US20060172963A1 - RNAi-mediated inhibition of ocular hypertension targets - Google Patents

RNAi-mediated inhibition of ocular hypertension targets Download PDF

Info

Publication number
US20060172963A1
US20060172963A1 US11/344,702 US34470206A US2006172963A1 US 20060172963 A1 US20060172963 A1 US 20060172963A1 US 34470206 A US34470206 A US 34470206A US 2006172963 A1 US2006172963 A1 US 2006172963A1
Authority
US
United States
Prior art keywords
seq
nucleotides
interfering rna
mrna
nucleotide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/344,702
Inventor
Allan Shepard
Jon Chatterton
Abbot Clark
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alcon Inc
Arrowhead Pharmaceuticals Inc
Original Assignee
Alcon Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alcon Inc filed Critical Alcon Inc
Priority to US11/344,702 priority Critical patent/US20060172963A1/en
Assigned to ALCON, INC. reassignment ALCON, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CLARK, ABBOT F., CHATTERTON, JON E., SHEPARD, ALLAN R.
Publication of US20060172963A1 publication Critical patent/US20060172963A1/en
Priority to US12/967,121 priority patent/US20110124708A1/en
Assigned to ARROWHEAD RESEARCH CORPORATION reassignment ARROWHEAD RESEARCH CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS AG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/01Carboxylic ester hydrolases (3.1.1)
    • C12Y301/01007Acetylcholinesterase (3.1.1.7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y306/00Hydrolases acting on acid anhydrides (3.6)
    • C12Y306/03Hydrolases acting on acid anhydrides (3.6) acting on acid anhydrides; catalysing transmembrane movement of substances (3.6.3)
    • C12Y306/03009Na+/K+-exchanging ATPase (3.6.3.9)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y402/00Carbon-oxygen lyases (4.2)
    • C12Y402/01Hydro-lyases (4.2.1)
    • C12Y402/01001Carbonate dehydratase (4.2.1.1), i.e. carbonic anhydrase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed

Definitions

  • the present invention relates to the field of interfering RNA compositions for inhibition of expression of ocular hypertension targets in glaucoma, particularly for primary open angle glaucoma.
  • Glaucoma is a heterogeneous group of optic neuropathies that share certain clinical features.
  • the loss of vision in glaucoma is due to the selective death of retinal ganglion cells in the neural retina that is clinically diagnosed by characteristic changes in the visual field, nerve fiber layer defects, and a progressive cupping of the optic nerve head (ONH).
  • One of the main risk factors for the development of glaucoma is the presence of ocular hypertension (elevated intraocular pressure, IOP).
  • IOP elevated intraocular pressure
  • An adequate intraocular pressure is needed to maintain the shape of the eye and to provide a pressure gradient to allow for the flow of aqueous humor to the avascular cornea and lens.
  • IOP levels may also be involved in the pathogenesis of normal tension glaucoma (NTG), as evidenced by patients benefiting from IOP lowering medications. Once adjustments for central corneal thickness are made to IOP readings in NTG patients, many of these patients may be found to be ocular hypertensive.
  • NTG normal tension glaucoma
  • the elevated IOP associated with glaucoma is due to elevated aqueous humor outflow resistance in the trabecular meshwork (TM), a small specialized tissue located in the iris-corneal angle of the ocular anterior chamber.
  • Glaucomatous changes to the TM include a loss in TM cells and the deposition and accumulation of extracellular debris including proteinaceous plaque-like material.
  • ONH glial cells In glaucomatous eyes, there are morphological and mobility changes in ONH glial cells.
  • IOP and/or transient ischemic insults there is a change in the composition of the ONH extracellular matrix and alterations in the glial cell and retinal ganglion cell axon morphologies.
  • POAG Primary open angle glaucoma
  • IOP Intraocular pressure
  • Glaucoma therapies include lowering IOP by the use of suppressants of aqueous humor formation or agents that enhance uveoscleral outflow, laser trabeculoplasty, or trabeculectomy, which is a filtration surgery to improve drainage.
  • Pharmaceutical anti-glaucoma approaches have exhibited various undesirable side effects. For example, miotics such as pilocarpine can cause blurring of vision and other negative visual side effects.
  • Systemically administered carbonic anhydrase inhibitors (CAIs) can also cause nausea, dyspepsia, fatigue, and metabolic acidosis. Further, certain beta-blockers have increasingly become associated with serious pulmonary side effects attributable to their effects on beta-2 receptors in pulmonary tissue.
  • Sympathomimetics cause tachycardia, arrhythmia and hypertension. Such negative side effects may lead to decreased patient compliance or to termination of therapy.
  • efficacy of current IOP lowering therapies is relatively short-lived requiring repeated dosing during each day and, in some cases, the efficacy decreases with time.
  • the present invention is directed to interfering RNAs that silence ocular hypertension target mRNA expression, thus lowering intraocular pressure in patients with open-angle glaucoma or ocular hypertension.
  • Ocular hypertension targets include carbonic anhydrase II, IV, and XII; ⁇ 1- and ⁇ 2 adrenergic receptors; acetylcholinesterase; Na + /K + -ATPase; and Na—K-2Cl cotransporter.
  • the interfering RNAs of the invention are useful for treating patients with open-angle glaucoma or ocular hypertension.
  • An embodiment of the present invention provides a method of attenuating expression of an ocular hypertension target mRNA such as carbonic anhydrase II, IV, or XII; ⁇ 1- or ⁇ 2 adrenergic receptors; acetylcholinesterase; Na + /K + -ATPase; or Na—K-2Cl cotransporter mRNA in a subject.
  • the method comprises administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier. Administration is to the eye of the subject for attenuating expression of an ocular hypertension target in a human.
  • the interfering RNA comprises a sense nucleotide strand, an antisense nucleotide strand and a region of at least near-perfect contiguous complementarity of at least 19 nucleotides.
  • the antisense strand hybridizes under physiological conditions to a portion of an mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134 which are sense cDNA sequences encoding carbonic anhydrase II and IV; ⁇ 1- and ⁇ 2 ad
  • the antisense strand has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, respectively.
  • the administration of such a composition attenuates the expression of an ocular hypertension target mRNA of the subject.
  • the ocular hypertension target mRNA encodes carbonic anhydrase II, IV or XII, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:101, or SEQ ID NO:134 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:101, or SEQ ID NO:134, respectively.
  • the ocular hypertension target mRNA encodes a ⁇ 1- or ⁇ 2-adrenergic receptor
  • the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4, respectively.
  • the ocular hypertension target mRNA encodes an acetylcholinesterase
  • the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123, respectively.
  • the ocular hypertension target mRNA encodes a subunit of Na + /K + -ATPase, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132, respectively.
  • the ocular hypertension target mRNA encodes a Na—K-2Cl cotransporter
  • the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:7 or SEQ ID NO:133 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:7 or SEQ ID NO:133, respectively.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:1 comprising nucleotide 232, 527, 721, 728, 809, 810, 855, 856, 921, 1139, 506, 547, 548, 740, 911, 1009, 1140, 1149, 1150, 1151, 1188, 1194, 1195, 1223, 1239, 1456, 1457, 1458, 100, 158, 166, 247, 286, 318, 322, 328, 371, 412, 482, 504, 505, 541, 734, 772, 777, 814, 972, 998, 1232, 317, or 401.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:2 comprising nucleotide 213, 252, 258, 266, 399, 457, 463, 490, 595, 1064, 109, 112, 125, 126, 150, 261, 265, 280, 398, 453, 459, 462, 467, 492, 534, 785, 801, 825, 827, 876, 1003, or 1012.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:101 comprising nucleotide 191, 239, 274, 275, 341, 389, 412, 413, 423, 687, 689, 695, 710, 791, 792, 794, 983, 993, 994, 995, 691, 1039, 1568, 2326, 2332, 2425, 2433, 2844, 2845, 2880, 2884, 2891, 2954, 2955, 2956, 2957, 2964, 2965, 3006, 3007, 3012, or 3026.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:134 comprising nucleotide 687, 1535, 2293, 2299, 2392, 2400, 2811, 2812, 2847, 2851, 2858, 2921, 2922, 2923, 2924, 2931, 2932, 2973, 2974, 2979, or 2993.
  • Another embodiment of the invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:3 comprising nucleotide 468, 523, 799, 1563, 1565, 1569, 1593, 1613, 1614, 1626, 310, 322, 726, 769, 772, 801, 802, 1501, 1576, 1577, 1579, 1580, 1581, 1586, 1590, 1592, 1594, 1615, 1616, 1632, 1633, or 1654.
  • a further embodiment of the invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:4 comprising nucleotide 329, 375, 1031, 1046, 1149, 1163, 1371, 1401, 1426, 1880, 283, 607, 608, 609, 619, 623, 722, 857, 1037, 1091, 1115, 1124, 1136, 1137, 1151, 1164, 1393, 1394, 1395, 1406, 1407, 1427, 1428, 1429, 1442, 1725, 1726, 1756, 1757, 1758, 1767, 1790, 1791, 1792, 1793, 1803, 1861, 1869, 1971, 1972, or 1979.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:123 comprising nucleotide 1875, 1890, 1891, 2011, 2012, 2133, or 2134.
  • Another embodiment of the invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:5 comprising nucleotide 366, 370, 384, 385, 525, 588, 768, 1045, 1046, 1061, 1090, 1232, 1314, 1316, 1460, 1461, 1462, 1528, 1607, 1705, 1713, 382, 393, 397, 622, 1131, 1459, 1530, 2251, 2885, 2886, 386, 1231, 1315, 2047, 2049, 2053, 2055, 2057, 2125, 2126, 2127, 2250, 2253, 2258, 2260, 2318, 2395, 2397, 2404, 2405, 2643, 2645, or 2887.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:124 comprising nucleotide 2208, 2275, 2307, 2526, 2538, 2592, 2628, 2979, 2985, 3093, 3474, 3504, 3505, 3506, 3518, 343, 442, 700, 707, 811, 907, 1059, 1363, 1594, 1662, 1758, 1760, 1896, 2037, or 2147.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:125 comprising nucleotide 436, 441, 443, 552, 617, 701, 702, 832, 2204, 2291, or 2495.
  • a further embodiment of the present invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:6 comprising nucleotide 471, 1990, 3080, 3797, 4037, 4093, 4225, 4323, 5213, 5285, 214, 467, 470, 472, 473, 632, 825, 946, 1693, 1767, 1768, 2157, 2263, 2589, 2590, 2765, 2988, 3094, 3144, 3145, 3344, 3345, 3418, 3666, 3828, 3850, 4040, 4041, 4061, 4882, 4894, 4900, 5040, 5114, 5115, 5128, 5129, 5253, 5296, 5375, 5384, or 5385.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:126 comprising nucleotide 240, 272, 362, 1836, 1851, 2103, 2137, 2138, 2139, 2157, 2158, 2160, 2425, 2580, 2601, 2646, 2650, 2794, 2803, 3116, 3124, 3126, 3129, or 3377.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:127 comprising nucleotide 113, 612, 702, 833, 1101, 1732, 1733, 1836, 2070, 2071, 2143, 2328, 2475, 2861, 2862, 2952, 3203, 3281, 3377, 3379, 3470, 3471, 3554, 3614, 3615, 3616, 3617, 3625, 3626, 3642, 3646, 3647, 3653, 3655, 3797, 3801, 3803, 3809 or 3810.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:128 comprising nucleotide 126, 251, 252, 253, 331, 427, 429, 520, 521, 530, 601, 602, 603, 604, 664, 665, 666, 667, 675, 676, 692, 696, 697, 702, 703, 705, 707, 847, 851, 853, 859, or 860.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:129 comprising nucleotide 1096, 1099, 1130, 1131, 1167, 1299, 1441, 1450, 1451, 1452, 1564, 1746, 1750, 1751, 1752, 1795, 203, 204, 214, 222, 224, 225, 226, 380, 525, 591, 612, 613, 615, 635, 636, 663, 664, 669, 699, 765, 790, 839, 840, 841, 900, 909, 933, or 947.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:130 comprising nucleotide 1063, 1102, 1106, 1107, 1108, 1109, 1111, or 1151.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:131 comprising nucleotide 653, 654, 771, 773, 841, 849, 853, 917, 918, 926, 927, 931, 981, 983, 984, 996, 998, 1022, 1023, 1160, 1214, 1355, 1356, 1381, 1394, 1425, 1474, 1550, 1620, 1707, 1740, 1753, 1825, 1956, 1965, 2598, 2599, 2608, 2828, 2829, 2888, 3012, or 3251.
  • an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:132 comprising nucleotide 292, 434, 438, 457, 459, 488, 490, 498, 499, 592, 639, 723, 774, 775, 788, 857, 858, 910, 911, 930, 931, 932, 1009, 1010, 1023, 1024, 1111, 1146, 1147, 1220, 1246, 1321, 1325, 1326, 1327, 1331, 1437, 1548, 1571, 1785, 1786, or 1787.
  • Another embodiment of the present invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:7 comprising nucleotide 675, 974, 1373, 1780, 2102, 2151, 2315, 2542, 2609, 3197, 67, 71, 73, 353, 405, 864, 911, 912, 913, 1409, 1748, 1811, 1935, 1937, 1993, 2012, 2346, 2388, 2437, 2586, 3007, 3008, 3022, 3130, 3210, 3237, or 3271.
  • Another embodiment of the present invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:133 comprising nucleotide 748, 749, 753, 1119, 1169, 1499, 1509, 1820, 2081, 2118, 2147, 2615, 2644, 2659, 2663, 2671, 2672, 2793, 2812, 2914, 2948, 3044, 3334, 3391, 3480, 3520, 3549, 3639, 3840, 3941, 3944, 4001, 4995, 4997, 5141, 5143, 5249, 5375, 5834, 5852, 5981, or 6678.
  • the present invention further provides for administering a second interfering RNA to a subject in addition to a first interfering RNA.
  • the method comprises administering to the subject a second interfering RNA having a length of 19 to 49 nucleotides and comprising a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect complementarity of at least 19 nucleotides; wherein the antisense strand of the second interfering RNA hybridizes under physiological conditions to a second portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ
  • a further embodiment of the invention is a method of treating ocular hypertension in a subject in need thereof.
  • the method comprises administering to the eye of the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect contiguous complementarity of at least 19 nucleotides.
  • the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ
  • Another embodiment of the invention is a method of attenuating expression of an ocular hypertension target mRNA in a subject comprising administering to the subject a composition comprising an effective amount of single-stranded interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier.
  • the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to the sequence identifiers and nucleotide positions cited supra for antisense strands.
  • Another embodiment of the invention is a method of attenuating expression of an ocular hypertension target mRNA in a subject, comprising administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, where the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:8, SEQ ID NO:14-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:717, SEQ ID NO:720, and SEQ ID NO:721, as follows.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:8, SEQ ID NO:14-SEQ ID NO:32, SEQ ID NO:83-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:219, SEQ ID NO:720, and SEQ ID NO:721.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:33-SEQ ID NO:52, and SEQ ID NO:220-SEQ ID NO:282.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:53-SEQ ID NO:62 and SEQ ID NO:283-333.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:334-SEQ ID NO:374.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:63-SEQ ID NO:72 and SEQ ID NO:375-SEQ ID NO:416.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:417-SEQ ID NO:440.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:441-SEQ ID NO:511.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:512-SEQ ID NO:563.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:564-SEQ ID NO:606.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:607-SEQ ID NO:648.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:73-SEQ ID NO:82 and SEQ ID NO:649-SEQ ID NO:675.
  • the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:676-SEQ ID NO:717.
  • the region of contiguous nucleotides is a region of at least 14 contiguous nucleotides having at least 85% sequence complementarity to, or at least 85% sequence identity with, the penultimate 14 nucleotides of the 3′ end of the sequence of the sequence identifier.
  • the region of contiguous nucleotides is a region of at least 15, 16, 17, or 18 contiguous nucleotides having at least 80% sequence complementarity to, or at least 80% sequence identity with, the penultimate 15, 16, 17, or 18 nucleotides, respectively, of the 3′ end of the sequence of the sequence identifier.
  • a composition comprising interfering RNA having a length of 19 to 49 nucleotides and having a nucleotide sequence of any one of SEQ ID NO's: 8, SEQ ID NO:14-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:717, SEQ ID NO:720, and SEQ ID NO:721, or a complement thereof, and a pharmaceutically acceptable carrier is an embodiment of the present invention.
  • the interfering RNA is isolated.
  • isolated means that the interfering RNA is free of its total natural mileau.
  • Another embodiment of the invention is a method of treating ocular hypertension in a subject in need thereof, the method comprising administering to an eye of the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:8, SEQ ID NO:14-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:717, SEQ ID NO:720, and SEQ ID NO:721, wherein the ocular hypertension is treated thereby.
  • a method of attenuating expression of an ocular hypertension target mRNA first variant without attenuating expression of an ocular hypertension target mRNA second variant in a subject is a further embodiment of the invention.
  • the method comprises administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of the first variant, wherein the expression of the first variant mRNA is attenuated without attenuating expression of the second variant mRNA, and wherein the first variant target mRNA is SEQ ID NO:101, SEQ ID NO:5, SEQ ID NO:124, SEQ ID NO:127, or SEQ ID NO:129, and the second variant target mRNA is SEQ ID NO:134, SEQ
  • the first variant target mRNA is SEQ ID NO:134, SEQ ID NO:123, SEQ ID NO:125, SEQ ID NO:128, or SEQ ID NO:130
  • the second variant target mRNA is SEQ ID NO:101, SEQ ID NO:5, SEQ ID NO:124, SEQ ID NO:127, or SEQ ID NO:129, respectively.
  • FIG. 1 provides a western blot, probed with antibodies against CA2 and actin, of HeLa cells transfected with CA2 siRNAs #1, #3, #4, and #5; a non-targeting control siRNA; and a buffer control ( ⁇ siRNA).
  • the siRNAs were at a concentration of 100 nM or 1 nM.
  • the arrows indicate the positions of the ⁇ 30-kDa CA2 protein and 42-kDa actin protein bands.
  • RNA interference is a process by which double-stranded RNA (dsRNA) is used to silence gene expression. While not wanting to be bound by theory, RNAi begins with the cleavage of longer dsRNAs into small interfering RNAs (siRNAs) by an RNaseIII-like enzyme, dicer. SiRNAs are dsRNAs that are usually about 19 to 28 nucleotides, or 20 to 25 nucleotides, or 21 to 22 nucleotides in length and often contain 2-nucleotide 3′ overhangs, and 5′ phosphate and 3′ hydroxyl termini.
  • RISC RNA-induced silencing complex
  • siRNA-induced silencing complex uses this siRNA strand to identify mRNA molecules that are at least partially complementary to the incorporated siRNA strand, and then cleaves these target mRNAs or inhibits their translation. Therefore, the siRNA strand that is incorporated into RISC is known as the guide strand or the antisense strand.
  • the other siRNA strand known as the passenger strand or the sense strand, is eliminated from the siRNA and is at least partially homologous to the target mRNA.
  • siRNA design e.g., decreased siRNA duplex stability at the 5′ end of the antisense strand
  • siRNA design can favor incorporation of the antisense strand into RISC.
  • RISC-mediated cleavage of mRNAs having a sequence at least partially complementary to the guide strand leads to a decrease in the steady state level of that mRNA and of the corresponding protein encoded by this mRNA.
  • RISC can also decrease expression of the corresponding protein via translational repression without cleavage of the target mRNA.
  • Other RNA molecules and RNA-like molecules can also interact with RISC and silence gene expression. Examples of other RNA molecules that can interact with RISC include short hairpin RNAs (shRNAs), single-stranded siRNAs, microRNAs (mRNAs), and dicer-substrate 27-mer duplexes.
  • RNA refers to a double-stranded interfering RNA unless otherwise noted.
  • RNA-like molecules that can interact with RISC include RNA molecules containing one or more chemically modified nucleotides, one or more deoxyribonucleotides, and/or one or more non-phosphodiester linkages.
  • interfering RNAs all RNA or RNA-like molecules that can interact with RISC and participate in RISC-mediated changes in gene expression will be referred to as “interfering RNAs.”
  • SiRNAs, shRNAs, mRNAs, and dicer-substrate 27-mer duplexes are, therefore, subsets of “interfering RNAs.”
  • Interfering RNA of embodiments of the invention appear to act in a catalytic manner for cleavage of target mRNA, i.e., interfering RNA is able to effect inhibition of target mRNA in substoichiometric amounts. As compared to antisense therapies, significantly less interfering RNA is required to provide a therapeutic effect under such cleavage conditions.
  • the present invention relates to the use of interfering RNA to inhibit the expression of ocular hypertension target mRNA, thus lowering intraocular pressure in patients with open-angle glaucoma or ocular hypertension.
  • Ocular hypertension targets include carbonic anhydrase II, IV, and XII; ⁇ 1- and ⁇ 2 adrenergic receptors; acetylcholinesterase; Na + /K + -ATPase subunits; and Na—K-2Cl cotransporter.
  • interfering RNAs provided exogenously or expressed endogenously effect silencing of ocular hypertension target mRNA in ocular tissue(s).
  • Carbonic anhydrase catalyzes reversible hydration of carbon dioxide and appears to play a role in the regulation of aqueous humor formation. Carbonic anhydrase inhibitors lower pressure in the eye by reducing the amount of fluid produced. Carbonic anhydrase inhibitors are available as eyedrops (dorzolamide, brinzolamide) or tablets/capsules (acetazolamide, methazolamide). The eyedrops are associated with fewer side effects than the tablets or capsules and are better tolerated by many patients. AZOPT® (brinzolamide) ophthalmic suspension 1% is a topical carbonic anhydrase inhibitor (Alcon Laboratories, Inc., Fort Worth, Tex.).
  • Ophthalmic ⁇ -blockers lower pressure in the eye by reducing the amount of fluid produced in the eye.
  • These drugs are divided into two classes: the nonselective beta-blockers (timolol, levobunolol, metipranolol, carteolol) and the ⁇ -1 selective blockers (betaxolol).
  • the usual dosage is one drop in each eye once or twice a day, depending on the drug used.
  • An example of this product is BETOPTIC S® (betaxolol HCl) ophthalmic suspension 0.25% (Alcon Laboratories, Inc., Fort Worth, Tex.).
  • Inhibitors of acetylcholinesterase preserve acetylcholine at the receptor site by blocking the enzyme responsible for its hydrolysis, acetylcholinesterase. Acetylcholine accumulates at the receptor, producing a reduction in intraocular pressure by contraction of the ciliary muscle, similar to the action of direct-acting cholinergic agonists.
  • Na + /K + -ATPase inhibitors such as ouabain, nitric oxide donors, and endothelin decrease the activity of Na + /K + -ATPase, the driving force for aqueous humour formation by the ciliary process.
  • Chloride transport inhibitors such as ethacrynic acid alter trabecular meshwork cell volume to increase outflow facility.
  • nucleic acid sequences cited herein are written in a 5′ to 3′ direction unless indicated otherwise.
  • the term “nucleic acid,” as used herein, refers to either DNA or RNA or a modified form thereof comprising the purine or pyrimidine bases present in DNA (adenine “A,” cytosine “C,” guanine “G,” thymine “T”) or in RNA (adenine “A,” cytosine “C,” guanine “G,” uracil “U”).
  • Interfering RNAs provided herein may comprise “T” bases, particularly at 3′ ends, even though “T” bases do not naturally occur in RNA.
  • Nucleic acid includes the terms “oligonucleotide” and “polynucleotide” and can refer to a single-stranded molecule or a double-stranded molecule.
  • a double-stranded molecule is formed by Watson-Crick base pairing between A and T bases, C and G bases, and between A and U bases.
  • the strands of a double-stranded molecule may have partial, substantial or full complementarity to each other and will form a duplex hybrid, the strength of bonding of which is dependent upon the nature and degree of complementarity of the sequence of bases.
  • SEQ ID NO:1 provides the sense strand sequence of DNA corresponding to the mRNA for carbonic anhydrase II.
  • the mRNA sequence is identical to the DNA sense strand sequence with the “T” bases replaced with “U” bases.
  • the mRNA sequence of carbonic anhydrase II is known from SEQ ID NO:1
  • the mRNA sequence of carbonic anhydrase IV is known from SEQ ID NO:2
  • the mRNA sequence of ⁇ 1-adrenergic receptor is known from SEQ ID NO:3
  • the mRNA sequence of ⁇ 2-adrenergic receptor is known from SEQ ID NO:4
  • the mRNA sequence of acetylcholinesterase splice variant E4-E5 is known from SEQ ID NO:5
  • the mRNA sequence of Na + /K + -ATPase ⁇ 2 is known from SEQ ID NO:6
  • the mRNA sequence of Na—K-2Cl cotransporter A1 is known from SEQ ID NO:7
  • the mRNA sequence of carbonic anhydrase XII, variant 1 is known from SEQ ID NO:101
  • the mRNA sequence of Na + /K + -ATPase al, variant 1 is known from SEQ ID NO:124
  • the mRNA sequence of Na + /K + -ATPase al, variant 2 is known from SEQ ID NO:125
  • the mRNA sequence of Na + /K + -ATPase ⁇ 3 is known from SEQ ID NO:126
  • the mRNA sequence of Na + /K + -ATPase ⁇ 4 variant 1 is known from SEQ ID NO:127
  • the mRNA sequence of Na + /K + -ATPase ⁇ 4, variant 2 is known from SEQ ID NO:128,
  • the mRNA sequence of Na + /K + -ATPase ⁇ 1, variant 1 is known from SEQ ID NO:129
  • the mRNA sequence of Na + /K + -ATPase ⁇ 1, variant 2 is known from SEQ ID NO:130
  • Carbonic anhydrases II, IV, and XII mRNA (CA2, CA4, and CA12): Carbonic anhydrases (CAs) II, IV and XII are members of a large family of zinc metalloenzymes that catalyze the reversible hydration of carbon dioxide as described by the GenBank database of the National Center for Biotechnology Information at ncbi.nlm.nih.gov.
  • Carbonic anhydrases are involved in crucial physiological processes such as respiration and transport of CO 2 /bicarbonate between metabolizing tissues and the lungs, pH and CO 2 homeostasis, electrolyte secretion in a variety of tissues and organs, biosynthetic reactions (such as gluconeogenesis, lipogenesis and ureagenesis), bone resorption, calcification, and tumorigenicity.
  • Carbonic anhydrase II is a cytosolic isozyme
  • carbonic anhydrases IV and XII are membrane-bound.
  • Two transcript variants encoding different isoforms have been identified for the CA-XII gene; variant 1 encodes the longer isoform while variant 2 is lacking one of the internal coding exons compared to transcript variant 1 thereby missing an 11 amino acid segment compared to isoform 1.
  • Systemic carbonic anhydrase inhibitors (CAIs) are useful in reducing the elevated intraocular pressure (IOP) that is characteristic of glaucoma.
  • Inhibition of the isozymes present in the ciliary process reduces the rate of bicarbonate and aqueous humor secretion, which leads to a 25-30% decrease in IOP.
  • inhibition of various CA isozymes present in extraocular tissues leads to side effects including numbness and tingling of extremities, metallic taste, depression, fatigue, malaise, weight loss, decreased libido, gastrointestinal irritation, metabolic acidosis, renal calculi, and transient myopia.
  • GenBank database provides the DNA sequence for CA2 as accession no. NM — 000067, provided in the “Sequence Listing” as SEQ ID NO:1.
  • SEQ ID NO:1 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAII (with the exception of “T” bases for “U” bases).
  • the coding sequence for CAII is from nucleotides 66-848.
  • CA2 nucleic acid sequences related to SEQ ID NO:1 include those having GenBank accession numbers M77181, X03251, BC011949, BC035424, CR536526, CR541875, J03037, M36532, S69526, and Y00339.
  • GenBank database provides the DNA sequence for CA4 as accession no. NM — 000717, provided in the “Sequence Listing” as SEQ ID NO:2.
  • SEQ ID NO:2 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAIV (with the exception of “T” bases for “U” bases).
  • the coding sequence for CAIV is from nucleotides 47-985.
  • CA4 mRNA sequences are alternative splice forms, allelic forms, isozymes, or a cognate thereof.
  • a cognate is a CA4 mRNA from another mammalian species that is homologous to SEQ ID NO:2 (i.e., an ortholog).
  • CA4 nucleic acid sequences related to SEQ ID NO:2 include those having GenBank accession numbers L10955, BC057792, BC069649, BC074768, CR541766, and M83670.
  • GenBank database provides the DNA sequence for CA12, variant 1, as accession no. NM — 001218, provided in the “Sequence Listing” as SEQ ID NO:101.
  • SEQ ID NO:101 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAXII, variant 1 (with the exception of “T” bases for “U” bases).
  • the coding sequence for CAXII, variant 1, is from nucleotides 157-1221.
  • CA12, variant 1 mRNA sequence are alternative splice forms, allelic forms, isozymes, or a cognate thereof.
  • a cognate is a CA12 mRNA from another mammalian species that is homologous to SEQ ID NO:101 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for CA12, variant 2, as accession no. NM — 206925, provided in the “Sequence Listing” as SEQ ID NO:134.
  • SEQ ID NO:134 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAXII, variant 2 (with the exception of “T” bases for “U” bases).
  • the coding sequence for CAXII, variant 2 is from nucleotides 157-1188.
  • Variant 2 lacks an internal coding exon compared to variant 1.
  • CA12, variant 2 mRNA sequence are alternative splice forms, allelic forms, isozymes, or a cognate thereof.
  • a cognate is a CA12 mRNA from another mammalian species that is homologous to SEQ ID NO:134 (i.e., an ortholog).
  • Adrenergic Receptors- ⁇ 1 and ⁇ 2 mRNA (ADRB1 and ADRB2): The adrenergic receptors (subtypes ⁇ 1, ⁇ 2, ⁇ 1, and ⁇ 2) are a prototypic family of G protein-coupled receptors that mediate the physiological effects of the hormone epinephrine and the neurotransmitter norepinephrine as described by the GenBank database of the National Center for Biotechnology Information at ncbi.nlm.nih.gov.
  • GenBank database provides the DNA sequence for ADRB1 as accession no. NM — 000684, provided in the “Sequence Listing” as SEQ ID NO:3.
  • SEQ ID NO:3 provides the sense strand sequence of DNA that corresponds to the mRNA encoding ⁇ 1-adrenergic receptor (with the exception of “T” bases for “U” bases).
  • the coding sequence for ⁇ 1-adrenergic receptor is from nucleotides 87-1520.
  • ADRB1 mRNA sequences are alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ADRB1 mRNA from another mammalian species that is homologous to SEQ ID NO:3 (i.e., an ortholog).
  • ADRB1 nucleic acid sequences related to SEQ ID NO:3 include those having GenBank accession numbers AF169006, AF169007, AY567837, and J03019.
  • GenBank database provides the DNA sequence for ADRB2 as accession no. NM — 000024, provided below as SEQ ID NO:4.
  • SEQ ID NO:4 provides the sense strand sequence of DNA that corresponds to the mRNA encoding ⁇ 2-adrenergic receptor (with the exception of “T” bases for “U” bases).
  • the coding sequence for ⁇ 2-adrenergic receptor is from nucleotides 220-1461.
  • ADRB2 nucleic acid sequences related to SEQ ID NO:4 include those having GenBank accession numbers AF022953, AF022954, AF022955, AF022956, AF169225, AF202305, AF203386, AY011291, J02960, Y00106, AY136741, BC012481, BC063486, BC073856, M15169, and X04827.
  • Acetylcholinesterase mRNA splice variants E4-E6 and E4-E5 As described by the GenBank database of the National Center for Biotechnology Information at ncbi.nlm.nih.gov, acetylcholinesterase hydrolyzes the neurotransmitter acetylcholine at neuromuscular junctions and brain cholinergic synapses, and thus terminates signal transmission. It is also found on red blood cell membranes, where it constitutes the Yt blood group antigen. Acetylcholinesterase exists in multiple molecular forms which possess similar catalytic properties, but differ in their oligomeric assembly and mode of cell attachment to the cell surface.
  • the major form of acetylcholinesterase found in brain, muscle and other tissues is the hydrophilic species, which forms disulfide-linked oligomers with collagenous, or lipid-containing structural subunits.
  • the other, alternatively spliced form expressed primarily in the erythroid tissues, differs at the C-terminal end, and contains a cleavable hydrophobic peptide with a GPI-anchor site. It associates with the membranes through the phosphoinositide (PI) moieties added post-translationally.
  • the splice variant E4-E6 is the major transcript and results from the splicing of exon 4 to exon 6.
  • the splice variant E4-E5 results from alternative splicing of exon 4 to exon 5.
  • GenBank database provides the DNA sequence for ACHE splice variant E4-E5 as accession no. NM — 015831, provided in the “Sequence Listing” as SEQ ID NO:5.
  • SEQ ID NO:5 provides the sense strand sequence of DNA that corresponds to the mRNA encoding acetylcholinesterase E4-E5 (with the exception of “T” bases for “U” bases).
  • the coding sequence for acetylcholinesterase E4-E5 is from nucleotides 95-1948.
  • ACHE nucleic acid sequences related to SEQ ID NO:5 include those having GenBank accession numbers AC011895, AF002993, AF312032, AY750146, CH236956, L06484, L42812, S71129, AF334270, BC026315, BC036813, M55040 and NM — 000665.
  • GenBank database provides the DNA sequence for ACHE splice variant E4-E6 as accession no. NM — 000665, provided in the “Sequence Listing” as SEQ ID NO:123.
  • SEQ ID NO:123 provides the sense strand sequence of DNA that corresponds to the mRNA encoding acetylcholinesterase E4-E6 variant (with the exception of “T” bases for “U” bases).
  • the coding sequence for acetylcholinesterase E4-E6 is from nucleotides 95-1939.
  • ACHE nucleic acid sequences related to SEQ ID NO:123 include those having GenBank accession numbers NM — 015831, AC011895, AF002993, AF312032, AY750146, CH236956, L06484, L42812, S71129, AF334270, BC026315, BC036813, and M55040.
  • Na + /K + -ATPase a and ⁇ mRNA (ATP1-A1 variant 1, -A1 variant 2, -A2, -A3, -A4 variant 1, -A4 variant 2, -B1 variant 1, -B1 variant 2, -B2, and -B3):
  • the proteins encoded by these genes belong to the family of P-type cation transport ATPases, and to the subfamily of Na + /K + -ATPases.
  • Na + /K + -ATPase is an integral membrane protein responsible for establishing and maintaining the electrochemical gradients of Na and K ions across the plasma membrane.
  • This enzyme is composed of two subunits, a large catalytic subunit (a or A) and a smaller glycoprotein subunit ( ⁇ or B).
  • the catalytic subunit of Na + /K + -ATPase is encoded by multiple genes.
  • GenBank database provides the DNA sequence for ATP1A1 variant 1 as accession no. NM — 000701, provided in the “Sequence Listing” as SEQ ID NO:124.
  • SEQ ID NO:124 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase subunit A1 variant 1 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase subunit A1 variant 1 is from nucleotides 299-3370.
  • ATP1A1 variant 1 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1A1 variant 1 mRNA from another mammalian species that is homologous to SEQ ID NO:124 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1A1 variant 2 as accession no. NM — 001001586, provided in the “Sequence Listing” as SEQ ID NO:125.
  • SEQ ID NO:125 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase subunit A1 variant 2 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase subunit A1 variant 2 is from nucleotides 299-2344.
  • ATP1A1 variant 2 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1A1 variant 2 mRNA from another mammalian species that is homologous to SEQ ID NO:125 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1A2 as accession no. NM — 000702, provided in the “Sequence Listing” as SEQ ID NO:6.
  • SEQ ID NO:6 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase A2 subunit (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase A2 subunit is from nucleotides 105-3167.
  • ATP1A2 nucleic acid sequences related to SEQ ID NO:6 include those having GenBank accession numbers J05096, M27578, AB018321, AK091617, AK124581, AK126573, AL831991, AL831997, BC013680, BC047533, BC052271, M16795, and Y07494.
  • GenBank database provides the DNA sequence for ATP1A3 as accession no. NM — 152296, provided in the “Sequence Listing” as SEQ ID NO:126.
  • SEQ ID NO:126 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase A3 subunit (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase A3 subunit is from nucleotides 155-3196.
  • ATP1A3 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1A3 mRNA from another mammalian species that is homologous to SEQ ID NO:126 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1A4 variant 1 as accession no. NM — 144699, provided in the “Sequence Listing” as SEQ ID NO:127.
  • SEQ ID NO:127 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase A4 subunit variant 1 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase A4 subunit variant 1 is from nucleotides 469-3558.
  • ATP1A4 variant 1 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1A4 variant 1 mRNA from another mammalian species that is homologous to SEQ ID NO:127 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1A4 variant 2 as accession no. NM — 001001734, provided in the “Sequence Listing” as SEQ ID NO:128.
  • SEQ ID NO:128 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase A4 subunit variant 2 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase A4 subunit variant 2 is from nucleotides 111-608.
  • ATP1A4 variant 2 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1A4 variant 2 mRNA from another mammalian species that is homologous to SEQ ID NO:128 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1B1 variant 1 as accession no. NM — 001677, provided in the “Sequence Listing” as SEQ ID NO:129.
  • SEQ ID NO:129 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase B1 subunit variant 1 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase B1 subunit variant 1 is from nucleotides 122-1033.
  • ATP1B1 variant 1 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1B1 variant 1 mRNA from another mammalian species that is homologous to SEQ ID NO:129 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1B1 variant 2 as accession no. NM — 001001787, provided in the “Sequence Listing” as SEQ ID NO:130.
  • SEQ ID NO:130 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase B1 subunit variant 2 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase B1 subunit variant 2 is from nucleotides 122-1027.
  • ATP1B1 variant 2 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1B1 variant 2 mRNA from another mammalian species that is homologous to SEQ ID NO:130 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1B2 as accession no. NM — 001678, provided in the “Sequence Listing” as SEQ ID NO:131.
  • SEQ ID NO:131 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase B2 subunit (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase B2 subunit is from nucleotides 584-1456.
  • ATP1B2 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1B2 mRNA from another mammalian species that is homologous to SEQ ID NO:131 (i.e., an ortholog).
  • GenBank database provides the DNA sequence for ATP1B3 as accession no. NM — 001679, provided in the “Sequence Listing” as SEQ ID NO:132.
  • SEQ ID NO:132 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na + /K + -ATPase B3 subunit (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na + /K + -ATPase B3 subunit is from nucleotides 175-1014.
  • ATP1B3 mRNA sequence is alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is an ATP1B3 mRNA from another mammalian species that is homologous to SEQ ID NO:132 (i.e., an ortholog).
  • Na—K-2Cl cotransporter mRNA SLC12A1 and SLC12A2: The sodium-potassium-chloride cotransporter (Na—K-2Cl cotransporter or NKCC) facilitates the coupled cotransport of Na + , K + , and Cl. ions across the plasma membrane.
  • Na—K-2Cl cotransporter or NKCC The sodium-potassium-chloride cotransporter
  • NKCC1 is expressed in most tissues, including the eye.
  • NKCC2 is expressed primarily in the kidney, however, there is evidence for lower level expression of this isoform in the eye as well.
  • NKCC1 is encoded by the SLC12A2 gene (solute carrier family 12, member 2) and NKCC2 is encoded by the SLC12A1 gene.
  • GenBank database provides the DNA sequence for SLC12A1 as accession no. NM — 000338, provided in the “Sequence Listing” as SEQ ID NO:7.
  • SEQ ID NO:7 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na—K-2Cl cotransporter NKCC2 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na—K-2Cl cotransporter NKCC2 is from nucleotides 20-3319.
  • Equivalents of the above cited Na—K-2Cl NKCC2 cotransporter mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is a Na—K-2Cl cotransporter NKCC2 mRNA from another mammalian species that is homologous to SEQ ID NO:7 (i.e., an ortholog).
  • SLC12A1 nucleic acid sequences related to SEQ ID NO:7 include those having GenBank accession numbers AJ005332, AJ005333, AB032525, AB032527, BC040138, BX647067, BX647484, and U58130.
  • GenBank database provides the DNA sequence for SLC12A2 as accession no. NM — 001046, provided in the “Sequence Listing” as SEQ ID NO:133.
  • SEQ ID NO:133 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na—K-2Cl cotransporter NKCC1 (with the exception of “T” bases for “U” bases).
  • the coding sequence for Na—K-2Cl cotransporter NKCC1 is from nucleotides 165-3803.
  • Equivalents of the above cited Na—K-2Cl cotransporter NKCC1 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof.
  • a cognate is a Na—K-2Cl cotransporter NKCC1 mRNA from another mammalian species that is homologous to SEQ ID NO:133 (i.e., an ortholog).
  • Attenuating expression of an mRNA means administering or expressing an amount of interfering RNA (e.g., an siRNA) to reduce translation of the target mRNA into protein, either through mRNA cleavage or through direct inhibition of translation.
  • interfering RNA e.g., an siRNA
  • the reduction in expression of the target mRNA or the corresponding protein is commonly referred to as “knock-down” and is reported relative to levels present following administration or expression of a non-targeting control RNA (e.g., a non-targeting control siRNA). Knock-down of expression of an amount including and between 50% and 100% is contemplated by embodiments herein.
  • a single interfering RNA targeting one of the ocular hypertension targets is administered to lower IOP.
  • two or more interfering RNAs targeting the same ocular hypertension target e.g., CA2
  • two or more interfering RNAs targeting multiple hypertension targets e.g., CA2 and ADRB2 are administered to lower IOP.
  • Knock-down is commonly assessed by measuring the mRNA levels using quantitative polymerase chain reaction (qPCR) amplification or by measuring protein levels by western blot or enzyme-linked immunosorbent assay (ELISA). Analyzing the protein level provides an assessment of both mRNA cleavage as well as translation inhibition. Further techniques for measuring knock-down include RNA solution hybridization, nuclease protection, northern hybridization, gene expression monitoring with a microarray, antibody binding, radioimmunoassay, and fluorescence activated cell analysis.
  • qPCR quantitative polymerase chain reaction
  • ELISA enzyme-linked immunosorbent assay
  • Targets cited herein are also inferred in a human or mammal by observing an improvement in a glaucoma symptom such as improvement in intraocular pressure, improvement in visual field loss, or improvement in optic nerve head changes, for example.
  • a glaucoma symptom such as improvement in intraocular pressure, improvement in visual field loss, or improvement in optic nerve head changes, for example.
  • Interfering RNA of embodiments of the invention appear to act in a catalytic manner for cleavage of target mRNA, i.e., interfering RNA is able to effect inhibition of target mRNA in substoichiometric amounts. As compared to antisense therapies, significantly less interfering RNA is required to provide a therapeutic effect under such cleavage conditions.
  • interfering RNA e.g., siRNA
  • the sense and antisense strands comprise a region of at least near-perfect contiguous complementarity of at least 19 nucleotides.
  • the interfering RNA comprises a region of at least 13, 14, 15, 16, 17, or 18 contiguous nucleotides having percentages of sequence complementarity to or, having percentages of sequence identity with, the penultimate 13, 14, 15, 16, 17, or 18 nucleotides, respectively, of the 3′ end of the corresponding target sequence within an mRNA.
  • each strand of the interfering RNA comprises 19 to 49 nucleotides, and may comprise a length of 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or 49 nucleotides.
  • the antisense strand of an siRNA is the active guiding agent of the siRNA in that the antisense strand is incorporated into RISC, thus allowing RISC to identify target mRNAs with at least partial complementarity to the antisense siRNA strand for cleavage or translational repression.
  • interfering RNA target sequences within a target mRNA sequence are selected using available design tools. Interfering RNAs corresponding to these target sequences are then tested by transfection of cells expressing the target mRNA followed by assessment of knockdown as described above. Interfering RNAs that produce a knockdown in expression of between 50% and 100% are selected for further analysis.
  • siRNA selection techniques are provided by Tuschl, T. et al., “The siRNA User Guide,” revised May 6, 2004, available on the Rockefeller University web site; by Technical Bulletin #506, “siRNA Design Guidelines,” Ambion Inc. at Ambion's web site; and by other web-based design tools at, for example, the Invitrogen, Dharmacon, Integrated DNA Technologies, Genscript, or Proligo web sites.
  • Initial search parameters can include G/C contents between 35% and 55% and siRNA lengths between 19 and 27 nucleotides.
  • the target sequence may be located in the coding region or in the 5′ or 3′ untranslated regions of the mRNA.
  • An embodiment of a 19-nucleotide DNA target sequence for carbonic anyhdrase II is present at nucleotides 232 to 250 of SEQ ID NO:1:
  • siRNA of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:8 and having 21-nucleotide strands and a 2-nucleotide 3′ overhang is: 5′-CCCUGAGGAUCCUCAACAANN-3′ SEQ ID NO:9 3′-NNGGGACUCCUAGGAGUUGUU-5′.
  • SEQ ID NO:10 Each “N” residue can be any nucleotide (A, C, G, U, T) or modified nucleotide. The 3′ end can have a number of “N” residues between and including 1, 2, 3, 4, 5, and 6.
  • the “N” residues on either strand can be the same residue (e.g., UU, AA, CC, GG, or TT) or they can be different (e.g., AC, AG, AU, CA, CG, CU, GA, GC, GU, UA, UC, or UG).
  • the 3′ overhangs can be the same or they can be different. In one embodiment, both strands have a 3′UU overhang.
  • siRNA of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:8 and having 21-nucleotide strands and a 3′UU overhang on each strand is: 5′-CCCUGAGGAUCCUCAACAAUU-3′ SEQ ID NO:11 3′-UUGGGACUCCUAGGAGUUGUU-5′.
  • the interfering RNA may also have a 5′ overhang of nucleotides or it may have blunt ends.
  • An siRNA of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:8 and having 19-nucleotide strands and blunt ends is: 5′-CCCUGAGGAUCCUCAACAA-3′ SEQ ID NO:722 3′-GGGACUCCUAGGAGUUGUU-5′. SEQ ID NO:723
  • a double-stranded interfering RNA e.g., an siRNA
  • a hairpin or stem-loop structure e.g., an shRNA
  • An shRNA of the invention targeting a corresponding mRNA sequence of SEQ ID NO:8 and having a 19 bp double-stranded stem region and a 3′UU overhang is: NNN / ⁇ 5′-CCCUGAGGAUCCUCAACAA N 3′-UUGGGACUCCUAGGAGUUGUU N SEQ ID NO:13 ⁇ ⁇ NNN.
  • N is a nucleotide A, T, C, G, U, or a modified form known by one of ordinary skill in the art.
  • the number of nucleotides N in the loop is a number between and including 3 to 23, or 5 to 15, or 7 to 13, or 4 to 9, or 9 to 11, or the number of nucleotides N is 9.
  • Some of the nucleotides in the loop can be involved in base-pair interactions with other nucleotides in the loop.
  • Examples of oligonucleotide sequences that can be used to form the loop include 5′-UUCAAGAGA-3′ (Brummelkamp, T. R. et al. (2002) Science 296: 550) and 5′-UUUGUGUAG-3′ (Castanotto, D.
  • RNAi machinery forms a stem-loop or hairpin structure comprising a double-stranded region capable of interacting with the RNAi machinery.
  • siRNA target sequence identified above can be extended at the 3′ end to facilitate the design of dicer-substrate 27-mer duplexes.
  • Extension of the 19-nucleotide DNA target sequence (SEQ ID NO:8) identified in the carbonic anhydrase II DNA sequence (SEQ ID NO:1) by 6 nucleotides yields a 25-nucleotide DNA target sequence present at nucleotides 232 to 256 of SEQ ID NO:1:
  • a dicer-substrate 27-mer duplex of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:724 is: 5′-CCCUGAGGAUCCUCAACAAUGGUCA-3′ SEQ ID NO:718 3′-UUGGGACUCCUAGGAGUUGUUACCAGU-5′.
  • SEQ ID NO:719 The two nucleotides at the 3′ end of the sense strand (i.e., the CA nucleotides of SEQ ID NO:718) may be deoxynucleotides for enhanced processing.
  • phosphorylation at the 5′ position of the nucleotide at the 5′ end of one or both strands can enhance siRNA efficacy and specificity of the bound RISC complex but is not required since phosphorylation can occur intracellularly.
  • Table 1 lists examples of siRNA target sequences within the CA2, CA4, and CA12 variant 1 and variant 2 DNA sequences (SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:101, and SEQ ID NO:134, respectively) from which siRNAs of the present invention are designed in a manner as set forth above.
  • CA2, CA4, and CA12 variant 1 and variant 2 encode carbonic anhydrase II, IV, and XII variant 1 and 2, respectively.
  • Table 2 lists examples of siRNA target sequences within the ADRB1 and ADRB2 DNA sequences (SEQ ID NO:3 and SEQ ID NO:4, respectively) from which siRNAs of the present invention are designed in a manner as set forth above.
  • ADRB1 and ADRB2 encode the ⁇ 1- and ⁇ 2-adrenergic receptors, respectively.
  • Table 3 lists examples of siRNA target sequences within the ACHE DNA sequences for splice variants E4-E5 and E4-E6 (SEQ ID NO:5 and SEQ ID NO:123, respectively) from which siRNAs of the present invention are designed in a manner as set forth above.
  • ACHE encodes acetylcholinesterase.
  • Table 4 lists examples of siRNA target sequences within the Na + /K + -ATPase A and B subunit DNA sequences (ATP1A1 variant 1, SEQ ID NO:124; ATP1A1 variant 2, SEQ ID NO:125; ATP1A2, SEQ ID NO:6; ATP1A3, SEQ ID NO:126; ATP1A4 variant 1, SEQ ID NO:127; ATP1A4 variant 2, SEQ ID NO:128; ATP1B1 variant 1, SEQ ID NO:129; ATP1B1 variant 2, SEQ ID NO:130; ATP1B2, SEQ ID NO:131; and ATP1B3, SEQ ID NO:132) from which siRNAs of the present invention are designed in a manner as set forth above.
  • Table 5 lists examples of siRNA target sequences within the SLC12A1 and SLC12A2 DNA sequences (SEQ ID NO:7 and SEQ ID NO:133, respectively) from which siRNAs of the present invention are designed in a manner as set forth above.
  • SLC12A1 and SLC12A2 encode the Na—K-2Cl cotransporter, NKCC2 and NKCC1, respectively.
  • siRNAs and other forms of interfering RNA is highly sequence specific.
  • siRNA containing a sense nucleotide strand identical in sequence to a portion of the target mRNA and an antisense nucleotide strand exactly complementary to a portion of the target mRNA are siRNA embodiments for inhibition of mRNAs cited herein.
  • 100% sequence complementarity between the antisense siRNA strand and the target mRNA, or between the antisense siRNA strand and the sense siRNA strand is not required to practice the present invention.
  • the invention allows for sequence variations that might be expected due to genetic mutation, strain polymorphism, or evolutionary divergence.
  • the antisense strand of the siRNA has at least near-perfect contiguous complementarity of at least 19 nucleotides with the target mRNA.
  • Near-perfect means the antisense strand of the siRNA is “substantially complementary to,” and the sense strand of the siRNA is “substantially identical” to at least a portion of the target mRNA.
  • Identity is the degree of sequence relatedness between nucleotide sequences as determined by matching the order and identity of nucleotides between the sequences.
  • the antisense strand of an siRNA having 80% and between 80% up to 100% complementarity, for example, 85%, 90% or 95% complementarity, to the target mRNA sequence are considered near-perfect complementarity and may be used in the present invention.
  • Perfect contiguous complementarity is standard Watson-Crick base pairing of adjacent base pairs.
  • At least near-perfect contiguous complementarity includes “perfect” complementarity as used herein.
  • Computer methods for determining identity or complementarity are designed to identify the greatest degree of matching of nucleotide sequences, for example, BLASTN (Altschul, S. F., et al. (1990) J. Mol. Biol. 215:403-410).
  • percent identity describes the percentage of contiguous nucleotides in a first nucleic acid molecule that is the same as in a set of contiguous nucleotides of the same length in a second nucleic acid molecule.
  • percent complementarity describes the percentage of contiguous nucleotides in a first nucleic acid molecule that can base pair in the Watson-Crick sense with a set of contiguous nucleotides in a second nucleic acid molecule.
  • the relationship between a target mRNA (sense strand) and one strand of an siRNA (the sense strand) is that of identity.
  • the sense strand of an siRNA is also called a passenger strand, if present.
  • the relationship between a target mRNA (sense strand) and the other strand of an siRNA (the antisense strand) is that of complementarity.
  • the antisense strand of an siRNA is also called a guide strand.
  • the penultimate base in a nucleic acid sequence that is written in a 5′ to 3′ direction is the next to the last base, i.e., the base next to the 3′ base.
  • the penultimate 13 bases of a nucleic acid sequence written in a 5′ to 3′ direction are the last 13 bases of a sequence next to the 3′ base and not including the 3′ base.
  • the penultimate 14, 15, 16, 17, or 18 bases of a nucleic acid sequence written in a 5′ to 3′ direction are the last 14, 15, 16, 17, or 18 bases of a sequence, respectively, next to the 3′ base and not including the 3′ base.
  • the region of contiguous nucleotides is a region of at least 14 contiguous nucleotides having at least 85% sequence complementarity to, or at least 85% sequence identity with, the penultimate 14 nucleotides of the 3′ end of the sequence identified by each sequence identifier.
  • the region of contiguous nucleotides is a region of at least 15, 16, 17, or 18 contiguous nucleotides having at least 80% sequence complementarity to, or at least 80% sequence identity with, the penultimate 14 nucleotides of the 3′ end of the sequence of the sequence identifier. Three nucleotide substitutions are included in such a phrase.
  • the target sequence in the mRNAs corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134 may be in the 5′ or 3′ untranslated regions of the mRNA as well as in the coding region of the mRNA.
  • One or both of the strands of double-stranded interfering RNA may have a 3′ overhang of from 1 to 6 nucleotides, which may be ribonucleotides or deoxyribonucleotides or a mixture thereof.
  • the nucleotides of the overhang are not base-paired.
  • the interfering RNA comprises a 3′ overhang of TT or UU.
  • the interfering RNA comprises at least one blunt end.
  • the termini usually have a 5′ phosphate group or a 3′ hydroxyl group.
  • the antisense strand has a 5′ phosphate group
  • the sense strand has a 5′ hydroxyl group.
  • the termini are further modified by covalent addition of other molecules or functional groups.
  • the sense and antisense strands of the double-stranded siRNA may be in a duplex formation of two single strands as described above or may be a single molecule where the regions of complementarity are base-paired and are covalently linked by a hairpin loop so as to form a single strand. It is believed that the hairpin is cleaved intracellularly by a protein termed dicer to form an interfering RNA of two individual base-paired RNA molecules.
  • Interfering RNAs may differ from naturally-occurring RNA by the addition, deletion, substitution or modification of one or more nucleotides.
  • Non-nucleotide material may be bound to the interfering RNA, either at the 5′ end, the 3′ end, or internally. Such modifications are commonly designed to increase the nuclease resistance of the interfering RNAs, to improve cellular uptake, to enhance cellular targeting, to assist in tracing the interfering RNA, to further improve stability, or to reduce the potential for activation of the interferon pathway.
  • interfering RNAs may comprise a purine nucleotide at the ends of overhangs. Conjugation of cholesterol to the 3′ end of the sense strand of an siRNA molecule by means of a pyrrolidine linker, for example, also provides stability to an siRNA.
  • Further modifications include a 3′ terminal biotin molecule, a peptide known to have cell-penetrating properties, a nanoparticle, a peptidomimetic, a fluorescent dye, or a dendrimer, for example.
  • Nucleotides may be modified on their base portion, on their sugar portion, or on the phosphate portion of the molecule and function in embodiments of the present invention. Modifications include substitutions with alkyl, alkoxy, amino, deaza, halo, hydroxyl, thiol groups, or a combination thereof, for example. Nucleotides may be substituted with analogs with greater stability such as replacing a ribonucleotide with a deoxyribonucleotide, or having sugar modifications such as 2′ OH groups replaced by 2′ amino groups, 2′ O-methyl groups, 2′ methoxyethyl groups, or a 2′-O, 4′-C methylene bridge, for example.
  • Examples of a purine or pyrimidine analog of nucleotides include a xanthine, a hypoxanthine, an azapurine, a methylthioadenine, 7-deaza-adenosine and O- and N-modified nucleotides.
  • the phosphate group of the nucleotide may be modified by substituting one or more of the oxygens of the phosphate group with nitrogen or with sulfur (phosphorothioates). Modifications are useful, for example, to enhance function, to improve stability or permeability, or to direct localization or targeting.
  • Non-complementary regions may be at the 3′, 5′ or both ends of a complementary region or between two complementary regions.
  • Interfering RNAs may be generated exogenously by chemical synthesis, by in vitro transcription, or by cleavage of longer double-stranded RNA with dicer or another appropriate nuclease with similar activity.
  • Chemically synthesized interfering RNAs produced from protected ribonucleoside phosphoramidites using a conventional DNA/RNA synthesizer, may be obtained from commercial suppliers such as Ambion Inc. (Austin, Tex.), Invitrogen (Carlsbad, Calif.), or Dharmacon (Lafayette, Colo.).
  • Interfering RNAs are purified by extraction with a solvent or resin, precipitation, electrophoresis, chromatography, or a combination thereof, for example. Alternatively, interfering RNA may be used with little if any purification to avoid losses due to sample processing.
  • Interfering RNAs can also be expressed endogenously from plasmid or viral expression vectors or from minimal expression cassettes, for example, PCR generated fragments comprising one or more promoters and an appropriate template or templates for the interfering RNA.
  • plasmid-based expression vectors for shRNA include members of the pSilencer series (Ambion, Austin, Tex.) and pCpG-siRNA (InvivoGen, San Diego, Calif.).
  • Viral vectors for expression of interfering RNA may be derived from a variety of viruses including adenovirus, adeno-associated virus, lentivirus (e.g., HIV, FIV, and EIAV), and herpes virus.
  • Examples of commercially available viral vectors for shRNA expression include pSilencer adeno (Ambion, Austin, Tex.) and pLenti6/BLOCK-iTTM-DEST (Invitrogen, Carlsbad, Calif.). Selection of viral vectors, methods for expressing the interfering RNA from the vector and methods of delivering the viral vector are within the ordinary skill of one in the art. Examples of kits for production of PCR-generated shRNA expression cassettes include Silencer Express (Ambion, Austin, Tex.) and siXpress (Mirus, Madison, Wis.).
  • Interfering RNAs may be expressed from a variety of eukaryotic promoters known to those of ordinary skill in the art, including pol III promoters, such as the U6 or HI promoters, or pol II promoters, such as the cytomegalovirus promoter. Those of skill in the art will recognize that these promoters can also be adapted to allow inducible expression of the interfering RNA.
  • an antisense strand of an interfering RNA hybridizes with an mRNA in vivo as part of the RISC complex.
  • Hybridization refers to a process in which single-stranded nucleic acids with complementary or near-complementary base sequences interact to form hydrogen-bonded complexes called hybrids. Hybridization reactions are sensitive and selective. In vitro, the specificity of hybridization (i.e., stringency) is controlled by the concentrations of salt or formamide in prehybridization and hybridization solutions, for example, and by the hybridization temperature; such procedures are well known in the art. In particular, stringency is increased by reducing the concentration of salt, increasing the concentration of form amide, or raising the hybridization temperature.
  • high stringency conditions could occur at about 50% formamide at 37° C. to 42° C.
  • Reduced stringency conditions could occur at about 35% to 25% formamide at 30° C. to 35° C.
  • Examples of stringency conditions for hybridization are provided in Sambrook, J., 1989 , Molecular Cloning. A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.
  • Further examples of stringent hybridization conditions include 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50° C. or 70° C. for 12-16 hours followed by washing, or hybridization at 70° C. in 1 ⁇ SSC or 50° C. in 1 ⁇ SSC, 50% formamide followed by washing at 70° C.
  • T m melting temperature
  • in vitro hybridization assay provides a method of predicting whether binding between a candidate siRNA and a target will have specificity.
  • specific cleavage of a target can also occur with an antisense strand that does not demonstrate high stringency for hybridization in vitro.
  • Single-stranded interfering RNA As cited above, interfering RNAs ultimately function as single strands. Single-stranded (ss) interfering RNA has been found to effect mRNA silencing, albeit less efficiently than double-stranded RNA.
  • embodiments of the present invention also provide for administration of a ss interfering RNA that hybridizes under physiological conditions to a portion of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO
  • the ss interfering RNA has a length of 19 to 49 nucleotides as for the ds interfering RNA cited above.
  • the ss interfering RNA has a 5′ phosphate or is phosphorylated in situ or in vivo at the 5′ position.
  • the term “5′ phosphorylated” is used to describe, for example, polynucleotides or oligonucleotides having a phosphate group attached via ester linkage to the C5 hydroxyl of the sugar (e.g., ribose, deoxyribose, or an analog of same) at the 5′ end of the polynucleotide or oligonucleotide.
  • SS interfering RNAs are synthesized chemically or by in vitro transcription or expressed endogenously from vectors or expression cassettes as for ds interfering RNAs. 5′ Phosphate groups may be added via a kinase, or a 5′ phosphate may be the result of nuclease cleavage of an RNA. Delivery is as for ds interfering RNAs. In one embodiment, ss interfering RNAs having protected ends and nuclease resistant modifications are administered for silencing. SS interfering RNAs may be dried for storage or dissolved in an aqueous solution. The solution may contain buffers or salts to inhibit annealing or for stabilization.
  • hairpin interfering RNA is a single molecule (e.g., a single oligonucleotide chain) that comprises both the sense and antisense strands of an interfering RNA in a stem-loop or hairpin structure (e.g., a shRNA).
  • shRNAs can be expressed from DNA vectors in which the DNA oligonucleotides encoding a sense interfering RNA strand are linked to the DNA oligonucleotides encoding the reverse complementary antisense interfering RNA strand by a short spacer. If needed for the chosen expression vector, 3′ terminal T's and nucleotides forming restriction sites may be added. The resulting RNA transcript folds back onto itself to form a stem-loop structure.
  • Interfering RNA may be delivered directly to the eye by ocular tissue injection such as periocular, conjunctival, subtenon, intracameral, intravitreal, intraocular, subretinal, subconjunctival, retrobulbar, or intracanalicular injections; by direct application to the eye using a catheter or other placement device such as a retinal pellet, intraocular insert, suppository or an implant comprising a porous, non-porous, or gelatinous material; by topical ocular drops or ointments; or by a slow release device in the cul-de-sac or implanted adjacent to the sclera (transscleral) or within the eye.
  • ocular tissue injection such as periocular, conjunctival, subtenon, intracameral, intravitreal, intraocular, subretinal, subconjunctival, retrobulbar, or intracanalicular injections
  • a catheter or other placement device such as a retinal
  • Intracanalicular injection may be through the cornea into the anterior chamber to allow the agent to reach the trabecular meshwork.
  • Intracanalicular injection may be into the venous collector channels draining Schlemm's canal or into Schlemm's canal.
  • Systemic or parenteral administration is contemplated including but not limited to intravenous, subcutaneous, and oral delivery.
  • a subject in need of treatment for ocular hypertension or at risk for developing ocular hypertension is a human or other mammal having ocular hypertension or at risk of having ocular hypertension associated with undesired or inappropriate expression or activity of targets as cited herein, i.e., carbonic anhydrase II, IV, or XII; ⁇ 1- or ⁇ 2-adrenergic receptors; acetylcholinesterase; Na + /K + -ATPase; or Na—K-2Cl cotransporter.
  • Ocular structures associated with such disorders may include the eye, retina, choroid, lens, cornea, trabecular meshwork, iris, optic nerve, optic nerve head, sclera, aqueous chamber, vitreous chamber, or ciliary body, for example.
  • a subject may also be an ocular cell, cell culture, organ or an ex vivo organ or tissue.
  • compositions comprise an interfering RNA, or salt thereof, of the invention up to 99% by weight mixed with a physiologically acceptable ophthalmic carrier medium such as water, buffer, saline, glycine, hyaluronic acid, mannitol, and the like.
  • a physiologically acceptable ophthalmic carrier medium such as water, buffer, saline, glycine, hyaluronic acid, mannitol, and the like.
  • Interfering RNAs of the present invention are administered as solutions, suspensions, or emulsions.
  • the following are examples of possible formulations embodied by this invention.
  • Purified water (RNase-free) qs 100 mL Phosphate Buffered Saline 1.0 Benzalkonium Chloride 0.01 Polysorbate 80 0.5 Purified water (RNase-free) q.s.
  • an effective amount of the interfering RNA of embodiments of the invention results in an extracellular concentration at the surface of the target cell of from 100 pM to 100 nM, or from 1 nM to 50 nM, or from 5 nM to about 10 nM, or to about 25 nM.
  • the dose required to achieve this local concentration will vary depending on a number of factors including the delivery method, the site of delivery, the number of cell layers between the delivery site and the target cell or tissue, whether delivery is local or systemic, etc.
  • the concentration at the delivery site may be considerably higher than it is at the surface of the target cell or tissue.
  • Topical compositions are delivered to the surface of the eye one to four times per day, or on an extended delivery schedule such as daily, weekly, bi-weekly, monthly, or longer, according to the routine discretion of a skilled clinician.
  • the pH of the formulation is about pH 4-9, or pH 4.5 to pH 7.4.
  • Therapeutic treatment of patients with siRNAs directed against the ocular hypertension target mRNAs is expected to be beneficial over small molecule topical ocular drops by increasing the duration of action, thereby allowing less frequent dosing and greater patient compliance.
  • interfering RNA may be administered by placing one drop in each eye as directed by the clinician.
  • An effective amount of a formulation may depend on factors such as the age, race, and sex of the subject, the severity of the ocular hypertension, the rate of target gene transcript/protein turnover, the interfering RNA potency, and the interfering RNA stability, for example.
  • the interfering RNA is delivered topically to the eye and reaches the trabecular meshwork, retina or optic nerve head at a therapeutic dose thereby ameliorating an ocular hypertension-associated disease process.
  • An ophthalmically acceptable carrier refers to those carriers that cause at most, little to no ocular irritation, provide suitable preservation if needed, and deliver one or more interfering RNAs of the present invention in a homogenous dosage.
  • An acceptable carrier for administration of interfering RNA of embodiments of the present invention include the cationic lipid-based transfection reagents TransIT®-TKO (Mirus Corporation, Madison, Wis.), LIPOFECTIN®, Lipofectamine, OLIGOFECTAMINETM (Invitrogen, Carlsbad, Calif.), or DHARMAFECTM (Dharmacon, Lafayette, Colo.); polycations such as polyethyleneimine; cationic peptides such as Tat, polyarginine, or Penetratin (Antp peptide); or liposomes.
  • Liposomes are formed from standard vesicle-forming lipids and a sterol, such as cholesterol, and may include a targeting molecule such as a monoclonal antibody having binding affinity for endothelial cell surface antigens, for example. Further, the liposomes may be PEGylated liposomes.
  • the interfering RNAs may be delivered in solution, in suspension, or in bioerodible or non-bioerodible delivery devices.
  • the interfering RNAs can be delivered alone, as components of covalent conjugates, complexed with cationic lipids, cationic peptides, or cationic polymers, or encapsulated in targeted or non-targeted nanoparticles.
  • an interfering RNA may be combined with ophthalmologically acceptable preservatives, co-solvents, surfactants, viscosity enhancers, penetration enhancers, buffers, sodium chloride, or water to form an aqueous, sterile ophthalmic suspension or solution.
  • Ophthalmic solution formulations may be prepared by dissolving the interfering RNA in a physiologically acceptable isotonic aqueous buffer. Further, the ophthalmic solution may include an ophthalmologically acceptable surfactant to assist in dissolving the inhibitor.
  • Viscosity building agents such as hydroxymethyl cellulose, hydroxyethyl cellulose, methylcellulose, polyvinylpyrrolidone, or the like may be added to the compositions of the present invention to improve the retention of the compound.
  • the interfering RNA is combined with a preservative in an appropriate vehicle, such as mineral oil, liquid lanolin, or white petrolatum.
  • an appropriate vehicle such as mineral oil, liquid lanolin, or white petrolatum.
  • Sterile ophthalmic gel formulations may be prepared by suspending the interfering RNA in a hydrophilic base prepared from the combination of, for example, CARBOPOL®-940 (BF Goodrich, Charlotte, N.C.), or the like, according to methods known in the art for other ophthalmic formulations.
  • VISCOAT® Alcon Laboratories, Inc., Fort Worth, Tex.
  • intraocular injection for example.
  • compositions of the present invention may contain penetration enhancing agents such as cremephor and TWEEN® 80 (polyoxyethylene sorbitan monolaureate, Sigma Aldrich, St. Louis, Mo.), in the event the interfering RNA is less penetrating in the eye.
  • penetration enhancing agents such as cremephor and TWEEN® 80 (polyoxyethylene sorbitan monolaureate, Sigma Aldrich, St. Louis, Mo.), in the event the interfering RNA is less penetrating in the eye.
  • kits Embodiments of the present invention provide a kit that includes reagents for attenuating the expression of an mRNA as cited herein in a cell.
  • the kit contains an siRNA or an shRNA expression vector.
  • the kit also may contain a transfection reagent or other suitable delivery vehicle.
  • the kit may contain the viral vector and/or the necessary components for viral vector production (e.g., a packaging cell line as well as a vector comprising the viral vector template and additional helper vectors for packaging).
  • the kit may also contain positive and negative control siRNAs or shRNA expression vectors (e.g., a non-targeting control siRNA or an siRNA that targets an unrelated mRNA).
  • the kit also may contain reagents for assessing knockdown of the intended target gene (e.g., primers and probes for quantitative PCR to detect the target mRNA and/or antibodies against the corresponding protein for western blots).
  • the kit may comprise an siRNA sequence or an shRNA sequence and the instructions and materials necessary to generate the siRNA by in vitro transcription or to construct an shRNA expression vector.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Printed instructions either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • TM cells for example, cell lines designated GTM-3 or HTM-3 (see Pang, I. H. et al., 1994 . Curr. Eye Res. 13:51-63), are plated 24 h prior to transfection in standard growth medium (e.g., DMEM supplemented with 10% fetal bovine serum). Transfection is performed using Dharmafect 1 (Dharmacon, Lafayette, Colo.) according to the manufacturer's instructions at interfering RNA concentrations ranging from 0.1 nM-100 nM.
  • standard growth medium e.g., DMEM supplemented with 10% fetal bovine serum
  • Non-targeting control interfering RNA and lamin A/C interfering RNA are used as controls.
  • Target mRNA levels are assessed by qPCR 24 h post-transfection using, for example, TAQMAN® forward and reverse primers and a probe set that encompasses the target site (Applied Biosystems, Foster City, Calif.).
  • Target protein levels may be assessed approximately 72 h post-transfection (actual time dependent on protein turnover rate) by western blot, for example. Standard techniques for RNA and/or protein isolation from cultured cells are well-known to those skilled in the art. To reduce the chance of non-specific, off-target effects, the lowest possible concentration of interfering RNA should be used that will produce the desired level of knock-down in target gene expression.
  • Example 1 The ability of interfering RNAs of the present invention to knock-down levels of CA2 protein expression is further exemplified in Example 1 as follows.
  • the present study examines the ability of CA2-interfering RNA to knock down the levels of endogenous CA2 expression in cultured HeLa cells.
  • Transfection of HeLa cells was accomplished using standard in vitro concentrations (100 nM and 1 nM) of CA2 siRNAs, or a non-targeting control siRNA and DharmaFECTTM 1 transfection reagent (Dharmacon, Lafayette, Colo.). All siRNAs were dissolved in 1 ⁇ siRNA buffer, an aqueous solution of 20 mM KCl, 6 mM HEPES (pH 7.5), 0.2 mM MgCl 2 . CA2 protein expression and actin protein expression (loading control) was evaluated by western blot analysis 72 hours post-transfection.
  • the CA2 siRNAs are double-stranded interfering RNAs having specificity for the following target sequences: siCA2#1 targets SEQ ID NO:721; siCA2#3 targets SEQ ID NO:15; siCA2#4 targets SEQ ID NO:720; siCA2#5 targets SEQ ID NO:141.
  • Each of the four CA2 siRNAs decreased CA2 expression significantly at both 100 nM and 1 nM relative to a non-targeting control siRNA as shown by the western blot data of FIG. 1 .
  • SiCA2#4 targeting SEQ ID NO:720 and siCA2#5 targeting SEQ ID NO:141 appeared to be particularly effective.

Abstract

RNA interference is provided for inhibition of ocular hypertension target mRNA expression for lowering elevated intraocular pressure in patients with open-angle glaucoma or ocular hypertension. Ocular hypertension targets include carbonic anhydrase II, IV, and XII; β1- and β2 adrenergic receptors; acetylcholinesterase; Na+/K+-ATPase; and Na—K-2Cl cotransporter. Ocular hypertension is treated by administering interfering RNAs of the present invention.

Description

  • The present application claims the benefit of co-pending U.S. Provisional Patent Applications having Ser. Nos. 60/648,926 filed Feb. 1, 2005, and 60/753,364 filed Dec. 22, 2005, the texts of which are specifically incorporated by reference herein.
  • FIELD OF THE INVENTION
  • The present invention relates to the field of interfering RNA compositions for inhibition of expression of ocular hypertension targets in glaucoma, particularly for primary open angle glaucoma.
  • BACKGROUND OF THE INVENTION
  • Glaucoma is a heterogeneous group of optic neuropathies that share certain clinical features. The loss of vision in glaucoma is due to the selective death of retinal ganglion cells in the neural retina that is clinically diagnosed by characteristic changes in the visual field, nerve fiber layer defects, and a progressive cupping of the optic nerve head (ONH). One of the main risk factors for the development of glaucoma is the presence of ocular hypertension (elevated intraocular pressure, IOP). An adequate intraocular pressure is needed to maintain the shape of the eye and to provide a pressure gradient to allow for the flow of aqueous humor to the avascular cornea and lens. IOP levels may also be involved in the pathogenesis of normal tension glaucoma (NTG), as evidenced by patients benefiting from IOP lowering medications. Once adjustments for central corneal thickness are made to IOP readings in NTG patients, many of these patients may be found to be ocular hypertensive.
  • The elevated IOP associated with glaucoma is due to elevated aqueous humor outflow resistance in the trabecular meshwork (TM), a small specialized tissue located in the iris-corneal angle of the ocular anterior chamber. Glaucomatous changes to the TM include a loss in TM cells and the deposition and accumulation of extracellular debris including proteinaceous plaque-like material. In addition, there are also changes that occur in the glaucomatous ONH. In glaucomatous eyes, there are morphological and mobility changes in ONH glial cells. In response to elevated IOP and/or transient ischemic insults, there is a change in the composition of the ONH extracellular matrix and alterations in the glial cell and retinal ganglion cell axon morphologies.
  • Primary glaucomas result from disturbances in the flow of intraocular fluid that has an anatomical or physiological basis. Secondary glaucomas occur as a result of injury or trauma to the eye or a preexisting disease. Primary open angle glaucoma (POAG), also known as chronic or simple glaucoma, represents ninety percent of all primary glaucomas. POAG is characterized by the degeneration of the trabecular meshwork, resulting in abnormally high resistance to fluid drainage from the eye. A consequence of such resistance is an increase in the IOP that is required to drive the fluid normally produced by the eye across the increased resistance.
  • Current anti-glaucoma therapies include lowering IOP by the use of suppressants of aqueous humor formation or agents that enhance uveoscleral outflow, laser trabeculoplasty, or trabeculectomy, which is a filtration surgery to improve drainage. Pharmaceutical anti-glaucoma approaches have exhibited various undesirable side effects. For example, miotics such as pilocarpine can cause blurring of vision and other negative visual side effects. Systemically administered carbonic anhydrase inhibitors (CAIs) can also cause nausea, dyspepsia, fatigue, and metabolic acidosis. Further, certain beta-blockers have increasingly become associated with serious pulmonary side effects attributable to their effects on beta-2 receptors in pulmonary tissue. Sympathomimetics cause tachycardia, arrhythmia and hypertension. Such negative side effects may lead to decreased patient compliance or to termination of therapy. In addition, the efficacy of current IOP lowering therapies is relatively short-lived requiring repeated dosing during each day and, in some cases, the efficacy decreases with time.
  • In view of the importance of ocular hypertension in glaucoma, and the inadequacies of prior methods of treatment, it would be desirable to have an improved method of treating ocular hypertension that would address the underlying causes of its progression.
  • SUMMARY OF THE INVENTION
  • The present invention is directed to interfering RNAs that silence ocular hypertension target mRNA expression, thus lowering intraocular pressure in patients with open-angle glaucoma or ocular hypertension. Ocular hypertension targets include carbonic anhydrase II, IV, and XII; β1- and β2 adrenergic receptors; acetylcholinesterase; Na+/K+-ATPase; and Na—K-2Cl cotransporter. The interfering RNAs of the invention are useful for treating patients with open-angle glaucoma or ocular hypertension.
  • An embodiment of the present invention provides a method of attenuating expression of an ocular hypertension target mRNA such as carbonic anhydrase II, IV, or XII; β1- or β2 adrenergic receptors; acetylcholinesterase; Na+/K+-ATPase; or Na—K-2Cl cotransporter mRNA in a subject. The method comprises administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier. Administration is to the eye of the subject for attenuating expression of an ocular hypertension target in a human.
  • In one embodiment of the invention, the interfering RNA comprises a sense nucleotide strand, an antisense nucleotide strand and a region of at least near-perfect contiguous complementarity of at least 19 nucleotides. Further, the antisense strand hybridizes under physiological conditions to a portion of an mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134 which are sense cDNA sequences encoding carbonic anhydrase II and IV; β1- and β2 adrenergic receptors; acetylcholinesterase (ACHE) variant E4-E5; Na+/K+-ATPase α2 polypeptide; Na—K-2Cl cotransporter NKCC2 (SLC12A1), carbonic anhydrase XII variant 1, acetylcholinesterase variant E4-E6, Na+/K+-ATPase al polypeptide variant 1 and variant 2, Na+/K+-ATPase α3 polypeptide, Na+/K+-ATPase α4 polypeptide variant 1 and variant 2, Na+/K+-ATPase β1 polypeptide variant 1 and 2, Na+/K+-ATPase β2 polypeptide, Na+/K+-ATPase β3 polypeptide, Na—K-2Cl cotransporter NKCC1 (SLC12A2), and carbonic anhydrase XII variant 2, respectively. The antisense strand has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, respectively. The administration of such a composition attenuates the expression of an ocular hypertension target mRNA of the subject.
  • In one embodiment, the ocular hypertension target mRNA encodes carbonic anhydrase II, IV or XII, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:101, or SEQ ID NO:134 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:101, or SEQ ID NO:134, respectively.
  • In another embodiment, the ocular hypertension target mRNA encodes a β1- or β2-adrenergic receptor, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4, respectively.
  • In a further embodiment, the ocular hypertension target mRNA encodes an acetylcholinesterase, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123, respectively.
  • In yet another embodiment, the ocular hypertension target mRNA encodes a subunit of Na+/K+-ATPase, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132, respectively.
  • In a further embodiment, the ocular hypertension target mRNA encodes a Na—K-2Cl cotransporter, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:7 or SEQ ID NO:133 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:7 or SEQ ID NO:133, respectively.
  • In one embodiment of the invention, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:1 comprising nucleotide 232, 527, 721, 728, 809, 810, 855, 856, 921, 1139, 506, 547, 548, 740, 911, 1009, 1140, 1149, 1150, 1151, 1188, 1194, 1195, 1223, 1239, 1456, 1457, 1458, 100, 158, 166, 247, 286, 318, 322, 328, 371, 412, 482, 504, 505, 541, 734, 772, 777, 814, 972, 998, 1232, 317, or 401.
  • In another embodiment of the invention, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:2 comprising nucleotide 213, 252, 258, 266, 399, 457, 463, 490, 595, 1064, 109, 112, 125, 126, 150, 261, 265, 280, 398, 453, 459, 462, 467, 492, 534, 785, 801, 825, 827, 876, 1003, or 1012.
  • In a further embodiment of the invention, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:101 comprising nucleotide 191, 239, 274, 275, 341, 389, 412, 413, 423, 687, 689, 695, 710, 791, 792, 794, 983, 993, 994, 995, 691, 1039, 1568, 2326, 2332, 2425, 2433, 2844, 2845, 2880, 2884, 2891, 2954, 2955, 2956, 2957, 2964, 2965, 3006, 3007, 3012, or 3026.
  • In another embodiment, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:134 comprising nucleotide 687, 1535, 2293, 2299, 2392, 2400, 2811, 2812, 2847, 2851, 2858, 2921, 2922, 2923, 2924, 2931, 2932, 2973, 2974, 2979, or 2993.
  • Another embodiment of the invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:3 comprising nucleotide 468, 523, 799, 1563, 1565, 1569, 1593, 1613, 1614, 1626, 310, 322, 726, 769, 772, 801, 802, 1501, 1576, 1577, 1579, 1580, 1581, 1586, 1590, 1592, 1594, 1615, 1616, 1632, 1633, or 1654.
  • A further embodiment of the invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:4 comprising nucleotide 329, 375, 1031, 1046, 1149, 1163, 1371, 1401, 1426, 1880, 283, 607, 608, 609, 619, 623, 722, 857, 1037, 1091, 1115, 1124, 1136, 1137, 1151, 1164, 1393, 1394, 1395, 1406, 1407, 1427, 1428, 1429, 1442, 1725, 1726, 1756, 1757, 1758, 1767, 1790, 1791, 1792, 1793, 1803, 1861, 1869, 1971, 1972, or 1979.
  • In another method of the invention, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:123 comprising nucleotide 1875, 1890, 1891, 2011, 2012, 2133, or 2134.
  • Another embodiment of the invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:5 comprising nucleotide 366, 370, 384, 385, 525, 588, 768, 1045, 1046, 1061, 1090, 1232, 1314, 1316, 1460, 1461, 1462, 1528, 1607, 1705, 1713, 382, 393, 397, 622, 1131, 1459, 1530, 2251, 2885, 2886, 386, 1231, 1315, 2047, 2049, 2053, 2055, 2057, 2125, 2126, 2127, 2250, 2253, 2258, 2260, 2318, 2395, 2397, 2404, 2405, 2643, 2645, or 2887.
  • In a further embodiment, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:124 comprising nucleotide 2208, 2275, 2307, 2526, 2538, 2592, 2628, 2979, 2985, 3093, 3474, 3504, 3505, 3506, 3518, 343, 442, 700, 707, 811, 907, 1059, 1363, 1594, 1662, 1758, 1760, 1896, 2037, or 2147.
  • In yet another embodiment, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:125 comprising nucleotide 436, 441, 443, 552, 617, 701, 702, 832, 2204, 2291, or 2495.
  • A further embodiment of the present invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:6 comprising nucleotide 471, 1990, 3080, 3797, 4037, 4093, 4225, 4323, 5213, 5285, 214, 467, 470, 472, 473, 632, 825, 946, 1693, 1767, 1768, 2157, 2263, 2589, 2590, 2765, 2988, 3094, 3144, 3145, 3344, 3345, 3418, 3666, 3828, 3850, 4040, 4041, 4061, 4882, 4894, 4900, 5040, 5114, 5115, 5128, 5129, 5253, 5296, 5375, 5384, or 5385.
  • In another embodiment of the invention, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:126 comprising nucleotide 240, 272, 362, 1836, 1851, 2103, 2137, 2138, 2139, 2157, 2158, 2160, 2425, 2580, 2601, 2646, 2650, 2794, 2803, 3116, 3124, 3126, 3129, or 3377.
  • In yet another embodiment of the invention, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:127 comprising nucleotide 113, 612, 702, 833, 1101, 1732, 1733, 1836, 2070, 2071, 2143, 2328, 2475, 2861, 2862, 2952, 3203, 3281, 3377, 3379, 3470, 3471, 3554, 3614, 3615, 3616, 3617, 3625, 3626, 3642, 3646, 3647, 3653, 3655, 3797, 3801, 3803, 3809 or 3810.
  • In another embodiment, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:128 comprising nucleotide 126, 251, 252, 253, 331, 427, 429, 520, 521, 530, 601, 602, 603, 604, 664, 665, 666, 667, 675, 676, 692, 696, 697, 702, 703, 705, 707, 847, 851, 853, 859, or 860.
  • In yet another embodiment, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:129 comprising nucleotide 1096, 1099, 1130, 1131, 1167, 1299, 1441, 1450, 1451, 1452, 1564, 1746, 1750, 1751, 1752, 1795, 203, 204, 214, 222, 224, 225, 226, 380, 525, 591, 612, 613, 615, 635, 636, 663, 664, 669, 699, 765, 790, 839, 840, 841, 900, 909, 933, or 947.
  • In another embodiment, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:130 comprising nucleotide 1063, 1102, 1106, 1107, 1108, 1109, 1111, or 1151.
  • In another embodiment, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:131 comprising nucleotide 653, 654, 771, 773, 841, 849, 853, 917, 918, 926, 927, 931, 981, 983, 984, 996, 998, 1022, 1023, 1160, 1214, 1355, 1356, 1381, 1394, 1425, 1474, 1550, 1620, 1707, 1740, 1753, 1825, 1956, 1965, 2598, 2599, 2608, 2828, 2829, 2888, 3012, or 3251.
  • In another embodiment of the invention, an interfering RNA is designed to target an mRNA corresponding to SEQ ID NO:132 comprising nucleotide 292, 434, 438, 457, 459, 488, 490, 498, 499, 592, 639, 723, 774, 775, 788, 857, 858, 910, 911, 930, 931, 932, 1009, 1010, 1023, 1024, 1111, 1146, 1147, 1220, 1246, 1321, 1325, 1326, 1327, 1331, 1437, 1548, 1571, 1785, 1786, or 1787.
  • Another embodiment of the present invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:7 comprising nucleotide 675, 974, 1373, 1780, 2102, 2151, 2315, 2542, 2609, 3197, 67, 71, 73, 353, 405, 864, 911, 912, 913, 1409, 1748, 1811, 1935, 1937, 1993, 2012, 2346, 2388, 2437, 2586, 3007, 3008, 3022, 3130, 3210, 3237, or 3271.
  • Another embodiment of the present invention provides an interfering RNA designed to target an mRNA corresponding to SEQ ID NO:133 comprising nucleotide 748, 749, 753, 1119, 1169, 1499, 1509, 1820, 2081, 2118, 2147, 2615, 2644, 2659, 2663, 2671, 2672, 2793, 2812, 2914, 2948, 3044, 3334, 3391, 3480, 3520, 3549, 3639, 3840, 3941, 3944, 4001, 4995, 4997, 5141, 5143, 5249, 5375, 5834, 5852, 5981, or 6678.
  • The present invention further provides for administering a second interfering RNA to a subject in addition to a first interfering RNA. The method comprises administering to the subject a second interfering RNA having a length of 19 to 49 nucleotides and comprising a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect complementarity of at least 19 nucleotides; wherein the antisense strand of the second interfering RNA hybridizes under physiological conditions to a second portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, and the antisense strand has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the second hybridizing portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, respectively. The second interfering RNA may target the same mRNA as the first interfering RNA or may target a different mRNA. Further, a third, fourth, or fifth, etc. interfering RNA may be administered in a similar manner.
  • A further embodiment of the invention is a method of treating ocular hypertension in a subject in need thereof. The method comprises administering to the eye of the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect contiguous complementarity of at least 19 nucleotides. The antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, respectively. The ocular hypertension is treated thereby.
  • Another embodiment of the invention is a method of attenuating expression of an ocular hypertension target mRNA in a subject comprising administering to the subject a composition comprising an effective amount of single-stranded interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier. For attenuating expression of an ocular hypertension target, the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to the sequence identifiers and nucleotide positions cited supra for antisense strands.
  • Another embodiment of the invention is a method of attenuating expression of an ocular hypertension target mRNA in a subject, comprising administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, where the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:8, SEQ ID NO:14-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:717, SEQ ID NO:720, and SEQ ID NO:721, as follows.
  • When the ocular hypertension target mRNA encodes carbonic anhydrase mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:8, SEQ ID NO:14-SEQ ID NO:32, SEQ ID NO:83-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:219, SEQ ID NO:720, and SEQ ID NO:721.
  • When the ocular hypertension target mRNA encodes a β-adrenergic receptor mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:33-SEQ ID NO:52, and SEQ ID NO:220-SEQ ID NO:282.
  • When the ocular hypertension target mRNA encodes ACHE mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:53-SEQ ID NO:62 and SEQ ID NO:283-333.
  • When the ocular hypertension target mRNA encodes ATP1A1 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:334-SEQ ID NO:374.
  • When the ocular hypertension target mRNA encodes ATP1A2 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:63-SEQ ID NO:72 and SEQ ID NO:375-SEQ ID NO:416.
  • When the ocular hypertension target mRNA encodes ATP1A3 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:417-SEQ ID NO:440.
  • When the ocular hypertension target mRNA encodes ATP1A4 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:441-SEQ ID NO:511.
  • When the ocular hypertension target mRNA encodes ATP1B1 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:512-SEQ ID NO:563.
  • When the ocular hypertension target mRNA encodes ATP1B2 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:564-SEQ ID NO:606.
  • When the ocular hypertension target mRNA encodes ATP1B3 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:607-SEQ ID NO:648.
  • When the ocular hypertension target mRNA encodes SLC12A1 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:73-SEQ ID NO:82 and SEQ ID NO:649-SEQ ID NO:675.
  • When the ocular hypertension target mRNA encodes SLC12A2 mRNA, the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:676-SEQ ID NO:717.
  • In a further embodiment of the present invention, the region of contiguous nucleotides is a region of at least 14 contiguous nucleotides having at least 85% sequence complementarity to, or at least 85% sequence identity with, the penultimate 14 nucleotides of the 3′ end of the sequence of the sequence identifier. In yet another embodiment of the invention, the region of contiguous nucleotides is a region of at least 15, 16, 17, or 18 contiguous nucleotides having at least 80% sequence complementarity to, or at least 80% sequence identity with, the penultimate 15, 16, 17, or 18 nucleotides, respectively, of the 3′ end of the sequence of the sequence identifier.
  • A composition comprising interfering RNA having a length of 19 to 49 nucleotides and having a nucleotide sequence of any one of SEQ ID NO's: 8, SEQ ID NO:14-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:717, SEQ ID NO:720, and SEQ ID NO:721, or a complement thereof, and a pharmaceutically acceptable carrier is an embodiment of the present invention. In one embodiment, the interfering RNA is isolated. The term “isolated” means that the interfering RNA is free of its total natural mileau.
  • Another embodiment of the invention is a method of treating ocular hypertension in a subject in need thereof, the method comprising administering to an eye of the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:8, SEQ ID NO:14-SEQ ID NO:100, SEQ ID NO:102-SEQ ID NO:122, SEQ ID NO:135-SEQ ID NO:717, SEQ ID NO:720, and SEQ ID NO:721, wherein the ocular hypertension is treated thereby.
  • A method of attenuating expression of an ocular hypertension target mRNA first variant without attenuating expression of an ocular hypertension target mRNA second variant in a subject is a further embodiment of the invention. The method comprises administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of the first variant, wherein the expression of the first variant mRNA is attenuated without attenuating expression of the second variant mRNA, and wherein the first variant target mRNA is SEQ ID NO:101, SEQ ID NO:5, SEQ ID NO:124, SEQ ID NO:127, or SEQ ID NO:129, and the second variant target mRNA is SEQ ID NO:134, SEQ ID NO:123, SEQ ID NO:125, SEQ ID NO:128, or SEQ ID NO:130, respectively.
  • In a further embodiment of the above-cited method, the first variant target mRNA is SEQ ID NO:134, SEQ ID NO:123, SEQ ID NO:125, SEQ ID NO:128, or SEQ ID NO:130, and the second variant target mRNA is SEQ ID NO:101, SEQ ID NO:5, SEQ ID NO:124, SEQ ID NO:127, or SEQ ID NO:129, respectively.
  • Use of any of the embodiments as described herein in the preparation of a medicament for attenuating expression of an ocular hypertension mRNA is also an embodiment of the present invention.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1 provides a western blot, probed with antibodies against CA2 and actin, of HeLa cells transfected with CA2 siRNAs #1, #3, #4, and #5; a non-targeting control siRNA; and a buffer control (−siRNA). The siRNAs were at a concentration of 100 nM or 1 nM. The arrows indicate the positions of the ˜30-kDa CA2 protein and 42-kDa actin protein bands.
  • DETAILED DESCRIPTION OF THE INVENTION
  • RNA interference (RNAi) is a process by which double-stranded RNA (dsRNA) is used to silence gene expression. While not wanting to be bound by theory, RNAi begins with the cleavage of longer dsRNAs into small interfering RNAs (siRNAs) by an RNaseIII-like enzyme, dicer. SiRNAs are dsRNAs that are usually about 19 to 28 nucleotides, or 20 to 25 nucleotides, or 21 to 22 nucleotides in length and often contain 2-nucleotide 3′ overhangs, and 5′ phosphate and 3′ hydroxyl termini. One strand of the siRNA is incorporated into a ribonucleoprotein complex known as the RNA-induced silencing complex (RISC). RISC uses this siRNA strand to identify mRNA molecules that are at least partially complementary to the incorporated siRNA strand, and then cleaves these target mRNAs or inhibits their translation. Therefore, the siRNA strand that is incorporated into RISC is known as the guide strand or the antisense strand. The other siRNA strand, known as the passenger strand or the sense strand, is eliminated from the siRNA and is at least partially homologous to the target mRNA. Those of skill in the art will recognize that, in principle, either strand of an siRNA can be incorporated into RISC and function as a guide strand. However, siRNA design (e.g., decreased siRNA duplex stability at the 5′ end of the antisense strand) can favor incorporation of the antisense strand into RISC.
  • RISC-mediated cleavage of mRNAs having a sequence at least partially complementary to the guide strand leads to a decrease in the steady state level of that mRNA and of the corresponding protein encoded by this mRNA. Alternatively, RISC can also decrease expression of the corresponding protein via translational repression without cleavage of the target mRNA. Other RNA molecules and RNA-like molecules can also interact with RISC and silence gene expression. Examples of other RNA molecules that can interact with RISC include short hairpin RNAs (shRNAs), single-stranded siRNAs, microRNAs (mRNAs), and dicer-substrate 27-mer duplexes. The term “siRNA” as used herein refers to a double-stranded interfering RNA unless otherwise noted. Examples of RNA-like molecules that can interact with RISC include RNA molecules containing one or more chemically modified nucleotides, one or more deoxyribonucleotides, and/or one or more non-phosphodiester linkages. For purposes of the present discussion, all RNA or RNA-like molecules that can interact with RISC and participate in RISC-mediated changes in gene expression will be referred to as “interfering RNAs.” SiRNAs, shRNAs, mRNAs, and dicer-substrate 27-mer duplexes are, therefore, subsets of “interfering RNAs.”
  • Interfering RNA of embodiments of the invention appear to act in a catalytic manner for cleavage of target mRNA, i.e., interfering RNA is able to effect inhibition of target mRNA in substoichiometric amounts. As compared to antisense therapies, significantly less interfering RNA is required to provide a therapeutic effect under such cleavage conditions.
  • The present invention relates to the use of interfering RNA to inhibit the expression of ocular hypertension target mRNA, thus lowering intraocular pressure in patients with open-angle glaucoma or ocular hypertension. Ocular hypertension targets include carbonic anhydrase II, IV, and XII; β1- and β2 adrenergic receptors; acetylcholinesterase; Na+/K+-ATPase subunits; and Na—K-2Cl cotransporter. According to the present invention, interfering RNAs provided exogenously or expressed endogenously effect silencing of ocular hypertension target mRNA in ocular tissue(s).
  • Carbonic anhydrase catalyzes reversible hydration of carbon dioxide and appears to play a role in the regulation of aqueous humor formation. Carbonic anhydrase inhibitors lower pressure in the eye by reducing the amount of fluid produced. Carbonic anhydrase inhibitors are available as eyedrops (dorzolamide, brinzolamide) or tablets/capsules (acetazolamide, methazolamide). The eyedrops are associated with fewer side effects than the tablets or capsules and are better tolerated by many patients. AZOPT® (brinzolamide) ophthalmic suspension 1% is a topical carbonic anhydrase inhibitor (Alcon Laboratories, Inc., Fort Worth, Tex.).
  • Ophthalmic β-blockers lower pressure in the eye by reducing the amount of fluid produced in the eye. These drugs are divided into two classes: the nonselective beta-blockers (timolol, levobunolol, metipranolol, carteolol) and the β-1 selective blockers (betaxolol). The usual dosage is one drop in each eye once or twice a day, depending on the drug used. An example of this product is BETOPTIC S® (betaxolol HCl) ophthalmic suspension 0.25% (Alcon Laboratories, Inc., Fort Worth, Tex.).
  • Inhibitors of acetylcholinesterase preserve acetylcholine at the receptor site by blocking the enzyme responsible for its hydrolysis, acetylcholinesterase. Acetylcholine accumulates at the receptor, producing a reduction in intraocular pressure by contraction of the ciliary muscle, similar to the action of direct-acting cholinergic agonists.
  • Na+/K+-ATPase inhibitors such as ouabain, nitric oxide donors, and endothelin decrease the activity of Na+/K+-ATPase, the driving force for aqueous humour formation by the ciliary process.
  • Chloride transport inhibitors such as ethacrynic acid alter trabecular meshwork cell volume to increase outflow facility.
  • Nucleic acid sequences cited herein are written in a 5′ to 3′ direction unless indicated otherwise. The term “nucleic acid,” as used herein, refers to either DNA or RNA or a modified form thereof comprising the purine or pyrimidine bases present in DNA (adenine “A,” cytosine “C,” guanine “G,” thymine “T”) or in RNA (adenine “A,” cytosine “C,” guanine “G,” uracil “U”). Interfering RNAs provided herein may comprise “T” bases, particularly at 3′ ends, even though “T” bases do not naturally occur in RNA. “Nucleic acid” includes the terms “oligonucleotide” and “polynucleotide” and can refer to a single-stranded molecule or a double-stranded molecule. A double-stranded molecule is formed by Watson-Crick base pairing between A and T bases, C and G bases, and between A and U bases. The strands of a double-stranded molecule may have partial, substantial or full complementarity to each other and will form a duplex hybrid, the strength of bonding of which is dependent upon the nature and degree of complementarity of the sequence of bases.
  • An mRNA sequence is readily deduced from the sequence of the corresponding DNA sequence. For example, SEQ ID NO:1 provides the sense strand sequence of DNA corresponding to the mRNA for carbonic anhydrase II. The mRNA sequence is identical to the DNA sense strand sequence with the “T” bases replaced with “U” bases.
  • Therefore, the mRNA sequence of carbonic anhydrase II is known from SEQ ID NO:1, the mRNA sequence of carbonic anhydrase IV is known from SEQ ID NO:2, the mRNA sequence of β1-adrenergic receptor is known from SEQ ID NO:3, the mRNA sequence of β2-adrenergic receptor is known from SEQ ID NO:4, the mRNA sequence of acetylcholinesterase splice variant E4-E5 is known from SEQ ID NO:5, the mRNA sequence of Na+/K+-ATPase α2 is known from SEQ ID NO:6, the mRNA sequence of Na—K-2Cl cotransporter A1 is known from SEQ ID NO:7, the mRNA sequence of carbonic anhydrase XII, variant 1 is known from SEQ ID NO:101, the mRNA sequence of acetylcholinesterase splice variant E4-E6 is known from SEQ ID. NO:123, the mRNA sequence of Na+/K+-ATPase al, variant 1, is known from SEQ ID NO:124, the mRNA sequence of Na+/K+-ATPase al, variant 2, is known from SEQ ID NO:125, the mRNA sequence of Na+/K+-ATPase α3 is known from SEQ ID NO:126, the mRNA sequence of Na+/K+-ATPase α4, variant 1, is known from SEQ ID NO:127, the mRNA sequence of Na+/K+-ATPase α4, variant 2, is known from SEQ ID NO:128, the mRNA sequence of Na+/K+-ATPase β1, variant 1, is known from SEQ ID NO:129, the mRNA sequence of Na+/K+-ATPase β1, variant 2, is known from SEQ ID NO:130, the mRNA sequence of Na+/K+-ATPase p2, is known from SEQ ID NO:131, the mRNA sequence of Na+/K+-ATPase β3 is known from SEQ ID NO:132, the mRNA sequence of Na—K-2Cl cotransporter A2 is known from SEQ ID NO:133, and the mRNA sequence of carbonic anhydrase XII, variant 2, is known from SEQ ID NO:134.
  • Carbonic anhydrases II, IV, and XII mRNA (CA2, CA4, and CA12): Carbonic anhydrases (CAs) II, IV and XII are members of a large family of zinc metalloenzymes that catalyze the reversible hydration of carbon dioxide as described by the GenBank database of the National Center for Biotechnology Information at ncbi.nlm.nih.gov. Carbonic anhydrases are involved in crucial physiological processes such as respiration and transport of CO2/bicarbonate between metabolizing tissues and the lungs, pH and CO2 homeostasis, electrolyte secretion in a variety of tissues and organs, biosynthetic reactions (such as gluconeogenesis, lipogenesis and ureagenesis), bone resorption, calcification, and tumorigenicity.
  • Fourteen different carbonic anhydrase isozymes have been identified with different subcellular localizations and tissue distributions. Carbonic anhydrase II is a cytosolic isozyme, whereas carbonic anhydrases IV and XII are membrane-bound. Two transcript variants encoding different isoforms have been identified for the CA-XII gene; variant 1 encodes the longer isoform while variant 2 is lacking one of the internal coding exons compared to transcript variant 1 thereby missing an 11 amino acid segment compared to isoform 1. Systemic carbonic anhydrase inhibitors (CAIs) are useful in reducing the elevated intraocular pressure (IOP) that is characteristic of glaucoma. Inhibition of the isozymes present in the ciliary process (the sulfonamide susceptible isozymes CA II and CA IV) reduces the rate of bicarbonate and aqueous humor secretion, which leads to a 25-30% decrease in IOP. However, inhibition of various CA isozymes present in extraocular tissues leads to side effects including numbness and tingling of extremities, metallic taste, depression, fatigue, malaise, weight loss, decreased libido, gastrointestinal irritation, metabolic acidosis, renal calculi, and transient myopia.
  • The GenBank database provides the DNA sequence for CA2 as accession no. NM000067, provided in the “Sequence Listing” as SEQ ID NO:1. SEQ ID NO:1 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAII (with the exception of “T” bases for “U” bases). The coding sequence for CAII is from nucleotides 66-848.
  • Equivalents of the above cited CA2 mRNA sequence are alternative splice forms, allelic forms, isozymes, or a cognate thereof. A cognate is a CA2 mRNA from another mammalian species that is homologous to SEQ ID NO:1 (i.e., an ortholog). CA2 nucleic acid sequences related to SEQ ID NO:1 include those having GenBank accession numbers M77181, X03251, BC011949, BC035424, CR536526, CR541875, J03037, M36532, S69526, and Y00339.
  • The GenBank database provides the DNA sequence for CA4 as accession no. NM000717, provided in the “Sequence Listing” as SEQ ID NO:2. SEQ ID NO:2 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAIV (with the exception of “T” bases for “U” bases). The coding sequence for CAIV is from nucleotides 47-985.
  • Equivalents of the above cited CA4 mRNA sequence are alternative splice forms, allelic forms, isozymes, or a cognate thereof. A cognate is a CA4 mRNA from another mammalian species that is homologous to SEQ ID NO:2 (i.e., an ortholog). CA4 nucleic acid sequences related to SEQ ID NO:2 include those having GenBank accession numbers L10955, BC057792, BC069649, BC074768, CR541766, and M83670.
  • The GenBank database provides the DNA sequence for CA12, variant 1, as accession no. NM001218, provided in the “Sequence Listing” as SEQ ID NO:101. SEQ ID NO:101 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAXII, variant 1 (with the exception of “T” bases for “U” bases). The coding sequence for CAXII, variant 1, is from nucleotides 157-1221.
  • Equivalents of the above cited CA12, variant 1 mRNA sequence are alternative splice forms, allelic forms, isozymes, or a cognate thereof. A cognate is a CA12 mRNA from another mammalian species that is homologous to SEQ ID NO:101 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for CA12, variant 2, as accession no. NM206925, provided in the “Sequence Listing” as SEQ ID NO:134. SEQ ID NO:134 provides the sense strand sequence of DNA that corresponds to the mRNA encoding CAXII, variant 2 (with the exception of “T” bases for “U” bases). The coding sequence for CAXII, variant 2, is from nucleotides 157-1188. Variant 2 lacks an internal coding exon compared to variant 1.
  • Equivalents of the above cited CA12, variant 2 mRNA sequence are alternative splice forms, allelic forms, isozymes, or a cognate thereof. A cognate is a CA12 mRNA from another mammalian species that is homologous to SEQ ID NO:134 (i.e., an ortholog).
  • Adrenergic Receptors-β1 and β2 mRNA (ADRB1 and ADRB2): The adrenergic receptors (subtypes α1, α2, β1, and β2) are a prototypic family of G protein-coupled receptors that mediate the physiological effects of the hormone epinephrine and the neurotransmitter norepinephrine as described by the GenBank database of the National Center for Biotechnology Information at ncbi.nlm.nih.gov.
  • The GenBank database provides the DNA sequence for ADRB1 as accession no. NM000684, provided in the “Sequence Listing” as SEQ ID NO:3. SEQ ID NO:3 provides the sense strand sequence of DNA that corresponds to the mRNA encoding β1-adrenergic receptor (with the exception of “T” bases for “U” bases). The coding sequence for β1-adrenergic receptor is from nucleotides 87-1520.
  • Equivalents of the above cited ADRB1 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ADRB1 mRNA from another mammalian species that is homologous to SEQ ID NO:3 (i.e., an ortholog). ADRB1 nucleic acid sequences related to SEQ ID NO:3 include those having GenBank accession numbers AF169006, AF169007, AY567837, and J03019.
  • The GenBank database provides the DNA sequence for ADRB2 as accession no. NM000024, provided below as SEQ ID NO:4. SEQ ID NO:4 provides the sense strand sequence of DNA that corresponds to the mRNA encoding β2-adrenergic receptor (with the exception of “T” bases for “U” bases). The coding sequence for β2-adrenergic receptor is from nucleotides 220-1461.
  • Equivalents of the above cited ADRB2 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ADRB2 mRNA from another mammalian species that is homologous to SEQ ID NO:4 (i.e., an ortholog). ADRB2 nucleic acid sequences related to SEQ ID NO:4 include those having GenBank accession numbers AF022953, AF022954, AF022955, AF022956, AF169225, AF202305, AF203386, AY011291, J02960, Y00106, AY136741, BC012481, BC063486, BC073856, M15169, and X04827.
  • Acetylcholinesterase mRNA splice variants E4-E6 and E4-E5 (ACHE): As described by the GenBank database of the National Center for Biotechnology Information at ncbi.nlm.nih.gov, acetylcholinesterase hydrolyzes the neurotransmitter acetylcholine at neuromuscular junctions and brain cholinergic synapses, and thus terminates signal transmission. It is also found on red blood cell membranes, where it constitutes the Yt blood group antigen. Acetylcholinesterase exists in multiple molecular forms which possess similar catalytic properties, but differ in their oligomeric assembly and mode of cell attachment to the cell surface. It is encoded by the single ACHE gene, and the structural diversity in the gene products arises from alternative mRNA splicing, and post-translational associations of catalytic and structural subunits. The major form of acetylcholinesterase found in brain, muscle and other tissues is the hydrophilic species, which forms disulfide-linked oligomers with collagenous, or lipid-containing structural subunits. The other, alternatively spliced form, expressed primarily in the erythroid tissues, differs at the C-terminal end, and contains a cleavable hydrophobic peptide with a GPI-anchor site. It associates with the membranes through the phosphoinositide (PI) moieties added post-translationally. The splice variant E4-E6 is the major transcript and results from the splicing of exon 4 to exon 6. The splice variant E4-E5 results from alternative splicing of exon 4 to exon 5.
  • The GenBank database provides the DNA sequence for ACHE splice variant E4-E5 as accession no. NM015831, provided in the “Sequence Listing” as SEQ ID NO:5. SEQ ID NO:5 provides the sense strand sequence of DNA that corresponds to the mRNA encoding acetylcholinesterase E4-E5 (with the exception of “T” bases for “U” bases). The coding sequence for acetylcholinesterase E4-E5 is from nucleotides 95-1948.
  • Equivalents of the above cited ACHE mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ACHE mRNA from another mammalian species that is homologous to SEQ ID NO:5 (i.e., an ortholog). ACHE nucleic acid sequences related to SEQ ID NO:5 include those having GenBank accession numbers AC011895, AF002993, AF312032, AY750146, CH236956, L06484, L42812, S71129, AF334270, BC026315, BC036813, M55040 and NM000665.
  • The GenBank database provides the DNA sequence for ACHE splice variant E4-E6 as accession no. NM000665, provided in the “Sequence Listing” as SEQ ID NO:123. SEQ ID NO:123 provides the sense strand sequence of DNA that corresponds to the mRNA encoding acetylcholinesterase E4-E6 variant (with the exception of “T” bases for “U” bases). The coding sequence for acetylcholinesterase E4-E6 is from nucleotides 95-1939.
  • Equivalents of the above cited ACHE mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ACHE mRNA from another mammalian species that is homologous to SEQ ID NO:123 (i.e., an ortholog). ACHE nucleic acid sequences related to SEQ ID NO:123 include those having GenBank accession numbers NM015831, AC011895, AF002993, AF312032, AY750146, CH236956, L06484, L42812, S71129, AF334270, BC026315, BC036813, and M55040.
  • Na+/K+-ATPase a and β mRNA (ATP1-A1 variant 1, -A1 variant 2, -A2, -A3, -A4 variant 1, -A4 variant 2, -B1 variant 1, -B1 variant 2, -B2, and -B3): As described by the GenBank database, the proteins encoded by these genes belong to the family of P-type cation transport ATPases, and to the subfamily of Na+/K+-ATPases. Na+/K+-ATPase is an integral membrane protein responsible for establishing and maintaining the electrochemical gradients of Na and K ions across the plasma membrane. These gradients are essential for osmoregulation, for sodium-coupled transport of a variety of organic and inorganic molecules, and for electrical excitability of nerve and muscle. This enzyme is composed of two subunits, a large catalytic subunit (a or A) and a smaller glycoprotein subunit (β or B). The catalytic subunit of Na+/K+-ATPase is encoded by multiple genes.
  • The GenBank database provides the DNA sequence for ATP1A1 variant 1 as accession no. NM000701, provided in the “Sequence Listing” as SEQ ID NO:124. SEQ ID NO:124 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase subunit A1 variant 1 (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase subunit A1 variant 1 is from nucleotides 299-3370.
  • Equivalents of the above cited ATP1A1 variant 1 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1A1 variant 1 mRNA from another mammalian species that is homologous to SEQ ID NO:124 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1A1 variant 2 as accession no. NM001001586, provided in the “Sequence Listing” as SEQ ID NO:125. SEQ ID NO:125 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase subunit A1 variant 2 (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase subunit A1 variant 2 is from nucleotides 299-2344.
  • Equivalents of the above cited ATP1A1 variant 2 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1A1 variant 2 mRNA from another mammalian species that is homologous to SEQ ID NO:125 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1A2 as accession no. NM000702, provided in the “Sequence Listing” as SEQ ID NO:6. SEQ ID NO:6 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase A2 subunit (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase A2 subunit is from nucleotides 105-3167.
  • Equivalents of the above cited ATP1A2 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1A2 mRNA from another mammalian species that is homologous to SEQ ID NO:6 (i.e., an ortholog). ATP1A2 nucleic acid sequences related to SEQ ID NO:6 include those having GenBank accession numbers J05096, M27578, AB018321, AK091617, AK124581, AK126573, AL831991, AL831997, BC013680, BC047533, BC052271, M16795, and Y07494.
  • The GenBank database provides the DNA sequence for ATP1A3 as accession no. NM152296, provided in the “Sequence Listing” as SEQ ID NO:126. SEQ ID NO:126 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase A3 subunit (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase A3 subunit is from nucleotides 155-3196.
  • Equivalents of the above cited ATP1A3 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1A3 mRNA from another mammalian species that is homologous to SEQ ID NO:126 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1A4 variant 1 as accession no. NM144699, provided in the “Sequence Listing” as SEQ ID NO:127. SEQ ID NO:127 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase A4 subunit variant 1 (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase A4 subunit variant 1 is from nucleotides 469-3558.
  • Equivalents of the above cited ATP1A4 variant 1 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1A4 variant 1 mRNA from another mammalian species that is homologous to SEQ ID NO:127 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1A4 variant 2 as accession no. NM001001734, provided in the “Sequence Listing” as SEQ ID NO:128. SEQ ID NO:128 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase A4 subunit variant 2 (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase A4 subunit variant 2 is from nucleotides 111-608.
  • Equivalents of the above cited ATP1A4 variant 2 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1A4 variant 2 mRNA from another mammalian species that is homologous to SEQ ID NO:128 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1B1 variant 1 as accession no. NM001677, provided in the “Sequence Listing” as SEQ ID NO:129. SEQ ID NO:129 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase B1 subunit variant 1 (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase B1 subunit variant 1 is from nucleotides 122-1033.
  • Equivalents of the above cited ATP1B1 variant 1 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1B1 variant 1 mRNA from another mammalian species that is homologous to SEQ ID NO:129 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1B1 variant 2 as accession no. NM001001787, provided in the “Sequence Listing” as SEQ ID NO:130. SEQ ID NO:130 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase B1 subunit variant 2 (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase B1 subunit variant 2 is from nucleotides 122-1027.
  • Equivalents of the above cited ATP1B1 variant 2 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1B1 variant 2 mRNA from another mammalian species that is homologous to SEQ ID NO:130 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1B2 as accession no. NM001678, provided in the “Sequence Listing” as SEQ ID NO:131. SEQ ID NO:131 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase B2 subunit (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase B2 subunit is from nucleotides 584-1456.
  • Equivalents of the above cited ATP1B2 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1B2 mRNA from another mammalian species that is homologous to SEQ ID NO:131 (i.e., an ortholog).
  • The GenBank database provides the DNA sequence for ATP1B3 as accession no. NM001679, provided in the “Sequence Listing” as SEQ ID NO:132. SEQ ID NO:132 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na+/K+-ATPase B3 subunit (with the exception of “T” bases for “U” bases). The coding sequence for Na+/K+-ATPase B3 subunit is from nucleotides 175-1014.
  • Equivalents of the above cited ATP1B3 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is an ATP1B3 mRNA from another mammalian species that is homologous to SEQ ID NO:132 (i.e., an ortholog).
  • Na—K-2Cl cotransporter mRNA (SLC12A1 and SLC12A2): The sodium-potassium-chloride cotransporter (Na—K-2Cl cotransporter or NKCC) facilitates the coupled cotransport of Na+, K+, and Cl. ions across the plasma membrane. There are two isoforms: NKCC1 and NKCC2. NKCC1 is expressed in most tissues, including the eye. In contrast, NKCC2 is expressed primarily in the kidney, however, there is evidence for lower level expression of this isoform in the eye as well. NKCC1 is encoded by the SLC12A2 gene (solute carrier family 12, member 2) and NKCC2 is encoded by the SLC12A1 gene. Trabecular meshwork cells possess a robust Na—K-2Cl cotransporter. The activity of this cotransporter is modulated by neurotransmitters and hormones such as norepinephrine, which reduces cotransport activity, or vasopressin, which increases cotransport activity.
  • The GenBank database provides the DNA sequence for SLC12A1 as accession no. NM000338, provided in the “Sequence Listing” as SEQ ID NO:7. SEQ ID NO:7 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na—K-2Cl cotransporter NKCC2 (with the exception of “T” bases for “U” bases). The coding sequence for Na—K-2Cl cotransporter NKCC2 is from nucleotides 20-3319.
  • Equivalents of the above cited Na—K-2Cl NKCC2 cotransporter mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is a Na—K-2Cl cotransporter NKCC2 mRNA from another mammalian species that is homologous to SEQ ID NO:7 (i.e., an ortholog). SLC12A1 nucleic acid sequences related to SEQ ID NO:7 include those having GenBank accession numbers AJ005332, AJ005333, AB032525, AB032527, BC040138, BX647067, BX647484, and U58130.
  • The GenBank database provides the DNA sequence for SLC12A2 as accession no. NM001046, provided in the “Sequence Listing” as SEQ ID NO:133. SEQ ID NO:133 provides the sense strand sequence of DNA that corresponds to the mRNA encoding Na—K-2Cl cotransporter NKCC1 (with the exception of “T” bases for “U” bases). The coding sequence for Na—K-2Cl cotransporter NKCC1 is from nucleotides 165-3803.
  • Equivalents of the above cited Na—K-2Cl cotransporter NKCC1 mRNA sequence are alternative splice forms, allelic forms, or a cognate thereof. A cognate is a Na—K-2Cl cotransporter NKCC1 mRNA from another mammalian species that is homologous to SEQ ID NO:133 (i.e., an ortholog).
  • Attenuating expression of an mRNA: The phrase, “attenuating expression of an mRNA,” as used herein, means administering or expressing an amount of interfering RNA (e.g., an siRNA) to reduce translation of the target mRNA into protein, either through mRNA cleavage or through direct inhibition of translation. The reduction in expression of the target mRNA or the corresponding protein is commonly referred to as “knock-down” and is reported relative to levels present following administration or expression of a non-targeting control RNA (e.g., a non-targeting control siRNA). Knock-down of expression of an amount including and between 50% and 100% is contemplated by embodiments herein. However, it is not necessary that such knock-down levels be achieved for purposes of the present invention. In one embodiment, a single interfering RNA targeting one of the ocular hypertension targets is administered to lower IOP. In other embodiments, two or more interfering RNAs targeting the same ocular hypertension target (e.g., CA2) are administered to lower IOP. In still other embodiments, two or more interfering RNAs targeting multiple hypertension targets (e.g., CA2 and ADRB2) are administered to lower IOP.
  • Knock-down is commonly assessed by measuring the mRNA levels using quantitative polymerase chain reaction (qPCR) amplification or by measuring protein levels by western blot or enzyme-linked immunosorbent assay (ELISA). Analyzing the protein level provides an assessment of both mRNA cleavage as well as translation inhibition. Further techniques for measuring knock-down include RNA solution hybridization, nuclease protection, northern hybridization, gene expression monitoring with a microarray, antibody binding, radioimmunoassay, and fluorescence activated cell analysis.
  • Inhibition of targets cited herein is also inferred in a human or mammal by observing an improvement in a glaucoma symptom such as improvement in intraocular pressure, improvement in visual field loss, or improvement in optic nerve head changes, for example.
  • Interfering RNA of embodiments of the invention appear to act in a catalytic manner for cleavage of target mRNA, i.e., interfering RNA is able to effect inhibition of target mRNA in substoichiometric amounts. As compared to antisense therapies, significantly less interfering RNA is required to provide a therapeutic effect under such cleavage conditions.
  • Interfering RNA: In one embodiment of the invention, interfering RNA (e.g., siRNA) has a sense strand and an antisense strand, and the sense and antisense strands comprise a region of at least near-perfect contiguous complementarity of at least 19 nucleotides. In a further embodiment of the invention, the interfering RNA comprises a region of at least 13, 14, 15, 16, 17, or 18 contiguous nucleotides having percentages of sequence complementarity to or, having percentages of sequence identity with, the penultimate 13, 14, 15, 16, 17, or 18 nucleotides, respectively, of the 3′ end of the corresponding target sequence within an mRNA.
  • The length of each strand of the interfering RNA comprises 19 to 49 nucleotides, and may comprise a length of 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or 49 nucleotides.
  • The antisense strand of an siRNA is the active guiding agent of the siRNA in that the antisense strand is incorporated into RISC, thus allowing RISC to identify target mRNAs with at least partial complementarity to the antisense siRNA strand for cleavage or translational repression.
  • In the present invention, interfering RNA target sequences (e.g., siRNA target sequences) within a target mRNA sequence are selected using available design tools. Interfering RNAs corresponding to these target sequences are then tested by transfection of cells expressing the target mRNA followed by assessment of knockdown as described above. Interfering RNAs that produce a knockdown in expression of between 50% and 100% are selected for further analysis.
  • Techniques for selecting target sequences for siRNAs are provided by Tuschl, T. et al., “The siRNA User Guide,” revised May 6, 2004, available on the Rockefeller University web site; by Technical Bulletin #506, “siRNA Design Guidelines,” Ambion Inc. at Ambion's web site; and by other web-based design tools at, for example, the Invitrogen, Dharmacon, Integrated DNA Technologies, Genscript, or Proligo web sites. Initial search parameters can include G/C contents between 35% and 55% and siRNA lengths between 19 and 27 nucleotides. The target sequence may be located in the coding region or in the 5′ or 3′ untranslated regions of the mRNA.
  • An embodiment of a 19-nucleotide DNA target sequence for carbonic anyhdrase II is present at nucleotides 232 to 250 of SEQ ID NO:1:
  • 5′-CCCTGAGGATCCTCAACAA-3′ SEQ ID NO:8.
  • An siRNA of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:8 and having 21-nucleotide strands and a 2-nucleotide 3′ overhang is:
    5′-CCCUGAGGAUCCUCAACAANN-3′ SEQ ID NO:9
    3′-NNGGGACUCCUAGGAGUUGUU-5′. SEQ ID NO:10

    Each “N” residue can be any nucleotide (A, C, G, U, T) or modified nucleotide. The 3′ end can have a number of “N” residues between and including 1, 2, 3, 4, 5, and 6. The “N” residues on either strand can be the same residue (e.g., UU, AA, CC, GG, or TT) or they can be different (e.g., AC, AG, AU, CA, CG, CU, GA, GC, GU, UA, UC, or UG). The 3′ overhangs can be the same or they can be different. In one embodiment, both strands have a 3′UU overhang.
  • An siRNA of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:8 and having 21-nucleotide strands and a 3′UU overhang on each strand is:
    5′-CCCUGAGGAUCCUCAACAAUU-3′ SEQ ID NO:11
    3′-UUGGGACUCCUAGGAGUUGUU-5′. SEQ ID NO:12
  • The interfering RNA may also have a 5′ overhang of nucleotides or it may have blunt ends. An siRNA of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:8 and having 19-nucleotide strands and blunt ends is:
    5′-CCCUGAGGAUCCUCAACAA-3′ SEQ ID NO:722
    3′-GGGACUCCUAGGAGUUGUU-5′. SEQ ID NO:723
  • The strands of a double-stranded interfering RNA (e.g., an siRNA) may be connected to form a hairpin or stem-loop structure (e.g., an shRNA). An shRNA of the invention targeting a corresponding mRNA sequence of SEQ ID NO:8 and having a 19 bp double-stranded stem region and a 3′UU overhang is:
                             NNN
                            /   \
      5′-CCCUGAGGAUCCUCAACAA     N
    3′-UUGGGACUCCUAGGAGUUGUU     N SEQ ID NO:13
                            \   \
                             NNN.
  • N is a nucleotide A, T, C, G, U, or a modified form known by one of ordinary skill in the art. The number of nucleotides N in the loop is a number between and including 3 to 23, or 5 to 15, or 7 to 13, or 4 to 9, or 9 to 11, or the number of nucleotides N is 9. Some of the nucleotides in the loop can be involved in base-pair interactions with other nucleotides in the loop. Examples of oligonucleotide sequences that can be used to form the loop include 5′-UUCAAGAGA-3′ (Brummelkamp, T. R. et al. (2002) Science 296: 550) and 5′-UUUGUGUAG-3′ (Castanotto, D. et al. (2002) RNA 8:1454). It will be recognized by one of skill in the art that the resulting single chain oligonucleotide forms a stem-loop or hairpin structure comprising a double-stranded region capable of interacting with the RNAi machinery.
  • The siRNA target sequence identified above can be extended at the 3′ end to facilitate the design of dicer-substrate 27-mer duplexes. Extension of the 19-nucleotide DNA target sequence (SEQ ID NO:8) identified in the carbonic anhydrase II DNA sequence (SEQ ID NO:1) by 6 nucleotides yields a 25-nucleotide DNA target sequence present at nucleotides 232 to 256 of SEQ ID NO:1:
  • 5′-CCCTGAGGATCCTCAACAATGGTCA-3′ SEQ ID NO:724.
  • A dicer-substrate 27-mer duplex of the invention for targeting a corresponding mRNA sequence of SEQ ID NO:724 is:
    5′-CCCUGAGGAUCCUCAACAAUGGUCA-3′ SEQ ID NO:718
    3′-UUGGGACUCCUAGGAGUUGUUACCAGU-5′. SEQ ID NO:719

    The two nucleotides at the 3′ end of the sense strand (i.e., the CA nucleotides of SEQ ID NO:718) may be deoxynucleotides for enhanced processing. Design of dicer-substrate 27-mer duplexes from 19-21 nucleotide target sequences, such as provided herein, is further discussed by the Integrated DNA Technologies (IDT) website and by Kim, D.-H. et al., (February, 2005) Nature Biotechnology 23:2; 222-226.
  • When interfering RNAs are produced by chemical synthesis, phosphorylation at the 5′ position of the nucleotide at the 5′ end of one or both strands (when present) can enhance siRNA efficacy and specificity of the bound RISC complex but is not required since phosphorylation can occur intracellularly.
  • Table 1 lists examples of siRNA target sequences within the CA2, CA4, and CA12 variant 1 and variant 2 DNA sequences (SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:101, and SEQ ID NO:134, respectively) from which siRNAs of the present invention are designed in a manner as set forth above. CA2, CA4, and CA12 variant 1 and variant 2 encode carbonic anhydrase II, IV, and XII variant 1 and 2, respectively.
    TABLE 1
    CA2, CA4, and CA12 Target Sequences for siRNAs
    # of Starting
    Nucleotide with SEQ
    reference to ID
    CA2 Target Sequence SEQ ID NO:1 NO:
    CCCTGAGGATCCTCAACAA 232 8
    GGGCCTTCAGAAAGTTGTT 527 14
    GCGAGCAGGTGTTGAAATT 721 15
    GGTGTTGAAATTCCGTAAA 728 16
    GCCACTGAAGAACAGGCAA 809 17
    CCACTGAAGAACAGGCAAA 810 18
    CCCATAGTCTGTATCCAAA 855 19
    CCATAGTCTGTATCCAAAT 856 20
    GGTGATTTGGACCCTGGTT 921 21
    GGGTGATGAGCACTCACAA 1139 22
    GAAGGTTGGCAGCGCTAAA 506 83
    ATGTGCTGGATTCCATTAA 547 84
    TGTGCTGGATTCCATTAAA 548 85
    CCGTAAACTTAACTTCAAT 740 86
    GATCTACCTTGGTGATTTG 911 87
    GACCAATTGTCATGCTTGA 1009 88
    GGTGATGAGCACTCACAAT 1140 89
    CACTCACAATTGTTGACTA 1149 90
    ACTCACAATTGTTGACTAA 1150 91
    CTCACAATTGTTGACTAAA 1151 92
    AGGAAAGTAGAATGGTTGA 1188 93
    GTAGAATGGTTGAGTGCAA 1194 94
    TAGAATGGTTGAGTGCAAA 1195 95
    CAAGATAAATTGAGCTAGT 1223 96
    AGTTAAGGCAAATCAGGTA 1239 97
    GAGTTGTGATACAGAGTAT 1456 98
    AGTTGTGATACAGAGTATA 1457 99
    GTTGTGATACAGAGTATAT 1458 100
    GACCTGAGCACTGGCATAA 100 135
    TGACATCGACACTCATACA 158 136
    ACACTCATACAGCCAAGTA 166 137
    ACAATGGTCATGCTTTCAA 247 138
    AGGACAAAGCAGTGCTCAA 286 139
    GATGGCACTTACAGATTGA 318 140
    GCACTTACAGATTGATTCA 322 141
    ACAGATTGATTCAGTTTCA 328 142
    ACAAGGTTCAGAGCATACT 371 143
    CAGAACTTCACTTGGTTCA 412 144
    ACTGGCCGTTCTAGGTATT 482 145
    TTGAAGGTTGGCAGCGCTA 504 146
    TGAAGGTTGGCAGCGCTAA 505 147
    TTGTTGATGTGCTGGATTC 541 148
    GAAATTCCGTAAACTTAAC 734 149
    CCGAAGAACTGATGGTGGA 772 150
    GAACTGATGGTGGACAACT 777 151
    TGAAGAACAGGCAAATCAA 814 152
    CTTACTTGATAGACTTACT 972 153
    TGTGAAGACTAGACCAATT 998 154
    TTGAGCTAGTTAAGGCAAA 1232 155
    GGATGGCACTTACAGATTG 317 720
    GAAATATGCTGCAGAACTT 401 721
    # of Starting
    Nucleotide with SEQ
    reference to ID
    CA4 Target Sequence SEQ ID NO:2 NO:
    TCGTCACCACCAAGGCAAA 213 23
    GCTTCTTCTTCTCTGGCTA 252 24
    TCTTCTCTGGCTACGATAA 258 25
    GGCTACGATAAGAAGCAAA 266 26
    GGTCCGACTTGCCATATAA 399 27
    GGAGATGCACATAGTACAT 457 28
    GCACATAGTACATGAGAAA 463 29
    GACATCGAGGAATGTGAAA 490 30
    GGTGGAGGCACTGTCTAAT 595 31
    GGGACTTTAGGCATGATTA 1064 32
    ACACTGGTGCTACGAGGTT 109 156
    CTGGTGCTACGAGGTTCAA 112 157
    GTTCAAGCCGAGTCCTCCA 125 158
    TTCAAGCCGAGTCCTCCAA 126 159
    CCTGCTTGGTGCCAGTCAA 150 160
    TCTCTGGCTACGATAAGAA 261 161
    TGGCTACGATAAGAAGCAA 265 162
    GCAAACGTGGACTGTCCAA 280 163
    TGGTCCGACTTGCCATATA 398 164
    CCATGGAGATGCACATAGT 453 165
    AGATGCACATAGTACATGA 459 166
    TGCACATAGTACATGAGAA 462 167
    ATAGTACATGAGAAAGAGA 467 168
    CATCGAGGAATGTGAAAGA 492 169
    TTGCGGTGCTGGCCTTTCT 534 170
    GAACAGATCCTGGCATTCT 785 171
    TCTCTCAGAAGCTGTACTA 801 172
    AGGAACAGACAGTGAGCAT 825 173
    GAACAGACAGTGAGCATGA 827 174
    GGCAGCGCACGGTGATAAA 876 175
    CAGCCTCTCTGTTGCCTCA 1003 176
    TGTTGCCTCAGCTCTCCAA 1012 177
    # of Starting
    CA12, variant 1 Nucleotide with SEQ
    and 2 Common reference to ID
    Target Sequences SEQ ID NO:101 NO:
    TCCTGCTGGTGATCTTAAA 191 102
    ACGGTTCCAAGTGGACTTA 239 103
    GAGAATAGCTGGTCCAAGA 274 104
    AGAATAGCTGGTCCAAGAA 275 105
    GTGACATCCTCCAGTATGA 341 106
    GCTACAATCTGTCTGCCAA 389 107
    CAGTTTCTCCTGACCAACA 412 108
    AGTTTCTCCTGACCAACAA 413 109
    GACCAACAATGGCCATTCA 423 110
    CTCCTTCAATCCGTCCTAT 687 111
    CCTTCAATCCGTCCTATGA 689 112
    ATCCGTCCTATGACAAGAT 695 113
    AGATCTTCAGTCACCTTCA 710 114
    CGGAGAGGACCGCTGAATA 791 115
    GGAGAGGACCGCTGAATAT 792 116
    AGAGGACCGCTGAATATTA 794 117
    AGGTCCAGAAGTTCGATGA 983 118
    GTTCGATGAGAGGCTGGTA 993 119
    TTCGATGAGAGGCTGGTAT 994 120
    TCGATGAGAGGCTGGTATA 995 121
    TTCAATCCGTCCTATGACA 691 178
    # of Starting
    CA12, Nucleotide with SEQ
    variant 1 Target reference to ID
    Sequence SEQ ID NO:101 NO:
    TGTACTGCGGCAGGACTGA 1039 122
    AGAGCGTGCTTTCAAGTGT 1568 179
    GATGTCAAATCGTGGTTTA 2326 180
    AAATCGTGGTTTAGATCAA 2332 181
    ATGGAATGCTACTAAGATA 2425 182
    CTACTAAGATACTCCATAT 2433 183
    ACAACGATGGCAAGCCTTA 2844 184
    CAACGATGGCAAGCCTTAT 2845 185
    TTGCTAGGCAAAGTTACAA 2880 186
    TAGGCAAAGTTACAAGTGA 2884 187
    AGTTACAAGTGACCTAATG 2891 188
    TGTGCACTCAAGACCTCTA 2954 189
    GTGCACTCAAGACCTCTAA 2955 190
    TGCACTCAAGACCTCTAAC 2956 191
    GCACTCAAGACCTCTAACA 2957 192
    AGACCTCTAACAGCCTCGA 2964 193
    GACCTCTAACAGCCTCGAA 2965 194
    TGCCATTAGCATGCCTCAT 3006 195
    GCCATTAGCATGCCTCATG 3007 196
    TAGCATGCCTCATGCATCA 3012 197
    CATCATCAGATGACAAGGA 3026 198
    # of Starting
    CA12, Nucleotide with SEQ
    variant 2 Target reference to ID
    Sequence SEQ ID NO:134 NO:
    CTCCTTCAATCCGTCCTAT 687 199
    AGAGCGTGCTTTCAAGTGT 1535 200
    GATGTCAAATCGTGGTTTA 2293 201
    AAATCGTGGTTTAGATCAA 2299 202
    ATGGAATGCTACTAAGATA 2392 203
    CTACTAAGATACTCCATAT 2400 204
    ACAACGATGGCAAGCCTTA 2811 205
    CAACGATGGCAAGCCTTAT 2812 206
    TTGCTAGGCAAAGTTACAA 2847 207
    TAGGCAAAGTTACAAGTGA 2851 208
    AGTTACAAGTGACCTAATG 2858 209
    TGTGCACTCAAGACCTCTA 2921 210
    GTGCACTCAAGACCTCTAA 2922 211
    TGCACTCAAGACCTCTAAC 2923 212
    GCACTCAAGACCTCTAACA 2924 213
    AGACCTCTAACAGCCTCGA 2931 214
    GACCTCTAACAGCCTCGAA 2932 215
    TGCCATTAGCATGCCTCAT 2973 216
    GCCATTAGCATGCCTCATG 2974 217
    TAGCATGCCTCATGCATCA 2979 218
    CATCATCAGATGACAAGGA 2993 219
  • Table 2 lists examples of siRNA target sequences within the ADRB1 and ADRB2 DNA sequences (SEQ ID NO:3 and SEQ ID NO:4, respectively) from which siRNAs of the present invention are designed in a manner as set forth above. As noted above, ADRB1 and ADRB2 encode the β1- and β2-adrenergic receptors, respectively.
    TABLE 2
    ADRE1 and ADRE2 Target Sequences for siRNAs
    # of Starting
    Nucleotide with SEQ
    reference to ID
    ADRE1 Target Sequence SEQ ID NO:3 NO:
    TCCTTCTTCTGCGAGCTGT 468 33
    TCGAGACCCTGTGTGTCAT 523 34
    GCATCATGGCCTTCGTGTA 799 35
    GAACGAGGAGATCTGTGTT 1563 36
    ACGAGGAGATCTGTGTTTA 1565 37
    GGAGATCTGTGTTTACTTA 1569 38
    GATAGCAGGTGAACTCGAA 1593 39
    CCCACAATCCTCGTCTGAA 1613 40
    CCACAATCCTCGTCTGAAT 1614 41
    TCTGAATCATCCGAGGCAA 1626 42
    GCAATGTGCTGGTGATCGT 310 220
    TGATCGTGGCCATCGCCAA 322 221
    AAGTGCTGCGACTTCGTCA 726 222
    CGTCCGTAGTCTCCTTCTA 769 223
    CCGTAGTCTCCTTCTACGT 772 224
    ATCATGGCCTTCGTGTACC 801 225
    TCATGGCCTTCGTGTACCT 802 226
    CCTCGGAATCCAAGGTGTA 1501 227
    TGTGTTTACTTAAGACCGA 1576 228
    GTGTTTACTTAAGACCGAT 1577 229
    GTTTACTTAAGACCGATAG 1579 230
    TTTACTTAAGACCGATAGC 1580 231
    TTACTTAAGACCGATAGCA 1581 232
    TAAGACCGATAGCAGGTGA 1586 233
    ACCGATAGCAGGTGAACTC 1590 234
    CGATAGCAGGTGAACTCGA 1592 235
    ATAGCAGGTGAACTCGAAG 1594 236
    CACAATCCTCGTCTGAATC 1615 237
    ACAATCCTCGTCTGAATCA 1616 238
    TCATCCGAGGCAAAGAGAA 1632 239
    CATCCGAGGCAAAGAGAAA 1633 240
    CCACGGACCGTTGCACAAA 1654 241
    # of Starting
    Nucleotide with SEQ
    reference to ID
    ADRE2 Target Sequence SEQ ID NO:4 NO:
    GCATCGTCATGTCTCTCAT 329 43
    GCTGGTCATCACAGCCATT 375 44
    CCCTCAAGACGTTAGGCAT 1031 45
    GCATCATCATGGGCACTTT 1046 46
    CCTAAATTGGATAGGCTAT 1149 47
    GCTATGTCAATTCTGGTTT 1163 48
    GGAAGACTTTGTGGGCCAT 1371 49
    GCCTAGCGATAACATTGAT 1401 50
    GGGAGGAATTGTAGTACAA 1426 51
    GCTGTGAACATGGACTCTT 1880 52
    CACGACGTCACGCAGCAAA 283 242
    GATCGCTACTTTGCCATTA 607 243
    ATCGCTACTTTGCCATTAC 608 244
    TCGCTACTTTGCCATTACT 609 245
    GCCATTACTTCACCTTTCA 619 246
    TTACTTCACCTTTCAAGTA 623 247
    CCATTCAGATGCACTGGTA 722 248
    TGATCATGGTCTTCGTCTA 857 249
    AGACGTTAGGCATCATCAT 1037 250
    TCGTTAACATTGTGCATGT 1091 251
    AGGATAACCTCATCCGTAA 1115 252
    TCATCCGTAAGGAAGTTTA 1124 253
    AAGTTTACATCCTCCTAAA 1136 254
    AGTTTACATCCTCCTAAAT 1137 255
    TAAATTGGATAGGCTATGT 1151 256
    CTATGTCAATTCTGGTTTC 1164 257
    GGTACTGTGCCTAGCGATA 1393 258
    GTACTGTGCCTAGCGATAA 1394 259
    TACTGTGCCTAGCGATAAC 1395 260
    GCGATAACATTGATTCACA 1406 261
    CGATAACATTGATTCACAA 1407 262
    GGAGGAATTGTAGTACAAA 1427 263
    GAGGAATTGTAGTACAAAT 1428 264
    AGGAATTGTAGTACAAATG 1429 265
    CAAATGACTCACTGCTGTA 1442 266
    GACCTGAGTCTGCTATATT 1725 267
    ACCTGAGTCTGCTATATTT 1726 268
    CCATGTATCTACCTCACTA 1756 269
    CATGTATCTACCTCACTAT 1757 270
    ATGTATCTACCTCACTATT 1758 271
    CCTCACTATTCAAGTATTA 1767 272
    TAATATATTGCTGCTGGTA 1790 273
    AATATATTGCTGCTGGTAA 1791 274
    ATATATTGCTGCTGGTAAT 1792 275
    TATATTGCTGCTGGTAATT 1793 276
    CTGGTAATTTGTATCTGAA 1803 277
    GAGTATCTCGGACCTTTCA 1861 278
    CGGACCTTTCAGCTGTGAA 1869 279
    CGAGCAAAGGTCTAAAGTT 1971 280
    GAGCAAAGGTCTAAAGTTT 1972 281
    GGTCTAAAGTTTACAGTAA 1979 282
  • Table 3 lists examples of siRNA target sequences within the ACHE DNA sequences for splice variants E4-E5 and E4-E6 (SEQ ID NO:5 and SEQ ID NO:123, respectively) from which siRNAs of the present invention are designed in a manner as set forth above. As noted above, ACHE encodes acetylcholinesterase.
    TABLE 3
    ACHE Target Sequences for siRNAs
    # of Starting
    Nucleotide with SEQ
    ACHE E4-E5 reference to ID
    Target Sequence SEQ ID NO:5 NO:
    CCAGAGTGTCTGCTACCAA 382 53
    GCTACCAATATGTGGACAC 393 54
    CCAATATGTGGACACCCTA 397 55
    GCTGGTGTCCATGAACTAC 622 56
    TCATCAACGCGGGAGACTT 1131 57
    GGTCTACGCCTACGTCTTT 1459 58
    GCTACGAGATCGAGTTCAT 1530 59
    GCTATAACGGTCAACCATT 2251 60
    GGCTGCAAATAAACTGTTA 2885 61
    GCTGCAAATAAACTGTTAC 2886 62
    AGTGTCTGCTACCAATATG 386 283
    AGACAACGAGTCTCTCATC 1231 284
    GGCTGTGGTCCTGCATTAC 1315 285
    CTTCCTCCTCAAACCGAGA 2047 286
    TCCTCCTCAAACCGAGAGA 2049 287
    CCTCAAACCGAGAGACTCA 2053 288
    TCAAACCGAGAGACTCACA 2055 289
    AAACCGAGAGACTCACACT 2057 290
    CCACGCCTTTGTTGTTTGA 2125 291
    CACGCCTTTGTTGTTTGAA 2126 292
    ACGCCTTTGTTGTTTGAAT 2127 293
    GGCTATAACGGTCAACCAT 2250 294
    TATAACGGTCAACCATTTC 2253 295
    CGGTCAACCATTTCTGTCT 2258 296
    GTCAACCATTTCTGTCTCT 2260 297
    CCGTCTTCCGGTCATTCTT 2318 298
    CCTCTCGTCTTTCGCACAT 2395 299
    TCTCGTCTTTCGCACATTC 2397 300
    TTTCGCACATTCTCCTGAT 2404 301
    TTCGCACATTCTCCTGATC 2405 302
    AGAACCAGTTCGACCACTA 2643 303
    AACCAGTTCGACCACTACA 2645 304
    CTGCAAATAAACTGTTACA 2887 305
    # of Starting
    ACHE E4-E5 and E4-E6 Nucleotide with SEQ
    Target Sequences reference to ID
    in Common SEQ ID NO:5 NO:
    TAGACGCTACAACCTTCCA 366 306
    CGCTACAACCTTCCAGAGT 370 307
    AGAGTGTCTGCTACCAATA 384 308
    GAGTGTCTGCTACCAATAT 385 309
    CTGTCCTCGTCTGGATCTA 525 310
    ATGGCCGCTTCTTGGTACA 588 311
    CGACATCAGTGACGCTGTT 768 312
    GCACGTGCTGCCTCAAGAA 1045 313
    CACGTGCTGCCTCAAGAAA 1046 314
    GAAAGCGTCTTCCGGTTCT 1061 315
    TGTGGTAGATGGAGACTTC 1090 316
    GACAACGAGTCTCTCATCA 1232 317
    AGGCTGTGGTCCTGCATTA 1314 318
    GCTGTGGTCCTGCATTACA 1316 319
    GTCTACGCCTACGTCTTTG 1460 320
    TCTACGCCTACGTCTTTGA 1461 321
    CTACGCCTACGTCTTTGAA 1462 322
    CGGCTACGAGATCGAGTTC 1528 323
    CAGCGACTGATGCGATACT 1607 324
    GGCTCAGCAGTACGTTAGT 1705 325
    AGTACGTTAGTCTGGACCT 1713 326
    # of Starting
    Nucleotide with SEQ
    ACHE E4-E6 Target reference to ID
    Sequence SEQ ID NO:123 NO:
    ACATGGTGCACTGGAAGAA 1875 327
    AGAACCAGTTCGACCACTA 1890 328
    GAACCAGTTCGACCACTAC 1891 329
    GGCTATAACACAGACGAGC 2011 330
    GCTATAACACAGACGAGCC 2012 331
    GCTGCAAATAAACTGTTAC 2133 332
    CTGCAAATAAACTGTTACA 2134 333
  • Table 4 lists examples of siRNA target sequences within the Na+/K+-ATPase A and B subunit DNA sequences (ATP1A1 variant 1, SEQ ID NO:124; ATP1A1 variant 2, SEQ ID NO:125; ATP1A2, SEQ ID NO:6; ATP1A3, SEQ ID NO:126; ATP1A4 variant 1, SEQ ID NO:127; ATP1A4 variant 2, SEQ ID NO:128; ATP1B1 variant 1, SEQ ID NO:129; ATP1B1 variant 2, SEQ ID NO:130; ATP1B2, SEQ ID NO:131; and ATP1B3, SEQ ID NO:132) from which siRNAs of the present invention are designed in a manner as set forth above.
    TABLE 4
    ATP1A and ATP1B Target Sequences for siRNAs
    # of Starting
    Nucleotide with SEQ
    ATP1A1 variant 1 reference to ID
    Target Sequence SEQ ID NO:124 NO:
    GCAATGAGACCGTGGAAGA 2208 334
    TGCCAAGQCCTGCGTAGTA 2275 335
    TAAAGGACATGACCTCCGA 2307 336
    AGCAAGCTGCTGACATGAT 2526 337
    ACATGATTCTTCTGGATGA 2538 338
    GTCGTCTGATCTTTGATAA 2592 339
    CTTATACCTTAACCAGTAA 2628 340
    GGATCAACGATGTGGAAGA 2979 341
    ACGATGTGGAAGACAGCTA 2985 342
    CCGACTTGGTCATCTGTAA 3093 343
    TAGGAAAGCACCGCAGCAT 3474 344
    AGACGTCCTGGAATGAAGC 3504 345
    GACGTCCTGGAATGAAGCA 3505 346
    ACGTCCTGGAATGAAGCAT 3506 347
    GAAGCATGTAGCTCTATGG 3518 348
    # of Starting
    ATP1A1 variant 1 Nucleotide with SEQ
    and variant 2 Common reference to ID
    Target Sequences SEQ ID NO:124 NO:
    TTCAGAACAAGGTGATAAA 343 349
    TGATGAACTTCATCGTAAA 442 350
    GGTGCTATCAGCCGTTGTA 700 351
    TCAGCCGTTGTAATCATAA 707 352
    GATTCGAAATGGTGAGAAA 811 353
    CAGAATCATATCTGCAAAT 907 354
    CACGTGGTATTGTTGTCTA 1059 355
    CTGCTTAGTGAAGAACTTA 1363 356
    GTTTCAGGCTAACCAGGAA 1594 357
    CACTCTTAAAGTGCATAGA 1662 358
    AGTACCAGTTGTCTATTCA 1758 359
    TACCAGTTGTCTATTCATA 1760 360
    AGCTGAAAGACGCCTTTCA 1896 361
    TCGATAATCTGTGCTTTGT 2037 362
    ACAGGAGACCATCCAATCA 2147 363
    # of Starting
    Nucleotide with SEQ
    ATP1A1 variant 2 reference to ID
    Target Sequence SEQ ID NO:125 NO:
    TAGCCTTGATGAACTTCAT 436 364
    TTGATGAACTTCATCGTAA 441 365
    GATGAACTTCATCGTAAAT 443 366
    CTACTCCTGAATGGATCAA 552 367
    GGAGCGATTCTTTGTTTCT 617 368
    GTGCTATCAGCCGTTGTAA 701 369
    TGCTATCAGCCGTTGTAAT 702 370
    GAGCATAAATGCGGAGGAA 832 371
    GAAGGCAATGGACCTATGA 2204 372
    CCGACTTGGTCATCTGTAA 2291 373
    TATATGACGAAGTCAGAAA 2495 374
    # of Starting
    Nucleotide with SEQ
    ATP1A2 reference to ID
    Target Sequence SEQ ID NO:6 NO:
    CCATCCAACGACAATCTAT 471 63
    GCATCATATCAGAGGGTAA 1990 64
    CCTCCTCATCTTCATCTAT 3080 65
    GGAAGTGAGGTAGTGCCAA 3797 66
    GGATGTCACTCATGTACTT 4037 67
    GCTCCATGCTGTTCTGAAA 4093 68
    GCTGGCCATTGGCTAGAAT 4225 69
    GGTCAGAACCTTTGGACAA 4323 70
    GCTAGAGGTGGCATGTTTA 5213 71
    GCGAGTGCATGGGCTAATT 5285 72
    TGGCAATGGATGACCACAA 214 375
    TGAACCATCCAACGACAAT 467 376
    ACCATCCAACGACAATCTA 470 377
    CATCCAACGACAATCTATA 472 378
    ATCCAACGACAATCTATAT 473 379
    GCAGATCAACGCAGAGGAA 632 380
    TGTTTCTTCTCCACCAACT 825 381
    CCATAGCAATGGAGATTGA 946 382
    AGATGCAAGATGCCTTTCA 1693 383
    CTGAATCTGCCATCTGGAA 1767 384
    TGAATCTGCCATCTGGAAA 1768 385
    ATCGTCTTTGCTCGAACGT 2157 386
    CTGCATTGAAGAAGGCTGA 2263 387
    ATGAAGCGGCAGCCACGAA 2589 388
    TGAAGCGGCAGCCACGAAA 2590 389
    GGATGACCGGACCATGAAT 2765 390
    GCTGCCTTTCTCTCTTACT 2988 391
    TCTATGATGAGGTCCGAAA 3094 392
    GTGGAGAAGGAGACATACT 3144 393
    TGGAGAAGGAGACATACTA 3145 394
    TAGACCTAACTGTGAACAA 3344 395
    AGACCTAACTGTGAACAAT 3345 396
    TCCACTATGTTGTCTATTT 3418 397
    TGAGTGCAAGAGCCTGAGA 3666 398
    TGACATGAGTCTCCAGATA 3828 399
    GTCGTGGACTCCAGCTCTA 3850 400
    TGTCACTCATGTACTTAAT 4040 401
    GTCACTCATGTACTTAATA 4041 402
    CACTTCACCTTCTGTAATA 4061 403
    GTAGAGAGAGACCTAGATA 4882 404
    CTAGATAGGTCATGCAAGT 4894 405
    AGGTCATGCAAGTGAGAAA 4900 406
    TATCAGAAGCAAGGAAGTA 5040 407
    TCCGATTAATTGGAGATTA 5114 408
    CCGATTAATTGGAGATTAC 5115 409
    GATTACTAACTGTGGACAA 5128 410
    ATTACTAACTGTGGACAAA 5129 411
    TCAGGCACTTTAGAAATAT 5253 412
    GGCTAATTATCATCAATCT 5296 413
    AGTTTGAGGTACTACCTAT 5375 414
    TACTACCTATGTACTTGAA 5384 415
    ACTACCTATGTACTTGAAA 5385 416
    # of Starting
    Nucleotide with SEQ
    ATP1A3 reference to ID
    Target Sequence SEQ ID NO:126 NO:
    TGGCTATGACAGAGCACAA 240 417
    GAGGTCTGCCGGAAATACA 272 418
    CTCACGCCACCGCCTACCA 362 419
    TCGACTGTGATGACGTGAA 1836 420
    TGAACTTCACCACGGACAA 1851 421
    CCAAGGCCTGCGTGATCCA 2103 422
    GGACTTCACCTCCGAGCAA 2137 423
    GACTTCACCTCCGAGCAAA 2138 424
    ACTTCACCTCCGAGCAAAT 2139 425
    TCGACGAGATCCTGCAGAA 2157 426
    CGACGAGATCCTGCAGAAT 2158 427
    ACGAGATCCTGCAGAATCA 2160 428
    GATCTTCGACAACCTAAAG 2425 429
    CCATCTCACTGGCGTACGA 2580 430
    CTGCCGAAAGCGACATCAT 2601 431
    CGGACAAATTGGTCAATGA 2646 432
    CAAATTGGTCAATGAGAGA 2650 433
    GGATGACCGCACCGTCAAT 2794 434
    CACCGTCAATGACCTGGAA 2803 435
    ATCTTCGTCTACGACGAAA 3116 436
    CTACGACGAAATCCGCAAA 3124 437
    ACGACGAAATCCGCAAACT 3126 438
    ACGAAATCCGCAAACTCAT 3129 439
    CCAAACCTCTCTCCTCTCT 3377 440
    # of Starting
    Nucleotide with SEQ
    ATP1A4 variant 1 reference to ID
    Target Sequence SEQ ID NO:127 NO:
    GGCACCTGGTTACGCTTCA 113 441
    CATGGATGATCACAAATTA 612 442
    AATCCTGACTCGAGATGGA 702 443
    CCTACAGCATCCAGATATA 833 444
    CCGGCTTATCTCTGCACAA 1101 445
    AGCTCTGATACCTGGTTTA 1732 446
    GCTCTGATACCTGGTTTAT 1733 447
    AGGTGATGCTTCCGAGTCA 1836 448
    GTACTCAATGAACGATGAA 2070 449
    TACTCAATGAACGATGAAA 2071 450
    GTGCTAGGCTTCTGCTTCT 2143 451
    CATGGTAACAGGAGATCAT 2328 452
    TGTGGTGCATGGTGCAGAA 2475 453
    TGTTCATCATCCTCGGTAT 2861 454
    GTTCATCATCCTCGGTATA 2862 455
    GGCTTATGAGTCAGCTGAA 2952 456
    GGACCTATGAGCAACGAAA 3203 457
    CGGATCTCATCATCTCCAA 3281 458
    TGGCTGCATTTCTGTCCTA 3377 459
    GCTGCATTTCTGTCCTACA 3379 460
    GTATTCTCATCTTCGTCTA 3470 461
    TATTCTCATCTTCGTCTAT 3471 462
    ACTAAACTCAGCAGATGAA 3554 463
    GGCCAGAGATTATAAGTTT 3614 464
    GCCAGAGATTATAAGTTTG 3615 465
    CCAGAGATTATAAGTTTGA 3616 466
    CAGAGATTATAAGTTTGAC 3617 467
    ATAAGTTTGACACAACATC 3625 468
    TAAGTTTGACACAACATCT 3626 469
    TCTGAGACACTAGGATGAA 3642 470
    AGACACTAGGATGAATTAT 3646 471
    GACACTAGGATGAATTATC 3647 472
    AGGATGAATTATCTTGGAT 3653 473
    GATGAATTATCTTGGATGA 3655 474
    CGTAGCCAGTCTAGACAGT 3797 475
    GCCAGTCTAGACAGTAAAT 3801 476
    CAGTCTAGACAGTAAATGT 3803 477
    AGACAGTAAATGTCTGGAA 3809 478
    GACAGTAAATGTCTGGAAA 3810 479
    # of Starting
    Nucleotide with SEQ
    ATP1A4 variant 2 reference to ID
    Target Sequence SEQ ID NO:128 NO:
    GCTGGATTCTTTACCTACT 126 480
    GTGGACCTATGAGCAACGA 251 481
    TGGACCTATGAGCAACGAA 252 482
    GGACCTATGAGCAACGAAA 253 483
    CGGATCTCATCATCTCCAA 331 484
    TGGCTGCATTTCTGTCCTA 427 485
    GCTGCATTTCTGTCCTACA 429 486
    GTATTCTCATCTTCGTCTA 520 487
    TATTCTCATCTTCGTCTAT 521 488
    CTTCGTCTATGATGAAATC 530 489
    ACTACTAAACTCAGCAGAT 601 490
    CTACTAAACTCAGCAGATG 602 491
    TACTAAACTCAGCAGATGA 603 492
    ACTAAACTCAGCAGATGAA 604 493
    GGCCAGAGATTATAAGTTT 664 494
    GCCAGAGATTATAAGTTTG 665 495
    CCAGAGATTATAAGTTTGA 666 496
    CAGAGATTATAAGTTTGAC 667 497
    ATAAGTTTGACACAACATC 675 498
    TAAGTTTGACACAACATCT 676 499
    TCTGAGACACTAGGATGAA 692 500
    AGACACTAGGATGAATTAT 696 501
    GACACTAGGATGAATTATC 697 502
    TAGGATGAATTATCTTGGA 702 503
    AGGATGAATTATCTTGGAT 703 504
    GATGAATTATCTTGGATGA 705 505
    TGAATTATCTTGGATGAGA 707 506
    CGTAGCCAGTCTAGACAGT 847 507
    GCCAGTCTAGACAGTAAAT 851 508
    CAGTCTAGACAGTAAATGT 853 509
    AGACAGTAAATGTCTGGAA 859 510
    GACAGTAAATGTCTGGAAA 860 511
    # of Starting
    Nucleotide with SEQ
    ATP1B1 variant 1 reference to ID
    Target Sequence SEQ ID NO:129 NO:
    ACCTACTAGTCTTGAACAA 1096 512
    TACTAGTCTTGAACAAACT 1099 513
    GGACCTACACTTAATCTAT 1130 514
    GACCTACACTTAATCTATA 1131 515
    CTGCATTTAATAGGTTAGA 1167 516
    CGTAACTGACTTGTAGTAA 1299 517
    AGCAAGGTTTGCTGTCCAA 1441 518
    TGCTGTCCAAGGTGTAAAT 1450 519
    GCTGTCCAAGGTGTAAATA 1451 520
    CTGTCCAAGGTGTAAATAT 1452 521
    TTAACATACTCCATAGTCT 1564 522
    GCCTTGTCCTCCGGTATGT 1746 523
    TGTCCTCCGGTATGTTCTA 1750 524
    GTCCTCCGGTATGTTCTAA 1751 525
    TCCTCCGGTATGTTCTAAA 1752 526
    CCATCACTTTGGCTAGTGA 1795 527
    # of Starting
    ATP1B1 variant 1 and Nucleotide with SEQ
    variant 2 Common reference to ID
    Target Sequences SEQ ID NO:129 NO:
    ACCGGTGGCAGTTGGTTTA 203 528
    CCGGTGGCAGTTGGTTTAA 204 529
    TTGGTTTAAGATCCTTCTA 214 530
    AGATCCTTCTATTCTACGT 222 531
    ATCCTTCTATTCTACGTAA 224 532
    TCCTTCTATTCTACGTAAT 225 533
    CCTTCTATTCTACGTAATA 226 534
    GAAATTTCCTTTCGTCCTA 380 535
    AACGAGGAGACTTTAATCA 525 536
    GAAATTGCTCTGGATTAAA 591 537
    ATGAAACTTATGGCTACAA 612 538
    TGAAACTTATGGCTACAAA 613 539
    AAACTTATGGCTACAAAGA 615 540
    GGCAAACCGTGCATTATTA 635 541
    GCAAACCGTGCATTATTAT 636 542
    ACCGAGTTCTAGGCTTCAA 663 543
    CCGAGTTCTAGGCTTCAAA 664 544
    TTCTAGGCTTCAAACCTAA 669 545
    ATGAGTCCTTGGAGACTTA 699 546
    GCAAGCGAGATGAAGATAA 765 547
    AGTTGGAAATGTGGAGTAT 790 548
    CTGCAGTATTATCCGTACT 839 549
    TGCAGTATTATCCGTACTA 840 550
    GCAGTATTATCCGTACTAT 841 551
    CCGTACAGTTCACCAATCT 900 552
    TCACCAATCTTACCATGGA 909 553
    AAATTCGCATAGAGTGTAA 933 554
    TGTAAGGCGTACGGTGAGA 947 555
    # of Starting
    Nucleotide with SEQ
    ATP1B1 variant 2 reference to ID
    Target Sequence SEQ ID NO:130 NO:
    TGTGTTATGCTTGTATTGA 1063 556
    GCCTTGTCCTCCGGTATGT 1102 557
    TGTCCTCCGGTATGTTCTA 1106 558
    GTCCTCCGGTATGTTCTAA 1107 559
    TCCTCCGGTATGTTCTAAA 1108 560
    CCTCCGGTATGTTCTAAAG 1109 561
    TCCGGTATGTTCTAAAGCT 1111 562
    CCATCACTTTGGCTAGTGA 1151 563
    # of Starting
    Nucleotide with SEQ
    ATP1B2 reference to ID
    Target Sequence SEQ ID NO:131 NO:
    CCGAGGACGCACCAGTTTA 653 564
    CGAGGACGCACCAGTTTAT 654 565
    TGCAGACTGTCTCCGACCA 771 566
    CAGACTGTCTCCGACCATA 773 567
    CAAGACTGAGAACCTTGAT 841 568
    AGAACCTTGATGTCATTGT 849 569
    CCTTGATGTCATTGTCAAT 853 570
    AAGTTCTTGGAGCCTTACA 917 571
    AGTTCTTGGAGCCTTACAA 918 572
    GAGCCTTACAACGACTCTA 926 573
    AGCCTTACAACGACTCTAT 927 574
    TTACAACGACTCTATCCAA 931 575
    GCTATTACGAACAGCCAGA 981 576
    TATTACGAACAGCCAGATA 983 577
    ATTACGAACAGCCAGATAA 984 578
    CAGATAATGGAGTCCTCAA 996 579
    GATAATGGAGTCCTCAACT 998 580
    AAACGTGCCTGCCAATTCA 1022 581
    AACGTGCCTGCCAATTCAA 1023 582
    AACCAGAGCATGAATGTTA 1160 583
    CTCGGCAACTTCGTCATGT 1214 584
    AATGTAGAATGTCGCATCA 1355 585
    ATGTAGAATGTCGCATCAA 1356 586
    CAACATCGCCACAGACGAT 1381 587
    GACGATGAGCGAGACAAGT 1394 588
    TGGCCTTCAAACTCCGCAT 1425 589
    CCATCTCTCTCCTGTGGAT 1474 590
    TTTGATAACAGAGCTATGA 1550 591
    CCATTGCGGTTCCGTCACT 1620 592
    AGGAGTTAGGAGCCTTTCT 1707 593
    TGTGAGAGCTATCCACTCT 1740 594
    CACTCTCCTGCCTGCATAT 1753 595
    CGCCACACACACACACAAA 1825 596
    TCTACACAGTCGCCATCTT 1956 597
    TCGCCATCTTGGTGACTTT 1965 598
    GGTTGACCTAGGCTGAATA 2598 599
    GTTGACCTAGGCTGAATAT 2599 600
    GGCTGAATATCCACTTTGT 2608 601
    AGCAAGTTATCAACTAATC 2828 602
    GCAAGTTATCAACTAATCA 2829 603
    CCAAATCTAGCCTCTGAAT 2888 604
    CTCCTGCTCTGAATATTCT 3012 605
    TGTGTCAGATCTACTGTAA 3251 606
    # of Starting
    Nucleotide with SEQ
    ATP1B3 reference to ID
    Target Sequence SEQ ID NO:132 NO:
    TTGCTCTTCTACCTAGTTT 292 607
    CAGTGACCGCATTGGAATA 434 608
    GACCGCATTGGAATATACA 438 609
    TTCAGTAGGTCTGATCCAA 457 610
    CAGTAGGTCTGATCCAACT 459 611
    GGTACATTGAAGACCTTAA 488 612
    TACATTGAAGACCTTAAGA 490 613
    AGACCTTAAGAAGTTTCTA 498 614
    GACCTTAAGAAGTTTCTAA 499 615
    GTTTATGTTGCATGTCAGT 592 616
    TGGTATGAATGATCCTGAT 639 617
    TGAAGGAGTGCCAAGGATA 723 618
    TGTAGCAGTTTATCCTCAT 774 619
    GTAGCAGTTTATCCTCATA 775 620
    CTCATAATGGAATGATAGA 788 621
    AGCCATTGGTTGCTGTTCA 857 622
    GCCATTGGTTGCTGTTCAG 858 623
    GTAACAGTTGAGTGCAAGA 910 624
    TAACAGTTGAGTGCAAGAT 911 625
    TGATGGATCAGCCAACCTA 930 626
    GATGGATCAGCCAACCTAA 931 627
    ATGGATCAGCCAACCTAAA 932 628
    GCATAGTATGAGTAGGATA 1009 629
    CATAGTATGAGTAGGATAT 1010 630
    GGATATCTCCACAGAGTAA 1023 631
    GATATCTCCACAGAGTAAA 1024 632
    AGAAAGGTGTGTGGTACAT 1111 633
    ATAACGTGCTTCCAGATCA 1146 634
    TAACGTGCTTCCAGATCAT 1147 635
    AGTGTACAGTCGCCAGATA 1220 636
    GTGAACACCTGATTCCAAA 1246 637
    AGCTTAATATGCCGTGCTA 1321 638
    TAATATGCCGTGCTATGTA 1325 639
    AATATGCCGTGCTATGTAA 1326 640
    ATATGCCGTGCTATGTAAA 1327 641
    GCCGTGCTATGTAAATATT 1331 642
    TGCAAGAAATGTGGTATGT 1437 643
    ATGCTGAATTAGCCTCGAT 1548 644
    TTGATTAAGAGCACAAACT 1571 645
    AGCAGACTGTGGACTGTAA 1785 646
    GCAGACTGTGGACTGTAAT 1786 647
    CAGACTGTGGACTGTAATA 1787 648
  • Table 5 lists examples of siRNA target sequences within the SLC12A1 and SLC12A2 DNA sequences (SEQ ID NO:7 and SEQ ID NO:133, respectively) from which siRNAs of the present invention are designed in a manner as set forth above. As noted above, SLC12A1 and SLC12A2 encode the Na—K-2Cl cotransporter, NKCC2 and NKCC1, respectively.
    TABLE 5
    SLC12A1 Target Sequences for siRNAs
    # of Starting
    Nucleotide with SEQ
    SLC12A1 reference to ID
    Target Sequence SEQ ID NO:7 NO:
    CCACCATAGTAACGACAAT 675 73
    GGAATGGAATGGGAGGCAA 974 74
    GGGATGAACTGCAATGGTT 1373 75
    CCATGCCTCTTATGCCAAA 1780 76
    CCTGCTCTCCTGGACATAA 2102 77
    GCATCTGCTGTGAAGTCTT 2151 78
    GCCTCAGGCTTAGGAAGAA 2315 79
    GGAAGCGACTATCAAAGAT 2542 80
    GCTGGCAAGTTGAACATTA 2609 81
    GCAAGAAAGGGATCCATAT 3197 82
    TAATACCAATCGCTTTCAA 67 649
    ACCAATCGCTTTCAAGTTA 71 650
    CAATCGCTTTCAAGTTAGT 73 651
    ATAGAGTACTATCGTAACA 353 652
    CCAGCCTGCTTGAGATTCA 405 653
    CTGTAGTAGATCTACTTAA 864 654
    ACCAATGACATCCGGATTA 911 655
    CCAATGACATCCGGATTAT 912 656
    CAATGACATCCGGATTATA 913 657
    GGCTATGACTTCTCAAGAT 1409 658
    GCCTCATATGCACTTATTA 1748 659
    AGACCTGCGTATGGAATTT 1811 660
    ACGTCTATGTGACTTGTAA 1935 661
    GTCTATGTGACTTGTAAGA 1937 662
    TTCCTACGTGAGTGCTTTA 1993 663
    GACAATGCTCTGGAATTAA 2012 664
    CTCTGGTGATTGGATATAA 2346 665
    TGACAGAGATTGAGAACTA 2388 666
    TGAGATTGGCGTGGTTATA 2437 667
    GCATCCGAGGCTTGTTTAA 2586 668
    ACCATATCGTCTCCATGAA 3007 669
    CCATATCGTCTCCATGAAA 3008 670
    TGAAAGCTGCAAAGATTTA 3022 671
    TCGACTGAATGAACTCTTA 3130 672
    CCATATCGGATTTGTTGTA 3210 673
    GGTTGGAAATCCTCACAAA 3237 674
    CTTACTAGTTAGAGGAAAT 3271 675
    # of Starting
    Nucleotide with SEQ
    SLC12A2 reference to ID
    Target Sequence SEQ ID NO:133 NO:
    ACCACCAGCACTACTATTA 748 676 /
    CCACCAGCACTACTATTAT 749 677
    CAGCACTACTATTATGATA 753 678
    CTATCAGTCCTTGTAATAA 1119 679
    ATTGTCTACTTCAGCAATA 1169 680
    TATTGGTGATTTCGTCATA 1499 681
    TTCGTCATAGGAACATTTA 1509 682
    TAATGACACTATCGTAACA 1820 683
    GATGTTTQCTAAAGGTTAT 2081 684
    CTTCGTGGCTACATCTTAA 2118 685
    TGCACTTGGATTCATCTTA 2147 686
    GATGATCTGTGGCCATGTA 2615 687
    CTCGAAGACAAGCCATGAA 2644 688
    TGAAAGAGATGTCCATCGA 2659 689
    AGAGATGTCCATCGATCAA 2663 690
    CCATCGATCAAGCCAAATA 2671 691
    CATCGATCAAGCCAAATAT 2672 692
    GGTCGTATGAAGCCAAACA 2793 693
    CACTTGTCCTTGGATTTAA 2812 694
    TAGTGGTTATTCGCCTAAA 2914 695
    ATCTCATCTTCAAGGACAA 2948 696
    CGATTTAGATACTTCCAAA 3044 697
    TCATTGGTGGAAAGATAAA 3334 698
    TTAGCAAGTTCCGGATAGA 3391 699
    GAAATCATTGAGCCATACA 3480 700
    AGCAAGATATTGCAGATAA 3520 701
    GATGAACCATGGCGAATAA 3549 702
    CATTCAAGCACAGCTAATA 3639 703
    TTCAGTGCCTAGTGTAGTA 3840 704
    AGGAAAGTTGCTCCATTGA 3941 705
    AAAGTTGCTCCATTGATAA 3944 706
    CAATCTTAATGGTGATTCT 4001 707
    TTGACATCATAGTCTAGTA 4995 708
    GACATCATAGTCTAGTAAA 4997 709
    GTGTGTGTGTGTGTATATA 5141 710
    GTGTGTGTGTGTATATATA 5143 711
    TAGGCAAACTTTGGTTTAA 5249 712
    GGAGAATACTTCGCCTAAA 5375 713
    TGAGTATGACCTAGGTATA 5834 714
    AGAGATCTGATAACTTGAA 5852 715
    GGTAAAGACAGTAGAAATA 5981 716
    TTTAAGCTCTGGTGGATGA 6678 717

    As cited in the examples above, one of skill in the art is able to use the target sequence information provided in Tables 1-5 to design interfering RNAs having a length shorter or longer than the sequences provided in Table 1-5 by referring to the sequence position in SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, and adding or deleting nucleotides complementary or near complementary to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, respectively.
  • The target RNA cleavage reaction guided by siRNAs and other forms of interfering RNA is highly sequence specific. In general, siRNA containing a sense nucleotide strand identical in sequence to a portion of the target mRNA and an antisense nucleotide strand exactly complementary to a portion of the target mRNA are siRNA embodiments for inhibition of mRNAs cited herein. However, 100% sequence complementarity between the antisense siRNA strand and the target mRNA, or between the antisense siRNA strand and the sense siRNA strand, is not required to practice the present invention. Thus, for example, the invention allows for sequence variations that might be expected due to genetic mutation, strain polymorphism, or evolutionary divergence.
  • In one embodiment of the invention, the antisense strand of the siRNA has at least near-perfect contiguous complementarity of at least 19 nucleotides with the target mRNA. “Near-perfect,” as used herein, means the antisense strand of the siRNA is “substantially complementary to,” and the sense strand of the siRNA is “substantially identical” to at least a portion of the target mRNA. “Identity,” as known by one of ordinary skill in the art, is the degree of sequence relatedness between nucleotide sequences as determined by matching the order and identity of nucleotides between the sequences. In one embodiment, the antisense strand of an siRNA having 80% and between 80% up to 100% complementarity, for example, 85%, 90% or 95% complementarity, to the target mRNA sequence are considered near-perfect complementarity and may be used in the present invention. “Perfect” contiguous complementarity is standard Watson-Crick base pairing of adjacent base pairs. “At least near-perfect” contiguous complementarity includes “perfect” complementarity as used herein. Computer methods for determining identity or complementarity are designed to identify the greatest degree of matching of nucleotide sequences, for example, BLASTN (Altschul, S. F., et al. (1990) J. Mol. Biol. 215:403-410).
  • The term “percent identity” describes the percentage of contiguous nucleotides in a first nucleic acid molecule that is the same as in a set of contiguous nucleotides of the same length in a second nucleic acid molecule. The term “percent complementarity” describes the percentage of contiguous nucleotides in a first nucleic acid molecule that can base pair in the Watson-Crick sense with a set of contiguous nucleotides in a second nucleic acid molecule.
  • The relationship between a target mRNA (sense strand) and one strand of an siRNA (the sense strand) is that of identity. The sense strand of an siRNA is also called a passenger strand, if present. The relationship between a target mRNA (sense strand) and the other strand of an siRNA (the antisense strand) is that of complementarity. The antisense strand of an siRNA is also called a guide strand.
  • The penultimate base in a nucleic acid sequence that is written in a 5′ to 3′ direction is the next to the last base, i.e., the base next to the 3′ base. The penultimate 13 bases of a nucleic acid sequence written in a 5′ to 3′ direction are the last 13 bases of a sequence next to the 3′ base and not including the 3′ base. Similarly, the penultimate 14, 15, 16, 17, or 18 bases of a nucleic acid sequence written in a 5′ to 3′ direction are the last 14, 15, 16, 17, or 18 bases of a sequence, respectively, next to the 3′ base and not including the 3′ base.
  • The phrase “a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of (a sequence identifier)” allows a one nucleotide substitution. Two nucleotide substitutions (i.e., 11/13=85% identity/complementarity) are not included in such a phrase.
  • In one embodiment of the invention, the region of contiguous nucleotides is a region of at least 14 contiguous nucleotides having at least 85% sequence complementarity to, or at least 85% sequence identity with, the penultimate 14 nucleotides of the 3′ end of the sequence identified by each sequence identifier. Two nucleotide substitutions (i.e., 12/14=86% identity/complementarity) are included in such a phrase.
  • In a further embodiment of the invention, the region of contiguous nucleotides is a region of at least 15, 16, 17, or 18 contiguous nucleotides having at least 80% sequence complementarity to, or at least 80% sequence identity with, the penultimate 14 nucleotides of the 3′ end of the sequence of the sequence identifier. Three nucleotide substitutions are included in such a phrase.
  • The target sequence in the mRNAs corresponding to SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, may be in the 5′ or 3′ untranslated regions of the mRNA as well as in the coding region of the mRNA.
  • One or both of the strands of double-stranded interfering RNA may have a 3′ overhang of from 1 to 6 nucleotides, which may be ribonucleotides or deoxyribonucleotides or a mixture thereof. The nucleotides of the overhang are not base-paired. In one embodiment of the invention, the interfering RNA comprises a 3′ overhang of TT or UU. In another embodiment of the invention, the interfering RNA comprises at least one blunt end. The termini usually have a 5′ phosphate group or a 3′ hydroxyl group. In other embodiments, the antisense strand has a 5′ phosphate group, and the sense strand has a 5′ hydroxyl group. In still other embodiments, the termini are further modified by covalent addition of other molecules or functional groups.
  • The sense and antisense strands of the double-stranded siRNA may be in a duplex formation of two single strands as described above or may be a single molecule where the regions of complementarity are base-paired and are covalently linked by a hairpin loop so as to form a single strand. It is believed that the hairpin is cleaved intracellularly by a protein termed dicer to form an interfering RNA of two individual base-paired RNA molecules.
  • Interfering RNAs may differ from naturally-occurring RNA by the addition, deletion, substitution or modification of one or more nucleotides. Non-nucleotide material may be bound to the interfering RNA, either at the 5′ end, the 3′ end, or internally. Such modifications are commonly designed to increase the nuclease resistance of the interfering RNAs, to improve cellular uptake, to enhance cellular targeting, to assist in tracing the interfering RNA, to further improve stability, or to reduce the potential for activation of the interferon pathway. For example, interfering RNAs may comprise a purine nucleotide at the ends of overhangs. Conjugation of cholesterol to the 3′ end of the sense strand of an siRNA molecule by means of a pyrrolidine linker, for example, also provides stability to an siRNA.
  • Further modifications include a 3′ terminal biotin molecule, a peptide known to have cell-penetrating properties, a nanoparticle, a peptidomimetic, a fluorescent dye, or a dendrimer, for example.
  • Nucleotides may be modified on their base portion, on their sugar portion, or on the phosphate portion of the molecule and function in embodiments of the present invention. Modifications include substitutions with alkyl, alkoxy, amino, deaza, halo, hydroxyl, thiol groups, or a combination thereof, for example. Nucleotides may be substituted with analogs with greater stability such as replacing a ribonucleotide with a deoxyribonucleotide, or having sugar modifications such as 2′ OH groups replaced by 2′ amino groups, 2′ O-methyl groups, 2′ methoxyethyl groups, or a 2′-O, 4′-C methylene bridge, for example. Examples of a purine or pyrimidine analog of nucleotides include a xanthine, a hypoxanthine, an azapurine, a methylthioadenine, 7-deaza-adenosine and O- and N-modified nucleotides. The phosphate group of the nucleotide may be modified by substituting one or more of the oxygens of the phosphate group with nitrogen or with sulfur (phosphorothioates). Modifications are useful, for example, to enhance function, to improve stability or permeability, or to direct localization or targeting.
  • There may be a region or regions of the antisense interfering RNA strand that is (are) not complementary to a portion of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134. Non-complementary regions may be at the 3′, 5′ or both ends of a complementary region or between two complementary regions.
  • Interfering RNAs may be generated exogenously by chemical synthesis, by in vitro transcription, or by cleavage of longer double-stranded RNA with dicer or another appropriate nuclease with similar activity. Chemically synthesized interfering RNAs, produced from protected ribonucleoside phosphoramidites using a conventional DNA/RNA synthesizer, may be obtained from commercial suppliers such as Ambion Inc. (Austin, Tex.), Invitrogen (Carlsbad, Calif.), or Dharmacon (Lafayette, Colo.). Interfering RNAs are purified by extraction with a solvent or resin, precipitation, electrophoresis, chromatography, or a combination thereof, for example. Alternatively, interfering RNA may be used with little if any purification to avoid losses due to sample processing.
  • Interfering RNAs can also be expressed endogenously from plasmid or viral expression vectors or from minimal expression cassettes, for example, PCR generated fragments comprising one or more promoters and an appropriate template or templates for the interfering RNA. Examples of commercially available plasmid-based expression vectors for shRNA include members of the pSilencer series (Ambion, Austin, Tex.) and pCpG-siRNA (InvivoGen, San Diego, Calif.). Viral vectors for expression of interfering RNA may be derived from a variety of viruses including adenovirus, adeno-associated virus, lentivirus (e.g., HIV, FIV, and EIAV), and herpes virus. Examples of commercially available viral vectors for shRNA expression include pSilencer adeno (Ambion, Austin, Tex.) and pLenti6/BLOCK-iT™-DEST (Invitrogen, Carlsbad, Calif.). Selection of viral vectors, methods for expressing the interfering RNA from the vector and methods of delivering the viral vector are within the ordinary skill of one in the art. Examples of kits for production of PCR-generated shRNA expression cassettes include Silencer Express (Ambion, Austin, Tex.) and siXpress (Mirus, Madison, Wis.).
  • Interfering RNAs may be expressed from a variety of eukaryotic promoters known to those of ordinary skill in the art, including pol III promoters, such as the U6 or HI promoters, or pol II promoters, such as the cytomegalovirus promoter. Those of skill in the art will recognize that these promoters can also be adapted to allow inducible expression of the interfering RNA.
  • Hybridization under Physiological Conditions. In certain embodiments of the present invention, an antisense strand of an interfering RNA hybridizes with an mRNA in vivo as part of the RISC complex.
  • “Hybridization” refers to a process in which single-stranded nucleic acids with complementary or near-complementary base sequences interact to form hydrogen-bonded complexes called hybrids. Hybridization reactions are sensitive and selective. In vitro, the specificity of hybridization (i.e., stringency) is controlled by the concentrations of salt or formamide in prehybridization and hybridization solutions, for example, and by the hybridization temperature; such procedures are well known in the art. In particular, stringency is increased by reducing the concentration of salt, increasing the concentration of form amide, or raising the hybridization temperature.
  • For example, high stringency conditions could occur at about 50% formamide at 37° C. to 42° C. Reduced stringency conditions could occur at about 35% to 25% formamide at 30° C. to 35° C. Examples of stringency conditions for hybridization are provided in Sambrook, J., 1989, Molecular Cloning. A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. Further examples of stringent hybridization conditions include 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50° C. or 70° C. for 12-16 hours followed by washing, or hybridization at 70° C. in 1×SSC or 50° C. in 1×SSC, 50% formamide followed by washing at 70° C. in 0.3×SSC, or hybridization at 70° C. in 4×SSC or 50° C. in 4×SSC, 50% formamide followed by washing at 67° C. in 1×SSC. The temperature for hybridization is about 5-10° C. less than the melting temperature (Tm) of the hybrid where Tm is determined for hybrids between 19 and 49 base pairs in length using the following calculation: Tm° C.=81.5+16.6(log10[Na+])+0.41 (% G+C)−(600/N) where N is the number of bases in the hybrid, and [Na+] is the concentration of sodium ions in the hybridization buffer.
  • The above-described in vitro hybridization assay provides a method of predicting whether binding between a candidate siRNA and a target will have specificity. However, in the context of the RISC complex, specific cleavage of a target can also occur with an antisense strand that does not demonstrate high stringency for hybridization in vitro.
  • Single-stranded interfering RNA: As cited above, interfering RNAs ultimately function as single strands. Single-stranded (ss) interfering RNA has been found to effect mRNA silencing, albeit less efficiently than double-stranded RNA. Therefore, embodiments of the present invention also provide for administration of a ss interfering RNA that hybridizes under physiological conditions to a portion of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:101, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, SEQ ID NO:133, or SEQ ID NO:134, respectively. The ss interfering RNA has a length of 19 to 49 nucleotides as for the ds interfering RNA cited above. The ss interfering RNA has a 5′ phosphate or is phosphorylated in situ or in vivo at the 5′ position. The term “5′ phosphorylated” is used to describe, for example, polynucleotides or oligonucleotides having a phosphate group attached via ester linkage to the C5 hydroxyl of the sugar (e.g., ribose, deoxyribose, or an analog of same) at the 5′ end of the polynucleotide or oligonucleotide.
  • SS interfering RNAs are synthesized chemically or by in vitro transcription or expressed endogenously from vectors or expression cassettes as for ds interfering RNAs. 5′ Phosphate groups may be added via a kinase, or a 5′ phosphate may be the result of nuclease cleavage of an RNA. Delivery is as for ds interfering RNAs. In one embodiment, ss interfering RNAs having protected ends and nuclease resistant modifications are administered for silencing. SS interfering RNAs may be dried for storage or dissolved in an aqueous solution. The solution may contain buffers or salts to inhibit annealing or for stabilization.
  • Hairpin interfering RNA: A hairpin interfering RNA is a single molecule (e.g., a single oligonucleotide chain) that comprises both the sense and antisense strands of an interfering RNA in a stem-loop or hairpin structure (e.g., a shRNA). For example, shRNAs can be expressed from DNA vectors in which the DNA oligonucleotides encoding a sense interfering RNA strand are linked to the DNA oligonucleotides encoding the reverse complementary antisense interfering RNA strand by a short spacer. If needed for the chosen expression vector, 3′ terminal T's and nucleotides forming restriction sites may be added. The resulting RNA transcript folds back onto itself to form a stem-loop structure.
  • Mode of administration: Interfering RNA may be delivered directly to the eye by ocular tissue injection such as periocular, conjunctival, subtenon, intracameral, intravitreal, intraocular, subretinal, subconjunctival, retrobulbar, or intracanalicular injections; by direct application to the eye using a catheter or other placement device such as a retinal pellet, intraocular insert, suppository or an implant comprising a porous, non-porous, or gelatinous material; by topical ocular drops or ointments; or by a slow release device in the cul-de-sac or implanted adjacent to the sclera (transscleral) or within the eye. Intracameral injection may be through the cornea into the anterior chamber to allow the agent to reach the trabecular meshwork. Intracanalicular injection may be into the venous collector channels draining Schlemm's canal or into Schlemm's canal. Systemic or parenteral administration is contemplated including but not limited to intravenous, subcutaneous, and oral delivery.
  • Subject: A subject in need of treatment for ocular hypertension or at risk for developing ocular hypertension is a human or other mammal having ocular hypertension or at risk of having ocular hypertension associated with undesired or inappropriate expression or activity of targets as cited herein, i.e., carbonic anhydrase II, IV, or XII; β1- or β2-adrenergic receptors; acetylcholinesterase; Na+/K+-ATPase; or Na—K-2Cl cotransporter. Ocular structures associated with such disorders may include the eye, retina, choroid, lens, cornea, trabecular meshwork, iris, optic nerve, optic nerve head, sclera, aqueous chamber, vitreous chamber, or ciliary body, for example. A subject may also be an ocular cell, cell culture, organ or an ex vivo organ or tissue.
  • Formulations and Dosage: Pharmaceutical formulations comprise an interfering RNA, or salt thereof, of the invention up to 99% by weight mixed with a physiologically acceptable ophthalmic carrier medium such as water, buffer, saline, glycine, hyaluronic acid, mannitol, and the like.
  • Interfering RNAs of the present invention are administered as solutions, suspensions, or emulsions. The following are examples of possible formulations embodied by this invention.
    Amount in weight %
    Interfering RNA up to 99; 0.1-99; 0.1-50; 0.5-10.0
    Hydroxypropylmethylcellulose 0.5
    Sodium chloride 0.8
    Benzalkonium Chloride 0.01
    EDTA 0.01
    NaOH/HCl qs pH 7.4
    Purified water (RNase-free) qs 100 mL
    Phosphate Buffered Saline 1.0
    Benzalkonium Chloride 0.01
    Polysorbate 80 0.5
    Purified water (RNase-free) q.s. to 100%
    Monobasic sodium phosphate 0.05
    Dibasic sodium phosphate 0.15
    (anhydrous)
    Sodium chloride 0.75
    Disodium EDTA 0.05
    Cremophor EL 0.1
    Benzalkonium chloride 0.01
    HCl and/or NaOH pH 7.3-7.4
    Purified water (RNase-free) q.s. to 100%
    Phosphate Buffered Saline 1.0
    Hydroxypropyl-β-cyclodextrin 4.0
    Purified water (RNase-free) q.s. to 100%
  • Generally, an effective amount of the interfering RNA of embodiments of the invention results in an extracellular concentration at the surface of the target cell of from 100 pM to 100 nM, or from 1 nM to 50 nM, or from 5 nM to about 10 nM, or to about 25 nM. The dose required to achieve this local concentration will vary depending on a number of factors including the delivery method, the site of delivery, the number of cell layers between the delivery site and the target cell or tissue, whether delivery is local or systemic, etc. The concentration at the delivery site may be considerably higher than it is at the surface of the target cell or tissue. Topical compositions are delivered to the surface of the eye one to four times per day, or on an extended delivery schedule such as daily, weekly, bi-weekly, monthly, or longer, according to the routine discretion of a skilled clinician. The pH of the formulation is about pH 4-9, or pH 4.5 to pH 7.4.
  • Therapeutic treatment of patients with siRNAs directed against the ocular hypertension target mRNAs is expected to be beneficial over small molecule topical ocular drops by increasing the duration of action, thereby allowing less frequent dosing and greater patient compliance.
  • While the precise regimen is left to the discretion of the clinician, interfering RNA may be administered by placing one drop in each eye as directed by the clinician. An effective amount of a formulation may depend on factors such as the age, race, and sex of the subject, the severity of the ocular hypertension, the rate of target gene transcript/protein turnover, the interfering RNA potency, and the interfering RNA stability, for example. In one embodiment, the interfering RNA is delivered topically to the eye and reaches the trabecular meshwork, retina or optic nerve head at a therapeutic dose thereby ameliorating an ocular hypertension-associated disease process.
  • Acceptable carriers: An ophthalmically acceptable carrier refers to those carriers that cause at most, little to no ocular irritation, provide suitable preservation if needed, and deliver one or more interfering RNAs of the present invention in a homogenous dosage. An acceptable carrier for administration of interfering RNA of embodiments of the present invention include the cationic lipid-based transfection reagents TransIT®-TKO (Mirus Corporation, Madison, Wis.), LIPOFECTIN®, Lipofectamine, OLIGOFECTAMINE™ (Invitrogen, Carlsbad, Calif.), or DHARMAFEC™ (Dharmacon, Lafayette, Colo.); polycations such as polyethyleneimine; cationic peptides such as Tat, polyarginine, or Penetratin (Antp peptide); or liposomes. Liposomes are formed from standard vesicle-forming lipids and a sterol, such as cholesterol, and may include a targeting molecule such as a monoclonal antibody having binding affinity for endothelial cell surface antigens, for example. Further, the liposomes may be PEGylated liposomes.
  • The interfering RNAs may be delivered in solution, in suspension, or in bioerodible or non-bioerodible delivery devices. The interfering RNAs can be delivered alone, as components of covalent conjugates, complexed with cationic lipids, cationic peptides, or cationic polymers, or encapsulated in targeted or non-targeted nanoparticles.
  • For ophthalmic delivery, an interfering RNA may be combined with ophthalmologically acceptable preservatives, co-solvents, surfactants, viscosity enhancers, penetration enhancers, buffers, sodium chloride, or water to form an aqueous, sterile ophthalmic suspension or solution. Ophthalmic solution formulations may be prepared by dissolving the interfering RNA in a physiologically acceptable isotonic aqueous buffer. Further, the ophthalmic solution may include an ophthalmologically acceptable surfactant to assist in dissolving the inhibitor. Viscosity building agents, such as hydroxymethyl cellulose, hydroxyethyl cellulose, methylcellulose, polyvinylpyrrolidone, or the like may be added to the compositions of the present invention to improve the retention of the compound.
  • In order to prepare a sterile ophthalmic ointment formulation, the interfering RNA is combined with a preservative in an appropriate vehicle, such as mineral oil, liquid lanolin, or white petrolatum. Sterile ophthalmic gel formulations may be prepared by suspending the interfering RNA in a hydrophilic base prepared from the combination of, for example, CARBOPOL®-940 (BF Goodrich, Charlotte, N.C.), or the like, according to methods known in the art for other ophthalmic formulations. VISCOAT® (Alcon Laboratories, Inc., Fort Worth, Tex.) may be used for intraocular injection, for example. Other compositions of the present invention may contain penetration enhancing agents such as cremephor and TWEEN® 80 (polyoxyethylene sorbitan monolaureate, Sigma Aldrich, St. Louis, Mo.), in the event the interfering RNA is less penetrating in the eye.
  • Kits: Embodiments of the present invention provide a kit that includes reagents for attenuating the expression of an mRNA as cited herein in a cell. The kit contains an siRNA or an shRNA expression vector. For siRNAs and non-viral shRNA expression vectors the kit also may contain a transfection reagent or other suitable delivery vehicle. For viral shRNA expression vectors, the kit may contain the viral vector and/or the necessary components for viral vector production (e.g., a packaging cell line as well as a vector comprising the viral vector template and additional helper vectors for packaging). The kit may also contain positive and negative control siRNAs or shRNA expression vectors (e.g., a non-targeting control siRNA or an siRNA that targets an unrelated mRNA). The kit also may contain reagents for assessing knockdown of the intended target gene (e.g., primers and probes for quantitative PCR to detect the target mRNA and/or antibodies against the corresponding protein for western blots). Alternatively, the kit may comprise an siRNA sequence or an shRNA sequence and the instructions and materials necessary to generate the siRNA by in vitro transcription or to construct an shRNA expression vector.
  • A pharmaceutical combination in kit form is further provided that includes, in packaged combination, a carrier means adapted to receive a container means in close confinement therewith and a first container means including an interfering RNA composition and an ophthalmically acceptable carrier. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Printed instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • The ability of interfering RNA to knock-down the levels of endogenous target gene expression in, for example, human trabecular meshwork (TM) cells is evaluated in vitro as follows. Transformed human TM cells, for example, cell lines designated GTM-3 or HTM-3 (see Pang, I. H. et al., 1994. Curr. Eye Res. 13:51-63), are plated 24 h prior to transfection in standard growth medium (e.g., DMEM supplemented with 10% fetal bovine serum). Transfection is performed using Dharmafect 1 (Dharmacon, Lafayette, Colo.) according to the manufacturer's instructions at interfering RNA concentrations ranging from 0.1 nM-100 nM. Non-targeting control interfering RNA and lamin A/C interfering RNA (Dharmacon) are used as controls. Target mRNA levels are assessed by qPCR 24 h post-transfection using, for example, TAQMAN® forward and reverse primers and a probe set that encompasses the target site (Applied Biosystems, Foster City, Calif.). Target protein levels may be assessed approximately 72 h post-transfection (actual time dependent on protein turnover rate) by western blot, for example. Standard techniques for RNA and/or protein isolation from cultured cells are well-known to those skilled in the art. To reduce the chance of non-specific, off-target effects, the lowest possible concentration of interfering RNA should be used that will produce the desired level of knock-down in target gene expression.
  • The ability of interfering RNAs of the present invention to knock-down levels of CA2 protein expression is further exemplified in Example 1 as follows.
  • EXAMPLE 1 Interfering RNA for Specifically Silencing CA2 in HeLa Cells
  • The present study examines the ability of CA2-interfering RNA to knock down the levels of endogenous CA2 expression in cultured HeLa cells.
  • Transfection of HeLa cells was accomplished using standard in vitro concentrations (100 nM and 1 nM) of CA2 siRNAs, or a non-targeting control siRNA and DharmaFECT™ 1 transfection reagent (Dharmacon, Lafayette, Colo.). All siRNAs were dissolved in 1×siRNA buffer, an aqueous solution of 20 mM KCl, 6 mM HEPES (pH 7.5), 0.2 mM MgCl2. CA2 protein expression and actin protein expression (loading control) was evaluated by western blot analysis 72 hours post-transfection. The CA2 siRNAs are double-stranded interfering RNAs having specificity for the following target sequences: siCA2#1 targets SEQ ID NO:721; siCA2#3 targets SEQ ID NO:15; siCA2#4 targets SEQ ID NO:720; siCA2#5 targets SEQ ID NO:141. Each of the four CA2 siRNAs decreased CA2 expression significantly at both 100 nM and 1 nM relative to a non-targeting control siRNA as shown by the western blot data of FIG. 1. SiCA2#4 targeting SEQ ID NO:720 and siCA2#5 targeting SEQ ID NO:141 appeared to be particularly effective.
  • The references cited herein, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated by reference.
  • Those of skill in the art, in light of the present disclosure, will appreciate that obvious modifications of the embodiments disclosed herein can be made without departing from the spirit and scope of the invention. All of the embodiments disclosed herein can be made and executed without undue experimentation in light of the present disclosure. The full scope of the invention is set out in the disclosure and equivalent embodiments thereof. The specification should not be construed to unduly narrow the full scope of protection to which the present invention is entitled.
  • As used herein and unless otherwise indicated, the terms “a” and “an” are taken to mean “one”, “at least one” or “one or more”.

Claims (102)

1. A method of attenuating expression of an ocular hypertension target mRNA in a subject, the method comprising:
administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:33-SEQ ID NO:82, and SEQ ID NO:220-SEQ ID NO:717.
2. The method of claim 1 wherein the composition is administered via a topical, intravitreal, transcleral, periocular, conjunctival, subtenon, intracameral, subretinal, subconjunctival, retrobulbar, or intracanalicular route.
3. The method of claim 1 wherein the composition is administered via in vivo expression from an expression vector capable of expressing the interfering RNA.
4. The method of claim 1 wherein the interfering RNA is an shRNA.
5. The method of claim 1 wherein the ocular hypertension target mRNA encodes a β-adrenergic receptor, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:33-SEQ ID NO:52, and SEQ ID NO:220-SEQ ID NO:282.
6. The method of claim 1 wherein the ocular hypertension target mRNA encodes acetylcholinesterase, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:53-SEQ ID NO:62 and SEQ ID NO:283-333.
7. The method of claim 1 wherein the ocular hypertension target mRNA encodes ATP1A1, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:334-SEQ ID NO:374.
8. The method of claim 1 wherein the ocular hypertension target mRNA encodes ATP1A2, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:63-SEQ ID NO:72 and SEQ ID NO:375-SEQ ID NO:416.
9. The method of claim 1 wherein the ocular hypertension target mRNA encodes ATP1A3, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:417-SEQ ID NO:440.
10. The method of claim 1 wherein the ocular hypertension target mRNA encodes ATP1A4, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:441-SEQ ID NO:511.
11. The method of claim 1 wherein the ocular hypertension target mRNA encodes ATP1B1, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:512-SEQ ID NO:563.
12. The method of claim 1 wherein the ocular hypertension target mRNA encodes ATP1B2, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:564-SEQ ID NO:606.
13. The method of claim 1 wherein the ocular hypertension target mRNA encodes ATP1B3, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:607-SEQ ID NO:648.
14. The method of claim 1 wherein the ocular hypertension target mRNA encodes SLC12A1, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:73-SEQ ID NO:82 and SEQ ID NO:649-SEQ ID NO:675.
15. The method of claim 1 wherein the ocular hypertension target mRNA encodes SLC12A2, and the interfering RNA comprises:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:676-SEQ ID NO:717.
16. The method of claim 1 wherein the region of contiguous nucleotides is a region of at least 14 contiguous nucleotides having at least 85% sequence complementarity to, or at least 85% sequence identity with, the penultimate 14 nucleotides of the 3′ end of the sequence of the sequence identifier.
17. The method of claim 1 wherein the region of contiguous nucleotides is a region of at least 15, 16, 17, or 18 contiguous nucleotides having at least 80% sequence complementarity to, or at least 80% sequence identity with, the penultimate 15, 16, 17, or 18 nucleotides, respectively, of the 3′ end of the sequence of the sequence identifier.
18. The method of claim 1 wherein the interfering RNA is an mRNA.
19. The method of claim 1 wherein the interfering RNA is an siRNA.
20. A method of treating ocular hypertension in a subject in need thereof, the method comprising:
administering to an eye of the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:33-SEQ ID NO:82, and SEQ ID NO:220-SEQ ID NO:717,
wherein the ocular hypertension is treated thereby.
21. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:33-SEQ ID NO:42, and SEQ ID NO:220-SEQ ID NO:241.
22. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:43-SEQ ID NO:52, and SEQ ID NO:242-SEQ ID NO:282.
23. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:53-SEQ ID NO:62 and SEQ ID NO:283-333.
24. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:334-SEQ ID NO:374.
25. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:63-SEQ ID NO:72 and SEQ ID NO:375-SEQ ID NO:416.
26. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:417-SEQ ID NO:440.
27. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:441-SEQ ID NO:511.
28. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:512-SEQ ID NO:563.
29. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:564-SEQ ID NO:606.
30. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:607-SEQ ID NO:648.
31. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:73-SEQ ID NO:82 and SEQ ID NO:649-SEQ ID NO:675.
32. The method of claim 20 wherein the interfering RNA comprises a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of any one of SEQ ID NO:676-SEQ ID NO:717.
33. The method of claim 20 wherein the interfering RNA is an shRNA.
34. The method of claim 20 wherein the composition is administered via a topical, intravitreal, transcleral, periocular, conjunctival, subtenon, intracameral, subretinal, subconjunctival, retrobulbar, or intracanalicular route.
35. The method of claim 20 wherein the composition is administered via in vivo expression from an expression vector capable of expressing the interfering RNA.
36. The method of claim 20 wherein the interfering RNA is an mRNA.
37. The method of claim 20 wherein the interfering RNA is an siRNA.
38. A method of attenuating expression of an ocular hypertension target mRNA first variant without attenuating expression of an ocular hypertension target mRNA second variant in a subject, comprising:
administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of the first variant,
wherein the expression of the first variant mRNA is attenuated without attenuating expression of the second variant mRNA, and wherein the first variant target mRNA is SEQ ID NO:5, SEQ ID NO:124, SEQ ID NO:127, or SEQ ID NO:129, and the second variant target mRNA is SEQ ID NO:123, SEQ ID NO:125, SEQ ID NO:128, or SEQ ID NO:130, respectively.
39. A method of attenuating expression of an ocular hypertension target mRNA first variant without attenuating expression of an ocular hypertension target mRNA second variant in a subject, comprising:
administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising:
a region of at least 13 contiguous nucleotides having at least 90% sequence complementarity to, or at least 90% sequence identity with, the penultimate 13 nucleotides of the 3′ end of the first variant,
wherein the expression of the first variant mRNA is attenuated without attenuating expression of the second variant mRNA, and wherein the first variant target mRNA is SEQ ID NO:123, SEQ ID NO:125, SEQ ID NO:128, or SEQ ID NO:130, and the second variant target mRNA is SEQ ID NO:5, SEQ ID NO:124, SEQ ID NO:127, or SEQ ID NO:129, respectively.
40. A method of attenuating expression of an ocular hypertension target mRNA of a subject, comprising:
administering to the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising:
a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect contiguous complementarity of at least 19 nucleotides;
wherein the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133, and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133, respectively,
wherein the expression of an ocular hypertension target mRNA is attenuated.
41. The method of claim 40 wherein the subject is a human and the human has ocular hypertension.
42. The method of claim 40 wherein the subject is a human and the human is at risk of developing ocular hypertension.
43. The method of claim 40 wherein the composition is administered via a topical, intravitreal, transcleral, periocular, conjunctival, subtenon, intracameral, subretinal, subconjunctival, retrobulbar, or intracanalicular route.
44. The method of claim 40 wherein the composition is administered via in vivo expression from an expression vector capable of expressing the interfering RNA.
45. The method of claim 40 wherein the ocular hypertension target mRNA encodes a β1- or β2-adrenergic receptor, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4, respectively.
46. The method of claim 40 wherein the ocular hypertension target mRNA encodes an acetylcholinesterase, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123, respectively.
47. The method of claim 40 wherein the ocular hypertension target mRNA encodes a subunit of Na+/K+-ATPase, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132, respectively.
48. The method of claim 40 wherein the ocular hypertension target mRNA encodes a Na—K-2Cl cotransporter, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:7 or SEQ ID NO:133 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:7 or SEQ ID NO:133, respectively.
49. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:3 comprising nucleotide 468, 523, 799, 1563, 1565, 1569, 1593, 1613, 1614, 1626, 310, 322, 726, 769, 772, 801, 802, 1501, 1576, 1577, 1579, 1580, 1581, 1586, 1590, 1592, 1594, 1615, 1616, 1632, 1633, or 1654.
50. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:4 comprising nucleotide 329, 375, 1031, 1046, 1149, 1163, 1371, 1401, 1426, 1880, 283, 607, 608, 609, 619, 623, 722, 857, 1037, 1091, 1115, 1124, 1136, 1137, 1151, 1164, 1393, 1394, 1395, 1406, 1407, 1427, 1428, 1429, 1442, 1725, 1726, 1756, 1757, 1758, 1767, 1790, 1791, 1792, 1793, 1803, 1861, 1869, 1971, 1972, or 1979.
51. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:123 comprising nucleotide 1875, 1890, 1891, 2011, 2012, 2133, or 2134.
52. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:5 comprising nucleotide 366, 370, 384, 385, 525, 588, 768, 1045, 1046, 1061, 1090, 1232, 1314, 1316, 1460, 1461, 1462, 1528, 1607, 1705, 1713, 382, 393, 397, 622, 1131, 1459, 1530, 2251, 2885, 2886, 386, 1231, 1315, 2047, 2049, 2053, 2055, 2057, 2125, 2126, 2127, 2250, 2253, 2258, 2260, 2318, 2395, 2397, 2404, 2405, 2643, 2645, or 2887.
53. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:124 comprising nucleotide 2208, 2275, 2307, 2526, 2538, 2592, 2628, 2979, 2985, 3093, 3474, 3504, 3505, 3506, 3518, 343, 442, 700, 707, 811, 907, 1059, 1363, 1594, 1662, 1758, 1760, 1896, 2037, or 2147.
54. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:125 comprising nucleotide 436, 441, 443, 552, 617, 701, 702, 832, 2204, 2291, or 2495.
55. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:6 comprising nucleotide 471, 1990, 3080, 3797, 4037, 4093, 4225, 4323, 5213, 5285, 214, 467, 470, 472, 473, 632, 825, 946, 1693, 1767, 1768, 2157, 2263, 2589, 2590, 2765, 2988, 3094, 3144, 3145, 3344, 3345, 3418, 3666, 3828, 3850, 4040, 4041, 4061, 4882, 4894, 4900, 5040, 5114, 5115, 5128, 5129, 5253, 5296, 5375, 5384, or 5385.
56. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:126 comprising nucleotide 240, 272, 362, 1836, 1851, 2103, 2137, 2138, 2139, 2157, 2158, 2160, 2425, 2580, 2601, 2646, 2650, 2794, 2803, 3116, 3124, 3126, 3129, or 3377.
57. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:127 comprising nucleotide 113, 612, 702, 833, 1101, 1732, 1733, 1836, 2070, 2071, 2143, 2328, 2475, 2861, 2862, 2952, 3203, 3281, 3377, 3379, 3470, 3471, 3554, 3614, 3615, 3616, 3617, 3625, 3626, 3642, 3646, 3647, 3653, 3655, 3797, 3801, 3803, 3809 or 3810.
58. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:128 comprising nucleotide 126, 251, 252, 253, 331, 427, 429, 520, 521, 530, 601, 602, 603, 604, 664, 665, 666, 667, 675, 676, 692, 696, 697, 702, 703, 705, 707, 847, 851, 853, 859, or 860.
59. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:129 comprising nucleotide 1096, 1099, 1130, 1131, 1167, 1299, 1441, 1450, 1451, 1452, 1564, 1746, 1750, 1751, 1752, 1795, 203, 204, 214, 222, 224, 225, 226, 380, 525, 591, 612, 613, 615, 635, 636, 663, 664, 669, 699, 765, 790, 839, 840, 841, 900, 909, 933, or 947.
60. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:130 comprising nucleotide 1063, 1102, 1106, 1107, 1108, 1109, 1111, or 1151.
61. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:131 comprising nucleotide 653, 654, 771, 773, 841, 849, 853, 917, 918, 926, 927, 931, 981, 983, 984, 996, 998, 1022, 1023, 1160, 1214, 1355, 1356, 1381, 1394, 1425, 1474, 1550, 1620, 1707, 1740, 1753, 1825, 1956, 1965, 2598, 2599, 2608, 2828, 2829, 2888, 3012, or 3251.
62. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:132 comprising nucleotide 292, 434, 438, 457, 459, 488, 490, 498, 499, 592, 639, 723, 774, 775, 788, 857, 858, 910, 911, 930, 931, 932, 1009, 1010, 1023, 1024, 1111, 1146, 1147, 1220, 1246, 1321, 1325, 1326, 1327, 1331, 1437, 1548, 1571, 1785, 1786, or 1787.
63. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:7 comprising nucleotide 675, 974, 1373, 1780, 2102, 2151, 2315, 2542, 2609, 3197, 67, 71, 73, 353, 405, 864, 911, 912, 913, 1409, 1748, 1811, 1935, 1937, 1993, 2012, 2346, 2388, 2437, 2586, 3007, 3008, 3022, 3130, 3210, 3237, or 3271.
64. The method of claim 40 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:133 comprising nucleotide 748, 749, 753, 1119, 1169, 1499, 1509, 1820, 2081, 2118, 2147, 2615, 2644, 2659, 2663, 2671, 2672, 2793, 2812, 2914, 2948, 3044, 3334, 3391, 3480, 3520, 3549, 3639, 3840, 3941, 3944, 4001, 4995, 4997, 5141, 5143, 5249, 5375, 5834, 5852, 5981, or 6678.
65. The method of claim 40 further comprising administering to the subject a second interfering RNA having a length of 19 to 49 nucleotides, and comprising
a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect complementarity of at least 19 nucleotides;
wherein the antisense strand of the second interfering RNA hybridizes under physiological conditions to a second portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133, and the antisense strand has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the second hybridizing portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133, respectively.
66. A method of treating ocular hypertension in a subject in need thereof, comprising:
administering to an eye of the subject a composition comprising an effective amount of interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier, the interfering RNA comprising:
a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect contiguous complementarity of at least 19 nucleotides;
wherein the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133, respectively,
wherein the ocular hypertension is treated thereby.
67. The method of claim 66 wherein the subject is a human.
68. The method of claim 66 wherein the composition is administered via a topical, intravitreal, transcleral, periocular, conjunctival, subtenon, intracameral, subretinal, subconjunctival, retrobulbar, or intracanalicular route.
69. The method of claim 66 wherein the composition is administered via in vivo expression from an expression vector capable of expressing the interfering RNA.
70. The method of claim 67 wherein the composition is administered via a topical, intravitreal, transcleral, periocular, conjunctival, subtenon, intracameral, subretinal, subconjunctival, retrobulbar, or intracanalicular route.
71. The method of claim 67 wherein the composition is administered via in vivo expression from an expression vector capable of expressing the interfering RNA.
72. The method of claim 66 wherein the ocular hypertension target mRNA encodes a β1- or β2-adrenergic receptor, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:3 or SEQ ID NO:4, respectively.
73. The method of claim 66 wherein the ocular hypertension target mRNA encodes an acetylcholinesterase, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:5 or SEQ ID NO:123, respectively.
74. The method of claim 66 wherein the ocular hypertension target mRNA encodes a subunit of Na+/K+-ATPase, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:6, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, or SEQ ID NO:132, respectively.
75. The method of claim 66 wherein the ocular hypertension target mRNA encodes a Na—K-2Cl cotransporter, and the antisense strand hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO: 7 or SEQ ID NO:133 and has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:7 or SEQ ID NO:133, respectively.
76. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:3 comprising nucleotide 468, 523, 799, 1563, 1565, 1569, 1593, 1613, 1614, 1626, 310, 322, 726, 769, 772, 801, 802, 1501, 1576, 1577, 1579, 1580, 1581, 1586, 1590, 1592, 1594, 1615, 1616, 1632, 1633, or 1654.
77. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:4 comprising nucleotide 329, 375, 1031, 1046, 1149, 1163, 1371, 1401, 1426, 1880, 283, 607, 608, 609, 619, 623, 722, 857, 1037, 1091, 1115, 1124, 1136, 1137, 1151, 1164, 1393, 1394, 1395, 1406, 1407, 1427, 1428, 1429, 1442, 1725, 1726, 1756, 1757, 1758, 1767, 1790, 1791, 1792, 1793, 1803, 1861, 1869, 1971, 1972, or 1979.
78. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:123 comprising nucleotide 1875, 1890, 1891, 2011, 2012, 2133, or 2134.
79. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:5 comprising nucleotide 366, 370, 384, 385, 525, 588, 768, 1045, 1046, 1061, 1090, 1232, 1314, 1316, 1460, 1461, 1462, 1528, 1607, 1705, 1713, 382, 393, 397, 622, 1131, 1459, 1530, 2251, 2885, 2886, 386, 1231, 1315, 2047, 2049, 2053, 2055, 2057, 2125, 2126, 2127, 2250, 2253, 2258, 2260, 2318, 2395, 2397, 2404, 2405, 2643, 2645, or 2887.
80. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:124 comprising nucleotide 2208, 2275, 2307, 2526, 2538, 2592, 2628, 2979, 2985, 3093, 3474, 3504, 3505, 3506, 3518, 343, 442, 700, 707, 811, 907, 1059, 1363, 1594, 1662, 1758, 1760, 1896, 2037, or 2147.
81. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:125 comprising nucleotide 436, 441, 443, 552, 617, 701, 702, 832, 2204, 2291, or 2495.
82. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:6 comprising nucleotide 471, 1990, 3080, 3797, 4037, 4093, 4225, 4323, 5213, 5285, 214, 467, 470, 472, 473, 632, 825, 946, 1693, 1767, 1768, 2157, 2263, 2589, 2590, 2765, 2988, 3094, 3144, 3145, 3344, 3345, 3418, 3666, 3828, 3850, 4040, 4041, 4061, 4882, 4894, 4900, 5040, 5114, 5115, 5128, 5129, 5253, 5296, 5375, 5384, or 5385.
83. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:126 comprising nucleotide 240, 272, 362, 1836, 1851, 2103, 2137, 2138, 2139, 2157, 2158, 2160, 2425, 2580, 2601, 2646, 2650, 2794, 2803, 3116, 3124, 3126, 3129, or 3377.
84. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:127 comprising nucleotide 113, 612, 702, 833, 1101, 1732, 1733, 1836, 2070, 2071, 2143, 2328, 2475, 2861, 2862, 2952, 3203, 3281, 3377, 3379, 3470, 3471, 3554, 3614, 3615, 3616, 3617, 3625, 3626, 3642, 3646, 3647, 3653, 3655, 3797, 3801, 3803, 3809 or 3810.
85. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:128 comprising nucleotide 126, 251, 252, 253, 331, 427, 429, 520, 521, 530, 601, 602, 603, 604, 664, 665, 666, 667, 675, 676, 692, 696, 697, 702, 703, 705, 707, 847, 851, 853, 859, or 860.
86. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:129 comprising nucleotide 1096, 1099, 1130, 1131, 1167, 1299, 1441, 1450, 1451, 1452, 1564, 1746, 1750, 1751, 1752, 1795, 203, 204, 214, 222, 224, 225, 226, 380, 525, 591, 612, 613, 615, 635, 636, 663, 664, 669, 699, 765, 790, 839, 840, 841, 900, 909, 933, or 947.
87. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:130 comprising nucleotide 1063, 1102, 1106, 1107, 1108, 1109, 1111, or 1151.
88. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:131 comprising nucleotide 653, 654, 771, 773, 841, 849, 853, 917, 918, 926, 927, 931, 981, 983, 984, 996, 998, 1022, 1023, 1160, 1214, 1355, 1356, 1381, 1394, 1425, 1474, 1550, 1620, 1707, 1740, 1753, 1825, 1956, 1965, 2598, 2599, 2608, 2828, 2829, 2888, 3012, or 3251.
89. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:132 comprising nucleotide 292, 434, 438, 457, 459, 488, 490, 498, 499, 592, 639, 723, 774, 775, 788, 857, 858, 910, 911, 930, 931, 932, 1009, 1010, 1023, 1024, 1111, 1146, 1147, 1220, 1246, 1321, 1325, 1326, 1327, 1331, 1437, 1548, 1571, 1785, 1786, or 1787.
90. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:7 comprising nucleotide 675, 974, 1373, 1780, 2102, 2151, 2315, 2542, 2609, 3197, 67, 71, 73, 353, 405, 864, 911, 912, 913, 1409, 1748, 1811, 1935, 1937, 1993, 2012, 2346, 2388, 2437, 2586, 3007, 3008, 3022, 3130, 3210, 3237, or 3271.
91. The method of claim 66 wherein the antisense strand is designed to target an mRNA corresponding to SEQ ID NO:133 comprising nucleotide 748, 749, 753, 1119, 1169, 1499, 1509, 1820, 2081, 2118, 2147, 2615, 2644, 2659, 2663, 2671, 2672, 2793, 2812, 2914, 2948, 3044, 3334, 3391, 3480, 3520, 3549, 3639, 3840, 3941, 3944, 4001, 4995, 4997, 5141, 5143, 5249, 5375, 5834, 5852, 5981, or 6678.
92. The method of claim 66 further comprising administering to the subject a second interfering RNA having a length of 19 to 49 nucleotides, and comprising
a sense nucleotide strand, an antisense nucleotide strand, and a region of at least near-perfect complementarity of at least 19 nucleotides;
wherein the antisense strand of the second interfering RNA hybridizes under physiological conditions to a second portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133, and the antisense strand has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the second hybridizing portion of mRNA corresponding to SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:123, SEQ ID NO:124, SEQ ID NO:125, SEQ ID NO:126, SEQ ID NO:127, SEQ ID NO:128, SEQ ID NO:129, SEQ ID NO:130, SEQ ID NO:131, SEQ ID NO:132, or SEQ ID NO:133, respectively.
93. The method of claim 66 wherein the sense nucleotide strand and the antisense nucleotide strand are connected by a loop nucleotide sequence.
94. A method of attenuating expression of an ocular hypertension target mRNA of a subject, comprising:
administering to the subject a composition comprising an effective amount of single-stranded interfering RNA having a length of 19 to 49 nucleotides and a pharmaceutically acceptable carrier,
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:3 comprising nucleotide 468, 523, 799, 1563, 1565, 1569, 1593, 1613, 1614, 1626, 310, 322, 726, 769, 772, 801, 802, 1501, 1576, 1577, 1579, 1580, 1581, 1586, 1590, 1592, 1594, 1615, 1616, 1632, 1633, or 1654, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:3; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:4 comprising nucleotide 329, 375, 1031, 1046, 1149, 1163, 1371, 1401, 1426, 1880, 283, 607, 608, 609, 619, 623, 722, 857, 1037, 1091, 1115, 1124, 1136, 1137, 1151, 1164, 1393, 1394, 1395, 1406, 1407, 1427, 1428, 1429, 1442, 1725, 1726, 1756, 1757, 1758, 1767, 1790, 1791, 1792, 1793, 1803, 1861, 1869, 1971, 1972, or 1979, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:4; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:123 comprising nucleotide 1875, 1890, 1891, 2011, 2012, 2133, or 2134, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:123; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:5 comprising nucleotide 366, 370, 384, 385, 525, 588, 768, 1045, 1046, 1061, 1090, 1232, 1314, 1316, 1460, 1461, 1462, 1528, 1607, 1705, 1713, 382, 393, 397, 622, 1131, 1459, 1530, 2251, 2885, 2886, 386, 1231, 1315, 2047, 2049, 2053, 2055, 2057, 2125, 2126, 2127, 2250, 2253, 2258, 2260, 2318, 2395, 2397, 2404, 2405, 2643, 2645, or 2887, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:5; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:124 comprising nucleotide 2208, 2275, 2307, 2526, 2538, 2592, 2628, 2979, 2985, 3093, 3474, 3504, 3505, 3506, 3518, 343, 442, 700, 707, 811, 907, 1059, 1363, 1594, 1662, 1758, 1760, 1896, 2037, or 2147, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:124; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:125 comprising nucleotide 436, 441, 443, 552, 617, 701, 702, 832, 2204, 2291, or 2495, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:125; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:6 comprising nucleotide 471, 1990, 3080, 3797, 4037, 4093, 4225, 4323, 5213, 5285, 214, 467, 470, 472, 473, 632, 825, 946, 1693, 1767, 1768, 2157, 2263, 2589, 2590, 2765, 2988, 3094, 3144, 3145, 3344, 3345, 3418, 3666, 3828, 3850, 4040, 4041, 4061, 4882, 4894, 4900, 5040, 5114, 5115, 5128, 5129, 5253, 5296, 5375, 5384, or 5385, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:6; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:126 comprising nucleotide 240, 272, 362, 1836, 1851, 2103, 2137, 2138, 2139, 2157, 2158, 2160, 2425, 2580, 2601, 2646, 2650, 2794, 2803, 3116, 3124, 3126, 3129, or 3377, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:126; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:127 comprising nucleotide 113, 612, 702, 833, 1101, 1732, 1733, 1836, 2070, 2071, 2143, 2328, 2475, 2861, 2862, 2952, 3203, 3281, 3377, 3379, 3470, 3471, 3554, 3614, 3615, 3616, 3617, 3625, 3626, 3642, 3646, 3647, 3653, 3655, 3797, 3801, 3803, 3809 or 3810, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:127; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:128 comprising nucleotide 126, 251, 252, 253, 331, 427, 429, 520, 521, 530, 601, 602, 603, 604, 664, 665, 666, 667, 675, 676, 692, 696, 697, 702, 703, 705, 707, 847, 851, 853, 859, or 860, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:128; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:129 comprising nucleotide 1096, 1099, 1130, 1131, 1167, 1299, 1441, 1450, 1451, 1452, 1564, 1746, 1750, 1751, 1752, 1795, 203, 204, 214, 222, 224, 225, 226, 380, 525, 591, 612, 613, 615, 635, 636, 663, 664, 669, 699, 765, 790, 839, 840, 841, 900, 909, 933, or 947, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:129; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:130 comprising nucleotide 1063, 1102, 1106, 1107, 1108, 1109, 1111, or 1151, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:130; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:131 comprising nucleotide 653, 654, 771, 773, 841, 849, 853, 917, 918, 926, 927, 931, 981, 983, 984, 996, 998, 1022, 1023, 1160, 1214, 1355, 1356, 1381, 1394, 1425, 1474, 1550, 1620, 1707, 1740, 1753, 1825, 1956, 1965, 2598, 2599, 2608, 2828, 2829, 2888, 3012, or 3251, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:131; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:132 comprising nucleotide 292, 434, 438, 457, 459, 488, 490, 498, 499, 592, 639, 723, 774, 775, 788, 857, 858, 910, 911, 930, 931, 932, 1009, 1010, 1023, 1024, 1111, 1146, 1147, 1220, 1246, 1321, 1325, 1326, 1327, 1331, 1437, 1548, 1571, 1785, 1786, or 1787, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:132; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:7 comprising nucleotide 675, 974, 1373, 1780, 2102, 2151, 2315, 2542, 2609, 3197, 67, 71, 73, 353, 405, 864, 911, 912, 913, 1409, 1748, 1811, 1935, 1937, 1993, 2012, 2346, 2388, 2437, 2586, 3007, 3008, 3022, 3130, 3210, 3237, or 3271, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:7; or
wherein the single-stranded interfering RNA hybridizes under physiological conditions to a portion of mRNA corresponding to SEQ ID NO:133 comprising nucleotide 748, 749, 753, 1119, 1169, 1499, 1509, 1820, 2081, 2118, 2147, 2615, 2644, 2659, 2663, 2671, 2672, 2793, 2812, 2914, 2948, 3044, 3334, 3391, 3480, 3520, 3549, 3639, 3840, 3941, 3944, 4001, 4995, 4997, 5141, 5143, 5249, 5375, 5834, 5852, 5981, or 6678, and the interfering RNA has a region of at least near-perfect contiguous complementarity of at least 19 nucleotides with the hybridizing portion of mRNA corresponding to SEQ ID NO:133;
wherein the expression of an ocular hypertension target mRNA is thereby attenuated.
95. The method of claim 94 wherein the composition is administered via a topical, intravitreal, transcleral, periocular, conjunctival, subtenon, intracameral, subretinal, subconjunctival, retrobulbar, or intracanalicular route.
96. The method of claim 94 wherein the composition is administered via in vivo expression from an expression vector capable of expressing the interfering RNA.
97. The method of claim 94 wherein the interfering RNA is an mRNA.
98. The method of claim 94 wherein the interfering RNA is an siRNA.
99. A composition comprising interfering RNA having a length of 19 to 49 nucleotides and having a nucleotide sequence of any one of SEQ ID NO:33-SEQ ID NO:82, and SEQ ID NO:220-SEQ ID NO:717, or a complement thereof, and a pharmaceutically acceptable carrier.
100. The composition of claim 99 wherein the interfering RNA is an shRNA.
101. The composition of claim 99 wherein the interfering RNA is an siRNA.
102. The composition of claim 99 wherein the interfering RNA is an miRNA.
US11/344,702 2005-02-01 2006-02-01 RNAi-mediated inhibition of ocular hypertension targets Abandoned US20060172963A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/344,702 US20060172963A1 (en) 2005-02-01 2006-02-01 RNAi-mediated inhibition of ocular hypertension targets
US12/967,121 US20110124708A1 (en) 2005-02-01 2010-12-14 Rnai-mediated inhibition of ocular hypertension targets

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US64892605P 2005-02-01 2005-02-01
US75336405P 2005-12-22 2005-12-22
US11/344,702 US20060172963A1 (en) 2005-02-01 2006-02-01 RNAi-mediated inhibition of ocular hypertension targets

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/967,121 Division US20110124708A1 (en) 2005-02-01 2010-12-14 Rnai-mediated inhibition of ocular hypertension targets

Publications (1)

Publication Number Publication Date
US20060172963A1 true US20060172963A1 (en) 2006-08-03

Family

ID=36587258

Family Applications (4)

Application Number Title Priority Date Filing Date
US11/344,702 Abandoned US20060172963A1 (en) 2005-02-01 2006-02-01 RNAi-mediated inhibition of ocular hypertension targets
US11/345,361 Expired - Fee Related US7592324B2 (en) 2005-02-01 2006-02-01 RNAi-mediated inhibition of ocular targets
US12/536,698 Abandoned US20090326044A1 (en) 2005-02-01 2009-08-06 RNAi-Mediated Inhibition of Ocular Targets
US12/967,121 Abandoned US20110124708A1 (en) 2005-02-01 2010-12-14 Rnai-mediated inhibition of ocular hypertension targets

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/345,361 Expired - Fee Related US7592324B2 (en) 2005-02-01 2006-02-01 RNAi-mediated inhibition of ocular targets
US12/536,698 Abandoned US20090326044A1 (en) 2005-02-01 2009-08-06 RNAi-Mediated Inhibition of Ocular Targets
US12/967,121 Abandoned US20110124708A1 (en) 2005-02-01 2010-12-14 Rnai-mediated inhibition of ocular hypertension targets

Country Status (14)

Country Link
US (4) US20060172963A1 (en)
EP (2) EP1848803A2 (en)
JP (2) JP2008528637A (en)
KR (2) KR20070099048A (en)
AR (2) AR052103A1 (en)
AU (2) AU2006210451B2 (en)
BR (2) BRPI0607096A2 (en)
CA (2) CA2595678A1 (en)
MX (2) MX2007009194A (en)
RU (2) RU2007132874A (en)
SA (1) SA06260487B1 (en)
TW (2) TW200639253A (en)
UY (2) UY29348A1 (en)
WO (2) WO2006083945A2 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060234970A1 (en) * 2004-08-23 2006-10-19 Jimenez Ana I Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US20080249048A1 (en) * 2005-03-14 2008-10-09 Sylentis S.A.U. Methods and Compositions for the Treatment of Intestinal Conditions
US20090087413A1 (en) * 2007-10-01 2009-04-02 Alcon Research, Ltd. Self-complementary aav-mediated delivery of interfering rna molecules to treat or prevent ocular disorders
US20090131358A1 (en) * 2007-11-15 2009-05-21 Alcon Research, Ltd. LOW DENSITY LIPOPROTEIN RECEPTOR-MEDIATED siRNA DELIVERY
US20090156470A1 (en) * 2007-12-18 2009-06-18 Chatterton Jon E Interfering rna delivery system and uses thereof
JP2010501188A (en) * 2006-08-24 2010-01-21 アルコン リサーチ, リミテッド RNAi-mediated inhibition of gremlin for the treatment of IOP-related conditions
US20100286230A1 (en) * 2005-10-20 2010-11-11 Sylentis S.A.U. Modulation of trpv expression levels
US20110160277A1 (en) * 2005-10-25 2011-06-30 Sylentis S.A.U. Modulation of 11 beta-hydroxysteriod dehydrogenase 1 expression for the treatment of ocular diseases
US7973019B1 (en) 2007-10-03 2011-07-05 Alcon Research, Ltd. Transferrin/transferrin receptor-mediated siRNA delivery
WO2012161677A1 (en) 2011-05-20 2012-11-29 Alcon Research, Ltd. TRANSFERRIN/TRANSFERRIN RECEPTOR-MEDIATED siRNA DELIVERY
WO2013103467A1 (en) 2012-01-06 2013-07-11 Alcon Research, Ltd. Interfering rna delivery system and uses thereof
US9550994B2 (en) 2005-03-11 2017-01-24 Arrowhead Pharmaceuticals, Inc. RNAI-mediated inhibition of frizzled related protein-1 for treatment of glaucoma
US9808479B2 (en) 2012-09-05 2017-11-07 Sylentis Sau SiRNA and their use in methods and compositions for the treatment and / or prevention of eye conditions
US10011832B2 (en) 2012-09-05 2018-07-03 Sylentis Sau SiRNA and their use in methods and compositions for the treatment and/or prevention of eye conditions
US10011837B2 (en) 2014-03-04 2018-07-03 Sylentis Sau SiRNAs and their use in methods and compositions for the treatment and/or prevention of eye conditions
WO2023239756A1 (en) 2022-06-07 2023-12-14 Generation Bio Co. Lipid nanoparticle compositions and uses thereof

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1977246B1 (en) * 2005-12-30 2010-01-20 Ventana Medical Systems, Inc. Na+, k+-atpase expression in cervical dysplasia and cancer
US20090054365A1 (en) * 2007-01-26 2009-02-26 Alcon Research, Ltd. RNAi-MEDIATED INHIBITION OF AQUAPORIN 1 FOR TREATMENT OF OCULAR NEOVASCULARIZATION
WO2008104978A2 (en) * 2007-02-28 2008-09-04 Quark Pharmaceuticals, Inc. Novel sirna structures
JP5646997B2 (en) * 2007-10-03 2014-12-24 クォーク ファーマシューティカルズ インコーポレーティッドQuark Pharmaceuticals,Inc. Novel siRNA structure
WO2009090639A2 (en) * 2008-01-15 2009-07-23 Quark Pharmaceuticals, Inc. Sirna compounds and methods of use thereof
SI2350279T1 (en) * 2008-10-22 2016-04-29 Quark Pharmaceuticals, Inc. Methods for treating eye disorders
CN109414414A (en) 2016-03-16 2019-03-01 戴维·格拉德斯通研究所 Method and composition for treating obesity and/or diabetes and for identifying candidate therapeutic agent
EP3388520A1 (en) * 2017-04-11 2018-10-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for reducing the expression of nkcc1 in a subject in need thereof
CN109557309B (en) * 2018-12-04 2021-09-10 九江学院附属医院 Application of carbonic anhydrase-2 as detection marker in diagnosis of kidney stones
KR102414025B1 (en) * 2019-06-27 2022-06-29 전북대학교 산학협력단 Novel ADP-ribosyl Cyclases and Their Inhibitors
EP4347836A2 (en) * 2021-05-27 2024-04-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing carbonic anhydrase 2 expression

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6326193B1 (en) * 1999-11-05 2001-12-04 Cambria Biosciences, Llc Insect control agent
US20020086356A1 (en) * 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
US20020114784A1 (en) * 1999-01-28 2002-08-22 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US20020162126A1 (en) * 2000-03-16 2002-10-31 David Beach Methods and compositions for RNA interference
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US20030027783A1 (en) * 1999-11-19 2003-02-06 Magdalena Zernicka-Goetz Inhibiting gene expression with dsRNA
US20030153524A1 (en) * 2001-12-11 2003-08-14 Hinton David R. Methods for inhibiting ocular processes
US20040053411A1 (en) * 2002-05-03 2004-03-18 Duke University Method of regulating gene expression
US20040175732A1 (en) * 2002-11-15 2004-09-09 Rana Tariq M. Identification of micrornas and their targets
US20040203145A1 (en) * 2002-08-07 2004-10-14 University Of Massachusetts Compositions for RNA interference and methods of use thereof
US20040259247A1 (en) * 2000-12-01 2004-12-23 Thomas Tuschl Rna interference mediating small rna molecules
US20050112118A1 (en) * 1999-12-02 2005-05-26 Myriad Genetics, Incorporated Compositions and methods for treating inflammatory disorders
US20050203043A1 (en) * 2004-01-23 2005-09-15 Dharmacon, Inc. Identification of toxic nucleotide sequences
US20050223427A1 (en) * 2004-04-01 2005-10-06 Dharmacon, Inc. Modified polynucleotides for reducing off-target effects in RNA interference
US20050246794A1 (en) * 2002-11-14 2005-11-03 Dharmacon Inc. Functional and hyperfunctional siRNA
US7022828B2 (en) * 2001-04-05 2006-04-04 Sirna Theraputics, Inc. siRNA treatment of diseases or conditions related to levels of IKK-gamma
US20060234970A1 (en) * 2004-08-23 2006-10-19 Jimenez Ana I Methods and compositions for the treatment of eye disorders with increased intraocular pressure

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
JP2004508019A (en) 2000-07-28 2004-03-18 コンピュジェン インコーポレイテッド Oligonucleotide libraries for detecting RNA transcripts and splice variants occupying locations in the transcriptome
EP1497658A2 (en) * 2002-04-11 2005-01-19 Oxford GlycoSciences (UK) Ltd Protein involved in cancer
US20050222061A1 (en) * 2002-04-18 2005-10-06 Schulte Ralf W Means and methods for the specific inhibition of genes in cells and tissue of the cns and/or eye
WO2004020583A2 (en) * 2002-08-27 2004-03-11 Bristol-Myers Squibb Company Polynucleotide predictor set for identifying protein tyrosine kinase modulators
WO2004022782A2 (en) 2002-09-04 2004-03-18 Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin Compositions and methods for tissue specific or inducible inhibition of gene expression
JP2005537790A (en) * 2002-09-05 2005-12-15 ガーバン インスティテュート オブ メディカル リサーチ Novel diagnostic and therapeutic methods and reagents therefor
US7521431B2 (en) 2002-11-01 2009-04-21 The Trustees Of The University Of Pennsylvania Compositions and methods for siRNA inhibition of HIF-1 alpha
EP1606300A4 (en) * 2003-02-11 2007-01-03 Immusol Inc Sirna libraries optimized for predetermined protein families
GB0403600D0 (en) 2004-02-18 2004-03-24 Trinity College Dublin Methods and reagents for treating disease
AU2005272816B2 (en) * 2004-08-10 2011-08-11 Alnylam Pharmaceuticals, Inc. Chemically modified oligonucleotides

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US20040147475A1 (en) * 1999-01-28 2004-07-29 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US20020114784A1 (en) * 1999-01-28 2002-08-22 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US6326193B1 (en) * 1999-11-05 2001-12-04 Cambria Biosciences, Llc Insect control agent
US20030027783A1 (en) * 1999-11-19 2003-02-06 Magdalena Zernicka-Goetz Inhibiting gene expression with dsRNA
US20050112118A1 (en) * 1999-12-02 2005-05-26 Myriad Genetics, Incorporated Compositions and methods for treating inflammatory disorders
US20020162126A1 (en) * 2000-03-16 2002-10-31 David Beach Methods and compositions for RNA interference
US20020086356A1 (en) * 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
US20040259247A1 (en) * 2000-12-01 2004-12-23 Thomas Tuschl Rna interference mediating small rna molecules
US7022828B2 (en) * 2001-04-05 2006-04-04 Sirna Theraputics, Inc. siRNA treatment of diseases or conditions related to levels of IKK-gamma
US20030153524A1 (en) * 2001-12-11 2003-08-14 Hinton David R. Methods for inhibiting ocular processes
US20040053411A1 (en) * 2002-05-03 2004-03-18 Duke University Method of regulating gene expression
US20040203145A1 (en) * 2002-08-07 2004-10-14 University Of Massachusetts Compositions for RNA interference and methods of use thereof
US20050246794A1 (en) * 2002-11-14 2005-11-03 Dharmacon Inc. Functional and hyperfunctional siRNA
US20050255487A1 (en) * 2002-11-14 2005-11-17 Dharmacon, Inc. Methods and compositions for selecting siRNA of improved functionality
US20040175732A1 (en) * 2002-11-15 2004-09-09 Rana Tariq M. Identification of micrornas and their targets
US20050203043A1 (en) * 2004-01-23 2005-09-15 Dharmacon, Inc. Identification of toxic nucleotide sequences
US20050223427A1 (en) * 2004-04-01 2005-10-06 Dharmacon, Inc. Modified polynucleotides for reducing off-target effects in RNA interference
US20060234970A1 (en) * 2004-08-23 2006-10-19 Jimenez Ana I Methods and compositions for the treatment of eye disorders with increased intraocular pressure

Cited By (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8389490B2 (en) 2004-08-23 2013-03-05 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US8247386B2 (en) 2004-08-23 2012-08-21 Sylentis Sau Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US8258110B2 (en) 2004-08-23 2012-09-04 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US8951982B2 (en) 2004-08-23 2015-02-10 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US20090099111A1 (en) * 2004-08-23 2009-04-16 Sylentis S.A.U. Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US20090099113A1 (en) * 2004-08-23 2009-04-16 Sylentis S.A.U Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US20090099112A1 (en) * 2004-08-23 2009-04-16 Sylentis S.A.U. Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US20090099114A1 (en) * 2004-08-23 2009-04-16 Sylentis S.A.U. Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US8252759B2 (en) 2004-08-23 2012-08-28 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US20090105175A1 (en) * 2004-08-23 2009-04-23 Sylentis S.A.U. Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US7902169B2 (en) 2004-08-23 2011-03-08 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US8030284B2 (en) 2004-08-23 2011-10-04 Sylentis S.A.U. Treatment of eye disorders characterized by an elevated intraocular pressure by siRNAs
US20090105176A1 (en) * 2004-08-23 2009-04-23 Sylentis S.A.U. Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US7592325B2 (en) * 2004-08-23 2009-09-22 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US8252758B2 (en) 2004-08-23 2012-08-28 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US20100022623A1 (en) * 2004-08-23 2010-01-28 Sylentis S.A.U. Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US20070270365A1 (en) * 2004-08-23 2007-11-22 Sylentis S.A. Treatment of Eye Disorders Characterized by an Elevated Intraocular Pressure by siRNAs
US8247387B2 (en) 2004-08-23 2012-08-21 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US8198250B2 (en) 2004-08-23 2012-06-12 Sylentis S.A.U. Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US20100331394A1 (en) * 2004-08-23 2010-12-30 Sylentis S.A.U. Methods and Compositions for the Treatment of Eye Disorders with Increased Intraocular Pressure
US20060234970A1 (en) * 2004-08-23 2006-10-19 Jimenez Ana I Methods and compositions for the treatment of eye disorders with increased intraocular pressure
US9550994B2 (en) 2005-03-11 2017-01-24 Arrowhead Pharmaceuticals, Inc. RNAI-mediated inhibition of frizzled related protein-1 for treatment of glaucoma
US8067577B2 (en) 2005-03-14 2011-11-29 Sylentis S.A.U. Methods and compositions for the treatment of intestinal conditions
US7807650B2 (en) 2005-03-14 2010-10-05 Sylentis S.A.U. Methods and compositions for the treatment of intestinal conditions
US20100317720A1 (en) * 2005-03-14 2010-12-16 Sytentis S.A.U. Methods and Compositions for the Treatment of Intestinal Conditions
US20080249048A1 (en) * 2005-03-14 2008-10-09 Sylentis S.A.U. Methods and Compositions for the Treatment of Intestinal Conditions
US20100286230A1 (en) * 2005-10-20 2010-11-11 Sylentis S.A.U. Modulation of trpv expression levels
US8354385B2 (en) 2005-10-20 2013-01-15 Sylentis S.A.U. Modulation of TRPV expression levels
US8188057B2 (en) 2005-10-25 2012-05-29 Sylentis S.A.U. Modulation of 11beta-hydroxysteriod dehydrogenase 1 expression for the treatment of ocular diseases
US20110160277A1 (en) * 2005-10-25 2011-06-30 Sylentis S.A.U. Modulation of 11 beta-hydroxysteriod dehydrogenase 1 expression for the treatment of ocular diseases
JP2010501188A (en) * 2006-08-24 2010-01-21 アルコン リサーチ, リミテッド RNAi-mediated inhibition of gremlin for the treatment of IOP-related conditions
US20090087413A1 (en) * 2007-10-01 2009-04-02 Alcon Research, Ltd. Self-complementary aav-mediated delivery of interfering rna molecules to treat or prevent ocular disorders
US8399653B2 (en) 2007-10-03 2013-03-19 Alcon Research, Ltd. Transferrin/transferrin receptor-mediated siRNA delivery
US10138483B2 (en) 2007-10-03 2018-11-27 Arrowhead Pharmaceuticals, Inc. Transferrin/transferrin receptor-mediated siRNA delivery
US9114152B2 (en) 2007-10-03 2015-08-25 Arrowhead Research Corporation Transferrin/transferrin receptor-mediated siRNA delivery
US9562230B2 (en) 2007-10-03 2017-02-07 Arrowhead Pharmaceuticals, Inc. Transferrin/transferrin receptor-mediated siRNA delivery
US7973019B1 (en) 2007-10-03 2011-07-05 Alcon Research, Ltd. Transferrin/transferrin receptor-mediated siRNA delivery
US9289514B2 (en) 2007-11-15 2016-03-22 Arrowhead Research Corporation Low density lipoprotein receptor-mediated siRNA delivery
US8937051B2 (en) 2007-11-15 2015-01-20 Alcon Research, Ltd. Low density lipoprotein receptor-mediated siRNA delivery
US20090131358A1 (en) * 2007-11-15 2009-05-21 Alcon Research, Ltd. LOW DENSITY LIPOPROTEIN RECEPTOR-MEDIATED siRNA DELIVERY
US8569258B2 (en) 2007-11-15 2013-10-29 Alcon Research, Ltd. Low density lipoprotein receptor-mediated siRNA delivery
US20110230410A1 (en) * 2007-11-15 2011-09-22 Alcon Research, Ltd. LOW DENSITY LIPOPROTEIN RECEPTOR-MEDIATED siRNA DELIVERY
US8664375B2 (en) 2007-12-18 2014-03-04 Alcon Research, Ltd. Interfering RNA delivery system and uses thereof
US9795684B2 (en) 2007-12-18 2017-10-24 Arrowhead Pharmaceuticals Interfering RNA delivery system and uses thereof
US9233170B2 (en) 2007-12-18 2016-01-12 Arrowhead Research Corporation Interfering RNA delivery system and uses thereof
US20090156470A1 (en) * 2007-12-18 2009-06-18 Chatterton Jon E Interfering rna delivery system and uses thereof
WO2009079635A1 (en) * 2007-12-18 2009-06-25 Alcon Research, Ltd. Interfering rna delivery system and uses thereof
US7879813B2 (en) 2007-12-18 2011-02-01 Alcon Research, Ltd. Interfering RNA delivery system and uses thereof
US20110082092A1 (en) * 2007-12-18 2011-04-07 Alcon Research, Ltd. Interfering rna delivery system and uses thereof
WO2012161677A1 (en) 2011-05-20 2012-11-29 Alcon Research, Ltd. TRANSFERRIN/TRANSFERRIN RECEPTOR-MEDIATED siRNA DELIVERY
WO2013103467A1 (en) 2012-01-06 2013-07-11 Alcon Research, Ltd. Interfering rna delivery system and uses thereof
US9932584B2 (en) 2012-01-06 2018-04-03 Arrowhead Pharmaceuticals, Inc. Interfering RNA delivery system and uses thereof
US9347062B2 (en) 2012-01-06 2016-05-24 Arrowhead Research Corporation Interfering RNA delivery system and uses thereof
US9808479B2 (en) 2012-09-05 2017-11-07 Sylentis Sau SiRNA and their use in methods and compositions for the treatment and / or prevention of eye conditions
US10011832B2 (en) 2012-09-05 2018-07-03 Sylentis Sau SiRNA and their use in methods and compositions for the treatment and/or prevention of eye conditions
US10011837B2 (en) 2014-03-04 2018-07-03 Sylentis Sau SiRNAs and their use in methods and compositions for the treatment and/or prevention of eye conditions
WO2023239756A1 (en) 2022-06-07 2023-12-14 Generation Bio Co. Lipid nanoparticle compositions and uses thereof

Also Published As

Publication number Publication date
WO2006084217A3 (en) 2007-09-13
RU2007132848A (en) 2009-03-10
EP1848803A2 (en) 2007-10-31
AU2006210451B2 (en) 2011-09-22
AR052102A1 (en) 2007-02-28
SA06260487B1 (en) 2011-02-23
JP2008528632A (en) 2008-07-31
US20090326044A1 (en) 2009-12-31
RU2007132874A (en) 2009-03-10
US20110124708A1 (en) 2011-05-26
EP1846557A2 (en) 2007-10-24
MX2007009200A (en) 2007-09-14
WO2006083945A8 (en) 2007-10-11
BRPI0606979A2 (en) 2009-07-28
WO2006084217A2 (en) 2006-08-10
UY29349A1 (en) 2006-05-31
KR20070099048A (en) 2007-10-08
US20060172965A1 (en) 2006-08-03
BRPI0607096A2 (en) 2009-08-04
CA2595790A1 (en) 2006-08-10
WO2006083945A3 (en) 2007-08-23
AR052103A1 (en) 2007-02-28
AU2006210846A1 (en) 2006-08-10
KR20070099673A (en) 2007-10-09
MX2007009194A (en) 2007-09-14
TW200639252A (en) 2006-11-16
CA2595678A1 (en) 2006-08-10
JP2008528637A (en) 2008-07-31
UY29348A1 (en) 2006-05-31
WO2006083945A2 (en) 2006-08-10
AU2006210451A1 (en) 2006-08-10
TW200639253A (en) 2006-11-16
US7592324B2 (en) 2009-09-22

Similar Documents

Publication Publication Date Title
US7592324B2 (en) RNAi-mediated inhibition of ocular targets
US9550994B2 (en) RNAI-mediated inhibition of frizzled related protein-1 for treatment of glaucoma
US8168609B2 (en) RNAi-mediated inhibition of Rho kinase for treatment of ocular disorders
US20190338287A1 (en) RNAi-MEDIATED INHIBITION OF HIF1A FOR TREATMENT OF OCULAR ANGIOGENESIS
US20120077864A1 (en) Rnai-mediated inhibition of gremlin for treatment of iop-related conditions
US20150057336A1 (en) RNAi-RELATED INHIBITION OF TNF alpha SIGNALING PATHWAY FOR TREATMENT OF GLAUCOMA
US9752147B2 (en) RNAi-mediated inhibition of connexin 43 for treatment of IOP-related conditions

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALCON, INC., SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SHEPARD, ALLAN R.;CHATTERTON, JON E.;CLARK, ABBOT F.;REEL/FRAME:017463/0136;SIGNING DATES FROM 20060330 TO 20060406

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: ARROWHEAD RESEARCH CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS AG;REEL/FRAME:035604/0484

Effective date: 20150420