used to close the wound. In order to promote development of neuropathic pain, no post-operative analgesics were administered.

Immunohistochemistry, Imaging, and Quantification of Transduction

Immunohistochemistry

[00192] Mice were euthanized with 100 μΐ Beuthanasia-D, and transcardially perfused with 10 ml of

4°C phosphate -buffered saline (IX PBS) and 10 ml of 4% paraformaldehyde (PFA). Bone rongeurs, spring scissors and forceps were used to carefully remove the sciatic nerve, associated dorsal root ganglia and the spinal cord together from the mouse. The feet were removed separately. All tissue was placed in 4% PFA overnight, stored at 4°C. Following this, samples were transferred to 30% sucrose (in IX PBS) and stored for varying lengths of time (at minimum 1 day). Samples were later dissected under microscopic guidance, and frozen separately in Tissue-Tek O.C.T. Samples were cut at 20 μηι thickness using a cryostat (Leica CM3050S), and mounted on slides. All samples were rinsed 3>< 10 min in IX PBS to remove any residual OCT. For all targets except myelin, samples were then blocked in 0.3% Triton-Xl OO, 2% Normal Donkey Serum (NDS), dissolved in IX PBS for 1 hour. Samples were then incubated overnight with primary antibody solutions with 0.3% Triton-XlOO, 5% NDS, dissolved in IX PBS. The next day, samples were rinsed 3x10 min with IX PBS, and then incubated for 1 hour with secondary antibody solutions dissolved in IX PBS . Samples were then rinsed 3x 10 min in 1 X PBS, and coverslipped with PVA DABCO. Primary antibodies used were Rat anti-Substance P (1 :500, #556312, BD Pharmingen), Biotin-IB4 (1 :50, #B-1205, Vector Laboratories), Rabbit anti-Somatostatin Receptor 2 (1 :250, #abl 34152, Abeam) and Rabbit anti-VRl (for TRPV1, 1 :500, #ab31895, Abeam). Secondary antibodies used were Cy5 Donkey anti-Rabbit (1 :500, # 711-175-152, Jackson

Laboratories), Cy3 Donkey anti-Rat (1 :500, #711-165-152, Jackson Laboratories) and Streptavidin- Texas Red (3: 100, #SA-5006, Vector Laboratories). For myelin, samples were permeabilized for 1 hour using 0.2% Triton-Xl OO dissolved in IX PBS. Samples were then incubated with FluoroMyelin Red (1 :300, #F34652, Molecular Probes) for 20 min. Following this, samples were rinsed 3 x10 min in IX PBS, and coverslipped with PVA DABCO.

Confocal Imaging

[00193] Samples were imaged using a Leica TCS SP5 confocal scanning laser microscope, using a 20X oil immersion objective, and analyzed using Leica LAS AF software. Images were later processed using Fiji 28 , which was used to stitch together z-stacks, balance image brightness and contrast, and modify colors to account for color-blindness. Quantification

[00194] DRGs from 3 different mice injected with AAV6-ChR2 were examined for co-expression with

Substance P, TRPV1, Somatostatin and IB4, and examined nerve samples from 3 different mice for co- expression with myelin. For each marker, the percentage of marker-expressing neurons/a ons that were YFP positive, and the percentage of YFP positive neurons/axons that were positive for the given marker, were quantified.

RESULTS

[00195] AAV6-hSyn-ChR2(H134R)-eYFP was injected into the sciatic nerve of mice (Fig. 1 a). Two to four weeks after injection, electrophysiological recordings from isolated ChR2 positive neurons in the dorsal root ganglia revealed that ChR2 was functional, and expressing cells could fire action potentials when stimulated at 5-1 OHz with 1 mW/mni 2 475 nm light (Fig. lb; Figure 6). In addition, ChR2 was expressed throughout the neuron, terminating in central projections to the dorsal horn of the spinal cord, with little expression seen in deeper laminae or in ascending dorsal columns, suggesting that the transduced neurons were nociceptors projecting to Rexed's lamina Ι/Π (Fig. lc). Immunohistochemistry showed considerable overlap between ChR2 expression and nociceptive markers, such as IB4, Substance P, TRPVl, and Somatostatin. No overlap was observed between ChR2 and myelin, indicating that no proprioceptors were transduced in the sciatic nerve; instead, expression was restricted to unmyelinated nociceptors (putative C-fibers) (Table 1, provided in Figure 9). Further, transduced dorsal root ganglia neurons were smaller in diameter than untransduced neurons (Figure 6), consistent with prior histological evidence regarding C-fiber size 17 .

[00196] The behavioral effect of optogenetic activation of these transduced nociceptors was examined.

Mice were allowed to freely explore a chamber with a transparent floor. After habituation, blue light (ImW/mm 2 ) was shown on the plantar hindpaws of ChR2 injected mice with the aim of optogenetically activating nociceptor nerve endings in the skin, and observed characteristic pain-like behavior (Fig. 2a). In response to blue light, mice flinched, engaged in prolonged foot-licking or vocalized: operant behaviors associated with pain 18 . To quantify this effect, the time between light onset and any paw withdrawal was measured, regardless of whether such withdrawal was due to pain or normal exploratory behavior. This latency reduced dramatically 2 weeks after injection (P = 0.034, effect size: 2.10), and remained low for 3 weeks thereafter (week 3: P = 0.027, effect size = 2.17; week 4: P = 0.026, effect size = 2.19), as compared with latencies recorded from YFP -injected mice, and ChR2 injected mice that received off-spectrum yellow light illumination (Fig. 2b). Latencies increased 5 weeks following injection, potentially due to shutdown of transgene expression in some tested mice, as has been previously reported with AAV6 in mice 15 . Despite stimulation being entirely transcutaneous, ChR2 injected mice were light sensitive, withdrawing in a few hundred milliseconds in response to low intensities of blue light (1 mW/mm 2 ). Lower levels of illumination showed progressively less of an effect, while an increase in illumination intensity past 1 mW/mm 2 did not result in any additional decrease in latency (Fig. 2c). [00197] To test if optogenetic induction of pain was tunable, it was asked if lower intensities of illumination (0.25 mW/mm 2 ) that did not prove immediately aversive would cause more subtle effects. A place aversion apparatus was constractetd, in which the floor of each chamber was illuminated with an LED array that emitted either off-spectrum (red, 625nm) or on-spectrum (blue, 475nm) light (Fig. 2d). ChR2 injected mice when exploring the blue chamber did not show any outward signs of pain, and did not engage in foot-licking or flinch from the light, but showed an 80-20% preference for the red chamber over the blue chamber (P— 0.0013, effect size = 3.1 1). YFP injected mice showed no significant preference (Fig. 2e,f). Such aversion is potentially caused by low levels of pain that do not rise to levels that induce reflexive withdrawal, but still cause changes in operant behavior.

[00198] It was reasoned that such low levels of optogenetic stimulation may also act to sensitize ChR2 injected mice to otherwise inoffensive stimuli. To demonstrate this, von Frey testing of the mechanical withdrawal threshold and Hargreaves testing of the thermal withdrawal latency was conduced, but with concurrent illumination of the relevant paw with low intensities of blue light (0.15 mW/mm 2 , Fig. 3a, b). While such illumination was insufficient to induce immediate aversion (Fig. 2c), it did significantly lower von Frey thresholds (Fig. 3b) by 50% (P = 0.027, effect size = 0.904), and lower Hargreaves latency (Fig. 3f, g) by 55% (P = 0.00038, effect size = 2.77). Such sensitization may occur through subthreshold depolarization induced in nociceptor free nerve endings, which may render them more sensitive to otherwise innocuous stimuli. Wild type mice showed no significant difference in behavior between illuminated and non-illuminated tests.

[00199] To complement our ability to optogenetically induce pain, methods were developed to

optogenetically inhibit action potential generation in nociceptors. Such inhibition could have great therapeutic value, and provide a type of spatially and temporally restricted control over action potential generation not possible with pharmacology or electrical stimulation.

[00200] Mice were injected in the sciatic nerve with AAV6-hSyn-eNpHR3.0-eYFP; and similar

transduction profiles to our ChR2 results were observed (Figure 7). Electrophysiological recordings of isolated cultured NpHR positive DRG neurons revealed strong hyperpolarization in response to constant yellow light illumination that was sufficient to block action potential initiation (Fig. 3c; Figure 8).

[00201] NpHR-injected mice were tested using similarly modified von Frey and Hargreaves apparatuses that emitted yellow (593nm) light. The NpHR-injected mice, when illuminated with 1.1-1.7 mW/mm 2 light, had a 69% increase (P— 0.0043, effect size— 0.802) in their von Frey withdrawal thresholds (Fig. 3d, e). Low intensities of yellow light (0.15 mW/mm 2 ) were sufficient to increase Hargreaves withdrawal latency by 97% (P = 0.00019, effect size = 2.05). Wild type mice showed no significant change in behavior upon illumination with yellow light.

[00202] Finally, whether the ability to optogenetically inhibit nociception was therapeutically relevant was determined by testing in an animal model of neuropathic pain. Baseline von Frey and Hargreaves testing were performed on NpHR-injected mice, replicating our initial findings that yellow light desensitized mice to mechanical and thermal stimuli (Fig 4a, 4b). A chronic constriction injury 20 was performed to induce symptoms of neuropathic pain in these mice. As expected, mice showed thermal and mechanical allodynia following the injury. Mice were then illuminated with yellow light while performing von Frey and Hargreaves testing. It was observed that optogenetic inhibition could reverse mechanical allodynia, increasing von Frey thresholds from 36 to 94% of pre-injury, non-illuminated levels (P = 0.0020, effect size = 0.920, Fig. 4a). Similarly, optogenetic inhibition also reversed thermal hyperalgesia in NpHR injected mice, increasing Hargreaves withdrawal latency from 55% to 128% of normal, non-illuminated levels (P = 0.012, effect size = 1.91, Fig. 4b). In both cases, YFP injected controls showed no significant changes with illumination, both before, and after chronic constriction injury.

[00203] Thus, the data show that opsins can be successfully expressed with high specificity in

nociceptors through a relatively simple injection procedure that does not require transgenesis.

Moreover, sufficiently strong opsin expression and trafficking to achieve robust behavioral effects through non-invasive transcutaneous illumination were observed. It is believed that this is due to expression of opsins in dermal and subdermal free nerve endings, which can be illuminated with minimal optical attenuation. Interestingly, effects of optogenetic illumination on mechanical and thermal thresholds were observed even when this illumination had low intensity. This may be due to the resting potential and baseline excitability of free nerve endings, which may allow relatively small optically induced membrane currents to still influence downstream neurotransmitter release.

Optogenetic effects were observed over a 3-week period, from 2 to 5 weeks following AAV6 injection.

[00204] The optogenetic capabilities reported here can be widely used by scientists who seek noninvasive ways to perturb nociceptive function. In particular, as opsin expression is specific to unmyelinated nociceptors, optogenetics could be used to understand the role of nociceptor activity in the genesis of neuropathic pain, through chronic bidirectional optogenetic control. Researchers who seek greater specificity and who do not wish to develop custom transgenic mice for each individual opsin could instead use Cre-dependent DIO-AAV6 22 in concert with any of the many different nociceptor-specific Cre lines, to achieve opsin expression restricted to sub-populations of nociceptors. Non-invasive, transcutaneous optogenetic inhibition can be used as a treatment for pain, e.g., intractable chronic pain.

[00205] Figure 1: Intra-sciatic injection of rAAV2/6-hSyn-ChR2(H134R)-eYFP transduced

unmyelinated nociceptors that projected to spinal cord lamina I. a) Injection schematic b)

Electrophysiology of dissociated ChR2+ DRG neurons. Representative whole-cell current-clamp recordings showing optogenetically induced action potentials in response to 475 nm light (5 Hz, 1 mW/mm 2 ) and representative whole-cell voltage-clamp recordings in response to a light pulse (1 s, 475 nm, lmW/mm 2 ). The cell is held at -50 mV. c) Expression profile at the spinal cord, DRG, nerve and foot; ChR2 is shown in green, cellular markers in magenta, overlay in white. ChR2 did not colocalize with myelinated neurons, but did colocalize with nociceptive markers including IB4, Somatostatin, TRPV1 and Substance P.

[00206] Figure 2: Transdermal illumination of ChR2+ mice resulted in tunable pain-like behavior, a)

Experimental schematic b) Time-dependence of light-sensitivity in ChR2+ mice (Light: 1 mw/mm 2 ) (n=A mice). Latencies in response to blue light were significantly lower compared to yellow light latencies, from week 2 to week 4 following injection (One-way ANOVA: F(6,21) = 3.98; P = 0.0082; Dunnett's test: P (week 2) = 0.034, P (week 3) = 0.027, P (week 4) = 0.026; effect size (week 2) = 2.10, effect size (week 3) = 2.17, effect size (week 4) = 2.19). Latencies recorded from YFP+ controls and from ChR2+ mice at week 1 and week 5 were not significant (P (week 1) = 0.55, P (week 5) = 0.49, P (YFP+) = 0.99). c) Light-sensitivity decreased with increase in intensity of blue light, steeply until intensity was 1 mW/mm 2 , stabilizing past this threshold, d) Place aversion schematic e) YFP+ mice showed a non-significant preference for the blue-lit areas (19.9% increase in time spent in blue-lit areas, P = 0.06, n = 5), while ChR2 mice were significantly averse to blue-lit areas (55.6% decrease in time spent in blue-lit areas, effect size = 3.11, P = 0.0013, n = 5). Inset: these two percent changes were statistically different from each other. (P = 0.00061) f) Individual traces for place aversion data. All grouped data are shown as mean ± s.e.m.

[00207] Figure 3: Blue light sensitized ChR2 expressing mice and yellow light desensitized NpHR expressing mice to mechanical and thermal stimuli, a) Schematic of ChR2 -mediated sensitization (0.15 mW/mm 2 blue light intensity) b) von Frey thresholds reduced 50% in ChR2+ mice (effect size = 0.904, P = 0.027, n— 10 paws), while wild-types showed no significant change {P = 0.50, n = 10 paws), c) i) Representative whole-cell voltage -clamp recording showing outward photocurrent in an NpHR- expressing DRG neuron, in response to a 1 s, 586 nm light pulse (indicated by yellow bar), ii) Representative whole-cell current-clamp recordings showing yellow (586 nm) light-mediated inhibition of electrically evoked spikes (400 pA current injection, 5 ms pulse width) in an NpHR-expressing DRG neuron, d) Schematic of NpHR-mediated inhibition (1.1-1.7 mW/mm 2 light intensity) e) von Frey thresholds increased 69% NpHR+ mice (effect size = 0.802, P = 0.0043, n = 24 paws) while wild-type mice showed no significant change (P = 0.71, n = 20 paws), f) Schematic of optogenetic modulation of thermal thresholds. Blue/Yellow light intensity (0.15 mW/mm 2 ) g) Withdrawal latency to infrared stimulus decreased 55% in ChR2+ mice during blue light illumination (effect size = 2.77, P = 0.00038, n = l paws) and increased 97% in NpHR+ mice during yellow light illumination (effect size = 2.05, P = 0.00019, n = 10 paws) (compared to off-spectrum illumination), while wild-type latencies did not significantly change (P = 0.91, n = 9 paws, controls for ChR2+ mice, P = 0.26, n = 10 paws, controls for NpHR+ mice). All grouped data are shown as mean ± s.e.m.

[00208] Figure 4: Yellow light stimulation of NpHR+ mice reversed mechanical allody ia and thermal hyperalgesia caused by a chronic constriction injury (CCI). a) Before CCI, yellow light significantly increased von Frey thresholds ofNpHR+ mice (78% increase, effect size = 1.43, P = 0.0020, n = 10 paws), but not of YFP+ mice (P = 0.41, n = 12 paws). After CCI, von Frey thresholds of all mice significantly reduced (NpHR+ mice, 64% reduction, effect size = 1.61, P = 0.0020, n = 10 paws; YFP+ mice, 59% reduction, effect size = 1.03, P = 0.00049, n = 12 paws). Yellow light significantly increased von Frey thresholds of NpHR+ mice to near pre-CCI levels (258% increase, effect size =

0.92. P = 0.0020, n = 10 paws), but did not significantly change the thresholds of YFP+ mice (P = 0.57, n = 12 paws), b) Before CCI, yellow light significantly increased withdrawal latency to infrared stimulus in NpHR+ mice (112% increase, effect size = 3.95, P = 0.00025, n = 1 paws, compared to off- spectrum illumination), while YFP+ mice showed no significant change (P = 0.97, n = 9 paws). After CCI, all mice showed a reduction in withdrawal latency to infrared stimulus during off-spectrum illumination (NpHR+ mice, 45% reduction, effect size = 3.75, P = 0.00077, n = 7 paws; YFP+ mice, 40%) reduction, effect size = 2.06, P = 0.0038, n = 9 paws). Yellow light significantly increased this latency in NpHR+ mice (132% increase, effect size = 1.91, P = 0.012, n = 1 paws) beyond initial pre- CCI latencies, but did not significantly change latency in YFP+ mice (P = 0.53, n = 9 paws). All grouped data are shown as mean ± s.e.m.

[00209] Figure 6: Size distribution of opsin transduction. Intra-neural injection of a) AAV6:ChR2 and b) AAV6:NpHR results in opsin transduction that is specific to smaller diameter nociceptors (n = 205 ChR2+ neurons, 666 ChR2- neurons, n = 217 NpHR+ neurons, 355 NpHR- neurons). AAV6:ChR2 transduced 80%) of neurons < 16 μιη, AAV6:NpHR transduced 75% of neurons < 16 μτη.

[00210] Figure 7: Representative images of NpHR transduction observed after intra-sciatic injection of

AAV2/6-hSyn-eNpHR3.0-eYFP.

[00211] Figure 8: Electrophysiological recording from ChR2+ and NpHR-i- DRG neurons, a) Image of

ChR2+ DRG neuron, scale bar 25 μιη. b) Sample whole-cell current-clamp recordings showing spikes evoked by 10 Hz blue (475 nm) light pulse trains (5 ms pulse widths) in a ChR2+ DRG neuron. Cell is held at -50 mV, light power density is 1 mW/mm 2 . c) Summary graph of peak and steady-state photocurrents in response to 1 s, 475 nm blue light. Mean ± SEM is plotted, n = 7. d) Image of NpHR+ DRG neuron, scale bar 25 μιη. e) Sample whole-cell current-clamp recordings showing yellow (586 nm) light-mediated hyperpolarization in an NpHR-i- DRG neuron. Cell is held at -48 mV, light power density is 1 mW/mm 2 . f) Summary graph of peak and steady-state photocurrents in response to 1 s, 586 nm yellow light. Mean ± SEM is plotted, n— 10.

[00212] Figure 9 presents Table 1. Analysis of opsin transduction profiles. Co-localization

percentages calculated from analysis of DRGs from 3 different ChR2+ and NphR+ mice each.

Somatostatin-l-, VR1+ and IB4+ neurons are the major components of the opsin+ pool.

References

1. Dubin, A. E. & Patapoutian, A. Review series Nociceptors: the sensors of the pain pathway.

The Journal of Clinical Investigation 120, 3760-3772 (2010).

2. Costigan, M., Scholz, J. & Woolf, C. J. Neuropathic pain: a maladaptive response of the

nervous system to damage. Annual Review of Neuroscience 32, 1-32 (2009). Fenno, L., Yizhar, O. & Deisseroth, K. The Development and Application of Optogenetics. Annual Review of Neuroscience 34, 389-412 (2011).

Liske, H. et al. Optical inhibition of motor nerve and muscle activity in vivo. Muscle & Nerve 47, 916-921 (2013).

Llewellyn, M. E., Thompson, K. R., Deisseroth, K. & Delp, S. L. Orderly recruitment of motor units under optical control in vivo. Nature Medicine 16, 1161-5 (2010).

Ji, Z.-G. et al. Light-evoked somatosensory perception of transgenic rats that express channelrhodopsin-2 in dorsal root ganglion cells. PLOS One 7, e32699 (2012).

Wang, H. & Zylka, M. J. Mrgprd-expressing polymodal nociceptive neurons innervate most known classes of substantia gelatinosa neurons. The Journal of Neuroscience: the official journal of the Society for Neuroscience 29, 13202-9 (2009).

Daou, I. et al. Light- induced nociception: Remote optogenetic control of peripheral pain pathways in freely moving mice. Society for Neuroscience Conference 575.06/1111 (2012). Mourot, A. et al. Rapid optical control of nociception with an ion-channel photoswitch. Nature Methods 9, 396-402 (2012).

Kokel, D. et al. Photochemical activation of TRPAl channels in neurons and animals. Nature Chemical Biology 9, 257-263 (2013).

Mattis, J. et al. Principles for applying optogenetic tools derived from direct comparative analysis of microbial opsins. Nature Methods 9, (2012).

Williams, J. C. & Denison, T. From optogenetic technologies to neuromodulation therapies.

Science Translational Medicine 5, 177ps6 (2013).

Chow, B. Y. & Boyden, E. S. Optogenetics and translational medicine. Science Translational Medicine 5, 177ps5 (2013).

Towne, C, Schneider, B. L., Kieran, D., Redmond, D. E. & Aebischer, P. Efficient transduction of non-human primate motor neurons after intramuscular delivery of recombinant AAV serotype 6. Gene Therapy (2009).

Asokan, A., Schaffer, D. V & Samulski, R. J. The AAV vector toolkit: poised at the clinical crossroads. Molecular Therapy 20, 699-708 (2012).

Towne, C, Pertin, M., Beggah, A. T., Aebischer, P. & Decosterd, I. Recombinant adeno- associated virus serotype 6 (rAAV2/6) -mediated gene transfer to nociceptive neurons through different routes of delivery. Molecular Pain 5, 52 (2009).

Ruscheweyh, R., Forsthuber, L. & Schoffhegger, D. Modification of Classical Neurochemical Markers in Identified Primary Afferent Neurons With Αβ- , Αδ- , and C-Fibers After Chronic Constriction Injury in. The Journal of Comparative Neurology 336, 325-336 (2007).

Mogil, J. S. Animal models of pain: progress and challenges. Nature Reviews Neuroscience 10, 283-294 (2009).

Mason, M. R. J. et al. Comparison of AAV serotypes for gene delivery to dorsal root ganglion neurons. Molecular Therapy: the journal of the American Society of Gene Therapy 18, 715-24 (2010).

Bennett, G. J. & Xie, Y. K. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 33, 87-107 (1988). 21. Gao, G.-P. et al. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proceedings of the National Academy of Sciences of the United States of America 99, 11854-9 (2002).

22. Sohal, V. S., Zhang, F., Yizhar, 0. & Deisseroth, K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature 459, 698-702 (2009).

23. Gold, M. S. Whole-Cell Recording in Isolated Primary Sensory Neurons. Pain Research:

Methods in Molecular Biology 851, 73-97 (2012).

24. Hentschke, H. & Stiittgen, M. C. Computation of measures of effect size for neuroscience data sets. The European Journal of Neuroscience 34, 1887-94 (201 1).

25. Chaplan, S. R., Bach, F. W., Pogrel, J. W., Chung, J. M. & Yaksh, T. L. Quantitative

assessment of tactile allodynia in the rat paw. Journal of Neuroscience Methods 53, 55-63 (1994).

26. Dixon, W. J. Efficient Analysis of Experimental Observations. Annual Review of

Pharmacology and Toxicology 20, 441-462 (1980).

27. Sommer, C. & Schafers, M. Painful mononeuropathy in C57BL/Wld mice with delayed

Wallerian degeneration: differential effects of cytokine production and nerve regeneration on thermal and mechanical hypersensitivity. Brain Research 784, 154-162

28. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nature

Methods 9, 676-82 (2012).

Example 2: Intraneurally injected AAV8 selectively transduces neurons that project to spinal cord dorsal columns and deep spinal cord laminae.

[00213] Large-diameter primary afferent neurons are responsible for mediating diverse sensory

processes including pressure, vibration, pleasurable touch, and painful touch. In the context of pain research, these neurons are known to be significantly modified in various chronic pain disorders. Spontaneous ('ectopic') firing in these neurons is thought to be one of the major contributors to the development of inflammatory and neuropathic pain, either through their directly driving central pain pathways, or through modifying spinal cord circuitry to induce central sensitization. Optogenetic inhibition of these afferent neurons would reduce symptoms of neuropathic pain. Optogenetic stimulation of these afferent neurons may also act to reduce pain in some conditions, through multi-step circuit processes in the spinal cord that form part of the 'pain gate'. The data shown below indicate that an adeno-associated virus vector (AAV8) can specifically infect large-diameter primary afferent neurons that mediate sensation of touch.

Methods

[00214] All surgical and behavioral procedures were approved by the Stanford University

Administrative Panel on Lab Animal Care. Under anesthesia, the sciatic nerve of C57BL/6 mice (6-8 weeks) was exposed; and 3-5μ1 of AAV8-CMV-GFP (1 -2E10 viral genomes (vg)) was injected into the exposed nerve. 2-4 weeks following injection, the mice were euthanized through transcardial perfusion. The mice were dissected and sectioned, and tissue from the spinal cords, nerves and paws was imaged. Results

[00215] Primary afferent neurons successfully infected by AAV8-CMV-GFP projected to deep spinal cord lammae (Fig. 10A) in the lumbar spinal cord. In more rostral regions of the cord, expression was seen primarily in the fasciculus gracilis, in the dorsal columns (Fig. 10B). This expression persisted until the brainstem.

Example 3: Delivery of optogenetic proteins to sensory neurons of the trigeminal ganglion

[00216] Neuropathic pain can arise in the trigeminal ganglion and is often characterized by episodic, lancinating, triggerable, often shock-like facial pain. The disease can result from infection (e.g. herpes virus), facial trauma, stroke or surgical nerve damage. The data in this example demonstrate delivery of the inhibitory opsin, eNpHR3.0, to the sensory neurons of the trigeminal ganglion in rats. The data presented in this example demonstrate that optogenetics can be used to control pain involving the trigeminal ganglion.

Methods

[00217] Under anesthesia, 10- week old Sprague Dawley rats were stereotaxically injected with 1 x 10 u vg of AAV5-hSyn-eNpHR3.0 into the trigeminal ganglion. Animals were euthanized 4 weeks later, and the trigeminal ganglion was dissected, sectioned and imaged using a confocal microscope.

Results

[00218] Four weeks following direct injection of AAV5-eNpHR3.0-YFP, strong expression of the inhibitory protein in sensory neurons of the trigeminal ganglion was observed. These results demonstrate that primary sensory neurons other than those of spinal cord dorsal root ganglia can be targeted with opsins and can be inhibited to control pain transmitted by these neurons. The data also demonstrate the feasibility of opsin delivery using direct injection. The data are depicted in Figure 1 1.

[00219] Figure 11. Expression of eNpHR3.0 in trigeminal ganglion sensory neurons following direct injection into ganglion. eNpHR3.0 was fused to yellow fluorescent protein to facilitate visualization (green). Neurons were stained with Nissl (red). All cell nuclei were labelled with DAPI (blue).

Example 4: Delivery of opsins and inhibition of pain in animals with prior neuropathic pain.

[00220] The data presented below demonstrate that AAV6 delivery of NpHR, following chronic

constriction injury (CCI) and development of pain, can reduce pain.

Methods

[00221] Naive C57B16 mice were habituated to the Von Frey mechanical testing procedure and then baseline mechanical thresholds recorded. Mice were then subjected to a CCI; 10 days following nerve injury, mechanical thresholds were recorded and only those animals that had developed neuropathic pain (as determined by a 25% or greater reduction in mechanical thresholds) were kept in the study. AAV6 expressing either NpHR or YFP under control of the human synapsin promoter (5 x 10 10 vg total) was delivered to the sciatic nerve of the mice. Thirty days following NpHR or YFP delivery, mechanical thresholds were recorded in the presence or absence of yellow light application to the targeted paw. Results

[00222] Thirty days following AAV6 delivery we observed that application of light to the targeted paw significantly increased mechanical threshold levels to pre-CCI levels in animals expressing NpHR but not YFP. The data are shown in Figure 12.

[00223] Figure 12. Mechanical thresholds for mice expressing GFP or eNpHR3.0 in nociceptive fibers using AAV6. Forty days following CCI thresholds were recorded in the presence or absence of yellow light. Animals in the eNpHR3.0 group had significantly higher mechanical thresholds in the presence of light (p<0.05). Mechanical thresholds in the YFP group were unchanged.

[00224] These results demonstrate that the optogenetic approach described here can be applied to nerves that have preexisting neuropathic pain.

[00225] While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.