TW201916883A - Treatment of sodium channel, voltage-gated, alpha subunit (SCNA) related diseases by inhibition of natural antisense transcript to SCNA - Google Patents

Treatment of sodium channel, voltage-gated, alpha subunit (SCNA) related diseases by inhibition of natural antisense transcript to SCNA Download PDF

Info

Publication number
TW201916883A
TW201916883A TW107132956A TW107132956A TW201916883A TW 201916883 A TW201916883 A TW 201916883A TW 107132956 A TW107132956 A TW 107132956A TW 107132956 A TW107132956 A TW 107132956A TW 201916883 A TW201916883 A TW 201916883A
Authority
TW
Taiwan
Prior art keywords
oligonucleotide
seq
antisense
scna
voltage
Prior art date
Application number
TW107132956A
Other languages
Chinese (zh)
Other versions
TWI734935B (en
Inventor
柯拉德約瑟夫
薛曼歐嘉 寇可維
里昂貝琳達 迪
柯多卡洛斯
H 蕭珍
Original Assignee
美商可娜公司
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 美商可娜公司 filed Critical 美商可娜公司
Priority to TW107132956A priority Critical patent/TWI734935B/en
Publication of TW201916883A publication Critical patent/TW201916883A/en
Application granted granted Critical
Publication of TWI734935B publication Critical patent/TWI734935B/en

Links

Abstract

The present invention relates to antisense oligonucleotides that modulate the expression of and/or function of Sodium channel, voltage-gated, alpha subunit (SCNA), in particular, by targeting natural antisense polynucleotides of Sodium channel, voltage-gated, alpha subunit (SCNA). The invention also relates to the identification of these antisense oligonucleotides and their use in treating diseases and disorders associated with the expression of SCNA.

Description

藉由抑制電壓門控鈉離子通道α次單元(SCNA)之天然反義轉錄物治療SCNA相關疾病Treatment of SCNA-related diseases by inhibiting the natural antisense transcript of voltage-gated sodium channel alpha subunit (SCNA)

本發明之實施例包含SCNA及相關分子之寡核苷酸調節表現及/或功能。 本申請案主張2010年6月23日申請之美國臨時專利申請案第61/357,774號之優先權,該臨時專利申請案以全文引用的方式併入本文中。Embodiments of the invention encompass oligonucleotide-regulated expression and/or function of SCNAs and related molecules. The present application claims priority to U.S. Provisional Patent Application Serial No. 61/357, file, filed on Jun.

DNA-RNA及RNA-RNA雜交對核酸功能之許多態樣(包括DNA複製、轉錄及轉譯)而言為重要的。對偵測特定核酸或改變其表現之多種技術而言,雜交亦為重要的。反義核苷酸例如藉由與目標RNA雜交,藉此干擾RNA剪接、轉錄、轉譯及複製而破壞基因表現。反義DNA具有附加特徵,即DNA-RNA雜交體充當核糖核酸酶H消化之受質(一種存在於大多數細胞類型中之活性)。與寡去氧核苷酸(ODN)之情況一樣,反義分子可傳遞入細胞中,或其可自內源性基因表現為RNA分子。FDA近來核准一種反義藥物VITRAVENE™(用於治療細胞巨大病毒視網膜炎),反映出反義具有治療效用。DNA-RNA and RNA-RNA hybridization are important for many aspects of nucleic acid function, including DNA replication, transcription, and translation. Hybridization is also important for a variety of techniques for detecting specific nucleic acids or altering their performance. Antisense nucleotides disrupt gene expression, for example, by hybridizing to a target RNA, thereby interfering with RNA splicing, transcription, translation, and replication. Antisense DNA has the additional feature that the DNA-RNA hybrid acts as a substrate for ribonuclease H digestion (an activity present in most cell types). As in the case of oligodeoxynucleotides (ODN), an antisense molecule can be delivered into a cell, or it can be expressed as an RNA molecule from an endogenous gene. The FDA recently approved an antisense drug, VITRAVENETM (for the treatment of giant viral retinitis), reflecting the therapeutic utility of antisense.

提供本[發明內容]以呈現簡要指示本發明之性質及實質之本發明的概述。其在應瞭解其不應用於解釋或限制申請專利範圍之範疇或意義之情況下呈遞。 在一實施例中,本發明提供藉由使用靶向天然反義轉錄物之任何區域,從而導致上調相應有義基因之反義寡核苷酸來抑制天然反義轉錄物之作用的方法,其中該有義基因係選自SCNA基因家族之至少一個成員及其變異體。本文亦涵蓋天然反義轉錄物可由視為在本發明之範疇內之siRNA、核糖核酸酶及小分子抑制。 一實施例提供一種活體內或活體外調節患者細胞或組織中之SCNA聚核苷酸之功能及/或表現的方法,其包含使該等細胞或組織與長度為5至30個核苷酸之反義寡核苷酸接觸,其中該寡核苷酸與包含以下核苷酸內之5至30個連續核苷酸之聚核苷酸的反向互補序列具有至少50%序列一致性:SEQ ID NO: 12之1至1123及SEQ ID NO: 13之1至2352、SEQ ID NO: 14之1至267、SEQ ID NO: 15之1至1080、SEQ ID NO: 16之1至173、SEQ ID NO: 17之1至618、SEQ ID NO: 18之1至871、SEQ ID NO: 19之1至304、SEQ ID NO: 20之1至293、SEQ ID NO: 21之1至892、SEQ ID NO: 22之1至260、SEQ ID NO: 23之1至982、SEQ ID NO: 24之1至906、SEQ ID NO: 25之1至476、SEQ ID NO: 26之1至185、SEQ ID NO: 27之1至162及SEQ ID NO: 28之1至94;藉此活體內或活體外調節患者細胞或組織中之SCNA聚核苷酸之功能及/或表現。 在一實施例中,寡核苷酸靶向SCNA聚核苷酸之天然反義序列(例如SEQ ID NO: 12至28中闡述之核苷酸)及其任何變異體、對偶基因、同源物、突變體、衍生物、片段及互補序列。反義寡核苷酸之實例係如SEQ ID NO: 29至94所闡述。 另一實施例提供一種活體內或活體外調節患者細胞或組織中之SCNA聚核苷酸之功能及/或表現的方法,其包含使該等細胞或組織與長度為5至30個核苷酸之反義寡核苷酸接觸,其中該寡核苷酸與該SCNA聚核苷酸之反義之反向互補序列具有至少50%序列一致性;藉此活體內或活體外調節患者細胞或組織中之SCNA聚核苷酸的功能及/或表現。 另一實施例提供一種活體內或活體外調節患者細胞或組織中之SCNA聚核苷酸之功能及/或表現的方法,其包含使該等細胞或組織與長度為5至30個核苷酸之反義寡核苷酸接觸,其中該寡核苷酸與SCNA天然反義轉錄物具有至少50%序列互補性;藉此活體內或活體外調節患者細胞或組織中之SCNA聚核苷酸的功能及/或表現。 在一實施例中,組合物包含一或多種與有義及/或反義SCNA聚核苷酸結合之反義寡核苷酸,其中該等聚核苷酸係選自由SCNA至SCN12A及其變異體組成之群。在一較佳實施例中,目標聚核苷酸係選自SCNA。 在一實施例中,寡核苷酸包含一或多個經修飾或經取代之核苷酸。 在一實施例中,寡核苷酸包含一或多個經修飾之鍵。 在另一實施例中,經修飾之核苷酸包含含有硫代磷酸酯、甲基膦酸酯、肽核酸、2'-O-甲基、氟-或碳、亞甲基或其他鎖核酸(locked nucleic acid,LNA)分子之經修飾之鹼基。較佳地,經修飾之核苷酸為鎖核酸分子,包括α-L-LNA。 在一實施例中,向患者皮下、肌肉內、靜脈內或腹膜內投與寡核苷酸。 在一實施例中,寡核苷酸係以醫藥組合物形式投與。治療方案包含向患者投與反義化合物至少1次;然而,此治療可經改進以包括歷時一段時期之多次劑量。治療可與一或多種其他類型之療法組合。 在一實施例中,寡核苷酸囊封於脂質體中或與載劑分子(例如膽固醇、TAT肽)連接。 下文描述其他態樣。The Summary of the Invention is provided to present an overview of the invention in which the nature and substance of the invention are described. It should be presented with the understanding that it should not be used to interpret or limit the scope or meaning of the scope of the patent application. In one embodiment, the invention provides a method of inhibiting the action of a natural antisense transcript by using any region that targets a natural antisense transcript, thereby causing upregulation of an antisense oligonucleotide of the corresponding sense gene, wherein The sense gene is selected from at least one member of the SCNA gene family and variants thereof. Also contemplated herein is a natural antisense transcript that can be inhibited by siRNA, ribonuclease, and small molecules that are considered to be within the scope of the invention. An embodiment provides a method of modulating the function and/or expression of a SCNA polynucleotide in a cell or tissue of a patient in vivo or ex vivo, comprising affixing the cell or tissue to a length of 5 to 30 nucleotides Antisense oligonucleotide contact, wherein the oligonucleotide has at least 50% sequence identity to a reverse complement comprising a polynucleotide of 5 to 30 contiguous nucleotides in the following nucleotide: SEQ ID NO: 12 to 1123 and SEQ ID NO: 13 to 1 to 2352, SEQ ID NO: 14 to 1 to 267, SEQ ID NO: 1 to 1 to 1080, SEQ ID NO: 1 to 1 to 173, SEQ ID NO: 17 to 1 618, 1 to 871 of SEQ ID NO: 18, 1 to 304 of SEQ ID NO: 19, 1 to 293 of SEQ ID NO: 20, 1 to 892 of SEQ ID NO: 21, SEQ ID NO: 22 to 1 to 260, 1 to 982 of SEQ ID NO: 23, 1 to 906 of SEQ ID NO: 24, 1 to 476 of SEQ ID NO: 25, 1 to 185 of SEQ ID NO: 26, SEQ ID NO: 27 to 1 162 and SEQ ID NO: 28 to 1 to 94; thereby regulating the function and/or expression of SCNA polynucleotides in a patient's cells or tissues in vivo or in vitro. In one embodiment, the oligonucleotide targets a natural antisense sequence of a SCNA polynucleotide (eg, the nucleotides set forth in SEQ ID NOs: 12-28) and any variants thereof, dual genes, homologs thereof , mutants, derivatives, fragments and complementary sequences. Examples of antisense oligonucleotides are set forth in SEQ ID NOs: 29-94. Another embodiment provides a method of modulating the function and/or expression of a SCNA polynucleotide in a cell or tissue of a patient in vivo or ex vivo, comprising affixing the cell or tissue to a length of 5 to 30 nucleotides The antisense oligonucleotide is contacted, wherein the oligonucleotide has at least 50% sequence identity to the antisense reverse complement of the SCNA polynucleotide; thereby modulating the patient's cells or tissues in vivo or in vitro The function and/or performance of the SCNA polynucleotide. Another embodiment provides a method of modulating the function and/or expression of a SCNA polynucleotide in a cell or tissue of a patient in vivo or ex vivo, comprising affixing the cell or tissue to a length of 5 to 30 nucleotides Antisense oligonucleotide contact, wherein the oligonucleotide has at least 50% sequence complementarity to the SCNA native antisense transcript; thereby modulating SCNA polynucleotides in a patient's cells or tissues in vivo or in vitro Function and / or performance. In one embodiment, the composition comprises one or more antisense oligonucleotides that bind to a sense and/or antisense SCNA polynucleotide, wherein the polynucleotides are selected from SCNA to SCN12A and variants thereof a group of body composition. In a preferred embodiment, the polynucleotide of interest is selected from the group consisting of SCNA. In one embodiment, the oligonucleotide comprises one or more modified or substituted nucleotides. In one embodiment, the oligonucleotide comprises one or more modified linkages. In another embodiment, the modified nucleotide comprises a phosphorothioate, a methylphosphonate, a peptide nucleic acid, a 2'-O-methyl, a fluoro- or carbon, a methylene group or other locked nucleic acid ( Locked nucleic acid, LNA) A modified base of a molecule. Preferably, the modified nucleotide is a locked nucleic acid molecule, including alpha-L-LNA. In one embodiment, the oligonucleotide is administered subcutaneously, intramuscularly, intravenously or intraperitoneally to the patient. In one embodiment, the oligonucleotide is administered as a pharmaceutical composition. The treatment regimen comprises administering to the patient an antisense compound at least once; however, the treatment can be modified to include multiple doses over a period of time. Treatment can be combined with one or more other types of therapies. In one embodiment, the oligonucleotide is encapsulated in a liposome or linked to a carrier molecule (eg, cholesterol, TAT peptide). Other aspects are described below.

以下參考用於說明之例示性應用來描述本發明之若干態樣。應瞭解闡述眾多特定詳情、關係及方法以全面瞭解本發明。然而,一般技術者將容易認識到本發明可在無一或多個特定詳情之情況下或用其他方法加以實施。本發明不受行為或事件之次序限制,因為某些行為可能以不同次序及/或與其他動作或事件並行發生。此外,並非所有經說明之動作或事件皆為實施本發明之方法所需。 本文揭示之所有基因、基因名稱及基因產物皆意欲對應於來自本文揭示之組合物及方法所適用之任何物種的同源物。因此,術語包括(但不限於)來自人類及小鼠之基因及基因產物。應瞭解當揭示來自特定物種之基因或基因產物時,除非其所出現之上下文明確指明,否則此揭示內容僅意欲具有例示性,且不應解釋為限制。因此,舉例而言,對於在一些實施例中與哺乳動物核酸及胺基酸序列相關之本文揭示之基因,其意欲涵蓋來自其他動物(包括(但不限於)其他哺乳動物、魚類、兩棲動物、爬行動物及鳥類)之同源及/或直系同源基因及基因產物。在一實施例中,基因或核酸序列為人類基因或核酸序列。定義 本文所用之術語係僅出於描述特定實施例之目的且不意欲限制本發明。如本文所用,除非上下文另外明確指明,否則單數形式「一」及「該」亦意欲包括複數形式。此外,就術語「包括」、「具有」或其變化形式用於[實施方式]及/或申請專利範圍中而言,此等術語意欲以與術語「包含」類似之方式具有包括性。 術語「約」意謂在如由一般技術者測定之特定值之可接受誤差範圍內,該誤差範圍將部分視如何量測或測定該值,亦即量測系統之限制而定。舉例而言,「約」可意謂根據此項技術中之慣例,在1或1以上之標準偏差內。或者,「約」可意謂既定值之至多20%、較佳至多10%、更佳至多5%、且仍然更佳至多1%之範圍。或者,特別就生物系統或方法而言,術語可意謂在某一值之一定數量級內,較佳在5倍內、且更佳在2倍內。當在本申請案及申請專利範圍中描述特定值時,除非另有陳述,否則術語「約」意味應假設在特定值之可接受誤差範圍內。 如本文所用,術語「mRNA」意謂靶向基因之目前已知mRNA轉錄物及可闡明之任何其他轉錄物。 就「反義寡核苷酸」或「反義化合物」而言,其意謂與另一RNA或DNA(目標RNA、DNA)結合之RNA或DNA分子。舉例而言,若其為RNA寡核苷酸,則其藉助於RNA-RNA相互作用與另一RNA目標結合且改變目標RNA之活性。反義寡核苷酸可上調或下調特定聚核苷酸之表現及/或功能。定義意欲包括就治療、診斷或其他觀點看來適用之任何外來RNA或DNA分子。此等分子包括例如反義RNA或DNA分子、干擾RNA(RNAi)、微RNA、誘餌RNA分子、siRNA、酶促RNA、治療性編輯RNA及促效劑及拮抗劑RNA、反義寡聚化合物、反義寡核苷酸、外部引導序列(external guide sequence,EGS)寡核苷酸、替代性剪接物(alternate splicer)、引子、探針、及與至少一部分目標核酸雜交之其他寡聚化合物。因而,此等化合物可以單股、雙股、部分單股、或環形寡聚化合物形式引入。 在本發明之情形下,術語「寡核苷酸」係指核糖核酸(RNA)或去氧核糖核酸(DNA)之寡聚物或聚合物或其模擬物。術語「寡核苷酸」亦包括天然及/或經修飾之單體或鍵(包括去氧核糖核苷、核苷、其經取代及α向差異構形式、肽核酸(PNA)、鎖核酸(LNA)、硫代磷酸酯、甲基膦酸酯及其類似物)之線性或環形寡聚物。寡核苷酸能夠藉由規則樣式之單體對單體相互作用(諸如華特生-克里克(Watson-Crick)類型之鹼基配對、胡格斯丁(Hoögsteen)或反向胡格斯丁類型之鹼基配對、或其類似相互作用)來特異性結合目標聚核苷酸。 寡核苷酸可為「嵌合的」,亦即,由不同區域構成。在本發明之情形下,「嵌合」化合物為寡核苷酸,其含有兩個或兩個以上化學區域,例如DNA區域、RNA區域、PNA區域等。各化學區域由至少一個單體單元,亦即在寡核苷酸化合物之情況下之核苷酸構成。此等寡核苷酸通常包含至少一個其中寡核苷酸經修飾以展現一或多種所要性質之區域。寡核苷酸之所要性質包括(但不限於)例如對核酸酶降解增加之抗性、增加之細胞攝取、及/或對目標核酸增加之結合親和力。因此,寡核苷酸之不同區域可具有不同性質。本發明之嵌合寡核苷酸可形成為兩種或兩種以上如上所述之寡核苷酸、經修飾之寡核苷酸、寡核苷及/或寡核苷酸類似物的混合結構。 寡核苷酸可由「互相對準(register)」連接(亦即,當單體如天然DNA中一樣連續連接時)或經由間隔子連接之區域構成。間隔子意欲在區域之間構成共價「橋」且在較佳情況中具有不超過約100個碳原子之長度。間隔子可帶有不同功能性,例如具有正電荷或負電荷、帶有特殊核酸結合性質(嵌入劑、凹槽結合劑(groove binder)、毒素、螢光團等)、具有親脂性、誘導特殊二級結構,如例如會誘導α-螺旋之含有丙胺酸之肽。 如本文所用,「SCN1A」包括所有家族成員、突變體、對偶基因、片段、物質、編碼及非編碼序列、有義及反義聚核苷酸股等。類似地,SCN2A-SCN12A包括所有突變體、對偶基因、片段等。 如本文所用,措詞「電壓門控鈉離子通道第I型α次單元」、SCN1A、FEB3、FEB3A、GEFSP2、HBSCI、NAC1、Nav1.1、SCN1、SMEI、鈉離子通道蛋白腦I次單元α、鈉離子通道蛋白第1型次單元α、鈉離子通道蛋白第I型次單元α及電壓門控鈉離子通道次單元α Nav1.1在文獻中視為相同且在本申請案中可互換使用。 如本文所用,術語「對…具有特異性之寡核苷酸」或「靶向…之寡核苷酸」係指具有(i)能夠與一部分靶向基因形成穩定複合物,或(ii)能夠與靶向基因之一部分mRNA轉錄物形成穩定雙螺旋的序列之寡核苷酸。複合物及雙螺旋之穩定性可藉由理論計算及/或活體外檢定來確定。用於確定雜交複合物及雙螺旋之穩定性之例示性檢定描述於以下實例中。 如本文所用,術語「目標核酸」涵蓋DNA、自此DNA轉錄之RNA(包含前mRNA(premRNA)及mRNA)、以及源於此RNA之cDNA、編碼序列、非編碼序列、有義或反義聚核苷酸。寡聚化合物與其目標核酸之特異性雜交會干擾核酸之正常功能。與目標核酸特異性雜交之化合物對該目標核酸功能之此調節通常稱為「反義(antisense)」。將會受到干擾之DNA功能包括例如複製及轉錄。將會受到干擾之RNA功能包括所有生命功能,諸如RNA向蛋白質轉譯位點之易位、蛋白質自RNA之轉譯、RNA之剪接以產生一或多種mRNA物質、及RNA可參與或促進之催化活性。對目標核酸功能之此干擾之總體效應為調節經編碼之產物或寡核苷酸之表現。 RNA干擾「RNAi」由與「目標」核酸序列具有序列特異性同源性之雙股RNA(dsRNA)分子介導。在本發明之某些實施例中,介體為含5-25個核苷酸之「小干擾」RNA雙螺旋(siRNA)。siRNA係由稱為Dicer之RNA酶對dsRNA進行加工產生。siRNA雙螺旋產物經募集進入稱為RISC(RNA誘導之靜止複合物)之多蛋白siRNA複合物中。在不希望受任何特定理論束縛之情況下,咸信RISC接著經引導至目標核酸(適合為mRNA),在此處,siRNA雙螺旋以序列特異性方式進行相互作用來以催化方式介導裂解。可根據本發明使用之小干擾RNA可根據此項技術中熟知且將為一般技術者所熟悉之程序合成並使用。用於本發明方法中之小干擾RNA適合地包含約1個至約50個之間的核苷酸(nt)。在非限制性實施例之實例中,siRNA可包含約5至約40個nt、約5至約30個nt、約10至約30個nt、約15至約25個nt、或約20-25個核苷酸。 藉由使用自動比對核酸序列且指示具有一致性或同源性之區域之電腦程式來促進對適當寡核苷酸之選擇。此等程式用於例如藉由搜尋諸如GenBank之資料庫或藉由對PCR產物定序來比較所得核酸序列。來自一定範圍之物種之核酸序列的比較允許對顯示物種之間之適當一致性程度的核酸序列進行選擇。在尚未經定序之基因之情況下,進行南方墨點分析(Southern blot)以允許確定目標物種中之基因與其他物種中之基因之間的一致性程度。如此項技術中所熟知,藉由在不同嚴格性程度下進行南方墨點分析,有可能獲得一致性之近似度量。此等程序使得可選擇與欲經控制之個體中之目標核酸序列具有高互補程度且與其他物種中之相應核酸序列具有較低互補程度的核酸序列。熟習此項技術者將認識到在選擇用於本發明中之基因之適當區域方面有很大的自由。 就「酶促RNA」而言,其意謂具有酶促活性之RNA分子(Cech, (1988)J. American. Med. Assoc . 260, 3030-3035)。酶促核酸(核糖核酸酶)藉由與目標RNA結合來起作用。此結合經由酶促核酸之目標結合部分發生,該目標結合部分保持密切鄰近於分子之起裂解目標RNA作用之酶促部分。因此,酶促核酸首先識別目標RNA且接著經由鹼基配對結合目標RNA,且一旦與正確位點結合,即以酶促方式起作用來切割目標RNA。 就「誘餌RNA」而言,其意謂模擬配位體之天然結合域之RNA分子。因此,誘餌RNA與天然結合目標競爭結合特定配位體。舉例而言,已顯示過度表現之HIV反式活化反應(trans-activation response;TAR)RNA可充當「誘餌」且高效結合HIV tat蛋白,藉此阻止其與HIV RNA中編碼之TAR序列結合。此意欲為一特定實例。此項技術中之相關人員應認識到此僅為一實例,且其他實施例容易使用此項技術中通常已知之技術產生。 如本文所用,術語「單體」通常指示經磷酸二酯鍵或其類似物連接以形成大小在數個單體單元(例如約3-4個單體單元)至約數百個單體單元之範圍內之寡核苷酸的單體。磷酸二酯鍵之類似物包括:硫代磷酸酯、二硫代磷酸酯、甲基膦酸酯、硒代磷酸酯、胺基磷酸酯及其類似物,如下更充分描述。 術語「核苷酸」涵蓋天然存在之核苷酸以及非天然存在之核苷酸。熟習此項技術者應清楚先前已視為「非天然存在」之各種核苷酸隨後已在自然界中發現。因此,「核苷酸」不僅包括已知之含有嘌呤及嘧啶雜環之分子,而且亦包括其雜環類似物及互變異構體。其他類型之核苷酸之說明性實例為含有以下之分子:腺嘌呤、鳥嘌呤、胸腺嘧啶、胞嘧啶、尿嘧啶、嘌呤、黃嘌呤、二胺基嘌呤、8-側氧基-N6-甲基腺嘌呤、7-去氮黃嘌呤、7-去氮鳥嘌呤、N4,N4-橋亞乙基胞嘧啶(N4,N4-ethanocytosin)、N6,N6-橋亞乙基-2,6-二胺基嘌呤、5-甲基胞嘧啶、5-(C3-C6)-炔基胞嘧啶、5-氟尿嘧啶、5-溴尿嘧啶、假異胞嘧啶、2-羥基‑5-甲基-4-三唑并吡啶、異胞嘧啶、異鳥嘌呤、肌苷,及Benner等人,美國專利第5,432,272號中描述之「非天然存在之」核苷酸。術語「核苷酸」意欲涵蓋每一及所有此等實例以及其類似物及互變異構體。尤其引起注意之核苷酸為含有腺嘌呤、鳥嘌呤、胸腺嘧啶、胞嘧啶及尿嘧啶之核苷酸,其視為與人類中之治療及診斷應用相關之天然存在的核苷酸。核苷酸包括例如如Kornberg及Baker, DNA Replication,第2版 (Freeman, San Francisco, 1992)中所述之天然2'-去氧及2'-羥基糖以及其類似物。 關於核苷酸之「類似物」包括具有經修飾之鹼基部分及/或經修飾之糖部分(參見例如通常由Scheit, Nucleotide Analogs, John Wiley, New York, 1980;Freier及Altmann, (1997)Nucl. Acid. Res ., 25(22), 4429-4443;Toulmé, J.J., (2001)Nature Biotechnology 19:17-18;Manoharan M., (1999)Biochemica et Biophysica Acta 1489:117-139;Freier S. M., (1997)Nucleic Acid Research , 25:4429-4443;Uhlman, E., (2000)Drug Discovery & Development , 3: 203-213;Herdewin P., (2000)Antisense & Nucleic Acid Drug Dev. , 10:297-310所述)的合成核苷酸;2'-O、3'-C連接之[3.2.0]雙環阿糖核苷。此等類似物包括經設計以增強結合性質,例如雙螺旋或三螺旋穩定性、特異性或其類似性質之合成核苷酸。 如本文所用,「雜交」意謂寡聚化合物之實質上互補股之配對。一種配對機制涉及寡聚化合物之股之互補核苷或核苷酸鹼基(核苷酸)之間的氫鍵結,其可為華特生-克里克、胡格斯丁或反向胡格斯丁氫鍵結。舉例而言,腺嘌呤及胸腺嘧啶為經由形成氫鍵而配對之互補核苷酸。雜交可在不同情況下發生。 當反義化合物與目標核酸之結合會干擾目標核酸之正常功能從而導致對功能及/或活性之調節,且存在足夠程度之互補性以避免該反義化合物與非目標核酸序列在特異性結合所要之條件下(亦即在活體內檢定或治療性治療之情況下為在生理條件下,及在活體外檢定之情況下為在進行檢定之條件下)進行非特異性結合時,該反義化合物為「可特異性雜交的」。 如本文所用,片語「嚴格雜交條件」或「嚴格條件」係指本發明化合物將與其目標序列但與最小數目之其他序列雜交的條件。嚴格條件具有序列依賴性且在不同情況下將不同,且在本發明之情形下,寡聚化合物與目標序列雜交之「嚴格條件」係由寡聚化合物之性質及組成以及對其進行研究之檢定決定。一般而言,嚴格雜交條件包含低濃度(<0.15 M)之具有無機陽離子(諸如Na++或K++)之鹽(亦即低離子強度)、在寡聚化合物:目標序列複合物之Tm以下高於20℃-25℃之溫度、諸如甲醯胺、二甲基甲醯胺、二甲亞碸或清潔劑十二烷基硫酸鈉(SDS)之變性劑之存在。舉例而言,對於每1%甲醯胺,雜交率減小1.1%。高嚴格性雜交條件之一實例為0.1×氯化鈉-檸檬酸鈉緩衝液(SSC)/0.1%(w/v)SDS,在60℃下,持續30分鐘。 如本文所用之「互補」係指一或兩個寡聚股上之兩個核苷酸之間精確配對的能力。舉例而言,若在反義化合物之某一位置處之核鹼基(nucleobase)能夠與在目標核酸(該目標核酸為DNA、RNA或寡核苷酸分子)之某一位置處之核鹼基氫鍵結,則寡核苷酸與目標核酸之間之氫鍵結的位置視為互補位置。當各分子中有足夠數目之互補位置由可彼此氫鍵結之核苷酸佔據時,寡聚化合物與其他DNA、RNA或寡核苷酸分子彼此互補。因此,「可特異性雜交」及「互補」為用於指示足夠數目之核苷酸足夠程度之精確配對或互補性以便在寡聚化合物與目標核酸之間發生穩定及特異性結合的術語。 在此項技術中應瞭解達成可特異性雜交無需寡聚化合物之序列100%互補於其目標核酸之序列。此外,寡核苷酸的一或多個區段可雜交以使介入或鄰近區段不涉及於雜交事件(例如環結構、錯配或髮夾結構)中。本發明之寡聚化合物包含與其靶向之目標核酸序列內之目標區域的至少約70%、或至少約75%、或至少約80%、或至少約85%、或至少約90%、或至少約95%、或至少約99%序列互補性。舉例而言,反義化合物之20個核苷酸中有18個核苷酸互補於目標區域且因此將特異性雜交之反義化合物將表示90%互補性。在此實例中,其餘非互補核苷酸可叢集或經互補核苷酸穿插且無需彼此鄰接或與互補核苷酸鄰接。因而,長度為18個核苷酸且具有經與目標核酸完全互補之兩個區域側接之4(四)個非互補核苷酸的反義化合物將與目標核酸具有77.8%總體互補性且因此將屬於本發明之範疇。具有目標核酸之區域之反義化合物的互補性百分比可使用此項技術中已知之BLAST程式(基本局部比對搜尋工具)及PowerBLAST程式按常規方式確定。同源性百分比、序列一致性或互補性可藉由例如使用Smith及Waterman演算法(Adv. Appl. Math ., (1981) 2, 482-489)之Gap程式(威斯康星序列分析套裝(Wisconsin Sequence Analysis Package), 用於Unix之第8版, Genetics Computer Group, University Research Park, Madison Wis.),使用預設設置加以確定。 如本文所用,術語「熱熔點(Tm)」係指在確定的離子強度、pH值及核酸濃度下,50%之互補於目標序列之寡核苷酸與處於均衡狀態的目標序列雜交之溫度。通常,嚴格條件將為以下條件:鹽濃度為至少約0.01至1.0 M Na離子濃度(或其他鹽),在pH 7.0至8.3下,且對於短寡核苷酸(例如10至50個核苷酸),溫度為至少約30℃。嚴格條件亦可在添加諸如甲醯胺之去穩定劑下達成。 如本文所用,「調節」意謂基因表現增加(刺激)或減小(抑制)。 當用於聚核苷酸序列之情形下時,術語「變異體」可涵蓋與野生型基因相關之聚核苷酸序列。此定義亦可包括例如「對偶基因」、「剪接」、「物種」或「多形性」變異體。剪接變異體可與參照分子具有顯著一致性,但歸因於mRNA加工期間外顯子之替代性剪接,通常將具有較大或較小數目之聚核苷酸。相應多肽可具有其他功能域或不存在功能域。物種變異體為自一物種至另一物種變化之聚核苷酸序列。在本發明中,野生型基因產物之變異體具有特定效用。變異體可由核酸序列中之至少一個突變產生且可產生改變之mRNA或結構或功能可能或可能不改變之多肽。任何既定天然或重組基因可不具有對偶基因形式或具有1個或許多對偶基因形式。產生變異體之常見突變變化通常歸因於核苷酸之天然缺失、添加或取代。每一此等類型之變化可單獨或與其他變化組合在既定序列中發生一或多次。 所得多肽相對於彼此通常將具有顯著胺基酸一致性。多形性變異體為既定物種之個體之間的特定基因之聚核苷酸序列有變化。多形性變異體亦可涵蓋「單核苷酸多形現象」(SNP)或聚核苷酸序列有1個鹼基發生變化之單鹼基突變。SNP之存在可指示例如某一群體具有疾病狀態傾向,亦即對抗性之敏感性。 衍生聚核苷酸包括經受化學修飾(例如用烷基、醯基或胺基置換氫)之核酸。例如衍生寡核苷酸之衍生物可包含非天然存在之部分,諸如改變之糖部分或糖間鍵。此等中之例示者為硫代磷酸酯及此項技術中已知之其他含硫物質。衍生核酸亦可含有標記,包括放射性核苷酸、酶、螢光劑、化學發光劑、顯色劑、受質、輔因子、抑制劑、磁性粒子及其類似物。 「衍生」多肽或肽為例如藉由糖基化、聚乙二醇化、磷酸化、硫酸化、還原/烷基化、醯化、化學偶合、或適度福馬林(formalin)處理加以修飾之多肽或肽。衍生物亦可直接或間接經修飾以含有可偵測標記,包括(但不限於)放射性同位素、螢光標記及酶標記。 如本文所用,術語「動物」或「患者」意欲包括例如人類、綿羊、麌、鹿、長耳鹿(mule deer)、貂、哺乳動物、猴、馬、牛、豬、山羊、犬、貓、大鼠、小鼠、鳥類、雞、爬行動物、魚類、昆蟲及蜘蛛類動物。 「哺乳動物」涵蓋通常在醫療照護下之溫血哺乳動物(例如人類及馴化動物)。實例包括貓科動物、犬科動物、馬科動物、牛科動物及人類,以及僅人類。 「治療」涵蓋治療哺乳動物中之疾病狀態,且包括:(a)預防該疾病狀態在哺乳動物中發生,特定言之,當此哺乳動物易感染該疾病狀態但尚未診斷為患有其時;(b)抑制該疾病狀態,例如遏止其發展;及/或(c)減輕該疾病狀態,例如導致該疾病狀態之消退直至達到所要終點。治療亦包括改善疾病之症狀(例如減輕疼痛或不適),其中此改善可能或可能不直接影響該疾病(例如病因、傳播、表現等)。 如本文所用,「神經疾病或病症」係指神經系統及/或視覺系統之任何疾病或病症。「神經疾病或病症」包括涉及中樞神經系統(腦、腦幹及小腦)、周邊神經系統(包括腦神經)及自主神經系統(其部分位於中樞神經系統與周邊神經系統兩者中)之疾病或病症。神經疾病或病症包括(但不限於)後天性癲癇狀失語症(acquired epileptiform aphasia);急性播散性腦脊髓炎(acute disseminated encephalomyelitis);腎上腺腦白質營養不良(adrenoleukodystrophy);年齡相關之黃斑部變性(age-related macular degeneration);胼胝體發育不全(agenesis of the corpus callosum);認知障礙症(agnosia);艾卡迪症候群(Aicardi syndrome);亞歷山大病(Alexander disease);阿爾珀斯病(Alpers' disease);交替性偏癱(alternating hemiplegia);阿茲海默氏病(Alzheimer's disease);血管性癡呆(Vascular dementia);肌萎縮性側索硬化(amyotrophic lateral sclerosis);無腦(anencephaly);安琪曼症候群(Angelman syndrome);血管瘤病(angiomatosis);缺氧症(anoxia);失語症(aphasia);精神性運動不能(apraxia);蛛網膜囊腫(arachnoid cysts);蛛網膜炎(arachnoiditis);阿諾尼-加里畸形(Anronl-Chiari malformation);動靜脈畸形(arteriovenous malformation);亞斯伯格症候群(Asperger syndrome);共濟失調毛細管擴張症(ataxia telegiectasia);注意力不足過動症(attention deficit hyperactivity disorder);自閉症(autism);自主功能障礙(autonomic dysfunction);背痛;巴登病(Batten disease);白塞氏病(Behcet's disease);貝爾氏麻痺(Bell's palsy);良性原發性瞼痙攣(benign essential blepharospasm);良性病灶(benign focal);肌萎縮(amyotrophy);良性顱內高壓;賓斯萬格氏病(Binswanger's disease);瞼痙攣(blepharospasm);布洛克-蘇茲貝格症候群(Bloch Sulzberger syndrome);臂叢損傷(brachial plexus injury);腦膿腫(brain abscess);腦損傷;腦腫瘤(包括多形性膠質母細胞瘤(glioblastoma multiforme));脊椎腫瘤;布朗-塞卡爾症候群(Brown-Sequard syndrome);卡納文病(Canavan disease);腕隧道徵候群(carpal tunnel syndrome);灼性神經痛(causalgia);中樞性疼痛症候群;中央腦橋脊髓溶解(central pontine myelinolysis);頭部病症;腦動脈瘤(cerebral aneurysm);腦動脈硬化(cerebral arteriosclerosis);腦萎縮(cerebral atrophy);大腦性巨人症(cerebral gigantism);大腦性麻痺(cerebral palsy);夏科-馬里-圖斯病(Charcot-Marie-Tooth disease);化學療法誘發之神經病及神經痛;加里畸形(Chiari malformation);舞蹈病(chorea);慢性發炎性脫髓鞘多發性神經病(chronic inflammatory demyelinating polyneuropathy);慢性疼痛;慢性區域疼痛症候群;科芬-勞里症候群(Coffin Lowry syndrome);昏迷(coma),包括持續性增長狀態;先天性兩側面癱(congenital facial diplegia);皮質基底核退化症(corticobasal degeneration);顱動脈炎(cranial arteritis);顱蓋骨折(craniosynostosis);庫賈氏病(Creutzfeldt-Jakob disease);累積性損傷病症;庫欣氏症候群(Cushing's syndrome);巨大細胞包涵體病(cytomegalic inclusion body disease);細胞巨大病毒感染;舞蹈眼-舞蹈足症候群(dancing eyes-dancing feet syndrome);丹尼沃克症候群(DandyWalker syndrome);道森病(Dawson disease);德莫斯特氏症候群(De Morsier's syndrome);代哲因-科魯姆克麻痺(Dejerine-Klumke palsy);癡呆;皮肌炎(dermatomyositis);糖尿病性神經病變;彌漫性硬化症(diffuse sclerosis);德拉威特氏自主神經障礙(Dravet's, dysautonomia);書寫困難(dysgraphia);閱讀困難(dyslexia);肌張力障礙(dystonias);早期嬰兒癲癇腦病(early infantile epileptic encephalopathy);空蝶鞍症候群(empty sella syndrome);腦炎(encephalitis);腦膨出(encephaloceles);腦三叉血管瘤病(encephalotrigeminal angiomatosis);癲癇症(epilepsy);厄博氏麻痺(Erb's palsy);原發性震顫(essential tremor);法布瑞氏病(Fabry's disease);華氏症候群(Fahr's syndrome);暈厥(fainting);家族性痙攣性麻痺(familial spastic paralysis);熱性發作(febrile seizures);費雪症候群(Fisher syndrome);弗里德棘希氏共濟失調(Friedreich's ataxia);額顳葉型癡呆及其他「τ病變」(fronto-temporal dementia and other「tauopathies」);高雪氏病(Gaucher's disease);格斯特曼氏症候群(Gerstmann's syndrome);巨細胞動脈炎(giant cell arteritis);巨細胞性包涵體病(giant cell inclusion disease);球狀細胞白血質障礙(globoid cell leukodystrophy);古立安-白瑞症候群(Guillain-Barre syndrome);HTLV-1相關之脊髓病(HTLV-1-associated myelopathy);哈勒沃登-施帕茨病(Hallervorden-Spatz disease);頭部損傷;頭痛;半面痙攣(hemifacial spasm);遺傳性痙孿性截癱(hereditary spastic paraplegia);多神經炎型遺傳性共濟失調(heredopathia atactic a polyneuritiformis);耳部帶狀疱疹(herpes zoster oticus);帶狀疱疹(herpes zoster);平山症候群(Hirayama syndrome);HIV相關之癡呆及神經病(亦為AIDS之神經學表現);前腦無裂畸形(holoprosencephaly);亨廷頓氏病(Huntington's disease)及其他聚麩醯胺酸重複疾病;腦內積水(hydranencephaly);腦積水(hydrocephalus);高皮質醇症(hypercortisolism);低氧(hypoxia);免疫介導之腦脊髓炎;包涵體肌炎;色素失調症(incontinentia pigmenti);嬰兒植烷酸儲積病(infantile phytanic acid storage disease);嬰兒雷夫蘇姆病(infantile refsum disease);嬰兒痙攣症(infantile spasms);發炎性肌病;顱內囊腫;顱內高壓;喬伯特症候群(Joubert syndrome);卡恩斯-塞爾症候群(Keams-Sayre syndrome);肯尼迪病(Kennedy disease)金斯布恩症候群(Kinsboume syndrome);克利佩爾-費爾症候群(Klippel Feil syndrome);克拉培病(Krabbe disease);庫格爾貝格-韋蘭德病(Kugelberg-Welander disease);庫魯症(kuru);拉弗拉病(Lafora disease);蘭伯特-伊頓重肌無力症候群(Lambert-Eaton myasthenic syndrome);蘭道-科萊弗勒症候群(Landau-Kleffner syndrome);側髓(瓦倫伯格)症候群(lateral medullary (Wallenberg) syndrome);學習困難(learning disabilities);雷氏病(Leigh's disease);勒諾克斯-古斯托特症候群(Lennox-Gustaut syndrome);萊施-奈恩症候群(Lesch-Nyhan syndrome);腦白質營養不良(leukodystrophy);路易體性癡呆(Lewy body dementia);無腦回症(Lissencephaly);閉鎖症候群(locked-in syndrome);盧•賈里格氏病(Lou Gehrig's disease)(亦即運動神經元病或肌萎縮性側索硬化);腰椎盤疾病;萊姆病(lyme disease)--神經後遺症;馬查多-約瑟夫病(Machado-Joseph disease);巨腦畸形(macrencephaly);巨腦畸形(megalencephaly);梅爾克遜-羅森塔爾症候群(Melkersson-Rosenthal syndrome);梅尼爾氏病(Menieres disease);腦膜炎;緬克斯病(Menkes disease);異染性腦白質營養不良(metachromatic leukodystrophy);小頭畸形(microcephaly);偏頭痛;米勒費雪症候群(Miller Fisher syndrome);小中風(mini-strokes);粒線體肌病(mitochondrial myopathies);默比厄斯症候群(Mobius syndrome);單肢肌萎縮;運動神經元病;毛毛樣腦血管病(Moyamoya disease);黏多醣病(mucopolysaccharidoses);多梗塞性癡呆(milti-infarct dementia);多病灶運動神經病(multifocal motor neuropathy);多發性硬化症及其他脫髓鞘病症;伴有體位性低血壓之多發性系統萎縮症(multiple system atrophy with postural hypotension);肌肉萎縮症;重症肌無力;脫髓鞘彌漫性硬化症(myelinoclastic diffuse sclerosis);嬰兒肌陣攣性腦病(myoclonic encephalopathy of infants);肌陣攣(myoclonus);肌病(myopathy);先天性肌強直(myotonia congenital);發作性睡病(narcolepsy);神經纖維瘤;精神抑制劑惡性症候群(neuroleptic malignant syndrome);AIDS之神經表現;狼瘡之神經後遺症(neurological sequelae of lupus);神經性肌強直(neuromyotonia);神經性類蠟質褐質病(neuronal ceroid lipofuscinosis);神經元遷移病症(neuronal migration disorders);尼曼-匹克病(Niemann-Pick disease);奧沙利文-麥克里德症候群(O'Sullivan-McLeod syndrome);枕神經痛(occipital neuralgia);潛隱性脊髓神經管縫合不全序列征(occult spinal dysraphism sequence);大田原症候群(Ohtahara syndrome);橄欖體腦橋小腦萎縮(olivopontocerebellar atrophy);斜視眼陣攣肌陣攣(opsoclonus myoclonus);視神經炎(optic neuritis);起立性低血壓(orthostatic hypotension);過度使用症候群(overuse syndrome);感覺異常;神經退化性疾病或病症(帕金森氏病(Parkinson's disease)、亨廷頓氏病(Huntington's disease)、阿茲海默氏病、肌萎縮性側索硬化(ALS)、癡呆、多發性硬化症及與神經元細胞死亡相關之其他疾病及病症);先天性肌強直病(paramyotonia congenital);副腫瘤疾病(paraneoplastic diseases);陣發性發作;帕里-羅格症候群(Parry Romberg syndrome);慢性兒童型腦硬化病(Pelizaeus-Merzbacher disease);週期性麻痺(periodic paralyses);周邊神經病(peripheral neuropathy);疼痛性神經病及神經痛;持續性增長狀態;滲透性發育病症(pervasive developmental disorders);感光性噴嚏反射(photic sneeze reflex);植烷酸儲積病;皮克氏病(Pick's disease);神經挫傷;垂體瘤;多發性肌炎;腦穿通畸形(porencephaly);小兒麻痺症後症候群(post-polio syndrome);帶狀疱疹後神經痛(postherpetic neuralgia);感染後腦脊髓炎(postinfectious encephalomyelitis);體位性低血壓;普威二氏症候群(Prader-Willi syndrome);原發性側索硬化(primary lateral sclerosis);朊病毒疾病(prion diseases);進行性面部單側萎縮(progressive hemifacial atrophy);進行性多病灶腦白質病(progressive multifocalleukoencephalopathy);進行性硬化性灰質萎縮(progressive sclerosing poliodystrophy);進行性核上眼神經麻痺症(progressive supranuclear palsy);腦假瘤(pseudotumor cerebri);拉姆西-亨特症候群(第I型及第11型);拉斯木森氏腦炎(Rasmussen's encephalitis);反射交感性營養不良症候群(reflex sympathetic dystrophy syndrome);雷夫蘇姆病;重複運動病症(repetitive motion disorders);重複應力損傷;腿不寧症候群(restless legs syndrome);反轉錄病毒相關之脊髓病;瑞特症候群(Rett syndrome);雷依氏症候群(Reye's syndrome);聖維特斯舞蹈病(Saint Vitus dance);山多夫病(Sandhoff disease);希爾逗氏病(Schilder's disease);腦裂(schizencephaly);中隔-眼發育不良(septo-optic dysplasia);搖晃嬰兒症候群(shaken baby syndrome);帶狀疱疹(shingles);夏-德里格症候群(Shy-Drager syndrome);休格連氏症候群(Sjogren's syndrome);睡眠呼吸暫停(sleep apnea);索特氏症候群(Soto's syndrome);痙攣(spasticity);脊椎裂;脊髓損傷;脊髓腫瘤;脊髓性肌萎縮;僵人症候群(Stiff-Person syndrome);中風;斯德奇-韋伯症候群(Sturge-Weber syndrome);亞急性硬化性全腦炎(subacute sclerosing panencephalitis);皮質下動脈硬化腦病(subcortical arteriosclerotic encephalopathy);西登哈姆舞蹈病(Sydenham chorea);昏厥(syncope);脊髓空洞症(syringomyelia);遲發性運動不能(tardive dyskinesia);泰-薩二氏病(Tay-Sachs disease);顳動脈炎(temporal arteritis);栓繫脊髓症候群(tethered spinal cord syndrome);托馬森病(Thomsen disease);胸部出口症候群(thoracic outlet syndrome);三叉神經痛(Tic Douloureux);托德氏麻痺(Todd's paralysis);妥瑞症候群(Tourette syndrome);短暫性缺血發作;傳染性海綿狀腦病(transmissible spongiform encephalopathies);橫貫性脊髓炎(transverse myelitis);創傷性腦損傷;震顫;三叉神經痛(trigeminal neuralgia);熱帶痙攣性後軀輕癱(tropical spastic paraparesis);結節性硬化症(tuberous sclerosis);血管性癡呆(多梗塞性癡呆);血管炎,包括顳動脈炎;逢希伯-林道病(Von Hippel-Lindau disease);瓦倫伯格氏症候群(Wallenberg's syndrome);偉-霍二氏病(Werdnig-Hoffman disease);韋斯特症候群(West syndrome);鞭抽式損傷(whiplash);威廉氏症候群(Williams syndrome);威爾頓氏病(Wildon's disease);及澤爾維格症候群(Zellweger syndrome)及本文敍述之其他神經病症。 心血管疾病或病症包括可導致缺血或由心臟再灌注導致之彼等病症。實例包括(但不限於)動脈粥樣硬化、冠狀動脈疾病、肉芽腫性心肌炎(granulomatous myocarditis)、慢性心肌炎(非肉芽腫性)、原發性肥厚型心肌病(primary hypertrophic cardiomyopathy)、周邊動脈疾病(PAD)、周邊血管疾病、靜脈血栓栓塞(venous thromboembolism)、肺栓塞(pulmonary embolism)、中風、心絞痛(angina pectoris)、心肌梗塞、由心跳驟停引起之心血管組織損傷、由心臟繞通(cardiac bypass)引起之心血管組織損傷、心原性休克、及將為一般技術者所知或涉及心臟或血管結構之功能障礙或組織損傷,尤其(但不限於)與SCNA活化相關之組織損傷的相關病狀。CVS疾病包括(但不限於)動脈粥樣硬化、肉芽腫性心肌炎、心肌梗塞、繼發於心臟瓣膜病之心肌纖維化、不伴有梗塞之心肌纖維化、原發性肥厚型心肌病、及慢性心肌炎(非肉芽腫性)。 與鈉離子通道功能障礙相關之疾病或病症之實例包括(但不侷限於)惡性高熱、肌無力、間歇性共濟失調、神經病性及發炎性疼痛、阿茲海默氏病、帕金森氏病、精神分裂症、過度驚跳症(hyperekplexia)、肌強直(諸如低血鉀性及高血鉀性週期性麻痺、先天性肌剛痙病(paramyotonia congenita)及鉀惡化之肌強直)以及心律不整(cardiac arrhythmias)(諸如長QT症候群)。聚核苷酸及寡核苷酸組合物及分子 目標: 在一實施例中,目標包含電壓門控鈉離子通道α次單元(SCNA)之核酸序列,包括(不限於)與SCNA相關之有義及/或反義非編碼及/或編碼序列。 電壓敏感性離子通道為一類跨膜蛋白,其提供細胞興奮性之基礎及經由離子產生之膜電位傳輸資訊之能力。回應膜電位之變化,此等分子介導離子穿過細胞膜中之選擇性通道快速流動。若通道密度足夠高,則產生稱為作用電位(action potential)之再生性去極化。 在大多數電可興奮細胞(包括神經元、心臟細胞及肌肉)中,電壓門控鈉離子通道負責產生及傳送作用電位。電活性藉由膜之去極化觸發,該去極化會打開穿過膜之對鈉離子具有高度選擇性之通道。離子接著由電化學梯度在細胞內驅動穿過開放通道。儘管不同組織中之基於鈉離子之作用電位類似,但電生理學研究已證明存在多種結構及功能不同之鈉離子通道,且已選殖編碼鈉離子通道之眾多基因。SCNA基因屬於電壓門控鈉離子通道之基因家族。 電壓門控鈉離子通道可根據Goldin等人(2000) Neuron 28:365-368中概述之標準化命名形式來命名。根據彼系統,電壓門控鈉離子通道集合成1個家族,已鑑別來自該家族之9個哺乳動物同功異型物且加以表現。此9個同功異型物給與名稱Navl.l至Navl.9。此外,各種同功異型物之剪接變異體係藉由在數字之後使用小寫字母(例如「Navl.la」)來加以區分。 電壓門控鈉離子通道在神經細胞及肌肉中之作用電位產生中起重要作用。α次單元(SCNA)為通道之主要組分,且在活體外表現於細胞中時將足以產生高效通道。反過來,β-1及2次單元需要α次單元以產生有效通道。此等次單元之作用將在於主要藉由使鈉離子流快速不活化來改進通道之動力學性質。相較於正常SCNB1,當與α次單元共表現時,於GEFS症候群中發現之於SCN1B基因上之突變顯示會降低鈉離子通道之快速不活化。 在一實施例中,反義寡核苷酸用於預防或治療與SCNA家族成員相關之疾病或病症。例示性電壓門控鈉離子通道α次單元(SCNA)介導之可用自使用反義化合物獲得之幹細胞再生之細胞/組織治療的疾病及病症包含:與SCNA之功能及/或表現異常相關之疾病或病症、神經疾病或病症、抽搐、疼痛(包括慢性疼痛)、涉及鈉離子通道功能障礙之受損電興奮性、與鈉離子通道功能障礙相關之疾病或病症、與電壓門控鈉離子通道α次單元活性之錯誤調控相關之疾病或病症(例如麻痺、高血鉀性週期性麻痺、先天性肌剛痙病、鉀惡化之肌強直、長Q-T症候群3、運動終板疾病(motor endplate disease)、共濟失調等)、歸因於腸神經系統之功能障礙之胃腸道疾病(例如結腸炎、迴腸炎、發炎性腸症候群等)、心血管疾病或病症(例如高血壓、充血性心臟衰竭等);涉及交感神經及副交感神經神經分佈之泌尿生殖道的疾病或病症(例如良性前列腺增生、陽痿);與神經肌肉系統相關之疾病或病症(例如肌肉萎縮症、多發性硬化症、癲癇症、自閉症、偏頭痛(例如偶發性及家族性偏癱性偏頭痛等)、嬰兒期重度肌痙攣癲癇症(SMEI或德拉威特氏症候群)、伴有熱性發作附加症之全身性癲癇症(GEFS+)等)及SCNA相關之發作病症。 本發明另外係關於一種包含至少一種寡核苷酸之醫藥組合物,該寡核苷酸靶向至少一或多個選自由以下組成之群之目標的天然反義轉錄物:SCN1A至SCN12A基因或其mRNA或同功異型物或變異體。本發明另外係關於一種治療神經疾病或病症之方法,其包含投與靶向至少一或多個選自由以下組成之群之目標的天然反義轉錄物之寡核苷酸:mRNA SCN1A、SCN2A、SCN3A、SCN4A、SCN5A、SCN6A、SCN7A、SCN8A、SCN9A、SCN10A、SCN11A及SCN12A或其變異體。在一較佳實施例中,選擇寡聚物以上調該SCNA家族之完全功能性表現產物的表現。在一較佳實施例中,本發明之寡聚物上調SCNXA基因家族之mRNA之任一者的轉錄及/或轉譯以在需要治療之患者中提供完全功能性鈉離子通道。在患有與突變形式之電壓門控鈉離子通道相關之疾病或病症的患者中,在一較佳實施例中,用包含靶向電壓門控鈉離子通道α基因或該種基因之mRNA的天然反義轉錄物之寡核苷酸的醫藥組合物投藥或治療會以大於由突變形式之基因產生之表現產物的上調之比率來上調完全功能性表現產物。在另一實施例中,本發明係關於靶向至少兩個SCNXA家族成員之至少一種天然反義轉錄物之寡核苷酸組合,其中X係選自1-12。舉例而言,在治療德拉威特氏症候群時,寡核苷酸之組合可用於上調例如SCN1A及SCN9A之表現產物。在另一實施例中,至少一種寡核苷酸可經選擇以靶向選自SCN1A至SCN12A之任一者之至少兩個基因的天然反義轉錄物。本發明之較佳寡核苷酸的長度介於約5至約30個核苷酸之間且至少50%互補於NAT之含5至約30個核苷酸之區段。SCNA基因或其轉錄產物之任一者之較佳NAT為在由本發明寡核苷酸靶向時會干擾且調節mRNA及/或該mRNA之轉譯產物之表現的NAT。在一較佳實施例中,寡核苷酸上調目標之功能性蛋白之表現以治療或減輕SCNA相關之疾病。在一較佳實施例中,此「上調」不與疾病(諸如癌症)之病因或促進相關。 SCNA基因中之改變可包括或涵蓋基因之編碼及/或非編碼區域中之許多或所有形式的基因突變,包括插入、缺失、重排及/或點突變。缺失可為整個基因或基因之一部分之缺失。點突變可產生胺基酸取代、框移位(frame shift)或終止密碼子。點突變亦可發生在SCNA基因之調控區域,諸如啟動子中,從而導致mRNA之表現損失或減少,或可導致對此mRNA之不當加工,從而引起穩定性或轉譯效率降低。人類中之此等改變可導致各種形式之疾病且有許多公開案描述SCNA基因中之改變與例如癲癇症或SMEI之關聯。此等改變可為「新生(de novo)」的或可遺傳。本發明不限於治療與SCNA基因中之改變相關之疾病且亦包括治療患者不具有或不必定具有SCNA基因中之改變或突變之SCNA相關的疾病或病狀。咸信功能性電壓門控鈉離子通道表現產物之任何調節或上調皆將導致減輕或治療需要治療之患者中的相關SCNA疾病或病狀。此減輕亦可包括臨床改善之至少一種可量測指標,包括較少之發作、不太頻繁之發作、不太嚴重之發作、顯現較少發作類型、神經發育改善或任何其他治療益處。 在一實施例中,向有需要之患者投與一或多種反義寡核苷酸對SCNA之調節以預防或治療與相較於正常對照為異常之SCNA表現、功能、活性相關的任何疾病或病症。 在一實施例中,寡核苷酸對包括(不限於)非編碼區域之SCNA之聚核苷酸具有特異性。SCNA目標包含SCNA之變異體;SCNA之突變體,包括SNP;SCNA之非編碼序列;對偶基因、片段及其類似物。較佳地,寡核苷酸為反義RNA分子。 根據本發明之實施例,目標核酸分子不僅僅限於SCNA聚核苷酸而係擴展至SCNA之同功異型物、受體、同源物、非編碼區域及其類似物中之任一者。 在一實施例中,寡核苷酸靶向SCNA目標之天然反義序列(編碼及非編碼區域之天然反義),包括(不限於)其變異體、對偶基因、同源物、突變體、衍生物、片段及互補序列。較佳地,寡核苷酸為反義RNA或DNA分子。 在一實施例中,本發明之寡聚化合物亦包括不同鹼基存在於化合物中之一或多個核苷酸位置處的變異體。舉例而言,若最初核苷酸為腺嘌呤,則可產生在此位置處含有胸苷、鳥苷、胞苷或其他天然或非天然核苷酸之變異體。此可在反義化合物之任何位置處進行。接著使用本文所述之方法測試此等化合物以確定其抑制目標核酸表現之能力。 在一些實施例中,反義化合物與目標之間的同源性、序列一致性或互補性為約50%至約60%。在一些實施例中,同源性、序列一致性或互補性為約60%至約70%。在一些實施例中,同源性、序列一致性或互補性為約70%至約80%。在一些實施例中,同源性、序列一致性或互補性為約80%至約90%。在一些實施例中,同源性、序列一致性或互補性為約90%、約92%、約94%、約95%、約96%、約97%、約98%、約99%或約100%。 當反義化合物與目標核酸之結合會干擾目標核酸之正常功能以致活性損失,且存在足夠程度之互補性以避免該反義化合物與非目標核酸序列在特異性結合所要之條件下進行非特異性結合時,該反義化合物為可特異性雜交的。此等條件包括亦即在活體內檢定或治療性治療之情況下的生理條件、及在活體外檢定之情況下進行檢定的條件。 當反義化合物(無論DNA、RNA、嵌合物、經取代之反義化合物等)與目標DNA或RNA分子之結合會干擾目標DNA或RNA之正常功能以致效用損失,且存在足夠程度之互補性以避免該反義化合物與非目標序列在特異性結合所要之條件下(亦即在活體內檢定或治療性治療之情況下為在生理條件下,及在活體外檢定之情況下為在進行檢定之條件下)進行非特異性結合時,該反義化合物為可特異性雜交的。 在一實施例中,靶向SCNA,包括(不限於)使用例如PCR、雜交等鑑別及擴增之反義序列;一或多種如SEQ ID NO: 12至28闡述之序列及其類似物會調節SCNA之表現或功能。在一實施例中,相較於對照,表現或功能經上調。在一實施例中,相較於對照,表現或功能經下調。 在一實施例中,寡核苷酸包含如SEQ ID NO: 29至94闡述之核酸序列,包括使用例如PCR、雜交等鑑別及擴增之反義序列。此等寡核苷酸可包含一或多個經修飾之核苷酸、較短或較長片段、經修飾之鍵及其類似物。經修飾之鍵或核苷酸間鍵聯之實例包含硫代磷酸酯、二硫代磷酸酯或其類似物。在一實施例中,核苷酸包含磷衍生物。可與本發明之經修飾之寡核苷酸中的糖或糖類似部分連接之磷衍生物(或經修飾之磷酸酯基)可為單磷酸酯、二磷酸酯、三磷酸酯、烷基磷酸酯、烷磷酸酯、硫代磷酸酯及其類似物。製備以上指示之磷酸酯類似物及將其併入核苷酸、經修飾之核苷酸及寡核苷酸中本身亦為已知的且無需在此處加以描述。 反義之特異性及敏感性亦由熟習此項技術者利用以達成治療用途。反義寡核苷酸已在動物及人類中之疾病狀態之治療中用作治療部分。反義寡核苷酸已向人類安全且有效投與且眾多臨床試驗目前正在進行中。因此確定寡核苷酸可為可經組態以適用於治療細胞、組織及動物,尤其人類之治療方案的適用治療模態(therapeutic modality)。 在本發明之實施例中,寡聚反義化合物,特定言之寡核苷酸與目標核酸分子結合且調節由目標基因編碼之分子之表現及/或功能。將會受到干擾之DNA功能包含例如複製及轉錄。將會受到干擾之RNA功能包含所有生命功能,諸如RNA向蛋白質轉譯之位點之易位、蛋白質自RNA之轉譯、RNA之剪接以產生一或多種mRNA物質、及RNA可參與或促進之催化活性。視所要功能而定,功能可經上調或抑制。 反義化合物包括反義寡聚化合物、反義寡核苷酸、外部引導序列(EGS)寡核苷酸、替代性剪接物、引子、探針、及與至少一部分目標核酸雜交之其他寡聚化合物。因而,此等化合物可以單股、雙股、部分單股、或環形寡聚化合物形式引入。 在本發明之情形下,使反義化合物靶向特定核酸分子可為多步驟過程。該過程通常以鑑別功能欲經調節之目標核酸開始。此目標核酸可為例如表現與特定病症或疾病狀態相關之細胞基因(或由基因轉錄之mRNA)、或來自感染物之核酸分子。在本發明中,目標核酸編碼電壓門控鈉離子通道α次單元(SCNA)。 靶向過程通常亦包括確定目標核酸內之用於發生反義相互作用以便將產生所要效應(例如調節表現)的至少一個目標區域、區段或位點。在本發明之情形內,術語「區域」定義為目標核酸之具有至少一種可鑑別結構、功能或特徵的一部分。區段在目標核酸之區域內。「區段」定義為目標核酸內之區域之較小部分或子部分。如本發明中使用之「位點」定義為目標核酸內之位置。 在一實施例中,反義寡核苷酸與電壓門控鈉離子通道α次單元(SCNA)之天然反義序列結合且調節SCNA(SEQ ID NO: 1至11)之表現及/或功能。天然反義序列之實例包括SEQ ID NO: 12至28。反義寡核苷酸之實例包括SEQ ID NO: 29至94。 在一實施例中,反義寡核苷酸與電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之一或多個區段結合且調節SCNA之表現及/或功能。區段包含SCNA有義或反義聚核苷酸之至少5個連續核苷酸。 在一實施例中,反義寡核苷酸對SCNA之天然反義序列具有特異性,其中寡核苷酸與SCNA之天然反義序列之結合會調節SCNA之表現及/或功能。 在一實施例中,寡核苷酸化合物包含如SEQ ID NO: 29至94闡述之序列,包括使用例如PCR、雜交等鑑別及擴增之反義序列。此等寡核苷酸可包含一或多個經修飾之核苷酸、較短或較長片段、經修飾之鍵及其類似物。經修飾之鍵或核苷酸間鍵聯之實例包含硫代磷酸酯、二硫代磷酸酯或其類似物。在一實施例中,核苷酸包含磷衍生物。可與本發明之經修飾之寡核苷酸中的糖或糖類似部分連接之磷衍生物(或經修飾之磷酸酯基)可為單磷酸酯、二磷酸酯、三磷酸酯、烷基磷酸酯、烷磷酸酯、硫代磷酸酯及其類似物。製備以上指示之磷酸酯類似物及將其併入核苷酸、經修飾之核苷酸及寡核苷酸中本身亦為已知的且無需在此處加以描述。 如此項技術中已知,因為轉譯起始密碼子通常為5'-AUG (在轉錄之mRNA分子中;在相應DNA分子中為5'-ATG),所以轉譯起始密碼子亦稱為「AUG密碼子」、「起始密碼子」或「AUG起始密碼子」。少數基因具有之轉譯起始密碼子具有RNA序列5'-GUG、5'-UUG或5'-CUG;且5'‑AUA、5'-ACG及5'-CUG已顯示在活體內起作用。因此,術語「轉譯起始密碼子」及「起始密碼子」可涵蓋許多密碼子序列,即使在各情況下之起始胺基酸通常為甲硫胺酸(在真核生物中)或甲醯甲硫胺酸(在原核生物中)。真核及原核基因可具有兩個或兩個以上替代性起始密碼子,其任一者可優先用於在特定細胞類型或組織中或在特定條件集合下起始轉譯。在本發明之情形下,「起始密碼子」及「轉譯起始密碼子」係指在活體內用於起始由編碼電壓門控鈉離子通道α次單元(SCNA)之基因所轉錄之mRNA的轉譯之密碼子,無論此等密碼子之序列如何。基因之轉譯終止密碼子(或「終止密碼子」)可具有以下三個序列之一:亦即5'-UAA、5'-UAG及5'-UGA(相應DNA序列分別為5'-TAA、5'-TAG及5'-TGA)。 術語「起始密碼子區域」及「轉譯起始密碼子區域」係指涵蓋自轉譯起始密碼子開始在任一方向(亦即5'或3')上之約25至約50個鄰接核苷酸之該種mRNA或基因的一部分。類似地,術語「終止密碼子區域」及「轉譯終止密碼子區域」係指涵蓋自轉譯終止密碼子開始在任一方向(亦即5'或3')上之約25至約50個鄰接核苷酸之該種mRNA或基因的一部分。因此,「起始密碼子區域」(或「轉譯起始密碼子區域」)及「終止密碼子區域」(或「轉譯終止密碼子區域」)為可用本發明之反義化合物有效靶向之所有區域。 此項技術中已知之指代轉譯起始密碼子與轉譯終止密碼子之間的區域之開放閱讀框架(ORF)或「編碼區域」亦為可經有效靶向之區域。在本發明之情形內,靶向區域為涵蓋基因之開放閱讀框架(ORF)之轉譯起始或終止密碼子的基因內區域。 另一目標區域包括此項技術中已知之指代自轉譯起始密碼子開始在5'方向上之mRNA之一部分,且因此包括mRNA之5'帽蓋位點(cap site)與轉譯起始密碼子之間的核苷酸(或基因上之相應核苷酸)之5'未轉譯區域(5'UTR)。另一目標區域包括此項技術中已知之指代自轉譯終止密碼子開始在3'方向上之mRNA之一部分,且因此包括mRNA之轉譯終止密碼子與3'端之間的核苷酸(或基因上之相應核苷酸)之3'未轉譯區域(3'UTR)。mRNA之5'帽蓋位點包含經由5'-5'三磷酸酯鍵與mRNA之最5'端殘基接合之N7-甲基化鳥苷殘基。認為mRNA之5'帽蓋區域包括5'帽蓋結構自身以及鄰近於帽蓋位點之前50個核苷酸。用於本發明之另一目標區域為5'帽蓋區域。 儘管一些真核mRNA轉錄物經直接轉譯,但許多含有一或多個稱為「內含子」之區域,其在轉譯之前自轉錄物切除。其餘(且因此經轉譯)區域稱為「外顯子」且一起剪接以形成連續mRNA序列。在一實施例中,靶向剪接位點,亦即內含子-外顯子接合點或外顯子-內含子接合點尤其適用於疾病中牽涉異常剪接或疾病中牽涉特定剪接產物之過度產生的情況中。歸因於重排或缺失之異常融合接合點為目標位點之另一實施例。經由來自不同基因來源之2個(或2個以上)mRNA之剪接過程產生之mRNA轉錄物稱為「融合轉錄物」。可使用靶向例如DNA或前mRNA之反義化合物有效靶向內含子。 在一實施例中,反義寡核苷酸與目標聚核苷酸之編碼及/或非編碼區域結合且調節目標分子之表現及/或功能。 在一實施例中,反義寡核苷酸與天然反義聚核苷酸結合且調節目標分子之表現及/或功能。 在一實施例中,反義寡核苷酸與有義聚核苷酸結合且調節目標分子之表現及/或功能。 替代性RNA轉錄物可自DNA之相同染色體組區域產生。此等替代性轉錄物通常稱為「變異體」。更詳言之,「前mRNA變異體」為自相同染色體組DNA產生之的轉錄物,不同於在其起始或終止位置中自相同染色體組DNA產生之其他轉錄物且含有內含子與外顯子序列兩者。 在剪接期間切除一或多個外顯子或內含子區域或其部分之時,前mRNA變異體產生較小「mRNA變異體」。因此,mRNA變異體為經加工之前mRNA變異體且各獨特前mRNA變異體由於剪接而必定始終產生獨特mRNA變異體。此等mRNA變異體亦稱為「替代性剪接變異體」。若不發生前mRNA變異體之剪接,則前mRNA變異體與mRNA變異體相同。 變異體可經由使用替代性信號以起始或終止轉錄來產生。前mRNA及mRNA可具有1個以上起始密碼子或終止密碼子。源於前mRNA或mRNA之使用替代性起始密碼子之變異體稱為彼前mRNA或mRNA之「替代性起始變異體」。使用替代性終止密碼子之彼等轉錄物稱為彼前mRNA或mRNA之「替代性終止變異體」。一種特定類型之替代性終止變異體為「polyA變異體」,其中產生之多個轉錄物由藉由轉錄機構對1個「polyA終止信號」進行替代性選擇,藉此產生在獨特polyA位點處終止之轉錄物來產生。在本發明之情形內,本文所述之變異體類型亦為目標核酸之實施例。 目標核酸上與反義化合物雜交之位置定義為目標區域之由活性反義化合物靶向之長度至少為5個核苷酸的部分。 儘管某些例示性目標區段之特定序列在本文中加以闡述,但熟習此項技術者應認識到此等序列用於說明及描述在本發明範疇內之特定實施例。其他目標區段容易由一般技術者鑒於本發明加以鑑別。 包含選自說明性較佳目標區段內之至少五個(5個)連續核苷酸之延伸的長度為5-100個核苷酸之目標區段視為亦適用於靶向。 目標區段可包括包含來自一個說明性較佳目標區段之5'端之至少5個連續核苷酸的DNA或RNA序列(其餘核苷酸為相同DNA或RNA之連續延伸,其緊接著目標區段之5'端之上游開始且延續直至DNA或RNA含有約5至約100個核苷酸)。類似較佳目標區段由包含來自一個說明性較佳目標區段之3'端之至少5個連續核苷酸的DNA或RNA序列表示(其餘核苷酸為相同DNA或RNA之連續延伸,其緊接著目標區段之3'端之下游開始且延續直至DNA或RNA含有約5至約100個核苷酸)。熟習本文說明之目標區段之相關技術者將能夠在不進行不容許實驗之情況下鑑別其他較佳目標區段。 一旦一或多個目標區域、區段或位點已經鑑別,即選擇充分互補於目標(亦即足夠良好雜交)且具有足夠特異性之反義化合物以產生所要效應。 在本發明之實施例中,寡核苷酸與特定目標之反義股結合。寡核苷酸的長度為至少5個核苷酸且可經合成以便各寡核苷酸皆靶向重疊序列以使寡核苷酸經合成來涵蓋目標聚核苷酸之整個長度。目標亦包括編碼以及非編碼區域。 在一實施例中,較佳用反義寡核苷酸靶向特定核酸。使反義化合物靶向特定核酸分子為多步驟過程。該過程通常以鑑別功能欲經調節之核酸序列開始。此可為例如表現與特定病症或疾病狀態相關之細胞基因(或由基因轉錄之mRNA)、或非編碼聚核苷酸,諸如非編碼RNA(ncRNA)。 RNA可分類成(1)信使RNA(mRNA),其轉譯成蛋白質,及(2)不編碼蛋白質之RNA(ncRNA)。ncRNA包含微RNA、反義轉錄物及其他含有高密度終止密碼子及缺乏任何延伸「開放閱讀框架」之轉錄單元(TU)。許多ncRNA似乎自編碼蛋白質之基因座之3'未轉譯區域(3'UTR)中的起始位點起始。ncRNA通常稀少且至少半數已由FANTOM協會定序之ncRNA似乎不經聚腺苷酸化。大多數研究人員已出於明顯原因集中於經加工且排出至細胞質中之聚腺苷酸化mRNA。近來,顯示非聚腺苷酸化核RNA之集合可極大,且許多此等轉錄物由所謂基因間區域產生。ncRNA可藉以調控基因表現之機制係根據與目標轉錄物之鹼基配對。藉由鹼基配對起作用之RNA可分組成(1)順式編碼RNA,其在相同遺傳位置處但在其所作用之RNA之相對股上編碼且因此顯示與其目標之完全互補性,及(2)反式編碼RNA,其在不同於其所作用之RNA之染色體位置處編碼且通常不展現與其目標之完全鹼基配對潛力。 在不希望受理論束縛之情況下,本文所述之反義寡核苷酸對反義聚核苷酸之擾動可改變相應有義信使RNA之表現。然而,此調控可為不一致的(反義阻斷基因表現導致信使RNA升高)或一致的(反義阻斷基因表現導致伴隨信使RNA降低)。在此等情況下,可使反義寡核苷酸靶向反義轉錄物之重疊或不重疊部分,從而導致其阻斷基因表現或螯合(sequestration)。編碼以及非編碼反義可以相同方式經靶向且彼任一種類能夠以一致或不一致方式調控相應有義轉錄物。用於鑑別用於對抗目標之新穎寡核苷酸之策略可基於藉由反義寡核苷酸或調節所要目標之任何其他手段阻斷反義RNA轉錄物之基因表現。策略 1 :在不一致調控之情況下,阻斷反義轉錄物基因表現會使習知(有義)基因之表現升高。若習知基因編碼已知或推定藥物目標,則可想像到阻斷其反義對應物之基因表現會模擬受體促效劑或酶刺激劑之作用。策略 2 :在一致調控之情況下,可相伴阻斷反義與有義轉錄物兩者之基因表現且藉此協同降低習知(有義)基因表現。若例如反義寡核苷酸用於阻斷基因表現,則此策略可用於施用一靶向有義轉錄物之反義寡核苷酸及另一靶向相應反義轉錄物之反義寡核苷酸,或同時靶向重疊有義及反義轉錄物之單一能量對稱反義寡核苷酸。 根據本發明,反義化合物包括反義寡核苷酸、核糖核酸酶、外部引導序列(EGS)寡核苷酸、siRNA化合物、單股或雙股RNA干擾(RNAi)化合物(諸如siRNA化合物)、及與至少一部分目標核酸雜交且調節其功能之其他寡聚化合物。因而,其可為DNA、RNA、DNA樣、RNA樣或其混合物,或可為此等之一或多者之模擬物。此等化合物可為單股、雙股、環形或髮夾寡聚化合物且可含有結構元件,諸如內部或端部突起、錯配或環。反義化合物以線性形式按常規方式製備但可經連接或另外製備成環形及/或分支狀。反義化合物可包括構築體,諸如雜交形成完全或部分雙股化合物之兩股或具有足夠自身互補性以允許雜交及形成完全或部分雙股化合物之單股。兩股可內部連接,從而留下自由3'或5'端;或可連接形成連續髮夾結構或環。髮夾結構可在5'或3'端上含有突出物,從而產生具有單股特性之延伸部分。雙股化合物視情況可在端上包括突出物。其他修飾可包括與一端、所選核苷酸位置、糖位置或1個核苷間鍵連接之結合基團。或者,兩股可經由非核酸部分或連接基團連接。當僅由一股形成時,dsRNA可採用在自身上對折以形成雙螺旋之自身互補性髮夾型分子的形式。因此,dsRNA可為完全或部分雙股。基因表現可藉由在轉殖基因細胞株中穩定表現dsRNA髮夾來特定調節,然而,在一些實施例中,基因表現或功能經上調。當由兩股、或採用在自身上對折以形成雙螺旋之自身互補性髮夾型分子形式的單股形成時,兩股(或單股之雙螺旋形成區域)為以華特生-克里克方式進行鹼基配對之互補RNA股。 一旦引入系統中,本發明化合物可引發一或多種酶或結構蛋白之作用以實現目標核酸之裂解或其他修飾或可經由基於占位性(occupancy-based)之機制起作用。一般而言,核酸(包括寡核苷酸)可描述為「DNA樣」(亦即通常具有一或多個2'-去氧糖且通常具有T鹼基而非U鹼基)或「RNA樣」(亦即通常具有一或多個2'-羥基或2'-經修飾之糖且通常具有U鹼基而非T鹼基)。核酸螺旋可採用一種以上類型之結構,最常見為A形式及B形式。咸信,一般而言,具有B形式樣結構之寡核苷酸為「DNA樣」且具有A形式樣結構之寡核苷酸為「RNA樣」。在一些(嵌合)實施例中,反義化合物可含有A形式區域與B形式區域兩者。 在一實施例中,所要寡核苷酸或反義化合物包含以下至少一者:反義RNA、反義DNA、嵌合反義寡核苷酸、包含經修飾之鍵之反義寡核苷酸、干擾RNA(RNAi)、短干擾RNA(siRNA);微型干擾RNA(miRNA);小時序RNA(small, temporal RNA)(stRNA);或短髮夾RNA(shRNA);小RNA誘導之基因活化(RNAa);小活化性RNA(saRNA)或其組合。 dsRNA亦可活化基因表現,一種已稱為「小RNA誘導之基因活化」或RNAa之機制。靶向基因啟動子之dsRNA誘導相關基因之強力轉錄活化。RNAa係使用稱為「小活化性RNA」(saRNA)之合成dsRNA在人類細胞中得以證明。當前未知在其他有機體中,RNAa是否保守。 已發現諸如小干擾RNA(siRNA)及微RNA(miRNA)之小雙股RNA(dsRNA)為稱為RNA干擾(RNAi)之進化保守機制的觸發物。RNAi總是經由重塑染色質以藉此抑制轉錄、降解互補mRNA、或阻斷蛋白質轉譯來導致基因靜止。然而,在隨後實例章節中詳述之情況下,寡核苷酸顯示會增加電壓門控鈉離子通道α次單元(SCNA)聚核苷酸及其編碼產物的表現及/或功能。dsRNA亦可充當小活化性RNA(saRNA)。在不希望受理論束縛之情況下,藉由靶向基因啟動子中之序列,saRNA將誘導目標基因表現,這種現象稱為dsRNA誘導之轉錄活化(RNAa)。 在另一實施例中,本文中鑑別之「較佳目標區段」可用於對調節電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之表現之其他化合物的篩檢中。「調節劑」為減少或增加編碼SCNA之核酸分子之表現且包含互補於較佳目標區段之至少含5個核苷酸之部分的彼等化合物。篩檢方法包含以下步驟:使編碼SCNA之有義或天然反義聚核苷酸之核酸分子的較佳目標區段與一或多種候選調節劑接觸,及選擇一或多種減少或增加編碼SCNA聚核苷酸之核酸分子之表現的候選調節劑,例如SEQ ID NO: 29至94。一旦顯示候選調節劑能夠調節(例如減少或增加)編碼SCNA聚核苷酸之核酸分子之表現,調節劑即可接著用於SCNA聚核苷酸之功能之其他探索研究中,或用作根據本發明之研究藥劑、診斷藥劑或治療藥劑。 靶向天然反義序列較佳調節目標基因之功能。舉例而言,SCNA基因(例如寄存編號NM_001165963、NM_021007、NM_006922、NM_000334、NM_198056、NM_002976、NM_014191、NM_002977、NM_006514、NM_014139、AF109737)。在一實施例中,目標為SCNA基因之反義聚核苷酸。在一實施例中,反義寡核苷酸靶向SCNA聚核苷酸(例如寄存編號NM_001165963、NM_021007、NM_006922、NM_000334、NM_198056、NM_002976、NM_014191、NM_002977、NM_006514、NM_014139、AF109737)之有義及/或天然反義序列、其變異體、對偶基因、同功異型物、同源物、突變體、衍生物、片段及互補序列。較佳地,寡核苷酸為反義分子且目標包括反義及/或有義SCNA聚核苷酸之編碼及非編碼區域。 本發明之較佳目標區段亦可與本發明之其各別互補反義化合物組合以形成穩定化雙股(雙螺旋)寡核苷酸。 在此項技術中,此等雙股寡核苷酸部分已顯示會經由反義機制調節目標表現且調控轉譯以及RNA加工。此外,雙股部分可經受化學修飾。舉例而言,此等雙股部分已顯示會藉由雙螺旋之反義股與目標進行經典雜交,藉此觸發目標之酶促降解來抑制目標。 在一實施例中,反義寡核苷酸靶向電壓門控鈉離子通道α次單元(SCNA)聚核苷酸(例如寄存編號NM_001165963、NM_021007、NM_006922、NM_000334、NM_198056、NM_002976、NM_014191、NM_002977、NM_006514、NM_014139、AF109737)、其變異體、對偶基因、同功異型物、同源物、突變體、衍生物、片段及互補序列。較佳地,寡核苷酸為反義分子。 根據本發明之實施例,目標核酸分子不僅僅限於SCNA而係擴展至SCNA分子之同功異型物、受體、同源物及其類似物之任一者。 在一實施例中,寡核苷酸靶向SCNA聚核苷酸之天然反義序列(例如如SEQ ID NO: 12至28闡述之聚核苷酸)及其任何變異體、對偶基因、同源物、突變體、衍生物、片段及互補序列。反義寡核苷酸之實例係如SEQ ID NO: 29至94所闡述。 在一實施例中,寡核苷酸互補於或結合SCNA反義之核酸序列,包括(不限於)與SCNA聚核苷酸相關之非編碼有義及/或反義序列,且調節SCNA分子之表現及/或功能。 在一實施例中,寡核苷酸互補於或結合如SEQ ID NO: 12至28闡述之SCNA天然反義之核酸序列,且調節SCNA分子之表現及/或功能。 在一實施例中,寡核苷酸包含SEQ ID NO: 29至94之至少5個連續核苷酸之序列且調節SCNA分子之表現及/或功能。 聚核苷酸目標包含SCNA,包括其家族成員、SCNA之變異體;SCNA之突變體,包括SNP;SCNA之非編碼序列;SCNA之對偶基因;物種變異體、片段及其類似物。較佳地,寡核苷酸為反義分子。 在一實施例中,靶向SCNA聚核苷酸之寡核苷酸包含:反義RNA、干擾RNA(RNAi)、短干擾RNA(siRNA);微型干擾RNA(miRNA);小時序RNA(stRNA);或短髮夾RNA(shRNA);小RNA誘導之基因活化(RNAa);或小活化性RNA(saRNA)。 在一實施例中,靶向電壓門控鈉離子通道α次單元(SCNA)聚核苷酸,例如SEQ ID NO: 1至11會調節此等目標之表現或功能。在一實施例中,相較於對照,表現或功能經上調。在一實施例中,相較於對照,表現或功能經下調。在另一實施例中,靶向天然反義轉錄物(例如SEQ ID NO: 12至28)以及此等目標聚核苷酸之任何其他目標NAT會導致該目標mRNA及相應蛋白質之上調。 在一實施例中,反義化合物包含如SEQ ID NO: 29至94闡述之序列。此等寡核苷酸可包含一或多個經修飾之核苷酸、較短或較長片段、經修飾之鍵及其類似物。 在一實施例中,SEQ ID NO: 29至94包含一或多個LNA核苷酸。表1展示適用於本發明方法中之例示性反義寡核苷酸。 1 *指示硫代磷酸酯鍵,+指示LNA,「r」指示RNA且「m」指示寡核苷酸之指定糖部分上之2'氧原子上的甲基。為避免歧義,此LNA具有下式:其中B為特定指定鹼基。 2 經靶向SCN1A特異性天然反義轉錄物之反義寡核苷酸處理之細胞中的SCN1A mRNA之相對表現。Avg-相較於經模擬轉染之對照,SCN1A表現之平均倍數差異;Std-標準偏差,P-經處理之樣品不異於模擬對照之機率。N-平行測定之總數。 2 可以此項技術中已知之若干方式調節所要目標核酸。舉例而言,反義寡核苷酸、siRNA等。酶促核酸分子(例如核糖核酸酶)為能夠催化多種反應之一或多者,包括能夠以核苷酸鹼基序列特異性方式重複裂解其他各別核酸分子之核酸分子。此等酶促核酸分子可用於例如靶向實際上任何RNA轉錄物。 反式裂解酶促核酸分子由於其序列特異性而有希望作為人類疾病之治療劑。酶促核酸分子可經設計以裂解在細胞RNA之背景內之特定RNA目標。該種裂解事件致使mRNA為非功能性的且取消自彼RNA表現蛋白質。以此方式,可選擇性抑制與疾病狀態相關之蛋白質之合成。 一般而言,具有RNA裂解活性之酶促核酸藉由首先與目標RNA結合而起作用。此結合經由酶促核酸之目標結合部分發生,該目標結合部分保持密切鄰近於分子之起裂解目標RNA作用之酶促部分。因此,酶促核酸首先識別目標RNA且接著經由互補鹼基配對結合目標RNA,且一旦與正確位點結合,即以酶促方式起作用來切割目標RNA。該種目標RNA之關鍵裂解將破壞其引導合成編碼蛋白質的能力。在酶促核酸已結合並裂解其RNA目標之後,其自彼RNA釋放以搜尋另一目標且可重複結合並裂解新目標。 諸如活體外選擇(進化)策略(Orgel, (1979) Proc. R. Soc. London, B 205, 435)之若干方法已用於開發能夠催化多種反應,諸如裂解及連接磷酸二酯鍵及醯胺鍵之新穎核酸催化劑。 開發催化活性最佳之核糖核酸酶將顯著有助於採用RNA裂解核糖核酸酶達成調控基因表現之目的的任何策略。錘頭狀核糖核酸酶例如在飽和(10 mM)濃度之Mg2+輔因子存在下以約1 min-1之催化速率(kcat)起作用。人工「RNA連接酶」核糖核酸酶已顯示會以約100 min-1之速率催化相應自身修飾反應。此外,已知具有由DNA構成之受質結合臂之某些經修飾之錘頭狀核糖核酸酶會以近似100 min-1之多個轉換速率催化RNA裂解。最後,用某些核苷酸類似物置換錘頭之催化核心內之特定殘基會產生顯示催化速率改良多達10倍之經修飾之核糖核酸酶。此等研究結果證明核糖核酸酶可以顯著大於大多數天然自身裂解核糖核酸酶在活體外顯示之催化速率的催化速率促進化學轉化。那麼有可能某些自身裂解核糖核酸酶之結構可經最佳化以產生最大催化活性,或可製備顯示顯著較快之RNA磷酸二酯裂解速率之完全新穎的RNA基元。 RNA催化劑對RNA受質之符合「錘頭」模型之分子間裂解首先顯示於1987年(Uhlenbeck, O. C. (1987) Nature, 328: 596-600)。回收RNA催化劑且使其與多個RNA分子反應,從而證明其確實起催化作用。 藉由在催化RNA中作適當鹼基變化以維持與目標序列之必要鹼基配對,基於「錘頭」基元設計出之催化RNA已用於裂解特定目標序列。此已允許使用催化RNA來裂解特定目標序列且指示根據「錘頭」模型設計出之催化RNA可能在活體內裂解特定受質RNA。 RNA干擾(RNAi)已成為調節哺乳動物及哺乳動物細胞中之基因表現之強力工具。此方法需要使用表現質體或病毒及經加工成siRNA之小髮夾RNA之編碼序列以自身RNA形式或以DNA形式傳遞小干擾RNA(siRNA)。此系統能夠高效轉運前siRNA至細胞質中,在細胞質中前siRNA具有活性且允許使用經調控且具組織特異性之啟動子以達成基因表現。 在一實施例中,寡核苷酸或反義化合物包含核糖核酸(RNA)及/或去氧核糖核酸(DNA)之寡聚物或聚合物、或其模擬物、嵌合體、類似物或同源物。此術語包括由天然存在之核苷酸、糖及共價核苷間(骨架)鍵構成之寡核苷酸以及具有非天然存在之以類似方式起作用之部分的寡核苷酸。此等經修飾或經取代之寡核苷酸常由於合乎需要之性質,諸如細胞攝取增強、對目標核酸之親和力增強及在核酸酶存在下之穩定性增加而超過天然形式被需要。 根據本發明,寡核苷酸或「反義化合物」包括反義寡核苷酸(例如其RNA、DNA、模擬物、嵌合體、類似物或同源物)、核糖核酸酶、外部引導序列(EGS)寡核苷酸、siRNA化合物、單股或雙股RNA干擾(RNAi)化合物(諸如siRNA化合物)、saRNA、aRNA、及與至少一部分目標核酸雜交且調節其功能之其他寡聚化合物。因而,其可為DNA、RNA、DNA樣、RNA樣或其混合物,或可為此等之一或多者之模擬物。此等化合物可為單股、雙股、環形或髮夾寡聚化合物且可含有結構元件,諸如內部或端部突起、錯配或環。反義化合物以線性形式按常規方式製備但可經連接或另外製備成環形及/或分支狀。反義化合物可包括構築體,諸如雜交形成完全或部分雙股化合物之兩股或具有足夠自身互補性以允許雜交及形成完全或部分雙股化合物之單股。兩股可內部連接,從而留下自由3'或5'端;或可連接形成連續髮夾結構或環。髮夾結構可在5'或3'端上含有突出物,從而產生具有單股特性之延伸部分。雙股化合物視情況可在端上包括突出物。其他修飾可包括與一端、所選核苷酸位置、糖位置或1個核苷間鍵連接之結合基團。或者,兩股可經由非核酸部分或連接基團連接。當僅由一股形成時,dsRNA可採用在自身上對折以形成雙螺旋之自身互補性髮夾型分子的形式。因此,dsRNAs可為完全或部分雙股。基因表現可藉由在轉殖基因細胞株中穩定表現dsRNA髮夾來特定調節。當由兩股、或採用在自身上對折以形成雙螺旋之自身互補性髮夾型分子之形式的單股形成時,兩股(或單股之雙螺旋形成區域)為以華特生-克里克方式進行鹼基配對之互補RNA股。 一旦引入系統中,本發明化合物可引發一或多種酶或結構蛋白之作用以實現目標核酸之裂解或其他修飾或可經由基於占位性之機制起作用。一般而言,核酸(包括寡核苷酸)可描述為「DNA樣」(亦即通常具有一或多個2'-去氧糖且通常具有T鹼基而非U鹼基)或「RNA樣」(亦即通常具有一或多個2'-羥基或2'-經修飾之糖且通常具有U鹼基而非T鹼基)。核酸螺旋可採用1種以上類型之結構,最常見為A形式及B形式。咸信,一般而言,具有B形式樣結構之寡核苷酸為「DNA樣」且具有A形式樣結構之寡核苷酸為「RNA樣」。在一些(嵌合)實施例中,反義化合物可含有A形式區域與B形式區域兩者。 本發明之反義化合物可包含長度約5至約80個核苷酸(亦即約5至約80個連接之核苷)之反義部分。此係指反義化合物之反義股或部分之長度。換言之,本發明之單股反義化合物包含5至約80個核苷酸,且本發明之雙股反義化合物(諸如dsRNA)包含長度為5至約80個核苷酸之有義及反義股或部分。一般技術者應瞭解此包括長度為5、6、7、8、9、10、11、12、13、14、15、16、17、18、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、49、50、51、52、53、54、55、56、57、58、59、60、61、62、63、64、65、66、67、68、69、70、71、72、73、74、75、76、77、78、79或80個核苷酸或其間任何範圍的反義部分。 在一實施例中,本發明之反義化合物具有長度為10至50個核苷酸之反義部分。一般技術者應瞭解此包括反義部分長度為10、11、12、13、14、15、16、17、18、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、49或50個核苷酸或其間任何範圍的寡核苷酸。在一些實施例中,寡核苷酸之長度為15個核苷酸。 在一實施例中,本發明之反義或寡核苷酸化合物具有長度為12或13至30個核苷酸之反義部分。一般技術者應瞭解此包括反義部分長度為12、13、14、15、16、17、18、19、20、21、22、23、24、25、26、27、28、29或30個核苷酸或其間任何範圍的反義化合物。 在一實施例中,本發明之寡聚化合物亦包括不同鹼基存在於化合物中之一或多個核苷酸位置處的變異體。舉例而言,若最初核苷酸為腺苷,則可產生在此位置處含有胸苷、鳥苷或胞苷之變異體。此可在反義或dsRNA化合物之任何位置處進行。接著使用本文所述之方法測試此等化合物以確定其抑制目標核酸表現之能力。 在一些實施例中,反義化合物與目標之間的同源性、序列一致性或互補性為約40%至約60%。在一些實施例中,同源性、序列一致性或互補性為約60%至約70%。在一些實施例中,同源性、序列一致性或互補性為約70%至約80%。在一些實施例中,同源性、序列一致性或互補性為約80%至約90%。在一些實施例中,同源性、序列一致性或互補性為約90%、約92%、約94%、約95%、約96%、約97%、約98%、約99%或約100%。 在一實施例中,諸如SEQ ID NO: 29至94中闡述之核酸分子之反義寡核苷酸包含一或多個取代或修飾。在一實施例中,核苷酸經鎖核酸(LNA)取代。 在一實施例中,寡核苷酸靶向與SCNA相關之編碼及/或非編碼序列之核酸分子有義及/或反義及如SEQ ID NO: 1至28闡述之序列的一或多個區域。亦使寡核苷酸靶向SEQ ID NO: 1至28之重疊區域。 本發明之某些較佳寡核苷酸為嵌合寡核苷酸。在本發明之情形下,「嵌合寡核苷酸」或「嵌合體」為含有兩個或兩個以上各自由至少一個核苷酸構成之化學性質不同區域的寡核苷酸。此等寡核苷酸通常含有經修飾之核苷酸之賦予一或多種有益性質(諸如核酸酶抗性增加、進入細胞之攝取增加、對目標之結合親和力增加)的至少一個區域及作為能夠裂解RNA:DNA或RNA:RNA雜交體之酶受質的區域。舉例而言,RNase H為一種會裂解RNA:DNA雙螺旋之RNA股的細胞核酸內切酶。因此,RNase H之活化會導致RNA目標之裂解,藉此極大增強反義調節基因表現之效率。因此,相較於硫代磷酸酯去氧寡核苷酸與相同目標區域之雜交,當使用嵌合寡核苷酸時,通常獲得用較短寡核苷酸所得到的類似結果。可藉由凝膠電泳及必要時此項技術中已知之相關核酸雜交技術按常規方式偵測RNA目標之裂解。在一實施例中,嵌合寡核苷酸包含至少一個經修飾以增加目標結合親和力之區域及通常充當RNAse H受質之區域。寡核苷酸對其目標(在此情況下,編碼ras之核酸)之親和力係藉由量測寡核苷酸/目標對之Tm按常規方式確定,Tm為寡核苷酸與目標解離之溫度;解離係以分光光度測定方式加以偵測。Tm愈高,寡核苷酸對目標之親和力愈大。 本發明之嵌合反義化合物可形成為兩個或兩個以上如上所述之寡核苷酸、經修飾之寡核苷酸、寡核苷及/或寡核苷酸模擬物之複合結構。此等化合物在此項技術中亦稱為雜交體或間隔體(gapmers)。教示製備此等雜交結構之代表性美國專利包含(但不限於)美國專利第5,013,830號;第5,149,797號;第5,220,007號;第5,256,775號;第5,366,878號;第5,403,711號;第5,491,133號;第5,565,350號;第5,623,065號;第5,652,355號;第5,652,356號;及第5,700,922號,各者以引用的方式併入本文中。 在一個實施例中,經修飾之寡核苷酸之區域包含至少一個在糖之2'位置經修飾之核苷酸,最佳為2'-O烷基、2'-O-烷基-O-烷基或2'-氟-修飾之核苷酸。在另一實施例中,RNA修飾包括嘧啶之核糖上2'-氟、2'-胺基及2' O-甲基修飾、無鹼基殘基或RNA之3'端之倒轉鹼基。此等修飾按常規方式併入寡核苷酸中,且此等寡核苷酸已顯示比2'-去氧寡核苷酸對於既定目標具有較高Tm(亦即較高目標結合親和力)。此增加之親和力之效應在於極大增強RNAi寡核苷酸抑制基因表現。RNAse H為一種裂解RNA:DNA雙螺旋之RNA股之細胞核酸內切酶;因此,此酶活化導致RNA目標之裂解,且因此可極大增強RNAi抑制之效率。RNA目標之裂解可按常規方式由凝膠電泳證明。在一個實施例中,嵌合寡核苷酸亦經修飾以增強核酸酶抗性。細胞含有多種可降解核酸之核酸外切酶及核酸內切酶。許多核苷酸及核苷修飾已顯示使得其所併入之寡核苷酸比天然寡去氧核苷酸對核酸酶消化具有更大抗性。核酸酶抗性係按常規方式測量,藉由寡核苷酸與細胞提取物或經分離之核酸酶溶液一起培育及通常藉由凝膠電泳測量完整寡核苷酸隨時間剩餘之程度。已經修飾以增強其核酸酶抗性之寡核苷酸完整存在的時間長於未經修飾之寡核苷酸。多種寡核苷酸修飾已經證明可增強或賦予核酸酶抗性。目前含有至少一個硫代磷酸酯修飾之寡核苷酸更佳。在一些情況下,增強目標結合親和力之寡核苷酸修飾亦獨立地能夠增強核酸酶抗性。 本發明所設想之一些較佳寡核苷酸之特定實例包括該等包含經修飾骨架,例如硫代磷酸酯、磷酸三酯、甲基膦酸酯、短鏈烷基或環烷基糖間鍵聯或短鏈雜原子或雜環糖間鍵聯者。最佳為具有硫代磷酸酯骨架之寡核苷酸,及具有雜原子骨架,特別是CH2--NH--O--CH2、CH,--N(CH3)--O--CH2[稱為亞甲基(甲基亞胺基)或MMI骨架]、CH2--O--N(CH3)--CH2、CH2-N(CH3)--N(CH3)--CH2及O--N(CH3)--CH2--CH2骨架,其中天然磷酸二酯骨架表示為O--P--O--CH者。De Mesmaeker等人(1995) Acc. Chem. Res. 28: 366-374揭示之醯胺骨架亦佳。具有嗎啉基骨架結構之寡核苷酸(Summerton及Weller, 美國專利第5,034,506號)亦佳。在另一實施例中,諸如寡核苷酸之肽核酸(PNA)骨架、磷酸二酯骨架經聚醯胺骨架置換,其中核苷酸直接或間接與聚醯胺骨架之氮雜氮原子結合。寡核苷酸亦可包含一或多個經取代之糖部分。較佳寡核苷酸在2'位置處包含以下之一:OH、SH、SCH3、F、OCN、OCH3 OCH3、OCH3 O(CH2)n CH3、O(CH2)n NH2或O(CH2)n CH3,其中n為1至約10;C1至C10低碳烷基、烷氧基烷氧基、經取代之低碳烷基、烷芳基或芳烷基;Cl;Br;CN;CF3;OCF3;O--、S--或N-烷基;O--、S--或N-烯基;SOCH3;SO2 CH3;ONO2;NO2;N3;NH2;雜環烷基;雜環烷芳基;胺基烷胺基;聚烷胺基;經取代之矽烷基;RNA裂解基團;報導基團;嵌入劑;改良寡核苷酸之藥物動力學性質之基團;或改良寡核苷酸之藥效學性質之基團及其他具有類似性質之取代基。較佳修飾包括2'-甲氧基乙氧基[2'-O-CH2 CH2 OCH3,亦稱為2'-O-(2-甲氧基乙基)]。其他較佳修飾包括2'-甲氧基(2'-O--CH3)、2'-丙氧基(2'-OCH2 CH2CH3)及2'-氟(2'-F)。亦可在寡核苷酸上之其他位置,特定言之3'端核苷酸上之糖之3'位置及5'端核苷酸之5'位置處進行類似修飾。寡核苷酸亦可具有糖模擬物,諸如替代戊呋喃糖基之環丁基。 寡核苷酸亦可或者或另外包括核鹼基(在此項技術中常簡稱為「鹼基」)修飾或取代。如本文所用,「未經修飾」或「天然」核苷酸包括腺嘌呤(A)、鳥嘌呤(G)、胸腺嘧啶(T)、胞嘧啶(C)及尿嘧啶(U)。經修飾之核苷酸包括僅偶爾或短暫見於天然核酸中之核苷酸,例如次黃嘌呤、6-甲基腺嘌呤、5-Me嘧啶、特定言之5-甲基胞嘧啶(亦稱為5-甲基-2'去氧胞嘧啶且在此項技術中常稱為5-Me-C)、5-羥甲基胞嘧啶(HMC)、糖基HMC及龍膽二糖基(gentobiosyl) HMC、以及合成核苷酸,例如2-胺基腺嘌呤、2-(甲胺基)腺嘌呤、2-(咪唑基烷基)腺嘌呤、2-(胺基烷胺基)腺嘌呤或其他雜取代之烷基腺嘌呤、2-硫尿嘧啶、2-硫胸腺嘧啶、5-溴尿嘧啶、5-羥甲基尿嘧啶、8-氮鳥嘌呤、7-去氮鳥嘌呤、N6 (6-胺己基)腺嘌呤及2,6-二胺基嘌呤。可包括此項技術中已知之「通用」鹼基,例如肌苷。5-Me-C取代已顯示會使核酸雙螺旋穩定性增加0.6-1.2℃且為目前較佳的鹼基取代。 本發明之寡核苷酸之另一修飾涉及使增強寡核苷酸之活性或細胞攝取之一或多個部分或結合物化學連接至寡核苷酸。此等部分包括(但不限於)脂質部分(諸如膽固醇部分、膽固醇基部分)、脂族鏈(例如十二烷二醇或十一基殘基)、聚胺或聚乙二醇鏈、或金剛烷(Adamantane)乙酸。包含親脂性部分之寡核苷酸及製備此等寡核苷酸之方法在此項技術,例如美國專利第5,138,045號、第5,218,105號及第5,459,255號中為已知的。 並非既定寡核苷酸中之所有位置皆必須經均一修飾,且實際上一個以上之以上提及之修飾可併入單一寡核苷酸中或甚至寡核苷酸內之單一核苷內。本發明亦包括作為如上文所定義之嵌合寡核苷酸的寡核苷酸。 在另一實施例中,本發明之核酸分子與另一部分結合,該另一部分包括(但不限於)無鹼基核苷酸、聚醚、聚胺、聚醯胺、肽、碳水化合物、脂質或聚烴化合物。熟習此項技術者應認識到此等分子可在糖、鹼基或磷酸基上之若干位置處與構成核酸分子之任何核苷酸的一或多者連接。 根據本發明使用之寡核苷酸可經由熟知固相合成技術方便且按常規方式製備。用於此合成之設備由包括Applied Biosystems之若干供應商銷售。亦可採用用於此合成之任何其他手段;寡核苷酸之實際合成完全在一般技術者之技能內。亦熟知使用類似技術來製備其他寡核苷酸,諸如硫代磷酸酯及烷基化衍生物。亦熟知使用類似技術及市售經修飾之胺基酸酯(amidites)及可控多孔性玻璃(controlled-pore glass;CPG)產品,諸如經生物素、螢光素、吖啶或補骨脂素(psoralen)修飾之胺基酸酯及/或CPG(可自Glen Research, Sterling VA獲得)來合成經螢光標記之寡核苷酸、經生物素標記之寡核苷酸或其他經修飾之寡核苷酸,諸如經膽固醇修飾之寡核苷酸。 根據本發明,使用修飾,諸如使用LNA單體以增強作用之效能、特異性及持續時間且拓寬包含當前化學組成(諸如MOE、ANA、FANA、PS等)之寡核苷酸的投藥途徑。此可藉由用LNA單體取代當前寡核苷酸中之一些單體達成。經LNA修飾之寡核苷酸可具有與母體化合物類似之尺寸或可較大或較佳較小。較佳地,此等經LNA修飾之寡核苷酸含有小於約70%、更佳小於約60%、最佳小於約50%之LNA單體且其尺寸介於約5與25個核苷酸之間、更佳介於約12與20個核苷酸之間。 較佳經修飾之寡核苷酸骨架包含(但不限於)硫代磷酸酯、對掌性硫代磷酸酯、二硫代磷酸酯、磷酸三酯、胺基烷基磷酸三酯、甲基膦酸酯及其他烷基膦酸酯(包含3'膦酸伸烷酯及對掌性膦酸酯)、亞膦酸酯、胺基磷酸酯(包含3'-胺基胺基磷酸酯及胺基烷基胺基磷酸酯)、硫羰基胺基磷酸酯、硫羰基烷基膦酸酯、硫羰基烷基磷酸三酯、及具有正常3'-5'鍵之硼烷磷酸酯(boranophosphate)、此等硼烷磷酸酯之2'-5'連接之類似物、及具有反極性之硼烷磷酸酯,其中鄰近核苷單元對係3'-5'至5'-3'或2'-5'至5'-2'連接。亦包括各種鹽、混合鹽及游離酸形式。 教示製備以上含磷鍵之代表性美國專利包含(但不限於)美國專利第3,687,808號;第4,469,863號;第4,476,301號;第5,023,243號;第5,177,196號;第5,188,897號;第5,264,423號;第5,276,019號;第5,278,302號;第5,286,717號;第5,321,131號;第5,399,676號;第5,405,939號;第5,453,496號;第5,455,233號;第5,466,677號;第5,476,925號;第5,519,126號;第5,536,821號;第5,541,306號;第5,550,111號;第5,563,253號;第5,571,799號;第5,587,361號及第5,625,050號,各專利以引用的方式併入本文中。 其中不包括磷原子之較佳經修飾之寡核苷酸骨架具有由短鏈烷基或環烷基核苷間鍵、混合雜原子及烷基或環烷基核苷間鍵、或一或多個短鏈雜原子或雜環核苷間鍵形成之骨架。此等寡核苷酸骨架包含具有以下之彼等寡核苷酸骨架:嗎啉基鍵(部分地由核苷之糖部分形成);矽氧烷骨架;硫化物、亞碸及碸骨架;甲乙醯基(formacetyl)及硫代甲乙醯基(thioformacetyl)骨架;亞甲基甲乙醯基及亞甲基硫代甲乙醯基骨架;含烯骨架;胺基磺酸酯骨架;亞甲基亞胺基及亞甲基肼基骨架;磺酸酯及磺醯胺骨架;醯胺骨架;及具有混合N、O、S及CH2組成部分之其他骨架。 教示製備以上寡核苷之代表性美國專利包含(但不限於)美國專利第5,034,506號;第5,166,315號;第5,185,444號;第5,214,134號;第5,216,141號;第5,235,033號;第5,264,562號;第5,264,564號;第5,405,938號;第5,434,257號;第5,466,677號;第5,470,967號;第5,489,677號;第5,541,307號;第5,561,225號;第5,596,086號;第5,602,240號;第5,610,289號;第5,602,240號;第5,608,046號;第5,610,289號;第5,618,704號;第5,623,070號;第5,663,312號;第5,633,360號;第5,677,437號;及第5,677,439號,各專利以引用的方式併入本文中。 在其他較佳寡核苷酸模擬物中,核苷酸單元之糖與核苷間鍵(亦即骨架)兩者均經新穎基團置換。保留鹼基單元以用於與適當核酸目標化合物雜交。一種此寡聚化合物,即已顯示具有極佳雜交性質之寡核苷酸模擬物稱為肽核酸(PNA)。在PNA化合物中,寡核苷酸之糖-骨架經含醯胺骨架,特定言之胺乙基甘胺酸骨架置換。核鹼基經保留且直接或間接與骨架醯胺部分之氮雜氮原子結合。教示製備PNA化合物之代表性美國專利包含(但不限於)美國專利第5,539,082號;第5,714,331號;及第5,719,262號,各專利以引用的方式併入本文中。PNA化合物之其他教示可見於Nielsen等人(1991) Science 254, 1497-1500中。 在本發明之一實施例中,寡核苷酸具有硫代磷酸酯骨架且寡核苷具有雜原子骨架,且特定言之CH2-NH-O-CH2-;稱為亞甲基(甲基亞胺基)或MMI骨架之-CH2-N (CH3)-O-CH2-;-CH2-O-N (CH3)-CH2-;-CH2N(CH3)-N(CH3) CH2-及-O-N(CH3)-CH2-CH2-,其中天然磷酸二酯骨架表示為以上引用之美國專利第5,489,677號之-O-P-O-CH2-;及以上引用之美國專利第5,602,240號之醯胺骨架。具有以上引用之美國專利第5,034,506號之嗎啉基骨架結構之寡核苷酸亦較佳。 經修飾之寡核苷酸亦可含有一或多個經取代之糖部分。較佳寡核苷酸在2'位置處包含以下之一:OH;F;O-、S-或N-烷基;O-、S-或N-烯基;O-、S-或N-炔基;或O烷基‑O-烷基,其中該烷基、烯基及炔基可為經取代或未經取代之C至CO烷基或C2至CO烯基及炔基。尤其較佳為O(CH2)n OmCH3、O(CH2)n,OCH3、O(CH2)nNH2、O(CH2)nCH3、O(CH2)nONH2及O(CH2nON(CH2)nCH3)2,其中n及m可為1至約10。其他較佳寡核苷酸在2'位置處包含以下之一:C至CO、低碳烷基、經取代之低碳烷基、烷芳基、芳烷基、O-烷芳基或O-芳烷基、SH、SCH3、OCN、Cl、Br、CN、CF3、OCF3、SOCH3、SO2CH3、ONO2、NO2、N3、NH2、雜環烷基、雜環烷芳基、胺基烷胺基、聚烷胺基、經取代之矽烷基、RNA裂解基團、報導基團、嵌入劑、改良寡核苷酸之藥物動力學性質之基團、或改良寡核苷酸之藥效學性質之基團、及具有類似性質之其他取代基。一較佳修飾包含2'-甲氧基乙氧基(2'‑O‑CH2CH2OCH3,亦稱為2'-O-(2-甲氧基乙基)或2'-MOE),亦即烷氧基烷氧基。另一較佳修飾包含2'-二甲胺基氧基乙氧基,亦即O(CH2)2ON(CH3)2基團,亦稱為2'‑DMAOE,如以下本文實例中所述;及2'-二甲胺基乙氧基乙氧基(在此項技術中亦稱為2'-O-二甲胺基乙氧基乙基或2'-DMAEOE),亦即2'-O-CH2-O-CH2-N (CH2)2。 其他較佳修飾包含2'-甲氧基(2'-O CH3)、2'-胺基丙氧基(2'-O CH2CH2CH2NH2)及2'-氟(2'-F)。亦可在寡核苷酸上之其他位置,特定言之3'端核苷酸上或2'-5'連接之寡核苷酸中之糖之3'位置及5'端核苷酸之5'位置處進行類似修飾。寡核苷酸亦可具有糖模擬物,諸如替代戊呋喃糖基糖之環丁基部分。教示製備此等經修飾之糖結構之代表性美國專利包含(但不限於)美國專利第4,981,957號;第5,118,800號;第5,319,080號;第5,359,044號;第5,393,878號;第5,446,137號;第5,466,786號;第5,514,785號;第5,519,134號;第5,567,811號;第5,576,427號;第5,591,722號;第5,597,909號;第5,610,300號;第5,627,053號;第5,639,873號;第5,646,265號;第5,658,873號;第5,670,633號;及第5,700,920號,各專利以引用的方式併入本文中。 寡核苷酸亦可包含核鹼基(在此項技術中常簡稱為「鹼基」)修飾或取代。如本文所用,「未經修飾」或「天然」核苷酸包含嘌呤鹼基腺嘌呤(A)及鳥嘌呤(G)、及嘧啶鹼基胸腺嘧啶(T)、胞嘧啶(C)及尿嘧啶(U)。經修飾之核苷酸包含其他合成及天然核苷酸,諸如 5-甲基胞嘧啶(5-me-C);5-羥甲基胞嘧啶;黃嘌呤;次黃嘌呤;2-胺基腺嘌呤;腺嘌呤及鳥嘌呤之6-甲基及其他烷基衍生物;腺嘌呤及鳥嘌呤之2-丙基及其他烷基衍生物;2-硫尿嘧啶;2-硫胸腺嘧啶及2-硫胞嘧啶;5-鹵尿嘧啶及5-鹵胞嘧啶;5-丙炔基尿嘧啶及5-丙炔基胞嘧啶;6-偶氮尿嘧啶、6-偶氮胞嘧啶及6-偶氮胸腺嘧啶;5-尿嘧啶(假尿嘧啶);4-硫尿嘧啶;8-鹵、8-胺基、8-硫醇、8-硫烷基、8-羥基及其他8取代腺嘌呤及鳥嘌呤;5-鹵(特定言之5-溴)、5-三氟甲基及其他5-取代尿嘧啶及胞嘧啶;7-甲基鳥嘌呤(7-methylquanine)及7-甲基腺嘌呤;8-氮鳥嘌呤及8-氮腺嘌呤;7-去氮鳥嘌呤及7-去氮腺嘌呤及3-去氮鳥嘌呤及3-去氮腺嘌呤。 另外,核苷酸包含以下中揭示之核苷酸:美國專利第3,687,808號、「The Concise Encyclopedia of Polymer Science And Engineering」,第858-859頁, Kroschwitz, J.I.編 John Wiley & Sons, 1990、Englisch等人,「Angewandle Chemie, International Edition」, 1991, 30,第613頁、及Sanghvi, Y.S.,第15章,「Antisense Research and Applications」,第289-302頁, Crooke, S.T.及Lebleu, B.編,CRC Press, 1993。某些此等核苷酸尤其適用於增加本發明之寡聚化合物之結合親和力。此等包含5-取代嘧啶、6-氮嘧啶及N-2、N-6及0-6取代嘌呤,包含2-胺丙基腺嘌呤、5-丙炔基尿嘧啶及5-丙炔基胞嘧啶。5-甲基胞嘧啶取代已顯示會使核酸雙螺旋穩定性增加0.6-1.2℃(Sanghvi, Y.S., Crooke, S.T.及Lebleu, B.編,「Antisense Research and Applications」, CRC Press, Boca Raton, 1993,第276-278頁)且為目前較佳的鹼基取代,當與2'-O甲氧基乙基糖修飾組合時甚至更特別較佳。 教示製備以上指示之經修飾之核苷酸以及其他經修飾之核苷酸之代表性美國專利包含(但不限於)美國專利第3,687,808號以及第4,845,205號;第5,130,302號;第5,134,066號;第5,175,273號;第5,367,066號;第5,432,272號;第5,457,187號;第5,459,255號;第5,484,908號;第5,502,177號;第5,525,711號第5,552,540號;第5,587,469號;第5,596,091號;第5,614,617號;第5,750,692號;及第5,681,941號,各專利以引用的方式併入本文中。 本發明之寡核苷酸之另一修飾涉及使增強寡核苷酸之活性,細胞分佈或細胞攝取之一或多個部分或結合物化學連接至寡核苷酸。 此等部分包含(但不限於)脂質部分(諸如膽固醇部分)、膽酸、硫醚(例如己基-S-三苯甲基硫醇)、硫代膽固醇、脂族鏈(例如十二烷二醇或十一基殘基)、磷脂(例如二-十六基-外消旋-甘油或1,2-二-O-十六基-外消旋-甘油基-3-H-膦酸三乙基銨)、聚胺或聚乙二醇鏈、或金剛烷乙酸、棕櫚基(palmityl)部分、或十八烷基胺或己胺基-羰基-羥膽固醇(hexylamino-carbonyl-t oxycholesterol)部分。 教示製備此等寡核苷酸結合物之代表性美國專利包含(但不限於)美國專利第4,828,979號;第4,948,882號;第5,218,105號;第5,525,465號;第5,541,313號;第5,545,730號;第5,552,538號;第5,578,717號;第5,580,731號;第5,580,731號;第5,591,584號;第5,109,124號;第5,118,802號;第5,138,045號;第5,414,077號;第5,486,603號;第5,512,439號;第5,578,718號;第5,608,046號;第4,587,044號;第4,605,735號;第4,667,025號;第4,762,779號;第4,789,737號;第4,824,941號;第4,835,263號;第4,876,335號;第4,904,582號;第4,958,013號;第5,082,830號;第5,112,963號;第5,214,136號;第5,082,830號;第5,112,963號;第5,214,136號;第5,245,022號;第5,254,469號;第5,258,506號;第5,262,536號;第5,272,250號;第5,292,873號;第5,317,098號;第5,371,241號;第5,391,723號;第5,416,203號;第5,451,463號;第5,510,475號;第5,512,667號;第5,514,785號;第5,565,552號;第5,567,810號;第5,574,142號;第5,585,481號;第5,587,371號;第5,595,726號;第5,597,696號;第5,599,923號;第5,599,928號及第5,688,941號,各專利以引用的方式併入本文中。藥物發現: 本發明化合物亦可應用於藥物發現及目標驗證之領域中。本發明包括使用本文鑑別出之化合物及較佳目標區段努力發現欲闡明存在於電壓門控鈉離子通道α次單元(SCNA)聚核苷酸與疾病狀態、表型或病狀之間的關係之藥物。此等方法包括偵測或調節SCNA聚核苷酸,包含使樣品、組織、細胞或有機體與本發明化合物接觸,在處理之後某時量測SCNA聚核苷酸之核酸或蛋白質含量及/或相關表型或化學終點,且視情況比較量測值與未處理樣品或經另一本發明化合物處理之樣品。此等方法亦可平行或與其他實驗組合進行以確定未知基因對於目標驗證方法之功能或確定特定基因產物作為用於治療或預防特定疾病、病狀或表型之目標的有效性。評估基因表現之上調或抑制: 可藉由直接偵測核酸在細胞或有機體中之存在來評估外源性核酸向宿主細胞或有機體中的轉移。此偵測可藉由此項技術中熟知之若干方法達成。舉例而言,外源性核酸之存在可藉由南方墨點分析或藉由使用特異性擴增與核酸相關之核苷酸序列之引子的聚合酶鏈反應(PCR)技術加以偵測。亦可使用包括基因表現分析之習知方法量測外源性核酸之表現。舉例而言,外源性核酸產生之mRNA可使用北方墨點分析及反轉錄PCR(RT-PCR)偵測及定量。 外源性核酸表現之RNA亦可藉由量測酶活性或報導蛋白活性加以偵測。舉例而言,反義調節活性可間接量測為目標核酸表現的減少或增加,其指示外源性核酸正在產生效應RNA。基於序列保守性,引子可經設計且用於擴增目標基因之編碼區域。最初,各基因之最高度表現之編碼區域可用於構築模型控制基因(model control gene),但可使用任何編碼或非編碼區域。各控制基因藉由在報導編碼區域與其聚(A)信號之間插入各編碼區域來裝配。此等質體將產生在基因之上游部分中具有報導基因且在3'非編碼區域中具有潛在RNAi目標的mRNA。個別反義寡核苷酸之效用將藉由調節報導基因加以檢定。適用於本發明方法中之報導基因包括乙醯羥基酸合成酶(AHAS)、鹼性磷酸酯酶(AP)、β半乳糖苷酶(LacZ)、β葡糖醛酸酶(GUS)、氯黴素乙醯轉移酶(chloramphenicol acetyltransferase;CAT)、綠色螢光蛋白(GFP)、紅色螢光蛋白(RFP)、黃色螢光蛋白(YFP)、藍螢光蛋白質(cyan fluorescent protein;CFP)、辣根過氧化酶(horseradish peroxidase)(HRP)、螢光素酶(luciferase)(Luc)、胭脂鹼合成酶(nopaline synthase;NOS)、章魚鹼合成酶(octopine synthase;OCS)及其衍生物。多種賦予對安比西林(ampicillin)、博來黴素(bleomycin)、氯黴素、健大黴素(gentamycin)、潮黴素(hygromycin)、康黴素(kanamycin)、林可黴素(lincomycin)、甲胺喋呤(methotrexate)、草胺膦(phosphinothricin)、嘌呤黴素(puromycin)及四環素(tetracycline)之抗性之可選擇標記可用。用於確定對報導基因之調節之方法在此項技術中為熟知的,且包括(但不限於)螢光分析法(例如螢光光譜分析、螢光活化之細胞揀選(FACS)、螢光顯微鏡術)、抗生素抗性確定。 亦可在基於細胞之檢定中偵測目標核酸區段。進行實驗以偵測HepG2中、帶有德拉威特症候群(Dravet syndrome)相關突變之初級人類纖維母細胞中以及人類睪丸中的Scn1a天然反義BG724147。對於HepG2以及帶有德拉威特症候群相關突變之初級人類纖維母細胞,使細胞生長且提取RNA。對於人類睪丸,購買並利用polyA分離之RNA。此實驗稱為RACE(cDNA端之快速擴增)且使用針對BG724147 RNA轉錄物之特定引子。 在來自HepG2之polyA分離之RNA及來自帶有德拉威特症候群相關突變之初級人類纖維母細胞的polyA分離之RNA中偵測到極類似之PCR產物但此產物未在來自人類睪丸之polyA分離之RNA中偵測到。此外,彼PCR產物未(或以極低量)在來自HepG2細胞之總RNA及來自帶有德拉威特症候群相關突變之初級人類纖維母細胞的總RNA中偵測到。結果表明Scn1a之稱為BG724147之天然反義存在於HepG2細胞及帶有德拉威特症候群相關突變之初級人類纖維母細胞中但不存在於人類睪丸中。 SCNA蛋白及mRNA表現可使用熟習此項技術者已知及在本文別處描述之方法加以檢定。舉例而言,諸如ELISA之免疫檢定可用於量測蛋白質含量。SCNA ELISA檢定套組可例如自R&D Systems(Minneapolis, MN)購得。在實施例中,使用本發明之反義寡核苷酸處理之樣品(例如活體內或活體外細胞或組織)中之SCNA表現(例如mRNA或蛋白質)係藉由與對照樣品中之SCNA表現進行比較來評估。舉例而言,蛋白質或核酸之表現可使用熟習此項技術者已知之方法與模擬處理或未處理樣品中之蛋白質或核酸之表現進行比較。或者,視所要資訊而定可與經對照反義寡核苷酸(例如具有改變或不同序列之反義寡核苷酸)處理之樣品進行比較。在另一實施例中,相對於未處理樣品,經處理樣品中之SCNA蛋白或核酸之表現差異可與相對於未處理樣品,經處理樣品中之不同核酸(包括由研究人員視為適當的任何標準物,例如管家基因)之表現差異進行比較。 觀測到之差異可按照需要加以表示,例如以比率或分數形式表示,以用於與對照進行比較。在實施例中,相對於未處理樣品或經對照核酸處理之樣品,經本發明之反義寡核苷酸處理之樣品中的SCNA mRNA或蛋白質之含量增加或減少約1.25倍至約10倍或10倍以上。在實施例中,SCNA mRNA或蛋白質之含量增加或減少至少約1.25倍、至少約1.3倍、至少約1.4倍、至少約1.5倍、至少約1.6倍、至少約1.7倍、至少約1.8倍、至少約2倍、至少約2.5倍、至少約3倍、至少約3.5倍、至少約4倍、至少約4.5倍、至少約5倍、至少約5.5倍、至少約6倍、至少約6.5倍、至少約7倍、至少約7.5倍、至少約8倍、至少約8.5倍、至少約9倍、至少約9.5倍或至少約10倍或10倍以上。套組、研究試劑、診斷劑及治療劑 本發明化合物可用於診斷、治療及預防,且可用作研究試劑及套組之組分。此外,能夠以強烈特異性抑制基因表現之反義寡核苷酸常由一般技術者用於闡明特定基因之功能或區分生物路徑之各個成員之功能。 對於在套組及診斷劑及各種生物系統中之使用,單獨或與其他化合物或治療劑組合之本發明化合物適用作差值分析及/或組合分析中之工具以闡明在細胞及組織內表現之基因之一部分或整個互補序列的表現樣式。 如本文所用,術語「生物系統」或「系統」定義為表現或使得有能力表現電壓門控鈉離子通道α次單元(SCNA)基因產物的任何有機體、細胞、細胞培養物或組織。此等包括(但不限於)人類、轉殖基因動物、細胞、細胞培養物、組織、異種移植物、移植物及其組合。 作為一非限制性實例,經一或多種反義化合物處理之細胞或組織內的表現樣式與未經反義化合物處理之對照細胞或組織進行比較且分析產生之樣式以獲得基因表現量差值,因為其係關於例如所檢查基因之疾病相關性、信號傳導路徑、細胞定位、表現量、尺寸、結構或功能。可在存在或不存在會影響表現樣式之其他化合物下對經刺激或未經刺激之細胞進行此等分析。 此項技術中已知之基因表現分析方法的實例包括DNA陣列或微陣列、基因表現之連續分析(serial analysis of gene expression;SAGE)、經消化之cDNA之限制酶擴增(restriction enzyme amplification of digested cDNAs;READS)、總基因表現分析(total gene expression analysis;TOGA)、蛋白質陣列及蛋白質組研究、表現之序列標籤(expressed sequence tag;EST)定序、差減RNA指紋(subtractive RNA fingerprinting;SuRF)、差減選殖、差異性呈現(differential display;DD)、比較染色體組雜交、螢光原位雜交(fluorescent in situ hybridization;FISH)技術及質譜方法。 本發明化合物適用於研究及診斷,因為此等化合物與編碼電壓門控鈉離子通道α次單元(SCNA)之核酸雜交。舉例而言,在如本文揭示之此等條件下以此效率作為有效SCNA調節劑雜交之寡核苷酸分別為在有利於基因擴增或偵測的條件下的有效引子或探針。此等引子及探針適用於需要特異性偵測編碼SCNA之核酸分子的方法中及用於擴增該等核酸分子以偵測SCNA或用於SCNA之其他研究中。本發明之反義寡核苷酸,特定言之引子及探針與編碼SCNA之核酸的雜交可藉由此項技術中已知之手段加以偵測。此等手段可包括使酶與寡核苷酸結合、對寡核苷酸進行放射性標記或任何其他適合偵測手段。亦可製備使用此等偵測手段用於偵測樣品中之SCNA含量的套組。 反義之特異性及敏感性亦由熟習此項技術者利用以達成治療用途。反義化合物已在包括人類之動物中之疾病狀態的治療中用作治療部分。反義寡核苷酸藥物已向人類安全且有效投與且眾多臨床試驗目前正在進行中。因此確定反義化合物可為可經組態以適用於治療細胞、組織及動物,尤其人類之治療方案的適用治療模態。 對於治療,懷疑患有可藉由調節SCNA聚核苷酸之表現加以治療之疾病或病症的動物,較佳人類係藉由投與本發明之反義化合物加以治療。舉例而言,在一非限制性實施例中,方法包含向需要治療之動物投與治療有效量之SCNA調節劑的步驟。本發明之SCNA調節劑有效調節SCNA之活性或調節SCNA蛋白之表現。在一實施例中,相較於對照,SCNA在動物中之活性或表現抑制約10%。較佳地,SCNA在動物中之活性或表現抑制約30%。更佳地,SCNA在動物中之活性或表現抑制50%或50%以上。因此,相較於對照,寡聚化合物調節電壓門控鈉離子通道α次單元(SCNA)mRNA之表現至少10%、至少50%、至少25%、至少30%、至少40%、至少50%、至少60%、至少70%、至少75%、至少80%、至少85%、至少90%、至少95%、至少98%、至少99%或100%。 在一實施例中,相較於對照,電壓門控鈉離子通道α次單元(SCNA)在動物中之活性或表現增加約10%。較佳地,SCNA在動物中之活性或表現增加約30%。更佳地,SCNA在動物中之活性或表現增加50%或50%以上。因此,相較於對照,寡聚化合物調節SCNA mRNA之表現至少10%、至少50%、至少25%、至少30%、至少40%、至少50%、至少60%、至少70%、至少75%、至少80%、至少85%、至少90%、至少95%、至少98%、至少99%或100%。 舉例而言,可量測動物之血清、血液、脂肪組織、肝或任何其他體液、組織或器官中之電壓門控鈉離子通道α次單元(SCNA)表現之降低。較佳地,所分析之該等流體、組織或器官內含有之細胞含有編碼SCNA肽之核酸分子及/或SCNA蛋白自身。 本發明化合物可藉由添加有效量之化合物至適合醫藥學上可接受之稀釋劑或載劑中而以醫藥組合物加以利用。使用本發明之化合物及方法在預防上亦可適用。結合物 本發明之寡核苷酸之另一修飾涉及使增強寡核苷酸之活性,細胞分佈或細胞攝取之一或多個部分或結合物化學連接至寡核苷酸。此等部分或結合物可包括與諸如一級或二級羥基之官能基共價結合的結合基團。本發明之結合基團包括嵌入劑、報導分子、聚胺、聚醯胺、聚乙二醇、聚醚、增強寡聚物之藥效學性質之基團及增強寡聚物之藥物動力學性質之基團。典型結合基團包括膽固醇、脂質、磷脂、生物素、吩嗪、葉酸酯、啡啶、蒽醌、吖啶、螢光素、若丹明(rhodamine)、香豆素(coumarin)及染料。在本發明之情形下,增強藥效學性質之基團包括改良攝取、增強對降解之抗性、及/或增強與目標核酸之序列特異性雜交的基團。在本發明之情形下,增強藥物動力學性質之基團包括改良本發明化合物之攝取、分佈、代謝或排泄的基團。代表性結合基團揭示於1992年10月23日申請之國際專利申請案第PCT/US92/09196號及美國專利第6,287,860號中,該等專利以引用的方式併入本文中。結合部分包括(但不限於)脂質部分(諸如膽固醇部分)、膽酸、硫醚(例如己基-5-三苯甲基硫醇)、硫代膽固醇、脂族鏈(例如十二烷二醇或十一基殘基)、磷脂(例如二-十六基-外消旋-甘油或1,2-二-O-十六基-外消旋-甘油基-3-H膦酸三乙基銨)、聚胺或聚乙二醇鏈、或金剛烷乙酸、棕櫚基部分、或十八烷基胺或己胺基-羰基-羥膽固醇部分。本發明之寡核苷酸亦可與活性原料藥結合,該等活性原料藥例如為阿司匹靈(aspirin)、華法林(warfarin)、苯基丁氮酮、布洛芬(ibuprofen)、舒洛芬(suprofen)、芬布芬(fenbufen)、酮洛芬(ketoprofen)、(S)-(+)-普拉洛芬(pranoprofen)、卡洛芬(carprofen)、丹磺醯基肌胺酸(dansylsarcosine)、2,3,5-三碘苯甲酸、氟芬那酸(flufenamic acid)、醛葉酸(folinic acid)、苯并噻二疊氮(benzothiadiazide)、氯噻嗪(chlorothiazide)、二氮呯(diazepine)、吲哚美辛(indomethicin)、巴比妥鹽(barbiturate)、頭孢菌素(cephalosporin)、磺胺藥、抗糖尿病藥、抗細菌劑或抗生素。 教示製備此等寡核苷酸結合物之代表性美國專利包括(但不限於)美國專利第4,828,979號;第4,948,882號;第5,218,105號;第5,525,465號;第5,541,313號;第5,545,730號;第5,552,538號;第5,578,717號;第5,580,731號;第5,580,731號;第5,591,584號;第5,109,124號;第5,118,802號;第5,138,045號;第5,414,077號;第5,486,603號;第5,512,439號;第5,578,718號;第5,608,046號;第4,587,044號;第4,605,735號;第4,667,025號;第4,762,779號;第4,789,737號;第4,824,941號;第4,835,263號;第4,876,335號;第4,904,582號;第4,958,013號;第5,082,830號;第5,112,963號;第5,214,136號;第5,082,830號;第5,112,963號;第5,214,136號;第5,082,830號;第5,112,963號;第5,214,136號;第5,245,022號;第5,254,469號;第5,258,506號;第5,262,536號;第5,272,250號;第5,292,873號;第5,317,098號;第5,371,241號;第5,391,723號;第5,416,203號;第5,451,463號;第5,510,475號;第5,512,667號;第5,514,785號;第5,565,552號;第5,567,810號;第5,574,142號;第5,585,481號;第5,587,371號;第5,595,726號;第5,597,696號;第5,599,923號;第5,599,928號及第5,688,941號。調配物 本發明化合物亦可與其他分子、分子結構或化合物之混合物,例如脂質體、受體靶向分子、經口調配物、經直腸調配物、局部調配物或其他調配物一起摻和、囊封、結合或另外締合以幫助攝取、分佈及/或吸收。教示製備此等有助於攝取、分佈及/或吸收之調配物之代表性美國專利包括(但不限於)美國專利第5,108,921號;第5,354,844號;第5,416,016號;第5,459,127號;第5,521,291號;第5,543,165號;第5,547,932號;第5,583,020號;第5,591,721號;第4,426,330號;第4,534,899號;第5,013,556號;第5,108,921號;第5,213,804號;第5,227,170號;第5,264,221號;第5,356,633號;第5,395,619號;第5,416,016號;第5,417,978號;第5,462,854號;第5,469,854號;第5,512,295號;第5,527,528號;第5,534,259號;第5,543,152號;第5,556,948號;第5,580,575號;及第5,595,756號,各專利以引用的方式併入本文中。 儘管,反義寡核苷酸無需在載體之情形下投與以調節目標表現及/或功能,但本發明之實施例係關於用於表現反義寡核苷酸之表現載體構築體,其包含啟動子、雜交啟動基因序列且具有強組成性啟動子活性或可在所要情況下經誘導之啟動子活性。 在一實施例中,發明實務涉及以適合核酸傳遞系統投與至少一種前述反義寡核苷酸。在一實施例中,彼系統包括非病毒載體可操作地與聚核苷酸連接。此等非病毒載體之實例包括單獨寡核苷酸(例如SEQ ID NO: 29至94之任何一或多者)或寡核苷酸與適合蛋白質、多醣或脂質調配物之組合。 另外適合之核酸傳遞系統包括病毒載體,序列通常來自腺病毒、腺病毒相關病毒(AAV)、輔助病毒依賴性腺病毒、反轉錄病毒或日本血球凝集蛋白病毒-脂質體(hemagglutinatin virus of Japan-liposome,HVJ)複合物中之至少一者。較佳地,病毒載體包含強真核啟動子,例如細胞巨大病毒(cytomegalovirus,CMV)啟動子可操作地與聚核苷酸連接。 另外較佳之載體包括病毒載體、融合蛋白及化學結合物。反轉錄病毒載體包括莫洛尼鼠類白血病病毒(Moloney murine leukemia viruses)及基於HIV之病毒。一種較佳基於HIV之病毒載體包含至少兩個載體,其中gag及pol基因來自於HIV基因組且env基因來自於另一病毒。DNA病毒載體較佳。此等載體包括痘載體,諸如正痘或鳥類痘載體;疱疹病毒載體,諸如單純疱疹I病毒(herpes simplex I virus,HSV)載體;腺病毒載體及腺相關病毒載體。 本發明之反義化合物涵蓋任何醫藥學上可接受之鹽、酯或此等酯之鹽、或在向包括人類之動物投與時能夠(直接或間接)提供生物活性代謝物或其殘餘物之任何其他化合物。 術語「醫藥學上可接受之鹽」係指本發明化合物之生理學上及醫藥學上可接受之鹽:亦即保留母體化合物之所要生物活性且不賦予不合需要之毒理學效應的鹽。對於寡核苷酸,醫藥學上可接受之鹽之較佳實例及其用途進一步描述於美國專利第6,287,860號中,該專利以引用的方式併入本文中。 本發明亦包括包括本發明之反義化合物之醫藥組合物及調配物。視需要局部治療或全身性治療及欲治療之區域而定,本發明之醫藥組合物可以許多方式投與。投藥可為局部投藥(包括眼用及向黏膜投藥,包括陰道及直腸傳遞);經肺投藥(例如藉由吸入或吹入散劑或氣霧劑,包括使用噴霧器;氣管內投藥;鼻內投藥;經表皮投藥及經皮投藥);經口投藥或非經腸投藥。非經腸投藥包括靜脈內、動脈內、皮下、腹膜內或肌肉內注射或輸注;或顱內,例如鞘內或室內投藥。 對於治療中樞神經系統中之組織,可藉由例如注射或輸注入腦脊髓液中來進行投藥。向腦脊髓液中投與反義RNA描述於例如美國專利申請公開案第2007/0117772號,「Methods for slowing familial ALS disease progression」中,該專利以全文引用的方式併入本文中。 當意欲本發明之反義寡核苷酸向中樞神經系統中之細胞投與時,可與一或多種能夠促進標的反義寡核苷酸跨越血腦障壁進行穿透之藥劑一起進行投藥。可例如在內嗅皮質(entorhinal cortex)或海馬(hippocampus)中進行注射。藉由向肌肉組織中之運動神經元投與腺病毒載體來傳遞神經營養因子描述於例如美國專利第6,632,427號,「Adenoviral-vector-mediated gene transfer into medullary motor neurons」中,該專利以引用的方式併入本文中。直接向腦,例如紋狀體(striatum)、丘腦(thalamus)、海馬或黑質(substantia nigra)中傳遞載體在此項技術中為已知的且描述於例如美國專利第6,756,523號,「Adenovirus vectors for the transfer of foreign genes into cells of the central nervous system particularly in brain」中,該專利以引用的方式併入本文中。當藉由注射進行時,投藥可為快速的,或當藉由緩慢輸注或投與緩慢釋放調配物時,投藥可歷時一段時期。 標的反義寡核苷酸亦可與會提供合乎需要之醫藥或藥效學性質之藥劑連接或結合。舉例而言,反義寡核苷酸可與此項技術中已知促進跨越血腦障壁進行穿透或轉運之任何物質,諸如轉鐵蛋白(transferrin)受體之抗體偶合,且藉由靜脈內注射進行投與。反義化合物可與例如使反義化合物更有效及/或增加反義化合物跨越血腦障壁進行轉運之病毒載體連接。滲透血腦障壁破壞亦可藉由例如輸注糖,包括(但不限於)內消旋赤藻糖醇(meso erythritol)、木糖醇(xylitol)、D(+)半乳糖、D(+)乳糖、D(+)木糖、半乳糖醇(dulcitol)、肌醇(myo-inositol)、L(-)果糖、D(-)甘露糖醇、D(+)葡萄糖、D(+)阿拉伯糖(D(+) arabinose)、D(-)阿拉伯糖、纖維二糖、D(+)麥芽糖、D(+)棉子糖(D(+) raffinose)、L(+)鼠李糖(L(+) rhamnose)、D(+)蜜二糖(D(+) melibiose)、D(-)核糖、核糖醇(adonitol)、D(+)阿拉伯糖醇、L(-)阿拉伯糖醇、D(+)海藻糖(D(+) fucose)、L(-)海藻糖、D(-)來蘇糖(D(-) lyxose)、L(+)來蘇糖及L(-)來蘇糖;或胺基酸,包括(但不限於)麩醯胺酸、離胺酸、精胺酸、天冬醯胺、天冬胺酸、半胱胺酸、麩胺酸、甘胺酸、組胺酸、白胺酸、甲硫胺酸、苯丙胺酸、脯胺酸、絲胺酸、蘇胺酸、酪胺酸、纈胺酸及牛磺酸來達成。增強血腦障壁穿透之方法及材料描述於例如美國專利第4,866,042號,「Method for the delivery of genetic material across the blood brain barrier」、第6,294,520號,「Material for passage through the blood-brain barrier」、及第6,936,589號,「Parenteral delivery systems」中,該等專利皆以全文引用之方式併入本文中。 標的反義化合物可與其他分子、分子結構或化合物之混合物,例如脂質體、受體靶向分子、經口調配物、經直腸調配物、局部調配物或其他調配物一起摻和、囊封、結合或另外締合以幫助攝取、分佈及/或吸收。舉例而言,調配物中可包括陽離子脂質以促進寡核苷酸攝取。一種顯示會促進攝取之此組合物為LIPOFECTIN(可自GIBCO-BRL, Bethesda, MD獲得)。 咸信具有至少一個2'-O-甲氧基乙基修飾之寡核苷酸特別適用於經口投藥。用於局部投藥之醫藥組合物及調配物可包括經皮貼片、軟膏、洗劑、乳膏、凝膠、滴劑、栓劑、噴霧劑、液體及散劑。習知醫藥載劑、水性基質、粉末基質或油性基質、增稠劑及其類似物可為必需的或合乎需要的。經塗敷之保險套(condom)、手套及其類似物亦可為適用的。 宜以單位劑型提供之本發明之醫藥調配物可根據醫藥工業中熟知之習知技術製備。此等技術包括使活性成分與醫藥載劑或賦形劑締合之步驟。一般而言,藉由使活性成分與液體載劑或細粉狀固體載劑或兩者均一且密切締合,且接著必要時使產物成形來製備調配物。 本發明之組合物可調配成任何許多可能之劑型,諸如(但不限於)錠劑、膠囊劑、凝膠膠囊劑、液體糖漿、軟凝膠劑、栓劑及灌腸劑。本發明之組合物亦可調配成於水性、非水性或混合介質中之懸浮液。水性懸浮液可另外含有會增加懸浮液黏度之物質,包括例如羧甲基纖維素鈉、山梨糖醇及/或葡聚糖。懸浮液亦可含有穩定劑。 本發明之醫藥組合物包括(但不限於)溶液、乳液、泡沫及含脂質體調配物。本發明之醫藥組合物及調配物可包含一或多種滲透增強劑、載劑、賦形劑或其他活性成分或非活性成分。 乳液通常為一種液體分散於另一液體中之呈直徑通常超過0.1 μm之小滴形式的異質系統。除分散相及可呈於水相、油相中之溶液或自身呈分離相(separate phase)存在的活性藥物之外,乳液亦可含有其他組分。微乳液作為本發明之一實施例而包括。乳液及其使用在此項技術中為熟知的且進一步描述於美國專利第6,287,860號中。 本發明之調配物包括脂質體調配物。如本發明中所用,術語「脂質體」意謂由以球狀雙層排列之兩親媒性脂質構成的微脂粒。脂質體為單層或多層微脂粒,其具有由親脂性物質形成之膜及含有欲傳遞之組合物之水性內部。陽離子脂質體為咸信與帶負電荷之DNA分子相互作用形成穩定複合物之帶正電荷的脂質體。咸信對pH值敏感或帶負電荷之脂質體會圈閉(entrap)DNA而非與其複合。陽離子脂質體與非陽離子脂質體兩者均已用於向細胞傳遞DNA。 脂質體亦包括「空間穩定」脂質體,如本文所用之該術語係指包含一或多種特殊化脂質之脂質體。當併入脂質體中時,此等特殊化脂質會產生相對於缺乏此等特殊化脂質之脂質體,循環壽命增加之脂質體。空間穩定脂質體之實例為以下脂質體:脂質體之形成微脂粒之脂質部分的一部分包含一或多種醣脂或經一或多種親水性聚合物,諸如聚乙二醇(PEG)部分衍生化。脂質體及其使用進一步描述於美國專利第6,287,860號中。 本發明之醫藥調配物及組合物亦可包括界面活性劑。界面活性劑在藥品、調配物及乳液中之使用在此項技術中為熟知的。界面活性劑及其使用進一步描述於美國專利第6,287,860號中,該專利以引用的方式併入本文中。 在一實施例中,本發明採用各種滲透增強劑來高效傳遞核酸,特定言之寡核苷酸。除幫助非親脂性藥物跨越細胞膜進行擴散之外,滲透增強劑亦增強親脂性藥物之滲透性。滲透增強劑可分類為屬於以下五大類之一:亦即界面活性劑、脂肪酸、膽汁鹽、螯合劑、及非螯合非界面活性劑。滲透增強劑及其使用進一步描述於美國專利第6,287,860號中,該專利以引用的方式併入本文中。 熟習此項技術者應認識到調配物係根據其預定用途,亦即投藥途徑按常規方式加以設計。 用於局部投藥之較佳調配物包括本發明之寡核苷酸與局部傳遞劑,諸如脂質、脂質體、脂肪酸、脂肪酸酯、類固醇、螯合劑及界面活性劑進行摻和的調配物。較佳脂質及脂質體包括中性(例如二油醯基-磷脂醯基乙醇胺DOPE、二肉豆蔻醯基磷脂醯基膽鹼DMPC、二硬脂醯基磷脂醯基膽鹼)、帶負電荷型(例如二肉豆蔻醯基磷脂醯基甘油DMPG)及陽離子型(例如二油醯基四甲基胺丙基DOTAP及二油醯基-磷脂醯基乙醇胺DOTMA)。 對於局部或其他投藥,本發明之寡核苷酸可囊封在脂質體內或可與脂質體,特定言之與陽離子脂質體形成複合物。或者,寡核苷酸可與脂質,特定言之與陽離子脂質複合。較佳脂肪酸及酯、其醫藥學上可接受之鹽及其使用進一步描述於美國專利第6,287,860號中。 用於經口投藥之組合物及調配物包括散劑或顆粒劑、微顆粒、奈米顆粒、於水或非水性介質中之懸浮液或溶液、膠囊劑、凝膠膠囊劑、藥囊、錠劑或小錠劑。增稠劑、調味劑、稀釋劑、乳化劑、分散助劑或黏合劑可合乎需要。較佳經口調配物為本發明之寡核苷酸連同一或多種滲透增強劑、界面活性劑及螯合劑投與的調配物。較佳界面活性劑包括脂肪酸及/或其酯或鹽、膽汁酸及/或其鹽。較佳膽汁酸/鹽及脂肪酸及其使用進一步描述於美國專利第6,287,860號中,該專利以引用的方式併入本文中。滲透增強劑之組合,例如脂肪酸/鹽與膽汁酸/鹽之組合亦較佳。一種特別較佳之組合為月桂酸、癸酸及UDCA之鈉鹽。其他滲透增強劑包括聚氧乙烯-9-月桂基醚、聚氧乙烯-20-十六基醚。本發明之寡核苷酸可以顆粒形式(包括噴霧乾燥粒子)或經複合形成微粒或奈米粒子來經口傳遞。寡核苷酸複合劑及其使用進一步描述於美國專利第6,287,860號中,該專利以引用的方式併入本文中。 用於非經腸、鞘內或室內投藥之組合物及調配物可包括亦可含有緩衝劑、稀釋劑及其他適合添加劑,諸如(但不限於)滲透增強劑、載體化合物及其他醫藥學上可接受之載劑或賦形劑之無菌水溶液。 本發明之某些實施例提供含有一或多種寡聚化合物及一或多種藉由非反義機制起作用之其他化學治療劑的醫藥組合物。此等化學治療劑實例包括(但不限於)癌症化學治療藥物,諸如道諾黴素(daunorubicin)、柔紅黴素(daunomycin)、放線菌素D(dactinomycin)、小紅莓(doxorubicin)、表柔比星(epirubicin)、黃膽素(idarubicin)、依索比星(esorubicin)、博來黴素(bleomycin)、馬磷醯胺(mafosfamide)、異環磷醯胺(ifosfamide)、阿糖胞苷(cytosine arabinoside)、雙氯乙基-亞硝脲(bischloroethyl-nitrosurea)、硫酸布他卡因(busulfan)、絲裂黴素C(mitomycin C)、放線菌素D(actinomycin D)、光神黴素(mithramycin)、潑尼松(prednisone)、羥孕酮(hydroxyprogesterone)、睪固酮(testosterone)、他莫昔芬(tamoxifen)、達卡巴嗪(dacarbazine)、丙卡巴肼(procarbazine)、六甲基三聚氰胺(hexamethylmelamine)、五甲基三聚氰胺、米托蒽醌(mitoxantrone)、安吖啶(amsacrine)、苯丁酸氮芥(chlorambucil)、甲基環己基亞硝脲、氮芥(nitrogen mustard)、美法侖(melphalan)、環磷醯胺、6-巰基嘌呤、6-硫代鳥嘌呤、阿糖胞苷(cytarabine)、5-氮胞苷、羥基脲、去氧柯福黴素(deoxycoformycin)、4-羥基過氧化環磷醯胺、5-氟尿嘧啶(5-FU)、5-氟去氧尿苷(5-FUdR)、甲胺喋呤(MTX)、秋水仙鹼(colchicine)、紫杉醇(taxol)、長春新鹼(vincristine)、長春鹼(vinblastine)、依託泊苷(VP‑16)、三甲曲沙(trimetrexate)、伊立替康(irinotecan)、拓朴替康(topotecan)、吉西他濱(gemcitabine)、替尼泊甙(teniposide)、順鉑(cisplatin)及己烯雌酚(diethylstilbestrol,DES)。當與本發明化合物一起使用時,此等化學治療劑可個別(例如5-FU及寡核苷酸)、依序(例如5-FU及寡核苷酸持續一段時間使用,隨後使用MTX及寡核苷酸)或與一或多種其他此等化學治療劑(例如5-FU、MTX及寡核苷酸,或5-FU、放射線療法及寡核苷酸)組合使用。本發明組合物中亦可組合消炎藥,包括(但不限於)非類固醇消炎藥及皮質類固醇;及抗病毒藥物,包括(但不限於)病毒唑(ribivirin)、阿糖腺苷(vidarabine)、阿昔洛韋(acyclovir)及更昔洛韋(ganciclovir)。反義化合物與其他非反義藥物之組合亦在本發明範疇內。兩種或兩種以上組合之化合物可一起或依序使用。 在另一相關實施例中,本發明組合物可含有一或多種靶向第一核酸之反義化合物,特定言之寡核苷酸;及一或多種靶向第二核酸目標之其他反義化合物。舉例而言,第一目標可為電壓門控鈉離子通道α次單元(SCNA)之特定反義序列,且第二目標可為來自另一核苷酸序列之區域。或者,本發明組合物可含有兩種或兩種以上靶向同一電壓門控鈉離子通道α次單元(SCNA)核酸目標之不同區域的反義化合物。反義化合物之眾多實例在本文中加以說明且其他實例可選自此項技術中已知之適合化合物中。兩種或兩種以上組合之化合物可一起或依序使用。給藥: 咸信治療組合物之調配及其隨後投藥(給藥)在此項技術中之相關人員的技能內。給藥視欲治療之疾病狀態之嚴重性及反應性而定,其中治療過程持續數天至數月,或直至治癒或減弱疾病狀態。最佳給藥時程可由藥物在患者體內之累積之量測結果加以計算。一般技術者可容易確定最佳劑量、給藥方法及重複率。最佳劑量可視個別寡核苷酸之相對效能而變化,且可通常基於在活體外及活體內動物模型中認為有效之EC50加以估計。一般而言,劑量為每公斤體重0.01 μg至100 mg,且可每日、每週、每月或每年給予一次或一次以上,或甚至每2至20年給予一次。一般技術者可容易基於量測藥物在體液或組織中之滯留時間及濃度來估計用於給藥的重複率。在成功治療之後,可能需要使患者經受維持療法以防止疾病狀態復發,其中以在每公斤體重0.01 μg至100 mg之範圍內之維持劑量,每日一次或一次以上至每20年一次投與寡核苷酸。 在實施例中,用以下藥物劑量治療患者:每公斤體重至少約1 mg、至少約2 mg、至少約3 mg、至少約4 mg、至少約5 mg、至少約6 mg、至少約7 mg、至少約8 mg、至少約9 mg、至少約10 mg、至少約15 mg、至少約20 mg、至少約25 mg、至少約30 mg、至少約35 mg、至少約40 mg、至少約45 mg、至少約50 mg、至少約60 mg、至少約70 mg、至少約80 mg、至少約90 mg或至少約100 mg。反義寡核苷酸之某些注射劑量描述於例如美國專利第7,563,884號,「Antisense modulation of PTP1B expression」中,該專利以全文引用的方式併入本文中。 儘管以上已描述本發明之各種實施例,但應瞭解其已僅藉由實例而非限制方式加以呈現。可根據本文之揭示內容在不脫離本發明之精神或範疇之情況下對揭示之實施例作眾多變化。因此,本發明之寬度及範疇不應由任何上述實施例限制。 本文提及之所有文獻皆以引用的方式併入本文中。本申請案中引用之所有公開案及專利文獻皆出於所有目的以引用的方式併入本文中,該引用的程度就如同各個別公開案或專利文獻經如此個別表示一般。就在本文件中引用各種參考文獻而言,申請人不承認任何特定參考文獻為其發明之「先前技術」。本發明組合物及方法之實施例在以下實例中加以說明。實例 以下非限制性實例用於說明本發明之所選實施例。應瞭解比例變化及所示組分之要素之替代物將為熟習此項技術者顯而易知且在本發明之實施例之範疇內。實例 1 :設計對電壓門控鈉離子通道 α 次單元 (SCNA) 之反義核酸分子及 / SCNA 聚核苷酸之有義股具有特異性的反義寡核苷酸 如上所指示,術語「對…具有特異性之寡核苷酸」或「靶向…之寡核苷酸」係指具有(i)能夠與一部分靶向基因形成穩定複合物,或(ii)能夠與靶向基因之一部分mRNA轉錄物形成穩定雙螺旋的序列之寡核苷酸。 藉由使用電腦程式(例如IDT AntiSense Design, IDT OligoAnalyzer)來促進對適當寡核苷酸之選擇,該電腦程式自動鑑別各既定序列中將以所要熔融溫度(通常50-60℃)與目標聚核苷酸序列形成雜交體且將不形成自身二聚體或其他複合物二級結構之含19-25個核苷酸之子序列。 藉由使用自動比對核酸序列且指示具有一致性或同源性之區域之電腦程式來進一步促進對適當寡核苷酸之選擇。此等程式用於例如藉由搜尋諸如GenBank之資料庫或藉由對PCR產物定序來比較所得核酸序列。比較來自既定基因組之一定範圍之基因及基因間區域的核酸序列使得可選擇對相關基因顯示適當特異性程度的核酸序列。此等程序使得可選擇展現與目標核酸序列具有高互補程度且與既定基因組中之其他核酸序列具有較低互補程度的寡核苷酸。熟習此項技術者將認識到在選擇用於本發明中之基因之適當區域方面有很大的自由。 當反義化合物與目標核酸之結合會干擾目標核酸之正常功能以致對功能及/或活性進行調節,且存在足夠程度之互補性以避免該反義化合物與非目標核酸序列在特異性結合所要之條件下(亦即在活體內檢定或治療性治療之情況下為在生理條件下,及在活體外檢定之情況下為在進行檢定之條件下)進行非特異性結合時,該反義化合物為「可特異性雜交的」。 本文所述之寡核苷酸之雜交性質可藉由如此項技術中已知之一或多種活體外檢定確定。舉例而言,本文所述之寡核苷酸之性質可藉由使用熔融曲線檢定測定目標天然反義與潛在藥物分子之間的結合強度獲得。 目標天然反義與潛在藥物分子(分子)之間的結合強度可使用任何確立之量測分子間相互作用強度之方法,例如熔融曲線檢定來估計。 熔融曲線檢定測定天然反義/分子複合物自雙股構形快速轉變為單股構形之溫度。此溫度廣泛接受為兩個分子之間相互作用強度的可靠量度。 熔融曲線檢定可使用實際天然反義RNA分子之cDNA複本或對應於分子之結合位點之合成DNA或RNA核苷酸進行。含有所有為進行此檢定所必需之試劑之多個套組可用(例如Applied Biosystems Inc. MeltDoctor套組)。此等套組包括含有一種雙股DNA(dsDNA)結合染料(諸如ABI HRM染料、SYBR Green、SYTO等)之適合緩衝溶液。dsDNA染料之性質為其幾乎不發射游離形式之螢光,但當與dsDNA結合時具有高度螢光性。 為了進行檢定,cDNA或相應寡核苷酸以由特定製造商方案確定之濃度與分子混合。加熱混合物至95℃以解離所有預先形成之dsDNA複合物,接著緩慢冷卻至室溫或由套組製造商確定之其他較低溫度以允許DNA分子黏接。接著緩慢加熱新形成之複合物至95℃,同時連續收集關於由反應產生之螢光量的資料。螢光強度與反應中存在之dsDNA量成反比。可使用與套組相容之即時PCR儀器(例如ABI之StepOne Plus即時PCR系統或lightTyper儀器,Roche Diagnostics, Lewes, UK)收集資料。 藉由使用適當軟體(例如lightTyper(Roche)或SDS Dissociation Curve, ABI)相對於溫度(x軸)繪製螢光相對於溫度之負導數(在y軸上-d(螢光)/dT)來構築熔融峰。分析資料以鑑別自dsDNA複合物快速轉變為單股分子之溫度。此溫度稱為Tm且與兩個分子之間相互作用強度成正比。通常,Tm將超過40℃。實例 2 :調節 SCNA 聚核苷酸 用反義寡核苷酸處理 HepG2 細胞 在37℃及5% CO2 下,使來自ATCC之HepG2細胞(目錄號HB-8065)在生長培養基(MEM/EBSS(Hyclone目錄號SH30024,或Mediatech目錄號MT-10-010-CV)+10% FBS (Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素(streptomycin)(Mediatech目錄號MT30-002-CI))中生長。在實驗之前1天,細胞以1.5×105 /ml之密度再接種於6孔盤中且在37℃及5% CO2 下培育。在實驗當天,6孔盤中之培養基換成新鮮生長培養基。所有反義寡核苷酸皆稀釋成20 μM之濃度。在室溫下,2 μl此溶液與400 μl Opti-MEM培養基(Gibco目錄號31985-070)及4 μl Lipofectamine 2000 (Invitrogen目錄號11668019)一起培育20分鐘且向6孔盤之各孔施加HepG2細胞。包括2 μl水替代寡核苷酸溶液的類似混合物用於模擬轉染之對照。在37℃及5% CO2 下培育3-18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且使用Promega之SV總RNA分離系統(目錄號Z3105)或來自Qiagen之RNeasy總RNA分離套組(目錄號74181)遵循製造商說明書自細胞提取RNA。添加600 ng RNA至使用Thermo Scientific之Verso cDNA套組(目錄號AB1453B)或高容量cDNA反轉錄套組(目錄號4368813)如製造商方案中所述進行之反轉錄反應中。來自此反轉錄反應之cDNA用於藉由使用ABI Taqman基因表現混合物(目錄號4369510)及由ABI設計之引子/探針進行即時PCR來監測基因表現(根據Applied Biosystems Inc., Foster City CA之Applied Biosystems Taqman基因表現檢定:針對人類SCNA之Hs00374696_m1、Hs00897350_m1或Hs00897341_m1)。使用以下PCR循環:50℃2分鐘;95℃10分鐘;40個循環(95℃15秒,60℃1分鐘),使用StepOne Plus即時PCR機(Applied Biosystems)。 基於經處理樣品與模擬轉染樣品之間的18S校正dCt值之差異計算在用反義寡核苷酸處理之後之基因表現的倍數變化。結果: 即時PCR結果顯示HepG2細胞中之SCN1A mRNA之含量在用SCN1A反義BG724147之反義寡核苷酸處理之後48小時顯著增加(圖1、4)。針對SCN1A反義BG724147及Hs.662210所設計之其他寡核苷酸不提高SCN1A含量(圖2、3)。實例 3 :藉由用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理來上調不同細胞株中之 SCNA mRNA 在實例3中,在一組各種細胞株中,在20 nM之最終濃度下篩檢靶向SCN1A特異性天然反義轉錄物之具有不同化學組成的反義寡核苷酸。所用細胞株來源於不同器官及不同動物物種。以下資料證實經由調節SCN1A特異性天然反義轉錄物之功能使SCN1A mRNA/蛋白質上調不限於單一寡核苷酸、組織或物種且因此表示一般性現象。材料及方法 帶有德拉威特症候群相關突變之初級人類纖維母細胞。 在37℃及5% CO2 下,使帶有由N.Kenyon博士(University of Miami)引入培養物中之德拉威特症候群相關突變E1099X的初級人類皮膚纖維母細胞在由MEM(Gibco,目錄號:12561-056)+10% FBS(Mediatech,目錄號:35-015 CV)+1%抗黴性抗生素(Gibco,目錄號:15240-062)組成之生長培養基中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約2×105 /孔之密度再接種於6孔盤中且在37℃及5% CO2 下培育隔夜。次日,6孔盤中之培養基換成新鮮生長培養基(1.5毫升/孔)且細胞用反義寡核苷酸進行給藥。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。所有寡核苷酸之序列皆列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了對1個孔給藥,在室溫下,2 μl此溶液與400 μl Opti-MEM培養基(Gibco目錄號31985-070)及4 μl Lipofectamine 2000 (Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之6孔盤之1個孔中逐滴施加。包括2 μl水替代寡核苷酸溶液之類似混合物用於模擬轉染之對照。另外,相同濃度下之非活性寡核苷酸CUR-1462用作對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且使用Promega之SV總RNA分離系統(目錄號Z3105)遵循製造商說明書自細胞提取RNA。添加600奈克經純化之總RNA至使用Invitrogen之SuperScript VILO cDNA合成套組(目錄號11754-250)如製造商方案中所述進行之反轉錄反應中。來自此反轉錄反應之cDNA用於藉由使用ABI Taqman基因表現混合物(目錄號4369510)及由ABI設計之引子/探針進行即時PCR來監測基因表現(針對人類SCN1A之檢定Hs00374696_m1、Hs00897350_m1或Hs00897341_m1)。使用所有3個檢定獲得之結果極類似。使用以下PCR循環:50℃2分鐘;95℃10分鐘;40個循環(95℃15秒,60℃1分鐘),使用StepOne Plus即時PCR系統(Applied Biosystems)。18S之檢定由ABI製造(目錄號4319413E)。基於經處理樣品與模擬轉染樣品之間的18S校正dCt值之差異計算在用反義寡核苷酸處理之後之基因表現的倍數變化。對於替代性同一天方法,所有程序皆類似地進行,但細胞在第一天在其分配至6孔盤中之後即刻用反義寡核苷酸進行給藥。SK-N-AS 細胞株 。在37℃及5% CO2 下,使來自ATCC之SK-N-AS人類神經母細胞瘤細胞(目錄號CRL-2137)在生長培養基(DMEM(Mediatech目錄號10-013-CV)+10% FBS (Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI)+非必需胺基酸(NEAA)(HyClone SH30238.01))中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約3×105 /孔之密度再接種於6孔盤中且在37℃及5% CO2 下培育隔夜。次日,6孔盤中之培養基換成新鮮生長培養基(1.5毫升/孔)且細胞用反義寡核苷酸進行給藥。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。所有寡核苷酸之序列皆列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了對1個孔給藥,在室溫下,2 μl此溶液與400 μl Opti-MEM培養基(Gibco目錄號31985-070)及4 μl Lipofectamine 2000 (Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之6孔盤之1個孔中逐滴施加。包括2 μl水替代寡核苷酸溶液之類似混合物用於模擬轉染之對照。另外,相同濃度下之非活性寡核苷酸CUR-1462用作對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且使用Promega之SV總RNA分離系統(目錄號Z3105)遵循製造商說明書自細胞提取RNA。添加600奈克經純化之總RNA至使用Invitrogen之SuperScript VILO cDNA合成套組(目錄號11754-250)如製造商方案中所述進行之反轉錄反應中。來自此反轉錄反應之cDNA用於藉由使用ABI Taqman基因表現混合物(目錄號4369510)及由ABI設計之引子/探針進行即時PCR來監測基因表現(針對人類SCN1A之檢定Hs00374696_m1、Hs00897350_m1或Hs00897341_m1)。使用所有3個檢定獲得之結果極類似。使用以下PCR循環:50℃2分鐘;95℃10分鐘;40個循環(95℃15秒,60℃1分鐘),使用StepOne Plus即時PCR系統(Applied Biosystems)。18S之檢定由ABI製造(目錄號4319413E)。基於經處理樣品與模擬轉染樣品之間的18S校正dCt值之差異計算在用反義寡核苷酸處理之後之基因表現的倍數變化。對於替代性同一天方法,所有程序皆類似地進行,但細胞在第一天在其分配至6孔盤中之後即刻用反義寡核苷酸進行給藥。CHP-212 細胞株。 在37℃及5% CO2 下,使來自ATCC之CHP-212人類神經母細胞瘤細胞(目錄號CRL-2273)在生長培養基(1: MEM及F12(分別為ATCC目錄號30-2003及Mediatech目錄號10-080-CV)之混合物+10% FBS(Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI))中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約2×105 /孔之密度再接種於6孔盤中且在37℃及5% CO2 下培育隔夜。次日,6孔盤中之培養基換成新鮮生長培養基(1.5毫升/孔)且細胞用反義寡核苷酸進行給藥。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。所有寡核苷酸之序列皆列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了對1個孔給藥,在室溫下,2 μl此溶液與400 μl Opti-MEM培養基(Gibco目錄號31985-070)及4 μl Lipofectamine 2000 (Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之6孔盤之1個孔中逐滴施加。包括2 μl水替代寡核苷酸溶液之類似混合物用於模擬轉染之對照。另外,相同濃度下之非活性寡核苷酸CUR-1462用作對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且使用Promega之SV總RNA分離系統(目錄號Z3105)遵循製造商說明書自細胞提取RNA。添加600奈克經純化之總RNA至使用Invitrogen之SuperScript VILO cDNA合成套組(目錄號11754-250)如製造商方案中所述進行之反轉錄反應中。來自此反轉錄反應之cDNA用於藉由使用ABI Taqman基因表現混合物(目錄號4369510)及由ABI設計之引子/探針進行即時PCR來監測基因表現(針對人類SCN1A之檢定Hs00374696_m1、Hs00897350_m1或Hs00897341_m1)。使用所有3個檢定獲得之結果極類似。使用以下PCR循環:50℃2分鐘;95℃10分鐘;40個循環(95℃15秒,60℃1分鐘),使用StepOne Plus即時PCR系統(Applied Biosystems)。18S之檢定由ABI製造(目錄號4319413E)。基於經處理樣品與模擬轉染樣品之間的18S校正dCt值之差異計算在用反義寡核苷酸處理之後之基因表現的倍數變化。對於替代性同一天方法,所有程序皆類似地進行,但細胞在第一天在其分配至6孔盤中之後即刻用反義寡核苷酸進行給藥。Vero76 細胞株。 在37℃及5% CO2 下,使來自ATCC之Vero76非洲綠猴胚腎細胞(目錄號CRL-1587)在生長培養基(達爾伯克氏改良伊格爾氏培養基(Dulbecco's Modified Eagle's Medium)(Cellgrow 10-013-CV)+5% FBS(Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI))中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約105 /孔之密度再接種於6孔盤中且在37℃及5% CO2 下培育隔夜。次日,6孔盤中之培養基換成新鮮生長培養基(1.5毫升/孔)且細胞用反義寡核苷酸進行給藥。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。所有寡核苷酸之序列皆列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了對1個孔給藥,在室溫下,2 μl此溶液與400 μl Opti-MEM培養基(Gibco目錄號31985-070)及4 μl Lipofectamine 2000(Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之6孔盤之1個孔中逐滴施加。包括2 μl水替代寡核苷酸溶液之類似混合物用於模擬轉染之對照。另外,相同濃度下之非活性寡核苷酸CUR-1462用作對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且使用Promega之SV總RNA分離系統(目錄號Z3105)遵循製造商說明書自細胞提取RNA。添加600奈克經純化之總RNA至使用Invitrogen之SuperScript VILO cDNA合成套組(目錄號11754-250)如製造商方案中所述進行之反轉錄反應中。來自此反轉錄反應之cDNA用於藉由使用ABI Taqman基因表現混合物(目錄號4369510)及由ABI設計之引子/探針進行即時PCR來監測基因表現(針對人類SCN1A之檢定Hs00374696_m1、Hs00897350_m1或Hs00897341_m1)。使用以下PCR循環:50℃2分鐘;95℃10分鐘;40個循環(95℃15秒,60℃1分鐘),使用StepOne Plus即時PCR系統(Applied Biosystems)。18S之檢定由ABI製造(目錄號4319413E)。基於經處理樣品與模擬轉染樣品之間的18S校正dCt值之差異計算在用反義寡核苷酸處理之後之基因表現的倍數變化。對於替代性同一天方法,所有程序皆類似地進行,但細胞在第一天在其分配至6孔盤中之後即刻用反義寡核苷酸進行給藥。3T3 細胞株。 在37℃及5% CO2 下,使來自ATCC之3T3小鼠胚胎纖維母細胞(目錄號CRL-1658)在生長培養基(達爾伯克氏改良伊格爾氏培養基(Cellgrow 10-013-CV)+10%胎牛血清(Cellgrow 35-22-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI))中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約105 /孔之密度再接種於6孔盤中且在37℃及5% CO2 下培育隔夜。次日,6孔盤中之培養基換成新鮮生長培養基(1.5毫升/孔)且細胞用反義寡核苷酸進行給藥。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。所有寡核苷酸之序列皆列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了對1個孔給藥,在室溫下,2 μl此溶液與400 μl Opti-MEM培養基(Gibco目錄號31985-070)及4 μl Lipofectamine 2000 (Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之6孔盤之1個孔中逐滴施加。包括2 μl水替代寡核苷酸溶液之類似混合物用於模擬轉染之對照。另外,相同濃度下之非活性寡核苷酸CUR-1462用作對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且使用Promega之SV總RNA分離系統(目錄號Z3105)遵循製造商說明書自細胞提取RNA。添加600奈克經純化之總RNA至使用Invitrogen之SuperScript VILO cDNA合成套組(目錄號11754-250)如製造商方案中所述進行之反轉錄反應中。來自此反轉錄反應之cDNA用於藉由使用ABI Taqman基因表現混合物(目錄號4369510)及由ABI設計之引子/探針進行即時PCR來監測基因表現(針對人類SCN1A之檢定Hs00374696_m1、Hs00897350_m1或Hs00897341_m1)。使用所有3個檢定獲得之結果極類似。使用以下PCR循環:50℃2分鐘;95℃10分鐘;40個循環(95℃15秒,60℃1分鐘),使用StepOne Plus即時PCR系統(Applied Biosystems)。18S之檢定由ABI製造(目錄號4319413E)。基於經處理樣品與模擬轉染樣品之間的18S校正dCt值之差異計算在用反義寡核苷酸處理之後之基因表現的倍數變化。對於替代性同一天方法,所有程序皆類似地進行,但細胞在第一天在其分配至6孔盤中之後即刻用反義寡核苷酸進行給藥。HepG2 細胞株。 來自ATCC之HepG2人類肝細胞癌細胞(目錄號HB-8065)在生長培養基(MEM/EBSS(Hyclone目錄號SH30024,或Mediatech目錄號MT-10-010-CV)+10% FBS (Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI))中在37℃及5% CO2 生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約3×105 個/孔之密度再接種於6孔盤中且在37℃及5% CO2 下培育隔夜。次日,6孔盤中之培養基換成新鮮生長培養基(1.5毫升/孔)且投與細胞反義寡核苷酸。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。所有寡核苷酸之序列列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之滅菌水中稀釋成20 μM濃度。為1孔給藥,2 μl此溶液與400 μl Opti-MEM培養基(Gibco目錄號31985-070)及4 μl Lipofectamine 2000(Invitrogen目錄號11668019)在室溫一起培育20分鐘且向具有細胞之6孔盤之1個孔中逐滴施加。包括2 μl水替代寡核苷酸溶液之類似混合物用於模擬轉染對照。另外,相同濃度之非活性寡核苷酸CUR-1462用作對照。在37℃及5% CO2 培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且使用Promega之SV總RNA分離系統(目錄號Z3105)遵循製造商說明書自細胞提取RNA。600奈克經純化之總RNA加入使用Invitrogen之SuperScript VILO cDNA合成套組(目錄號11754-250)如製造商方案中所述進行之反轉錄反應中。使用來自此反轉錄反應之cDNA藉由使用ABI Taqman基因表現混合物(目錄號4369510)及ABI設計之引子/探針進行即時PCR來監測基因表現(針對人類SCN1A之檢定Hs00374696_m1、Hs00897350_m1或Hs00897341_m1)。使用所有3個檢定所獲得之結果極類似。使用以下PCR循環:50℃2分鐘,95℃10分鐘,40個循環(95℃15秒,60℃1分鐘),使用StepOne Plus即時PCR系統(Applied Biosystems)。18S之檢定由ABI製造(目錄號4319413E)。基於經處理樣品與模擬轉染樣品之間18S校正dCt值之差異計算以反義寡核苷酸處理之後基因表現的倍數變化。對於替代性同一天方法,所有程序類似地進行,但在第一天細胞分配至6孔盤之後即刻投與反義寡核苷酸。結果。 相較於模擬轉染之對照,在用20 nM反義寡核苷酸處理之後SCN1A mRNA於不同細胞株中之含量展示於表2中。如自資料所見,當以20 nM施加時,一些寡核苷酸在上調SCN1A mRNA含量方面具有高度活性且在若干物種(人類、非洲綠猴及小鼠)中、在源於不同器官/細胞類型(肝、腎、腦、胚胎纖維母細胞)之細胞株及帶有SCN1A突變之初級皮膚纖維母細胞中一致顯示上調。帶有德拉威特突變之細胞中之SCN1A蛋白的上調支持用於治療與SCN1A基因突變相關之疾病的方法之適合性。針對天然反義序列設計之一些寡核苷酸不影響或僅微小影響所有或一些測試細胞株中之SCN1A mRNA含量。此等差異與指示寡核苷酸之結合可能取決於寡核苷酸之目標序列之二級及三級結構的文獻資料一致。值得注意的是用與SCN1A天然反義序列無同源性但化學組成類似之寡核苷酸(CUR-1462)處理之細胞中的SCN1A含量不顯著不同於模擬轉染之對照,此證實靶向寡核苷酸之影響不取決於此等分子之非特異性毒性。實例 4 :藉由用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理來上調不同細胞株中之 SCNA mRNA 的劑量依賴性 在實例4中,在一組各種細胞株中,在5至80 nM之範圍內之最終濃度下篩檢靶向SCNA特異性天然反義轉錄物之具有不同化學組成的反義寡核苷酸。所用細胞株來源於不同器官及不同動物物種。以下資料證實經由調節SCNA特異性天然反義轉錄物之功能來上調SCNA mRNA之程度可藉由施加不同量之活性寡核苷酸來改變。材料及方法。 SK-N-AS、Vero 76及帶有德拉威特突變之初級人類纖維母細胞如實例2中所述用反義寡核苷酸處理,例外之處為用於處理各孔之寡核苷酸及Lipofectamine 2000濃度不同。調整寡核苷酸及Lipofectamine 2000濃度以便確保5、10、20、40及80 nM之最終寡核苷酸濃度及2:1(v:v)之Lipofectamine 2000與20 μM寡核苷酸儲備溶液的比率。結果。 劑量反應實驗之結果已證實靶向SCN1A特異性天然反義RNA之反義寡核苷酸可誘導SCN1A mRNA之劑量依賴性上調(圖1-3)。在一些情況下,在較高劑量下,此上調極為強力(多達60倍)(圖1-3)。由同一核苷酸在不同細胞株中誘導之上調程度似乎不同,例如在40 nM下在初級纖維母細胞中達成之上調程度為10-40倍,而在相同濃度下由同一寡核苷酸在Vero 76細胞中達成之上調為2-6倍(圖1對圖3)。此等差異可歸因於不同細胞株之不同轉染效率及/或由其表現之各種回饋路徑。大多數寡核苷酸之影響在約40 nM下達到平穩狀態,例外為CUR-1764及CUR-1770在SCN1A纖維母細胞中及在Vero 76細胞中測試之所有寡核苷酸,在以上情況下在最高測試濃度下未達到平穩狀態(圖1-3)。實例 5 :藉由靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸上調 SCNA mRNA 之序列特異性 在實例5中,在經設計以證實由寡核苷酸引起之SCN1A上調獨立於與所用寡核苷酸化學組成相關之非特異性毒性的實驗中,測試靶向SCN1A特異性天然反義轉錄物之反義寡核苷酸。以下資料證實經由調節SCN1A特異性天然反義轉錄物之功能來上調SCN1A mRNA之程度僅取決於活性寡核苷酸之量,而非化學組成類似之分子的總量。材料及方法 :Vero 76及帶有德拉威特突變之初級人類纖維母細胞如實例2中所述用反義寡核苷酸處理,例外之處為用於處理各孔之寡核苷酸濃度不同。活性寡核苷酸與化學組成類似但在人類基因組中無已知目標且對測試之多個基因之表現無影響(資料未展示)的非活性寡核苷酸(CUR-1462)共投與。寡核苷酸之總量以及Lipofectamine 2000之量保持恆定,而改變活性寡核苷酸在混合物中之比例。調整寡核苷酸濃度以便確保5、10、20及40 nM之最終活性寡核苷酸濃度及40 nM之總寡核苷酸濃度(活性+非活性)。 如自資料(圖7)所見,靶向SCN1A天然反義之寡核苷酸之劑量依賴性影響不由與此等分子潛在相關之非特異性毒性產生。SCN1A mRNA含量視用於處理其之活性寡核苷酸之劑量而定(圖7)。實例 6 :藉由靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸上調 SCNA mRNA 之目標特異性 在實例6中,靶向SCN1A特異性天然反義轉錄物之反義寡核苷酸在經設計以證實其目標(亦即SCN1A)之特異性的實驗中加以測試。以下資料證實經由調節SCN1A特異性天然反義轉錄物之功能來上調SCN1A mRNA限於SCN1A mRNA且不影響相關鈉離子通道SCN9A、SCN8A、SCN7A、SCN3A及SCN2A。材料及方法。 Vero 76及帶有德拉威特突變之初級人類纖維母細胞如實例3中所述用反義寡核苷酸處理。在處理後,如實例2中所述分析經分離之RNA,例外之處為使用Taqman基因表現檢定來即時PCR偵測SCN9A、SCN8A、SCN7A、SCN3A及SCN2A通道之mRNA。用於人類SCN9A、SCN8A、SCN7A、SCN3A及SCN2A通道之α次單元之檢定係自ABI Inc.(分別為目錄號Hs00161567_m1、Hs00274075_m1、Hs00161546_m1、Hs00366902_m1及Hs00221379_m1)獲得。結果 。如圖8中所示,用寡核苷酸CUR-1916及CUR-1770處理不顯著影響SCN8A及SCN9A通道在帶有德拉威特突變之人類纖維母細胞中之表現。在處理之前或之後,SCN7A、SCN3A及SCN2A通道之表現在此等細胞中不可偵測(資料未展示)。資料證實使用針對既定基因之天然反義RNA之寡核苷酸進行基因表現調節的特異性。實例 7 :靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸之穩定性 在實例7中,兩批靶向SCN1A特異性天然反義轉錄物之反義寡核苷酸在經設計以檢查其在4℃下於稀釋(1 mM)水溶液中儲存之後之穩定性的實驗中加以測試。以下資料顯示寡核苷酸可在此等條件中持續至少6個月之時期穩定而不顯著損失活性。材料及方法。 Vero 76細胞如實例2中所述用兩批不同反義寡核苷酸處理。該等批料係在2010年8月及2011年3月合成。2010年8月合成之寡核苷酸在4℃下以1 mM水溶液形式儲存。2011年3月合成之寡核苷酸在合成之後3天內以凍乾形式運送且在到達後即刻加以測試。結果: 如圖9中所示,在寡核苷酸在4℃下於水溶液中儲存6個月之久之後,無顯著生物活性損失。實例 8 :用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理之帶有德拉威特症候群相關突變的初級人類纖維母細胞中之 SCNA 蛋白上調 此實驗之目的在於根據反義寡核苷酸CUR-1740、CUR-1770及CUR-1916上調SCNA蛋白在帶有德拉威特症候群相關突變之纖維母細胞中的表現之能力對其進行分級。材料及方法。 在37℃及5% CO2 下,使帶有由N.Kenyon博士(University of Miami)引入培養物中之德拉威特症候群相關突變的纖維母細胞在由MEM(Gibco,目錄號:12561-056)+10% FBS(Mediatech,目錄號:35-015 CV)+1%抗黴性抗生素(Gibco,目錄號:15240-062)組成之生長培養基中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約4×104 /孔之密度再接種於24孔盤中且在37℃及5% CO2 下培育隔夜。次日,24孔盤中之培養基換成新鮮生長培養基(1毫升/孔)且細胞用反義寡核苷酸CUR-1740、CUR-1770及CUR-1916進行給藥。所有反義寡核苷酸皆由IDT Inc. (Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。寡核苷酸CUR-1740、CUR-1770及CUR-1916之序列列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了以20 nM之最終濃度對1個孔給藥,在室溫下,1 μl 20 μM寡核苷酸儲備溶液與200 μl Opti-MEM培養基(Gibco目錄號31985-070)及2 μl Lipofectamine 2000(Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之24孔盤之1個孔中逐滴施加。為了達成5、10、40及80 nM之最終濃度,所用20 μM寡核苷酸儲備溶液之體積經相應調整。20 μM寡核苷酸儲備溶液與Lipofectamine 2000之比率為1:2(v:v)。包括8 μl水替代寡核苷酸溶液及相應體積之Lipofectamine 2000的類似混合物用於模擬轉染之對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且細胞用無鈣及鎂之達爾伯克氏磷酸鹽緩衝鹽水(PBS)(Mediatech目錄號21-031-CV)洗滌3次。接著丟棄PBS且在-20℃下使用300 μl 100%甲醇於24孔盤中固定細胞15分鐘。在移除甲醇且用PBS洗滌之後,在21℃下細胞與3%過氧化氫(Fisher Chemical目錄號H325-100)一起培育5分鐘。細胞用PBS洗滌3次每次5分鐘,接著在21℃下與300 μl含0.1%牛血清白蛋白(BSA)(Sigma目錄號A-9647)之PBS一起培育30分鐘。細胞用PBS洗滌3次每次5分鐘,接著在21℃下與300 μl抗生蛋白(avidin)溶液(Vector Laboratories目錄號SP-2001)一起培育30分鐘。細胞用PBS簡短沖洗3次,接著在21℃下與生物素溶液(Vector Laboratories目錄號SP-2001)一起培育30分鐘。細胞用PBS洗滌3次且接著在4℃下與每孔300 μl以250倍稀釋於PBS/0.1% BSA中之對抗人類SCN1A之兔抗體(Abcam目錄號ab24820)一起培育隔夜。在21℃下使板平衡5分鐘之後,細胞用PBS洗滌3次每次持續5分鐘,接著在21℃下與以200倍稀釋於PBS/0.1% BSA中之山羊抗兔抗體一起培育30分鐘。細胞用PBS洗滌3次每次5分鐘且接著與300 μl Vectastain Elite ABC試劑A+B溶液(Vector Laboratories目錄號PK-6101)一起培育30分鐘;Vectastain Elite ABC試劑A+B溶液係在21℃下在與細胞一起培育之前30分鐘藉由依次向5 ml PBS中添加並混合2滴試劑A及2滴試劑B加以製備。在21℃下細胞用PBS洗滌3次每次持續5分鐘,且接著與二胺基聯苯胺(DAB)過氧化酶受質溶液(Vector Laboratories目錄號SK-4105)一起培育直至細胞染色;DAB過氧化酶受質溶液係在添加至細胞中之前藉由混合1 ml ImmPACT™DAB稀釋液與30 μl ImmPACT™ DAB色素原濃縮物加以復原。此時,細胞用PBS簡短洗滌3次且各孔中留有300 μl PBS。在Dell Latitude D630膝上型電腦之螢幕上使用配備有與Nikon數位觀測設備耦合之Nikon DS-Ri1攝影機的反向Nikon Eclipse TS100顯微鏡,直接在24孔盤之各孔內部分析細胞之染色。個別孔之相片係使用具有Nikon攝影機之軟體NIS-Elements D 3.0拍攝。結果。 所有測試之反義寡核苷酸皆高效上調SCN1A蛋白,其中CUR-1770及CUR-1916為兩種最佳反義寡核苷酸(圖10)。實例 9 :用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理之 SK-N-AS 細胞中的 SCNA 蛋白上調 此實驗之目的在於根據反義寡核苷酸CUR-1740、CUR-1764、CUR-1770及CUR-1916上調SCN1A蛋白在SK-N-AS細胞中之表現的能力對其進行分級。SK-N-AS為人類神經母細胞瘤細胞株。材料及方法 :在37℃及5% CO2 下使來自ATCC之SK-N-AS人類神經母細胞瘤細胞(目錄號CRL-2137)在生長培養基(DMEM(Mediatech目錄號10-013-CV)+10% FBS(Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI)+非必需胺基酸(NEAA)(HyClone SH30238.01))中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約5×104 /孔之密度再接種於24孔盤中且在37℃及5% CO2 下培育隔夜。次日,24孔盤中之培養基換成新鮮生長培養基(1毫升/孔)且細胞用反義寡核苷酸CUR-1740、CUR-1764、CUR-1770及CUR-1916進行給藥。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon(Vedbaek, Denmark)製造。寡核苷酸CUR-1740、CUR-1764、CUR-1770及CUR-1916之序列列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了以20 nM之最終濃度對1個孔給藥,在室溫下,1 μl 20 μM寡核苷酸儲備溶液與200 μl Opti-MEM培養基(Gibco目錄號31985-070)及2 μl Lipofectamine 2000(Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之24孔盤之1個孔中逐滴施加。為了達成5、10、40及80 nM之最終濃度,所用20 μM寡核苷酸儲備溶液之體積經相應調整。20 μM寡核苷酸儲備溶液與Lipofectamine 2000之比率為1:2(v:v)。包括8 μl水替代寡核苷酸溶液及相應體積之Lipofectamine 2000的類似混合物用於模擬轉染之對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且細胞用無鈣及鎂之達爾伯克氏磷酸鹽緩衝鹽水(PBS)(Mediatech目錄號21-031-CV)洗滌3次。接著丟棄PBS且在-20℃下使用300 μl 100%甲醇於24孔盤中固定細胞15分鐘。在移除甲醇且用PBS洗滌之後,在21℃下細胞與3%過氧化氫(Fisher Chemical目錄號H325-100)一起培育5分鐘。細胞用PBS洗滌3次每次5分鐘,接著在21℃下與300 μl含0.1%牛血清白蛋白(BSA)(Sigma目錄號A-9647)之PBS一起培育30分鐘。細胞用PBS洗滌3次每次5分鐘,接著在21℃下與300 μl抗生蛋白溶液(Vector Laboratories目錄號SP-2001)一起培育30分鐘。細胞用PBS簡短沖洗3次,接著在21℃下與生物素溶液(Vector Laboratories目錄號SP-2001)一起培育30分鐘。細胞用PBS洗滌3次且接著在4℃下與每孔300 μl以250倍稀釋於PBS/0.1% BSA中之對抗人類SCN1A之兔抗體(Abcam目錄號ab24820)一起培育隔夜。在21℃下使培養盤平衡5分鐘之後,細胞用PBS洗滌3次每次5分鐘,接著在21℃下與以200倍稀釋於PBS/0.1% BSA中之山羊抗兔抗體一起培育30分鐘。細胞用PBS洗滌3次每次5分鐘且接著與300 μl Vectastain Elite ABC試劑A+B溶液(Vector Laboratories目錄號PK-6101)一起培育30分鐘;Vectastain Elite ABC試劑A+B溶液係在21℃下在與細胞一起培育之前30分鐘藉由依次向5 ml PBS中添加並混合2滴試劑A及2滴試劑B加以製備。在21℃下細胞用PBS洗滌3次每次5分鐘,且接著與二胺基聯苯胺(DAB)過氧化酶受質溶液(Vector Laboratories目錄號SK-4105)一起培育直至細胞染色;DAB過氧化酶受質溶液係在添加至細胞中之前藉由混合1 ml ImmPACT™DAB稀釋液與30 μl ImmPACT™ DAB色素原濃縮物加以復原。此時,細胞用PBS簡短洗滌3次且各孔中留有300 μl PBS。在Dell Latitude D630膝上型電腦之螢幕上使用配備有與Nikon數位觀測設備耦合之Nikon DS-Ri1攝影機的反向Nikon Eclipse TS100顯微鏡,直接在24孔盤之各孔內部分析細胞之染色。個別孔之相片係使用具有Nikon攝影機之軟體NIS-Elements D 3.0拍攝。結果: 所有測試之反義寡核苷酸皆上調SCN1A蛋白,其中CUR-1764及CUR-1770為兩種最佳反義寡核苷酸(圖11)。實例 10 :用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理之 Vero 76 細胞中的 SCNA 蛋白上調 此實驗之目的在於根據反義寡核苷酸CUR-1740、CUR-1770、CUR-1916、CUR-1924及CUR-1945上調SCN1A蛋白在Vero 76細胞中之表現的能力對其進行分級。Vero76為草原猴(Cercopithecus aethiops )(綠色猴(vervet)或非洲綠猴)腎細胞株。材料及方法: 在37℃及5% CO2 下,使來自ATCC之Vero76非洲綠猴胚腎細胞(目錄號CRL-1587)在生長培養基(達爾伯克氏改良伊格爾氏培養基(Cellgrow 10-013-CV)+5% FBS(Mediatech目錄號MT35-011-CV)+青黴素/鏈黴素(Mediatech目錄號MT30-002-CI))中生長。使用以下方法之一用反義寡核苷酸處理細胞。對於次日方法,在實驗之前1天,將細胞於生長培養基中以約4×104 /孔之密度再接種於24孔盤中且在37℃及5% CO2 下培育隔夜。次日,24孔盤中之培養基換成新鮮生長培養基(1毫升/孔)且細胞用反義寡核苷酸CUR-1740、CUR-1770及CUR-1916進行給藥。所有反義寡核苷酸皆由IDT Inc.(Coralville, IA)或Exiqon (Vedbaek, Denmark)製造。寡核苷酸CUR-1740、CUR-1770、CUR-1916、CUR-1924及CUR-1945之序列列於表1中。寡核苷酸之儲備溶液於無DNAse/RNAse之無菌水中稀釋成20 μM之濃度。為了以20 nM之最終濃度對1個孔給藥,在室溫下,1 μl 20 μM寡核苷酸溶液與200 μl Opti-MEM培養基(Gibco目錄號31985-070)及2 μl Lipofectamine 2000 (Invitrogen目錄號11668019)一起培育20分鐘且向具有細胞之24孔盤之1個孔中逐滴施加。為了達成5、10、40及80 nM之最終濃度,所用20 μM寡核苷酸儲備溶液之體積經相應調整。20 μM寡核苷酸儲備溶液與Lipofectamine 2000之比率為1:2(v:v)。包括8 μl水替代寡核苷酸溶液及相應體積之Lipofectamine 2000的類似混合物用於模擬轉染之對照。在37℃及5% CO2 下培育約18小時之後,培養基換成新鮮生長培養基。在添加反義寡核苷酸之後48小時,移除培養基且細胞用無鈣及鎂之達爾伯克氏磷酸鹽緩衝鹽水(PBS)(Mediatech目錄號21-031-CV)洗滌3次。丟棄PBS且在-20℃下使用300 μl 100%甲醇於24孔盤中固定Vero 76細胞15分鐘。在移除甲醇且用PBS洗滌細胞之後,在21℃下細胞與3%過氧化氫(Fisher Chemical目錄號H325-100)一起培育5分鐘。細胞用PBS洗滌3次每次5分鐘,接著在21℃下與300 μl含0.1%牛血清白蛋白(BSA)(Sigma目錄號A-9647)之PBS一起培育30分鐘。細胞用PBS洗滌3次每次5分鐘,接著在21℃下與300 μl抗生蛋白溶液(Vector Laboratories目錄號SP-2001)一起培育30分鐘。細胞用PBS簡短沖洗3次,接著在21℃下與生物素溶液(Vector Laboratories目錄號SP‑2001)一起培育30分鐘。細胞用PBS洗滌3次且接著在4℃下與每孔300 μl以250倍稀釋於PBS/0.1% BSA中之對抗人類SCN1A之兔抗體(Abcam目錄號ab24820)一起培育隔夜。在21℃下使培養盤平衡5分鐘之後,細胞用PBS洗滌3次每次5分鐘,接著在21℃下與以200倍稀釋於PBS/0.1% BSA中之山羊抗兔抗體一起培育30分鐘。細胞用PBS洗滌3次每次5分鐘且接著與300 μl Vectastain Elite ABC試劑A+B溶液(Vector Laboratories目錄號PK-6101)一起培育30分鐘;Vectastain Elite ABC試劑A+B溶液係在21℃下在與細胞一起培育之前30分鐘藉由依次向5 ml PBS中添加並混合2滴試劑A及2滴試劑B加以製備。在21℃下細胞用PBS洗滌3次每次5分鐘,且接著與二胺基聯苯胺(DAB)過氧化酶受質溶液(Vector Laboratories目錄號SK-4105)一起培育直至細胞染色;DAB過氧化酶受質溶液係在添加至細胞中之前藉由混合1 ml ImmPACT™DAB稀釋液與30 μl ImmPACT™ DAB色素原濃縮物加以復原。此時,細胞用PBS簡短洗滌3次且各孔中留有300 μl PBS。在Dell Latitude D630膝上型電腦之螢幕上使用配備有與Nikon數位觀測設備耦合之Nikon DS-Ri1攝影機的反向Nikon Eclipse TS100顯微鏡,直接在24孔盤之各孔內部分析細胞之染色。個別孔之相片係使用具有Nikon攝影機之軟體NIS-Elements D 3.0拍攝。結果。 所有測試之反義寡核苷酸皆上調SCN1A蛋白,其中CUR-1764及CUR-1770產生最高上調(圖12)。實例 11 :強力上調 SCNA mRNA 之靶向 SCNA 特異性天然反義轉錄物之寡核苷酸不上調 Vero76 細胞中之肌動蛋白 (actin)mRNA 此實驗之目的在於檢查顯示會上調SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物的反義寡核苷酸(CUR-1924、CUR-1740、CUR-1838)是否能夠調控Vero76非洲綠猴胚腎細胞中之其他非相關基因(諸如肌動蛋白)之mRNA。材料及方法。 來自ATCC之Vero76非洲綠猴胚腎細胞株(目錄號CRL-1587)在與實例2中所述相同之條件中進行給藥。肌動蛋白mRNA係藉由如實例2中所述之即時PCR加以定量,例外之處為此次由ABI設計之引子/探針對肌動蛋白(目錄號Hs99999903_m1)具有特異性。資料呈現於圖13中。結果。 如圖13中所示,測試在實例3及10中顯示會上調Vero76細胞中之SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物的寡核苷酸(CUR-1924、CUR-1740、CUR-1838)對Vero 76細胞中之肌動蛋白mRNA表現之影響。圖13中之資料證實靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調非相關基因,諸如肌動蛋白。因此,此等寡核苷酸在上調SCN1A方面具有特異性。實例 12 :顯示會上調 SCNA mRNA 及蛋白質之靶向 SCNA 特異性天然反義轉錄物之寡核苷酸不上調帶有德拉威特相關突變之初級纖維母細胞中的肌動蛋白 mRNA 此實驗之目的在於檢查顯示會上調SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物的反義寡核苷酸(CUR-1916、CUR-1945)是否能夠調控帶有德拉威特症候群相關突變E1099X之初級人類皮膚纖維母細胞中的其他非相關基因(諸如肌動蛋白)之mRNA。材料及方法。 帶有由N.Kenyon博士(University of Miami)引入培養物中之德拉威特症候群相關突變E1099X的初級人類皮膚纖維母細胞在與實例3中所述相同之條件中進行給藥。肌動蛋白mRNA係藉由如實例3中所述之即時PCR加以定量,例外之處為此次由ABI設計之引子/探針對肌動蛋白(目錄號Hs99999903_m1)具有特異性。資料呈現於圖14中。結果 :如圖14中所示,靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調非相關基因,諸如肌動蛋白。因此,此等寡核苷酸在上調SCN1A方面具有特異性。實例 13 :顯示會上調 SCNA mRNA 及蛋白質之靶向 SCNA 特異性天然反義轉錄物之寡核苷酸不上調 SK-N-AS 細胞中的肌動蛋白 mRNA 此實驗之目的在於檢查顯示會上調SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物的反義寡核苷酸(CUR-1740、CUR-1764、CUR-1770、CUR-1838、CUR-1916)是否能夠調控SK-N-AS人類神經母細胞瘤細胞中之其他非相關基因(諸如肌動蛋白)之mRNA。材料及方法。 來自ATCC之SK-N-AS人類神經母細胞瘤細胞(目錄號CRL-2137)在與實例2中所述相同之條件中進行給藥。肌動蛋白mRNA係藉由如實例2中所述之即時PCR加以定量,例外之處為此次由ABI設計之引子/探針對肌動蛋白(目錄號Hs99999903_m1)具有特異性。資料呈現於圖15中。結果。 如圖15中所示,靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調非相關基因,諸如肌動蛋白。因此,此等寡核苷酸在上調SCN1A方面具有特異性。實例 14 :肌動蛋白在用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理之 SK-N-AS 細胞中未上調 此實驗之目的在於確定靶向SCN1A特異性天然反義轉錄物且能夠上調SCN1A蛋白之寡核苷酸(CUR-1740、CUR-1764、CUR-1770及CUR-1916)是否亦能夠調控SK-N-AS細胞中之非相關蛋白(諸如肌動蛋白)之表現。SK-N-AS為人類神經母細胞瘤細胞株。材料及方法: 使來自ATCC之SK-N-AS人類神經母細胞瘤細胞(目錄號CRL-2137)在與實例9中所述相同之條件中生長。細胞在與實例8中所述相同之條件中精確固定且染色,例外之處為第一抗體為稀釋500倍使用之兔抗肌動蛋白(Abcam目錄號ab1801)。細胞之染色係使用與實例9中所述相同之方法直接在24孔盤之各孔內部加以分析。結果: 如圖16中所示,所測試之反義寡核苷酸中無一使肌動蛋白上調。因此,此等寡核苷酸在上調SCN1A蛋白方面具有特異性。實例 15 :肌動蛋白在用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理之 Vero 76 細胞中未上調 此實驗之目的在於確定靶向SCN1A特異性天然反義轉錄物且能夠上調SCN1A蛋白之特異性反義寡核苷酸(CUR-1740、CUR-1770、CUR-1916、CUR-1924及CUR-1945)是否亦能夠調控Vero76細胞中之非相關基因之蛋白質(諸如肌動蛋白)表現。Vero76為草原猴(綠色猴或非洲綠猴)腎細胞株。材料及方法。 使來自ATCC之Vero76非洲綠猴胚腎細胞株(目錄號CRL-1587)在實例10中所述之相同條件中生長。細胞在與實例10中所述相同之條件中精確固定且染色,例外之處為第一抗體為稀釋500倍使用之兔抗肌動蛋白(Abcam目錄號ab1801)。細胞之染色係使用與實例10中所述相同之方法直接在24孔盤之各孔內部加以分析。結果。 如圖17中所示,所測試之反義寡核苷酸中無一使肌動蛋白上調。因此,此等寡核苷酸在上調SCN1A蛋白方面具有特異性。實例 16 :肌動蛋白在用靶向 SCNA 特異性天然反義轉錄物之反義寡核苷酸處理之帶有德拉威特症候群相關突變的初級人類纖維母細胞中未上調 此實驗之目的在於確定靶向SCN1A特異性天然反義轉錄物且能夠上調SCNA蛋白之寡核苷酸(CUR-1740、CUR-1764、CUR-1770、CUR-1838及CUR-1916)是否亦能夠調控帶有德拉威特症候群相關突變之初級人類纖維母細胞中的非相關基因之蛋白質(諸如肌動蛋白)表現。材料及方法。 使帶有由N.Kenyon博士(University of Miami)引入培養物中之德拉威特症候群相關突變的纖維母細胞在與實例8中所述相同之條件中生長。細胞在與實例8中所述相同之條件中精確固定且染色,例外之處為第一抗體為稀釋500倍使用之兔抗肌動蛋白(Abcam目錄號ab1801)。細胞之染色係使用與實例8中所述相同之方法直接在24孔盤之各孔內部加以分析。結果: 如圖18中所示,所測試之反義寡核苷酸中無一使肌動蛋白上調。因此,此等寡核苷酸在上調SCN1A蛋白方面具有特異性。實例 17 :使用 ELISA 對用靶向 SCNA 特異性天然反義轉錄物之寡核苷酸處理之帶有德拉威特症候群相關突變的初級人類纖維母細胞中之 SCNA 蛋白進行定量 此實驗之目的在於使用ELISA定量歸因於用靶向SCN1A特異性天然反義轉錄物之寡核苷酸(CUR-1740、CUR-1770及CUR-1916)處理引起之帶有德拉威特症候群相關突變之初級人類纖維母細胞中的SCN1A蛋白上調之程度。材料及方法: 使帶有由N.Kenyon博士(University of Miami)引入培養物中之德拉威特症候群相關突變的纖維母細胞在與實例8中所述相同之條件中生長,但僅0及80 nM濃度之寡核苷酸用於給藥。接著對細胞計數且再接種於96孔盤中。在24小時之後,細胞在與實例8及16中所述相同之條件中精確固定,例外之處為所有300 μl體積皆減少至100 μl。平行測定孔如實例8中所述用肌動蛋白及SCN1A抗體染色,例外之處為所有反應皆以100 μl體積進行。抗肌動蛋白抗體稀釋度為1:500,抗SCN1A稀釋度為1:250且抗小鼠稀釋度為1:250。此外,替代二胺基聯苯胺(DAB)過氧化酶受質溶液,使用四甲基聯苯胺(TMB)過氧化酶受質溶液(Thermo Scientific目錄號N301)。在上清液變藍之後,將其轉移至新96孔盤(Greiner bio one目錄號65121)中且添加1 M硫酸。使用Multiskan Spectrum分光光度計(Thermo Scientific)在450 nm下讀取吸光度。自所有SCN1A及肌動蛋白讀數中減去背景信號(在用作為一次抗體之抗小鼠染色之孔中讀取)。接著將各條件之SCN1A信號相對於肌動蛋白信號進行校正。結果: 圖19顯示所有測試之反義寡核苷酸(CUR-1740、CUR-1770及CUR-1916)皆高效上調SCN1A蛋白多達40%。實例 18 :使用 ELISA 對用靶向 SCNA 特異性天然反義轉錄物之寡核苷酸處理之 Vero76 細胞中的 SCNA 蛋白進行定量 此實驗之目的在於使用ELISA定量歸因於用靶向SCN1A特異性天然反義轉錄物之寡核苷酸(CUR-1740、CUR-1770、CUR-1916、CUR-1924、CUR-1945)處理引起之帶有德拉威特症候群相關突變之初級人類纖維母細胞中的SCN1A蛋白上調之程度。材料及方法: 使Vero76非洲綠猴胚腎細胞在實例10中所述之相同條件中生長,但僅0及80 nM濃度之寡核苷酸用於給藥。接著對細胞計數且再接種於96孔盤中。在24小時之後,細胞在與實例8中所述相同之條件中精確固定,例外之處為所有300 μl體積皆減少至100 μl。平行測定孔如實例10及15中所述用肌動蛋白及SCN1A抗體染色,例外之處為所有反應皆在100 μl下進行,抗肌動蛋白抗體稀釋度為1:500,抗SCN1A稀釋度為1:250且抗小鼠稀釋度為1:250。此外,替代使用二胺基聯苯胺(DAB)過氧化酶受質溶液,使用四甲基聯苯胺(TMB)過氧化酶受質溶液(Thermo Scientific目錄號N301)。在上清液變藍之後,將其轉移至新96孔盤(Greiner bio one目錄號651201)中且添加1 M硫酸。使用Multiskan Spectrum分光光度計(Thermo Scientific)在450 nm下讀取吸光度。自所有SCN1A及肌動蛋白讀數中減去背景信號(在用作為一次抗體之抗小鼠染色之孔中讀取)。接著各條件之SCN1A信號相對於肌動蛋白信號進行校正。結果: 圖20顯示所有測試之反義寡核苷酸(CUR-1740、CUR-1770、CUR-1916、CUR-1924、CUR-1945)皆高效上調SCN1A蛋白多達300%。實例 19 :使用 ELISA 對用靶向 SCNA 特異性天然反義轉錄物之寡核苷酸處理之 SK-N-AS 細胞中的 SCNA 蛋白進行定量 此實驗之目的在於定量歸因於用靶向SCN1A特異性天然反義轉錄物之寡核苷酸(CUR-1740、CUR-1770、CUR-1924及CUR-1945)處理引起之SK-N-AS細胞中的SCN1A蛋白上調之程度。材料及方法: 使來自ATCC之SK-N-AS人類神經母細胞瘤細胞(目錄號CRL-2137)在與實例10中所述相同之條件中生長,但僅0及20 nM濃度之寡核苷酸用於給藥。接著對細胞計數且再接種於96孔盤中。在24小時之後,細胞在與實例9中所述相同之條件中精確固定,例外之處為所有300 μl體積皆減少至100 μl。平行測定孔如實例9及13中所述用肌動蛋白及SCN1A抗體染色,例外之處為所有反應皆在100 μl下進行,抗肌動蛋白抗體稀釋度為1:500,抗SCN1A稀釋度為1:250且抗小鼠稀釋度為1:250。此外,替代二胺基聯苯胺(DAB)過氧化酶受質溶液,使用四甲基聯苯胺(TMB)過氧化酶受質溶液(Thermo Scientific目錄號N301)。在上清液變藍之後,將其轉移至新96孔盤(Greiner bio one目錄號651201)中且添加1 M硫酸。使用Multiskan Spectrum (Thermo Scientific)在450 nm下讀取吸光度。自所有SCN1A及肌動蛋白讀數中減去背景信號(在用作為一次抗體之抗小鼠染色之孔中讀取)。接著各條件之SCN1A信號相對於肌動蛋白信號進行校正。結果: 圖21顯示所有測試之反義寡核苷酸(CUR-1740、CUR-1770、CUR-1924及CUR-1945)皆高效上調SK-N-AS細胞中之SCN1A蛋白多達500%。實例 20 :偵測 HepG2 細胞及帶有德拉威特症候群相關突變之初級人類纖維母細胞中的天然反義 BG724147 此實驗之目的在於確定天然反義BG724147是否存在於人類肝細胞癌HepG2細胞株及帶有德拉威特症候群相關突變之初級人類纖維母細胞中。為了達成此目的,使用自各細胞類型分離得到之兩種不同RNA(poly A RNA及總RNA)。在使用兩種細胞類型進行連續兩輪PCR之後獲得PCR產物,將其在凝膠上加以分析。使用BG724147特異性引子獲得之尺寸類似之帶的擴增證實在兩種細胞類型中均存在BG724147。材料及方法 分離總 RNA 在75 cm2 培養瓶中生長之80%匯合之HepG2細胞或帶有德拉威特症候群相關突變的初級人類纖維母細胞用PBS AccuGENE 1×(Lonza Rockeland Inc., Rockeland, ME)洗滌2次。在丟棄PBS之後,添加5 ml 含有b-巰基乙醇(QIAGEN Inc.-USA, Valencia, CA)之RLT緩衝液至此等細胞中且細胞溶解產物以1 ml等分試樣在-80℃下儲存於微量離心管中直至分離總RNA。使用RNeasy中型套組(QIAGEN Inc.-USA, Valencia, CA)遵循製造商方案來自此等細胞分離總RNA。簡而言之,細胞溶解產物在3000×g下離心5分鐘以使溶解產物澄清且丟棄任何離心塊。澄清細胞溶解產物經由QIAshredder管柱(在2 ml微量離心管內部)在14800×g下離心且所得均質化溶解產物與等體積之70%乙醇混合。與乙醇混合之細胞溶解產物施加至RNeasy中型管柱(在15 ml錐形管內部)中且在3000×g下離心5分鐘。管柱用4 ml RW1緩衝液洗滌1次且接著經受15分鐘用140 μl含無RNase之DNase之RDD緩衝液進行的管柱上DNase消化。藉由添加4 ml RW1緩衝液來終止DNase消化且在3000×g下離心管柱。管柱用RPE緩衝液洗滌2次且與過濾器結合之總RNA用150 μl無DNase及RNAse之水溶離。總RNA儲存在-80℃下直至下一步驟。 HepG2 細胞之總 RNA 分離 poly-A RNA 使用來自Ambion之用磁珠分離poly-A之套組(Applied Biosystems/ Ambion, Austin, TX)遵循製造商方案自HepG2細胞及具有德拉威特症候群相關突變之初級人類纖維母細胞之總RNA分離poly A RNA。基本上,100 μg總RNA經再懸浮以使得於無DNase/RNAse之水中之最終濃度為600 μg/ml且添加等體積之2×結合溶液。在此期間,將10 μl Oligo (dT)磁珠置放於微量離心管中,藉由將此管置放在磁性台架(magnetic stand)上進行捕捉且丟棄儲存緩衝液。添加50 μl洗滌溶液1至珠粒中且自磁性台架移除管並丟棄洗滌溶液。此時,含來自HepG2細胞之總RNA之1×結合緩衝液與磁珠混合且在70℃下加熱5分鐘,接著在室溫下在溫和攪拌下培育60分鐘。與磁珠結合之poly A RNA藉由使用磁性台架捕捉5分鐘。丟棄上清液。Oligo(dT)磁珠用洗滌溶液1洗滌2次且用洗滌溶液2洗滌1次以移除非特異性結合之RNA。用磁性台架捕捉磁珠且添加200 μl溫熱RNA儲存溶液(在70℃下預加熱5分鐘)至珠粒中。磁珠由磁性台架捕捉,儲存上清液(poly A RNA之第一溶離液)。接著添加第二200 μl溫熱RNA儲存溶液(在70℃下預加熱5分鐘)至珠粒中。polyA RNA之第二溶離液添加至第一溶離液中。此時,在-20℃下使用5 M乙酸銨、肝醣及100%乙醇使溶離之RNA沈澱隔夜。聚RNA在4℃下在14800×g下離心30分鐘。丟棄上清液且用1 ml 70%乙醇洗滌RNA離心塊3次,每次藉由在4℃下離心10分鐘回收RNA離心塊。最後,poly A RNA離心塊再懸浮於加熱至70℃以較佳溶解RNA之RNA儲存溶液中。poly A RNA儲存在-80℃下。 添加腺苷至RNA轉錄物之3'端。來自HepG2細胞或帶有德拉威特症候群相關突變之初級人類纖維母細胞的總RNA(40 μg)與2單位之RNA Poly (A)聚合酶混合,最終反應體積100 μl(Ambion, Applied Biosystems, St. Austin TX)。聚腺苷酸化反應中使用之ATP來自Invitrogen。在聚腺苷酸化之後,使用苯酚/氯仿技術,隨後進行肝醣/乙酸鈉沈澱來純化RNA。此RNA再懸浮於40 μl無DNAse/RNAse之水中且用於3'RACE反應中(來自Ambion, Applied Biosystems, St. Austin TX之FirstChoice RLM-RACE套組)。 SCN1A之BG724147天然反義轉錄物之3'延伸。使用來自Ambion, Applied Biosystems(St. Austin, TX)之FirstChoice RLM-RACE套組進行兩組不同cDNA端之3'快速擴增(RACE)反應。一組使用poly A RNA且另一組使用添加有1個腺苷之總RNA,poly A RNA及總RNA來自HepG2細胞或帶有德拉威特症候群相關突變的初級人類纖維母細胞。進行連續兩輪PCR。第一PCR係使用套組中供應之3'外部引子及由OPKO CURNA設計之對BG724147具有特異性之5'引子(5' GATTCTCCTACA GCAATTGGTA 3')進行。第二輪PCR係使用套組中供應之3'外部引子及由OPKO CURNA設計之對BG724147具有特異性之不同5'引子(5' GACATGTAATCACTTTCATCAA 3')進行。第二PCR反應之產物在1%瓊脂糖-1×TAE凝膠上操作。結果 :圖22展示使用來自HepG2細胞之poly A RNA及添加有腺苷之總RNA及來自帶有德拉威特症候群相關突變之初級人類纖維母細胞的poly A RNA及添加有腺苷之總RNA所進行之3'RACE實驗的第二輪PCR反應之產物。在來自HepG2細胞及帶有德拉威特症候群相關突變之初級人類纖維母細胞的poly A RNA中觀測到1個相同亮帶。結論: 使用對SCN1A之BG724147天然反義轉錄物具有特異性之引子進行的PCR擴增在兩種不同細胞(HepG2細胞及帶有德拉威特症候群相關突變之初級人類纖維母細胞)中產生共同PCR亮帶。此外,靶向SCN1A天然反義BG724147之反義寡核苷酸已顯示會上調如實例2、7及16中所示之此等細胞中的SCN1A mRNA及蛋白質。此資料指示BG724147實際上存在於此兩種細胞(HepG2細胞及帶有德拉威特症候群相關突變之初級人類纖維母細胞)中。實例 21 SCNA 天然反義序列 BG724147 之延伸 此實驗之目的在於藉由對SCN1A天然反義BG724147之所有序列定序來延伸其已知序列。原始BG724147 RNA轉錄物係獲自由Miklos Palkovits取得之人類睪丸。由Michael J. Brownstein(在NHGRI)、Shiraki Toshiyuki及Piero Carninci(在RIKEN)製備於pBluescriptR載體中之cDNA文庫。cDNA文庫由I.M.A.G.E.協會(或LLNL)進行排列且純系由Incyte Genomics, Inc.在2001年5月進行定序。BG724147純系可在Open Biosystems(Open Biosystems Products, Huntsville, AL)獲得。在2001年,插入BG724147純系中之cDNA未完全定序。OPKO-CURNA獲得BG724147純系且對全部插入物定序。為達成此目的,自Open Biosystems獲得含有具有BG724147插入物之質體之細菌純系且接種於含有安比西林之盧里亞伯坦尼(Luria Bertani,LB)瓊脂盤中以分離個別群落。接著在5 ml LB培養液中擴增群落。接著自此等細菌分離含有BG724147插入物之質體並遞送至Davis Sequencing(Davis, CA)進行定序。 材料及方法: 分離並定序含有 SCNA 天然反義 BG724147 cDNA 之質體。 自Open Biosystems購買含有BG724147質體之冷凍細菌之懸浮液(Open Biosystems Products,目錄號4829512),稀釋10、100、1000、10000、100000倍,接著接種在含有100 μg/ml安比西林(Calbiochem,目錄號171254)之盧里亞伯坦尼(Luria Bertani;LB)(BD,目錄號244520)瓊脂盤(Falcon,目錄號351005)上。在15小時之後,自稀釋100000倍之培養盤分離20個個別細菌群落且各別地在5 ml LB培養液(Fisher Scientific,目錄號BP1426-2)中生長15小時-24小時。此時,細菌集結且使用Promega之PureYieldTM質體小規模純化系統套組(Promega, 目錄號A1222)遵循製造商方案來分離質體(含有BG724147 RNA轉錄物之cDNA)。分離之DNA稀釋至200 ng/ml且12 μl來自各群落之質體遞送至Davis sequencing(Davis, CA)進行定序。結果: 自Davis sequencing獲得之序列提供延伸之BG724147(SEQ ID NO: 12)。結論: 已知BG724147序列成功延伸403個核苷酸充當設計對抗SCN1A天然反義轉錄物BG724147之反義寡核苷酸的基礎。 儘管本發明已關於一或多個實施例加以說明及描述,但熟習此項技術者在閱讀及理解本說明書及附圖後將想到等效變化及修改。此外,儘管本發明之特定特徵可能已僅關於若干實施例之一加以揭示,但當可能為任何既定或特定應用所需及對任何既定或特定應用有利時,此特徵可與其他實施例之一或多個其他特徵組合。 本發明之摘要將允許讀者快速探明技術揭示內容之性質。應瞭解其不應用於解釋或限制以下申請專利範圍之範疇或意義。Several aspects of the invention are described below with reference to illustrative applications for illustration. It should be appreciated that numerous specific details, relationships, and methods are described in order to fully understand the invention. However, one of ordinary skill in the art will readily recognize that the invention can be practiced without one or more specific details. The present invention is not limited by the order of acts or events, as some acts may occur in a different order and/or in parallel with other acts or events. In addition, not all illustrated acts or events may be required to practice a method of the invention. All of the genes, gene names, and gene products disclosed herein are intended to correspond to homologs of any species from which the compositions and methods disclosed herein are applicable. Thus, terms include, but are not limited to, genes and gene products from humans and mice. It is to be understood that the disclosure of the present invention is intended to be illustrative only and not to be construed as limiting. Thus, for example, for the genes disclosed herein associated with mammalian nucleic acid and amino acid sequences in some embodiments, it is intended to encompass other animals (including but not limited to other mammals, fish, amphibians, Homologous and/or orthologous genes and gene products of reptiles and birds. In one embodiment, the gene or nucleic acid sequence is a human gene or nucleic acid sequence.definition The terminology used herein is for the purpose of describing particular embodiments and is not intended to limit the invention. As used herein, the singular forms "" In addition, the terms "including", "having" or variations thereof are used in the context of the [embodiments] and/or claims, and such terms are intended to be inclusive in a manner similar to the term "comprising." The term "about" means that within the acceptable error range of a particular value as determined by one of ordinary skill, the error range will depend in part on how the value is measured or measured, i.e., the limits of the measurement system. For example, "about" can mean within one or more standard deviations according to the practice in the art. Alternatively, "about" may mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and still more preferably up to 1% of the established value. Alternatively, particularly with respect to biological systems or methods, the term may mean within a certain order of magnitude of a value, preferably within 5 times, and more preferably within 2 times. When a particular value is recited in the present application and the scope of the claims, the term "about" is intended to be within the range of acceptable tolerances of the particular value. As used herein, the term "mRNA" means a currently known mRNA transcript of a targeted gene and any other transcript that can be elucidated. In the case of "antisense oligonucleotide" or "antisense compound", it means an RNA or DNA molecule that binds to another RNA or DNA (target RNA, DNA). For example, if it is an RNA oligonucleotide, it binds to another RNA target by means of an RNA-RNA interaction and alters the activity of the target RNA. Antisense oligonucleotides can upregulate or downregulate the performance and/or function of a particular polynucleotide. The definition is intended to include any foreign RNA or DNA molecule that appears to be therapeutic, diagnostic or otherwise. Such molecules include, for example, antisense RNA or DNA molecules, interfering RNA (RNAi), microRNAs, decoy RNA molecules, siRNA, enzymatic RNA, therapeutically editable RNA and agonist and antagonist RNA, antisense oligomeric compounds, Antisense oligonucleotides, external guide sequence (EGS) oligonucleotides, alternative splicers, primers, probes, and other oligomeric compounds that hybridize to at least a portion of the target nucleic acid. Thus, such compounds can be introduced as single, double, partially single, or cyclic oligomeric compounds. In the context of the present invention, the term "oligonucleotide" refers to an oligo or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or a mimetic thereof. The term "oligonucleotide" also includes natural and/or modified monomers or bonds (including deoxyribonucleosides, nucleosides, substituted and alpha-directed forms, peptide nucleic acids (PNA), locked nucleic acids ( Linear or cyclic oligomers of LNA), phosphorothioates, methylphosphonates and the like). Oligonucleotides can interact with monomers by regular patterning (such as Watson-Crick type base pairing, Hoögsteen or reverse Hugues) Base pairing of the D-type, or a similar interaction thereof) to specifically bind to the polynucleotide of interest. Oligonucleotides can be "chimeric", that is, composed of different regions. In the context of the present invention, a "chimeric" compound is an oligonucleotide comprising two or more chemical regions, such as a DNA region, an RNA region, a PNA region, and the like. Each chemical region consists of at least one monomeric unit, i.e., a nucleotide in the case of an oligonucleotide compound. Such oligonucleotides typically comprise at least one region in which the oligonucleotide has been modified to exhibit one or more desired properties. Desirable properties of the oligonucleotide include, but are not limited to, increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity to the target nucleic acid. Thus, different regions of the oligonucleotide can have different properties. The chimeric oligonucleotide of the present invention can be formed into a mixed structure of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide analogs as described above. . Oligonucleotides can be constructed by "registered" linkages (i.e., when monomers are contiguously joined as in native DNA) or by regions joined by spacers. The spacers are intended to form a covalent "bridge" between the regions and, preferably, have a length of no more than about 100 carbon atoms. Spacers can have different functionalities, such as having a positive or negative charge, with specific nucleic acid binding properties (intercalators, groove binders, toxins, fluorophores, etc.), lipophilic, induction specific Secondary structure, such as, for example, an alanine-containing peptide that induces an alpha-helix. As used herein, "SCN1A" includes all family members, mutants, dual genes, fragments, materials, coding and non-coding sequences, sense and antisense polynucleotide strands, and the like. Similarly, SCN2A-SCN12A includes all mutants, dual genes, fragments, and the like. As used herein, the phrase "voltage-gated sodium ion channel type I alpha subunit", SCN1A, FEB3, FEB3A, GEFSP2, HBSCI, NAC1, Nav1.1, SCN1, SMEI, sodium ion channel protein brain I subunit The sodium ion channel protein type 1 subunit α, the sodium ion channel protein type I subunit α, and the voltage-gated sodium ion channel subunit α Nav1.1 are considered identical in the literature and are used interchangeably in this application. As used herein, the term "oligonucleotide specific for" or "an oligonucleotide that targets" refers to having (i) capable of forming a stable complex with a portion of a targeted gene, or (ii) capable of An oligonucleotide that forms a stable duplex sequence with a portion of the mRNA transcript of the targeted gene. The stability of the complex and the double helix can be determined by theoretical calculations and/or in vitro assays. Exemplary assays for determining the stability of hybridization complexes and duplexes are described in the Examples below. As used herein, the term "target nucleic acid" encompasses DNA, RNA transcribed from such DNA (including pre-mRNA (premRNA) and mRNA), and cDNA derived from the RNA, coding sequence, non-coding sequence, sense or antisense polymerization. Nucleotide. The specific hybridization of an oligomeric compound to its target nucleic acid interferes with the normal function of the nucleic acid. This modulation of the function of the target nucleic acid by a compound that specifically hybridizes to the target nucleic acid is commonly referred to as "antisense." DNA functions that will be disturbed include, for example, replication and transcription. RNA functions that will be interfered with include all life functions, such as translocation of RNA to protein translation sites, translation of proteins from RNA, splicing of RNA to produce one or more mRNA species, and catalytic activity in which RNA can participate or promote. The overall effect of this interference with the function of the target nucleic acid is to modulate the performance of the encoded product or oligonucleotide. RNA interference "RNAi" is mediated by a double-stranded RNA (dsRNA) molecule with sequence-specific homology to a "target" nucleic acid sequence. In certain embodiments of the invention, the mediator is a "small interference" RNA duplex (siRNA) containing 5-25 nucleotides. siRNA is produced by processing dsRNA with an RNase called Dicer. The siRNA duplex product is recruited into a polyprotein siRNA complex called RISC (RNA-induced resting complex). Without wishing to be bound by any particular theory, the RISC is then directed to a target nucleic acid (suitable for mRNA), where the siRNA duplexes interact in a sequence-specific manner to catalyze cleavage in a catalytic manner. Small interfering RNAs that can be used in accordance with the present invention can be synthesized and used according to procedures well known in the art and which will be familiar to those of ordinary skill. The small interfering RNA used in the methods of the invention suitably comprises between about 1 and about 50 nucleotides (nt). In an example of a non-limiting embodiment, the siRNA can comprise from about 5 to about 40 nt, from about 5 to about 30 nt, from about 10 to about 30 nt, from about 15 to about 25 nt, or from about 20-25 Nucleotides. Selection of appropriate oligonucleotides is facilitated by the use of computer programs that automatically align nucleic acid sequences and indicate regions of homology or homology. Such programs are used to compare the resulting nucleic acid sequences, for example, by searching a library such as GenBank or by sequencing the PCR products. Comparison of nucleic acid sequences from a range of species allows selection of nucleic acid sequences that exhibit the appropriate degree of identity between species. Southern blots are performed in the absence of sequenced genes to allow for the determination of the degree of identity between genes in the target species and genes in other species. As is well known in the art, it is possible to obtain an approximate measure of consistency by performing Southern blot analysis at varying degrees of stringency. Such procedures allow for the selection of nucleic acid sequences that have a high degree of complementarity with a target nucleic acid sequence in an individual to be controlled and that have a lower degree of complementarity with corresponding nucleic acid sequences in other species. Those skilled in the art will recognize that there is a great deal of freedom in selecting the appropriate region for the genes used in the present invention. In the case of "enzymatic RNA", it means an enzymatically active RNA molecule (Cech, (1988)J. American. Med. Assoc 260, 3030-3035). An enzymatic nucleic acid (ribonuclease) functions by binding to a target RNA. This binding occurs via a target binding moiety of the enzymatic nucleic acid that remains in close proximity to the enzymatic portion of the molecule that functions to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes the target RNA and then binds to the target RNA via base pairing, and once bound to the correct site, acts enzymatically to cleave the target RNA. By "bait RNA" it is meant an RNA molecule that mimics the natural binding domain of a ligand. Thus, the decoy RNA competes with the natural binding target for binding to a particular ligand. For example, an over-expressed HIV trans-activation response (TAR) RNA has been shown to act as a "bait" and efficiently binds to the HIV tat protein, thereby preventing its binding to the TAR sequence encoded in HIV RNA. This is intended to be a specific example. Those skilled in the art will recognize that this is merely an example, and that other embodiments are readily produced using techniques generally known in the art. As used herein, the term "monomer" generally indicates that it is linked via a phosphodiester bond or an analog thereof to form a size in a number of monomer units (eg, from about 3-4 monomer units) to about several hundred monomer units. Monomers of oligonucleotides within the range. Analogs of phosphodiester bonds include: phosphorothioates, dithiophosphates, methylphosphonates, selenophosphates, aminophosphates, and the like, as described more fully below. The term "nucleotide" encompasses naturally occurring nucleotides as well as non-naturally occurring nucleotides. Those skilled in the art should be aware that the various nucleotides previously considered "non-naturally occurring" have subsequently been discovered in nature. Thus, "nucleotide" includes not only molecules known to contain purine and pyrimidine heterocycles, but also heterocyclic analogs and tautomers thereof. Illustrative examples of other types of nucleotides are molecules containing adenine, guanine, thymine, cytosine, uracil, guanidine, xanthine, diamine guanidine, 8-sided oxy-N6-A Adenine, 7-deazapurine, 7-deazaguanine, N4, N4-ethanocytosin, N6, N6-bridged ethylene-2,6-di Aminoguanidine, 5-methylcytosine, 5-(C3-C6)-alkynylcytosine, 5-fluorouracil, 5-bromouracil, pseudoisomeromide, 2-hydroxy-5-methyl-4- Triazolopyridine, isocytosine, isoguanine, inosine, and "non-naturally occurring" nucleotides described in U.S. Patent No. 5,432,272, to Benner et al. The term "nucleotide" is intended to cover every and every such instance as well as analogs and tautomers thereof. Particularly interesting nucleotides are nucleotides containing adenine, guanine, thymine, cytosine, and uracil, which are considered to be naturally occurring nucleotides associated with therapeutic and diagnostic applications in humans. Nucleotides include, for example, natural 2'-deoxy and 2'-hydroxy sugars as described in Kornberg and Baker, DNA Replication, 2nd Edition (Freeman, San Francisco, 1992) and analogs thereof. "Analogs" with respect to nucleotides include portions having modified bases and/or modified sugar moieties (see, for example, commonly, Scheit, Nucleotide Analogs, John Wiley, New York, 1980; Freier and Altmann, (1997)).Nucl. Acid. Res ., 25(22), 4429-4443; Toulmé, J.J., (2001)Nature Biotechnology 19:17-18; Manoharan M., (1999)Biochemica et Biophysica Acta 1489: 117-139; Freier S. M., (1997)Nucleic Acid Research , 25:4429-4443; Uhlman, E., (2000)Drug Discovery & Development , 3: 203-213; Herdewin P., (2000)Antisense & Nucleic Acid Drug Dev. , 10:297-310) synthetic nucleotide; 2'-O, 3'-C linked [3.2.0] bicyclic glucosinolate. Such analogs include synthetic nucleotides designed to enhance binding properties, such as duplex or triple helix stability, specificity, or the like. As used herein, "hybridization" means the pairing of substantially complementary strands of an oligomeric compound. A pairing mechanism involves hydrogen bonding between complementary nucleosides or nucleotide bases (nucleotides) of a strand of an oligomeric compound, which may be a Watson-Crick, Huguestin, or Reversed Hu Gastin hydrogen bonding. For example, adenine and thymine are complementary nucleotides paired by the formation of hydrogen bonds. Hybridization can occur in different situations. When the binding of the antisense compound to the target nucleic acid interferes with the normal function of the target nucleic acid, resulting in modulation of function and/or activity, and there is a sufficient degree of complementarity to avoid specific binding of the antisense compound to the non-target nucleic acid sequence. Antisense compound under conditions (ie, under in vivo conditions or in the case of therapeutic treatment, under physiological conditions, and in the case of in vitro assays, under conditions of assay) It is "specifically hybridizable". As used herein, the phrase "stringent hybridization conditions" or "stringent conditions" refers to conditions under which a compound of the invention will hybridize to its target sequence but to a minimum number of other sequences. Stringent conditions are sequence dependent and will vary from case to case, and in the context of the present invention, the "stringent conditions" at which an oligomeric compound hybridizes to a target sequence are determined by the nature and composition of the oligomeric compound and its investigation. Decide. In general, stringent hybridization conditions include low concentrations (<0.15 M) of salts with inorganic cations (such as Na++ or K++) (ie, low ionic strength), below the Tm of the oligomeric compound: target sequence complex. The presence of a denaturant such as formamide, dimethylformamide, dimethylhydrazine or detergent sodium dodecyl sulfate (SDS) at a temperature of °C-25 °C. For example, for every 1% metformin, the hybridization rate is reduced by 1.1%. An example of a high stringency hybridization condition is 0.1 x sodium chloride-sodium citrate buffer (SSC) / 0.1% (w/v) SDS at 60 ° C for 30 minutes. As used herein, "complementary" refers to the ability to precisely pair between two nucleotides on one or two oligomeric strands. For example, if the nucleobase at a position of the antisense compound is capable of interacting with a nucleobase at a position of the target nucleic acid (the target nucleic acid is a DNA, RNA or oligonucleotide molecule) Hydrogen bonding, the position of the hydrogen bond between the oligonucleotide and the target nucleic acid is considered to be a complementary position. When a sufficient number of complementary positions in each molecule are occupied by nucleotides that are hydrogen-bondable to each other, the oligomeric compound and other DNA, RNA or oligonucleotide molecules are complementary to each other. Thus, "specifically hybridizable" and "complementary" are terms used to indicate a sufficient degree of precise matching or complementarity of a sufficient number of nucleotides for stable and specific binding between an oligomeric compound and a target nucleic acid. It is understood in the art that a sequence that achieves specific hybridization without the need for an oligomeric compound is 100% complementary to its target nucleic acid. Furthermore, one or more segments of the oligonucleotide can hybridize such that the intervening or adjacent segments are not involved in a hybridization event (eg, a loop structure, a mismatch, or a hairpin structure). The oligomeric compound of the invention comprises at least about 70%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least of the target region within the target nucleic acid sequence to which it is targeted. About 95%, or at least about 99%, of sequence complementarity. For example, 18 of the 20 nucleotides of the antisense compound are complementary to the region of interest and thus the antisense compound that specifically hybridizes will represent 90% complementarity. In this example, the remaining non-complementary nucleotides may be clustered or interspersed with complementary nucleotides and need not be contiguous with each other or contiguous with complementary nucleotides. Thus, an antisense compound of 18 nucleotides in length and having 4 (four) non-complementary nucleotides flanked by two regions that are fully complementary to the target nucleic acid will have 77.8% overall complementarity with the target nucleic acid and thus It will fall within the scope of the invention. The percent complementarity of the antisense compound having the region of the target nucleic acid can be determined in a conventional manner using the BLAST program (basic local alignment search tool) and the PowerBLAST program known in the art. Percent homology, sequence identity or complementarity can be achieved, for example, by using the Smith and Waterman algorithms (Adv. Appl. Math . (1981) 2, 482-489) Gap program (Wisconsin Sequence Analysis Package, 8th Edition for Unix, Genetics Computer Group, University Research Park, Madison Wis.), using presets Settings are determined. As used herein, the term "thermal melting point (Tm)" refers to the temperature at which 50% of the oligonucleotide complementary to the target sequence hybridizes to the target sequence in equilibrium at a defined ionic strength, pH, and nucleic acid concentration. Generally, stringent conditions will be those having a salt concentration of at least about 0.01 to 1.0 M Na ion concentration (or other salt) at pH 7.0 to 8.3 and for short oligonucleotides (eg, 10 to 50 nucleotides) The temperature is at least about 30 °C. Stringent conditions can also be achieved with the addition of destabilizing agents such as formamide. As used herein, "modulate" means increased (stimulated) or reduced (inhibited) gene expression. When used in the context of a polynucleotide sequence, the term "variant" can encompass a polynucleotide sequence associated with a wild-type gene. This definition may also include, for example, "dual genes", "splicing", "species" or "polymorphic" variants. Splice variants may have significant identity to a reference molecule, but will typically have a larger or smaller number of polynucleotides due to alternative splicing of exons during mRNA processing. The corresponding polypeptide may have other functional domains or no functional domains. A species variant is a sequence of nucleotides that varies from one species to another. In the present invention, variants of the wild type gene product have a specific utility. A variant may be produced by at least one mutation in a nucleic acid sequence and may produce a polypeptide whose altered mRNA or structure or function may or may not be altered. Any given natural or recombinant gene may not have a dual gene form or have one or many dual gene forms. Common mutational changes that produce variants are often attributed to natural deletions, additions or substitutions of nucleotides. Each such type of change can occur one or more times in a given sequence, either alone or in combination with other variations. The resulting polypeptides will typically have significant amino acid identity relative to each other. A polymorphic variant is a change in the nucleotide sequence of a particular gene between individuals of a given species. Polymorphic variants may also encompass single-base mutations in which a single nucleotide polymorphism (SNP) or a polynucleotide sequence has one base change. The presence of a SNP may indicate, for example, that a population has a predisposition to disease status, i.e., sensitivity to resistance. Derivatized polynucleotides include nucleic acids that undergo chemical modification (e.g., replacement of hydrogen with an alkyl, sulfhydryl or amine group). For example, derivatives of the derivatized oligonucleotide may comprise non-naturally occurring moieties such as altered sugar moieties or intersaccharide linkages. Illustrative of these are phosphorothioates and other sulfur-containing materials known in the art. Derivatized nucleic acids can also contain labels, including radionucleotides, enzymes, fluorescers, chemiluminescent agents, chromogenic agents, receptors, cofactors, inhibitors, magnetic particles, and the like. A "derived" polypeptide or peptide is, for example, a polypeptide modified by glycosylation, pegylation, phosphorylation, sulfation, reduction/alkylation, deuteration, chemical coupling, or moderate formalin treatment or Peptide. Derivatives may also be modified, directly or indirectly, to contain detectable labels including, but not limited to, radioisotopes, fluorescent labels, and enzyme labels. As used herein, the term "animal" or "patient" is intended to include, for example, humans, sheep, donkeys, deer, mule deer, baboons, mammals, monkeys, horses, cows, pigs, goats, dogs, cats, Rats, mice, birds, chickens, reptiles, fish, insects and arachnids. "Mammal" encompasses warm-blooded mammals (such as humans and domesticated animals) that are usually under medical care. Examples include felines, canines, equines, bovines, and humans, as well as humans only. "Treatment" encompasses the treatment of a disease state in a mammal and includes: (a) preventing the disease state from occurring in a mammal, in particular, when the mammal is susceptible to the disease state but has not yet been diagnosed as having it; b) inhibiting the disease state, for example, arresting its development; and/or (c) mitigating the disease state, for example, causing the disease state to subside until the desired end point is reached. Treatment also includes amelioration of the symptoms of the disease (eg, alleviating pain or discomfort), where the improvement may or may not directly affect the disease (eg, etiology, transmission, performance, etc.). As used herein, "neurological disease or condition" refers to any disease or condition of the nervous system and/or the visual system. "Nervous diseases or conditions" include diseases involving the central nervous system (brain, brainstem, and cerebellum), the peripheral nervous system (including the cranial nerves), and the autonomic nervous system (which is partially located in both the central nervous system and the peripheral nervous system) or Illness. Neurological diseases or conditions include, but are not limited to, acquired epileptiform aphasia; acute disseminated encephalomyelitis; adrenoleukodystrophy; age-related macular degeneration ( Age-related macular degeneration); adenesis of the corpus callosum; agnosia; Aicardi syndrome; Alexander disease; Alpers' disease Alternating hemiplegia; Alzheimer's disease; Vascular dementia; amyotrophic lateral sclerosis; anencephaly; Angieman syndrome (Angelman syndrome); angiomatosis; anoxia; aphasia; apraxia; arachnoid cysts; arachnoiditis; -Anronl-Chiari malformation; arteriovenous malformation; Asperger syndrome; ataxia telegiectasia; attention deficit hyperactivity disorder; autism; autonomic dysfunction; back pain; Batten disease; Behcet's disease; Bell's palsy; benign essential blepharospasm; benign focal; amyotrophy; Benign intracranial hypertension; Binswanger's disease; blepharospasm; Bloch Sulzberger syndrome; brachial plexus injury; brain abscess Brain damage; brain tumors (including glioblastoma multiforme); spinal tumors; Brown-Sequard syndrome; Canavan disease; carpal tunnel syndrome (carpal tunnel syndrome); causalgia (causalgia); central pain syndrome; central pontine myelinolysis; Cerebral aneurysm; cerebral arteriosclerosis; cerebral atrophy; cerebral gigantism; cerebral palsy; Charcoal-Mari-Tus Charcot-Marie-Tooth disease; chemotherapy-induced neuropathy and neuralgia; Chiari malformation; chorea; chronic inflammatory demyelinating polyneuropathy; chronic pain Chronic regional pain syndrome; Coffin Lowry syndrome; coma (coma), including persistent growth; congenital facial diplegia; corticobasal degeneration; Cranial arteritis; craniosynostosis; Creutzfeldt-Jakob disease; cumulative injury disorder; Cushing's syndrome; cytomegalic inclusion body disease ; cell giant virus infection; dance eye - dance eyes syndrome (dancing eyes- Dancing feet syndrome);DandyWalker syndrome; Dawson disease; De Morsier's syndrome; Dejerine-Klumke palsy; Dementia; dermatomyositis; diabetic neuropathy; diffuse sclerosis; Dravet's dysautonomia; dysgraphia; dyslexia; Dystonias; early infantile epileptic encephalopathy; empty sella syndrome; encephalitis; encephaloceles; encephalotrigeminal angiomatosis; Epilepsy; Erb's palsy; essential tremor; Fabry's disease; Fahr's syndrome; fainting; familial spasticity Familial spastic paralysis; febrile seizures; Fisher syndrome; Friedrich's syndrome (Friedreich's ataxia); fronto-temporal dementia and other "tauopathies"; Gaucher's disease; Gerstmann's syndrome; giant cells Giant cell arteritis; giant cell inclusion disease; globular cell leukodystrophy; Guillain-Barre syndrome; HTLV-1 Related skeletal disease (HTLV-1-associated myelopathy); Hallervorden-Spatz disease; head injury; headache; hemifacial spasm; hereditary spastic paraplegia (hereditary Spastic paraplegia; heredopathia atactic a polyneuritiformis; herpes zoster oticus; herpes zoster; Hirayama syndrome; HIV-related dementia And neuropathy (also a neurological manifestation of AIDS); holoprosencephaly; Huntington's disease and other polyglutamic acid Repeated disease; hydranencephaly in the brain; hydrocephalus; hypercortisolism; hypoxia; immune-mediated encephalomyelitis; inclusion body myositis; incontinentia pigmenti Infantile phytanic acid storage disease; infantile refsum disease; infantile spasms; inflammatory myopathy; intracranial cyst; intracranial hypertension; Joubert syndrome; Keams-Sayre syndrome; Kennedy disease Kinsboume syndrome; Klippel Feil syndrome; Krabbe disease; Kugelberg-Welander disease; Kuru; Lafora disease; Lambert-Eaton myasthenia gravis syndrome (Lambert) -Eaton myasthenic syndrome); Landau-Kleffner syndrome; lateral medullary (Wallenberg) syndrome; learning difficulties (learnin g disabilities); Leigh's disease; Lennox-Gustaut syndrome; Lesch-Nyhan syndrome; leukodystrophy; Lewy body dementia; Lissencephaly; locked-in syndrome; Lou Gehrig's disease (also known as motor neuron disease or muscular atrophy) Lateral cord sclerosis; lumbar disc disease; lyme disease - neurological sequelae; Machado-Joseph disease; macrocephalon malformation; megalencephaly; Melkersson-Rosenthal syndrome; Menieres disease; meningitis; Menkes disease; metachromatic leukodystrophy; small Head deformity (microcephaly); migraine; Miller Fisher syndrome; mini-strokes; mitochondrial myopathies; Mobius syndrome; Muscular atrophy Motor neuron disease; Moyamoya disease; mucopolysaccharidoses; milti-infarct dementia; multifocal motor neuropathy; multiple sclerosis and others Demyelinating disorder; multiple system atrophy with postural hypotension; muscular dystrophy; myasthenia gravis; myelinoclastic diffuse sclerosis; infant myoblast Myoclonic encephalopathy of infants; myoclonus; myopathy; myotonia congenital; narcolepsy; neurofibromatosis; psychiatric inhibitor malignant syndrome ( Neuroleptic malignant syndrome); neurological manifestations of AIDS; neurological sequelae of lupus; neuromytonia; neuronal ceroid lipofuscinosis; neuronal migration Disorder); Niemann-Pick disease; Osali - O'Sullivan-McLeod syndrome; occipital neuralgia; occult spinal dysraphism sequence; Ohtahara syndrome; olive pons cerebellum Atrophy (olivopontocerebellar atrophy); strabismus myoclonus; optic neuritis; orthostatic hypotension; overuse syndrome; paresthesia; neurodegenerative diseases or Conditions (Parkinson's disease, Huntington's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), dementia, multiple sclerosis and associated with neuronal cell death Other diseases and conditions); paramyotonia congenital; paraneoplastic diseases; paroxysmal seizures; Parry Romberg syndrome; chronic childhood sclerosis (Pelizaeus- Merzbacher disease); periodic paralyses; peripheral neuropathy Hy); painful neuropathy and neuralgia; persistent growth state; pervasive developmental disorders; photic sneeze reflex; phytanic acid storage disease; Pick's disease; Nerve contusion; pituitary tumor; polymyositis; pore penetrating (porencephaly); post-polio syndrome; postherpetic neuralgia; postinfectious encephalomyelitis; Orthostatic hypotension; Prader-Willi syndrome; primary lateral sclerosis; prion diseases; progressive hemifacial atrophy; Progressive multifocalleukoencephalopathy; progressive sclerosing poliodystrophy; progressive supranuclear palsy; pseudotumor cerebri; Ramsey-Heng Special syndrome (type I and type 11); Rasmussen encephalitis (Rasmussen 's encephalitis); reflex sympathetic dystrophy syndrome; Lefsum disease; repetitive motion disorders; repetitive stress impairment; restless legs syndrome; retrovirus Related myelopathy; Rett syndrome; Reye's syndrome; Saint Vitus dance; Sandhoff disease; Schilder's disease ); schizencephaly; septo-optic dysplasia; shaken baby syndrome; shingles; shy-Drager syndrome; Sjogren's syndrome; sleep apnea; Soto's syndrome; spasticity; spina bifida; spinal cord injury; spinal cord tumor; spinal muscular atrophy; -Person syndrome); stroke; Sturge-Weber syndrome; subacute sclerosing panenceph Alitis); subcortical arteriosclerotic encephalopathy; Sydenham chorea; syncope; syringomyelia; tardive dyskinesia; Tay-Sachs disease; temporal arteritis; tethered spinal cord syndrome; Thomsen disease; thoracic outlet syndrome; trigeminal neuralgia (Tic Douloureux); Todd's paralysis; Tourette syndrome; transient ischemic attack; transmissible spongiform encephalopathies; transverse myelitis; traumatic Brain injury; tremor; trigeminal neuralgia; tropical spastic paraparesis; tuberous sclerosis; vascular dementia (multiple infarct dementia); vasculitis, including sputum Arteritis; Von Hippel-Lindau disease; Wallenberg's syndrome (Wallenberg) 's syndrome); Werdnig-Hoffman disease; West syndrome; whiplash; Williams syndrome; Wilton's disease Disease); and Zellweger syndrome and other neurological disorders described herein. Cardiovascular diseases or conditions include those that result in ischemia or reperfusion from the heart. Examples include, but are not limited to, atherosclerosis, coronary artery disease, granulomatous myocarditis, chronic myocarditis (non-granulomatous), primary hypertrophic cardiomyopathy, peripheral arterial disease (PAD), peripheral vascular disease, venous thromboembolism, pulmonary embolism, stroke, angina pectoris, myocardial infarction, cardiovascular tissue damage caused by cardiac arrest, bypass by the heart ( Cardiac bypass caused by cardiovascular bypass, cardiogenic shock, and dysfunction or tissue damage known to the general practitioner or involving cardiac or vascular structures, particularly but not limited to tissue damage associated with SCNA activation Related conditions. CVS diseases include, but are not limited to, atherosclerosis, granulomatous myocarditis, myocardial infarction, myocardial fibrosis secondary to valvular heart disease, myocardial fibrosis without infarction, primary hypertrophic cardiomyopathy, and Chronic myocarditis (non-granulomatous). Examples of diseases or conditions associated with sodium ion channel dysfunction include, but are not limited to, malignant hyperthermia, myasthenia gravis, intermittent ataxia, neuropathic and inflammatory pain, Alzheimer's disease, Parkinson's disease , schizophrenia, hyperekplexia, myotonia (such as hypokalemia and hyperkalemia periodic paralysis, paramyotonia congenita and potassium-induced muscle rigidity) and arrhythmia (cardiac arrhythmias) (such as long QT syndrome).Polynucleotide and oligonucleotide compositions and molecules aims: In one embodiment, the target comprises a nucleic acid sequence of a voltage-gated sodium ion channel alpha subunit (SCNA), including, without limitation, a sense and/or antisense non-coding and/or coding sequence associated with SCNA. Voltage-sensitive ion channels are a class of transmembrane proteins that provide the basis for cell excitability and the ability to transmit information via membrane potential generated by ions. In response to changes in membrane potential, these molecules mediate the rapid flow of ions across selective channels in the cell membrane. If the channel density is sufficiently high, a regenerative depolarization called an action potential is produced. In most electrically excitable cells, including neurons, heart cells, and muscles, voltage-gated sodium ion channels are responsible for generating and transmitting action potentials. Electroactivity is triggered by depolarization of the membrane, which opens a channel that is highly selective to sodium ions across the membrane. The ions are then driven through the open channel within the cell by an electrochemical gradient. Although the action potentials based on sodium ions in different tissues are similar, electrophysiological studies have demonstrated the existence of a variety of sodium ion channels with different structures and functions, and have cloned numerous genes encoding sodium ion channels. The SCNA gene belongs to the gene family of voltage-gated sodium ion channels. The voltage-gated sodium ion channel can be named according to the standardized nomenclature outlined in Goldin et al. (2000) Neuron 28:365-368. According to the system, voltage-gated sodium ion channels are grouped into one family, and nine mammalian isoforms from the family have been identified and expressed. These 9 isoforms are given the names Navl.l to Navl.9. In addition, the splice variants of various isoforms are distinguished by using lowercase letters (eg, "Navl.la") after the numbers. Voltage-gated sodium ion channels play an important role in the generation of action potentials in nerve cells and muscle. The alpha subunit (SCNA) is the major component of the channel and will be sufficient to produce efficient channels when expressed in vitro in cells. In turn, the alpha-1 and second order units require alpha subunits to create an effective channel. The role of these secondary units will be to improve the dynamic properties of the channels primarily by rapidly inactivating the sodium ion stream. Compared to normal SCNB1, mutations found in the SCN1B gene in GEFS syndrome were shown to reduce rapid inactivation of sodium ion channels when co-expressed with alpha subunits. In one embodiment, an antisense oligonucleotide is used to prevent or treat a disease or condition associated with a member of the SCNA family. Exemplary voltage-gated sodium channel alpha subunit (SCNA) mediated diseases and conditions that can be treated with cells/tissues of stem cell regeneration obtained using antisense compounds include: diseases associated with abnormal function and/or performance of SCNA Or a condition, a neurological disease or condition, convulsions, pain (including chronic pain), impaired electrical excitability involving sodium ion channel dysfunction, a disease or condition associated with sodium ion channel dysfunction, and voltage-gated sodium ion channel alpha Diseases or conditions associated with mis-regulation of subunit activity (eg paralysis, hyperkalemia, periodic paralysis, congenital myocardium, potassium tonic muscle rigidity, long QT syndrome 3, motor endplate disease) , ataxia, etc.), gastrointestinal diseases (such as colitis, ileitis, inflammatory bowel syndrome, etc.), cardiovascular diseases or conditions (such as hypertension, congestive heart failure, etc.) attributed to dysfunction of the enteric nervous system a disease or condition involving the genitourinary and parasympathetic nerve distribution of the genitourinary tract (eg, benign prostatic hyperplasia, impotence); and the neuromuscular system a disease or condition (eg, muscular dystrophy, multiple sclerosis, epilepsy, autism, migraine (eg, sporadic and familial hemiplegic migraine), severe episodic epilepsy in infants (SMEI or German) Laweit's syndrome), systemic epilepsy (GEFS+) with thermal seizures, and SCNA-related seizures. The invention further relates to a pharmaceutical composition comprising at least one oligonucleotide that targets at least one or more natural antisense transcripts selected from the group consisting of: SCN1A to SCN12A genes or Its mRNA or isoform or variant. The invention further relates to a method of treating a neurological disease or disorder comprising administering an oligonucleotide that targets at least one or more natural antisense transcripts selected from the group consisting of: mRNA SCN1A, SCN2A, SCN3A, SCN4A, SCN5A, SCN6A, SCN7A, SCN8A, SCN9A, SCN10A, SCN11A and SCN12A or variants thereof. In a preferred embodiment, the oligomer is selected to upregulate the performance of the fully functional performance product of the SCNA family. In a preferred embodiment, the oligomers of the invention upregulate transcription and/or translation of any of the mRNAs of the SCNXA gene family to provide a fully functional sodium ion channel in a patient in need of treatment. In a patient having a disease or condition associated with a mutated form of a voltage-gated sodium ion channel, in a preferred embodiment, a natural one comprising a targeted voltage-gated sodium ion channel alpha gene or mRNA of the gene is used. Administration or treatment of a pharmaceutical composition of an oligonucleotide of an antisense transcript will upregulate a fully functional performance product at a greater rate than the upregulation of the performance product produced by the mutated form of the gene. In another embodiment, the invention relates to an oligonucleotide combination that targets at least one natural antisense transcript of at least two members of the SCNXA family, wherein the X is selected from 1-12. For example, in the treatment of Dravid's syndrome, a combination of oligonucleotides can be used to upregulate performance products such as SCN1A and SCN9A. In another embodiment, at least one oligonucleotide can be selected to target a natural antisense transcript of at least two genes selected from any one of SCN1A to SCN12A. Preferred oligonucleotides of the invention are between about 5 and about 30 nucleotides in length and at least 50% complementary to a segment of 5 to about 30 nucleotides of NAT. A preferred NAT for any of the SCNA gene or its transcript is a NAT that interferes with and modulates the expression of mRNA and/or translation products of the mRNA when targeted by the oligonucleotides of the invention. In a preferred embodiment, the oligonucleotide upregulates the performance of the target's functional protein to treat or ameliorate SCNA-related diseases. In a preferred embodiment, this "upregulation" is not associated with the etiology or promotion of a disease, such as cancer. Alterations in the SCNA gene may include or encompass many or all forms of genetic mutations in the coding and/or non-coding regions of the gene, including insertions, deletions, rearrangements, and/or point mutations. A deletion can be a deletion of a whole gene or part of a gene. Point mutations can result in amino acid substitutions, frame shifts or stop codons. Point mutations can also occur in regulatory regions of the SCNA gene, such as in promoters, resulting in loss or reduction in the performance of mRNA, or can result in improper processing of this mRNA, resulting in reduced stability or reduced translation efficiency. Such changes in humans can lead to various forms of disease and there are numerous publications describing the association of changes in the SCNA gene with, for example, epilepsy or SMEI. Such changes may be "de novo" or heritable. The invention is not limited to the treatment of diseases associated with alterations in the SCNA gene and also includes treating SCNA-associated diseases or conditions in which the patient does not have or does not have an alteration or mutation in the SCNA gene. Any modulation or up-regulation of the performance of a functional voltage-gated sodium ion channel will result in a reduction or treatment of the relevant SCNA disease or condition in the patient in need of treatment. This alleviation may also include at least one measurable indicator of clinical improvement, including fewer episodes, less frequent episodes, less severe episodes, less seizure types, neurodevelopmental improvements, or any other therapeutic benefit. In one embodiment, administration of one or more antisense oligonucleotides to a patient in need thereof modulates SCNA to prevent or treat any disease associated with abnormal SCNA performance, function, activity compared to a normal control or Illness. In one embodiment, the oligonucleotide is specific for a polynucleotide comprising, without limitation, a non-coding region of SCNA. SCNA targets include variants of SCNA; mutants of SCNA, including SNPs; non-coding sequences of SCNA; dual genes, fragments and analogs thereof. Preferably, the oligonucleotide is an antisense RNA molecule. According to an embodiment of the invention, the target nucleic acid molecule is not limited to the SCNA polynucleotide but extends to any of the isoforms, receptors, homologs, non-coding regions and analogs of the SCNA. In one embodiment, the oligonucleotide targets a natural antisense sequence of the SCNA target (natural antisense of the coding and non-coding regions), including, without limitation, variants thereof, dual genes, homologs, mutants, Derivatives, fragments and complementary sequences. Preferably, the oligonucleotide is an antisense RNA or DNA molecule. In one embodiment, the oligomeric compounds of the invention also include variants in which different bases are present at one or more nucleotide positions in the compound. For example, if the initial nucleotide is adenine, a variant containing thymidine, guanosine, cytidine or other natural or non-natural nucleotides at this position can be produced. This can be done at any position of the antisense compound. These compounds are then tested using the methods described herein to determine their ability to inhibit the performance of the target nucleic acid. In some embodiments, the homology, sequence identity, or complementarity between the antisense compound and the target is from about 50% to about 60%. In some embodiments, the homology, sequence identity, or complementarity is from about 60% to about 70%. In some embodiments, the homology, sequence identity, or complementarity is from about 70% to about 80%. In some embodiments, the homology, sequence identity, or complementarity is from about 80% to about 90%. In some embodiments, the homology, sequence identity, or complementarity is about 90%, about 92%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%. When the binding of the antisense compound to the target nucleic acid interferes with the normal function of the target nucleic acid to cause loss of activity, and there is a sufficient degree of complementarity to avoid non-specificity of the antisense compound and the non-target nucleic acid sequence under the conditions required for specific binding. When combined, the antisense compound is specifically hybridizable. Such conditions include physiological conditions in the case of in vivo assays or therapeutic treatments, and conditions for assays in the case of in vitro assays. When an antisense compound (whether DNA, RNA, chimeric, substituted antisense compound, etc.) binds to a target DNA or RNA molecule, it interferes with the normal function of the target DNA or RNA for loss of utility, and there is a sufficient degree of complementarity. To avoid the specific binding of the antisense compound to the non-target sequence (ie, in the case of in vivo assay or therapeutic treatment, under physiological conditions, and in the case of in vitro assays, the assay is performed) The antisense compound is specifically hybridizable when subjected to non-specific binding. In one embodiment, targeting SCNA includes, without limitation, antisense sequences identified and amplified using, for example, PCR, hybridization, etc.; one or more of the sequences set forth in SEQ ID NOs: 12 to 28 and analogs thereof are modulated The performance or function of SCNA. In one embodiment, the performance or function is up-regulated compared to the control. In one embodiment, the performance or function is down-regulated compared to the control. In one embodiment, the oligonucleotide comprises the nucleic acid sequences set forth in SEQ ID NOs: 29-94, including antisense sequences identified and amplified using, for example, PCR, hybridization, and the like. Such oligonucleotides may comprise one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. Examples of modified linkages or internucleotide linkages include phosphorothioates, dithiophosphates, or the like. In one embodiment, the nucleotide comprises a phosphorus derivative. Phosphorus derivatives (or modified phosphate groups) which may be attached to a sugar or sugar analog moiety in a modified oligonucleotide of the invention may be monophosphate, diphosphate, triphosphate, alkyl phosphate Esters, alkyl phosphates, phosphorothioates and the like. The preparation of the above indicated phosphate analogs and their incorporation into nucleotides, modified nucleotides and oligonucleotides are also known per se and need not be described herein. The specificity and sensitivity of antisense are also utilized by those skilled in the art to achieve therapeutic use. Antisense oligonucleotides have been used as therapeutic moieties in the treatment of disease states in animals and humans. Antisense oligonucleotides have been safely and efficiently administered to humans and numerous clinical trials are currently underway. Thus, it is determined that the oligonucleotide can be a therapeutic modality that can be configured to be useful in the treatment of cells, tissues, and animals, particularly humans. In an embodiment of the invention, an oligomeric antisense compound, in particular an oligonucleotide, binds to a target nucleic acid molecule and modulates the expression and/or function of the molecule encoded by the gene of interest. DNA functions that will be interfered with include, for example, replication and transcription. The RNA function that will be interfered with contains all life functions, such as translocation of RNA to protein translation sites, translation of proteins from RNA, splicing of RNA to produce one or more mRNA species, and catalytic activity in which RNA can participate or promote . Depending on the function you want, the function can be up or down. Antisense compounds include antisense oligomeric compounds, antisense oligonucleotides, external leader sequence (EGS) oligonucleotides, alternative splicers, primers, probes, and other oligomeric compounds that hybridize to at least a portion of the target nucleic acid. . Thus, such compounds can be introduced as single, double, partially single, or cyclic oligomeric compounds. In the context of the present invention, targeting an antisense compound to a particular nucleic acid molecule can be a multi-step process. This process typically begins with the identification of a target nucleic acid whose function is to be modulated. The target nucleic acid can be, for example, a nucleic acid molecule that exhibits a cellular gene (or mRNA transcribed from a gene) associated with a particular disorder or disease state, or from an infectious agent. In the present invention, the target nucleic acid encodes a voltage-gated sodium ion channel alpha subunit (SCNA). The targeting process also typically includes determining at least one target region, segment or site within the target nucleic acid for the antisense interaction to occur to produce the desired effect (e.g., to modulate the expression). In the context of the present invention, the term "region" is defined as a portion of a target nucleic acid having at least one identifiable structure, function or feature. The segment is within the region of the target nucleic acid. A "segment" is defined as a smaller portion or sub-portion of a region within a target nucleic acid. A "site" as used in the present invention is defined as a position within a target nucleic acid. In one embodiment, the antisense oligonucleotide binds to the natural antisense sequence of a voltage-gated sodium channel alpha subunit (SCNA) and regulates the performance and/or function of SCNA (SEQ ID NOS: 1 to 11). Examples of natural antisense sequences include SEQ ID NOS: 12 to 28. Examples of antisense oligonucleotides include SEQ ID NOs: 29 to 94. In one embodiment, the antisense oligonucleotide binds to one or more segments of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide and modulates the performance and/or function of the SCNA. The segment comprises at least 5 contiguous nucleotides of a SCNA sense or antisense polynucleotide. In one embodiment, the antisense oligonucleotide is specific for the natural antisense sequence of SCNA, wherein binding of the oligonucleotide to the natural antisense sequence of SCNA modulates the performance and/or function of the SCNA. In one embodiment, the oligonucleotide compound comprises the sequences set forth in SEQ ID NOs: 29-94, including antisense sequences identified and amplified using, for example, PCR, hybridization, and the like. Such oligonucleotides may comprise one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. Examples of modified linkages or internucleotide linkages include phosphorothioates, dithiophosphates, or the like. In one embodiment, the nucleotide comprises a phosphorus derivative. Phosphorus derivatives (or modified phosphate groups) which may be attached to a sugar or sugar analog moiety in a modified oligonucleotide of the invention may be monophosphate, diphosphate, triphosphate, alkyl phosphate Esters, alkyl phosphates, phosphorothioates and the like. The preparation of the above indicated phosphate analogs and their incorporation into nucleotides, modified nucleotides and oligonucleotides are also known per se and need not be described herein. As is known in the art, since the translation initiation codon is typically 5'-AUG (in the transcribed mRNA molecule; 5'-ATG in the corresponding DNA molecule), the translation initiation codon is also referred to as "AUG". Codon, Start Codon or AUG Start Codon. A few genes have a translation initiation codon with an RNA sequence of 5'-GUG, 5'-UUG or 5'-CUG; and 5'-AUA, 5'-ACG and 5'-CUG have been shown to function in vivo. Thus, the terms "translation initiation codon" and "start codon" can encompass a number of codon sequences, even if the starting amino acid in each case is usually methionine (in eukaryotes) or Methionine (in prokaryotes). Eukaryotic and prokaryotic genes can have two or more alternative initiation codons, either of which can be preferentially used to initiate translation in a particular cell type or tissue or under a particular set of conditions. In the context of the present invention, "start codon" and "translation initiation codon" refer to mRNA transcribed in vivo for the initiation of a gene encoding a voltage-gated sodium ion channel alpha subunit (SCNA). The translated codon, regardless of the sequence of such codons. The gene translation stop codon (or "stop codon") can have one of three sequences: 5'-UAA, 5'-UAG, and 5'-UGA (the corresponding DNA sequence is 5'-TAA, respectively). 5'-TAG and 5'-TGA). The terms "start codon region" and "translation initiation codon region" refer to about 25 to about 50 contiguous nucleosides covering either the start of the translation start codon in either direction (ie, 5' or 3'). A portion of the mRNA or gene of the acid. Similarly, the terms "stop codon region" and "translatation stop codon region" refer to about 25 to about 50 contiguous nucleosides covering either the start of the translation stop codon in either direction (ie, 5' or 3'). A portion of the mRNA or gene of the acid. Therefore, the "start codon region" (or "transition initiation codon region") and the "stop codon region" (or "transition termination codon region") are all effectively targeted by the antisense compounds of the present invention. region. An open reading frame (ORF) or "coding region", which is known in the art to refer to the region between the translation initiation codon and the translation stop codon, is also an area that can be effectively targeted. In the context of the present invention, the targeting region is an intragenic region encompassing the translation initiation or termination codon of the open reading frame (ORF) of the gene. Another target region includes a portion of the mRNA known in the art that refers to the start of the translation start codon in the 5' direction, and thus includes the 5' cap site of the mRNA and the translation initiation password. The 5' untranslated region (5' UTR) of the nucleotide (or corresponding nucleotide on the gene) between the children. Another region of interest includes a portion of the mRNA known in the art that refers to the start of the translation stop codon in the 3' direction, and thus includes the nucleotide between the translation stop codon of the mRNA and the 3' end (or 3' untranslated region (3' UTR) of the corresponding nucleotide on the gene. The 5' capping site of the mRNA comprises an N7-methylated guanosine residue joined to the most 5' end of the mRNA via a 5'-5' triphosphate linkage. The 5' cap region of the mRNA is considered to comprise the 5' cap structure itself and 50 nucleotides adjacent to the cap site. Another target area for use in the present invention is the 5' cap region. Although some eukaryotic mRNA transcripts are directly translated, many contain one or more regions called "introns" that are excised from the transcript prior to translation. The remaining (and thus translated) regions are referred to as "exons" and are spliced together to form a continuous mRNA sequence. In one embodiment, targeting a splice site, ie, an intron-exon junction or an exon-intron junction, is particularly useful in diseases involving abnormal splicing or disease involving a particular splicing product. In the case of production. Another embodiment of the target site is due to an abnormal fusion junction due to rearrangement or deletion. mRNA transcripts produced via a splicing process from two (or more) mRNAs of different gene sources are referred to as "fusion transcripts." Introns can be efficiently targeted using antisense compounds that target, for example, DNA or pre-mRNA. In one embodiment, the antisense oligonucleotide binds to the coding and/or non-coding region of the polynucleotide of interest and modulates the expression and/or function of the target molecule. In one embodiment, the antisense oligonucleotide binds to a natural antisense polynucleotide and modulates the expression and/or function of the target molecule. In one embodiment, the antisense oligonucleotide binds to a sense polynucleotide and modulates the expression and/or function of the target molecule. Alternative RNA transcripts can be produced from the same genomic region of DNA. Such alternative transcripts are often referred to as "variants." More specifically, a "pre-mRNA variant" is a transcript produced from the same genomic DNA, unlike other transcripts produced from the same genomic DNA in its initiation or termination position and containing introns and Both of the sequence of the show. Pre-mRNA variants produce smaller "mRNA variants" when one or more exons or intron regions or portions thereof are excised during splicing. Thus, mRNA variants are mRNA variants prior to processing and each unique pre-mRNA variant must always produce unique mRNA variants due to splicing. These mRNA variants are also referred to as "alternative splice variants". If the splicing of the pre-mRNA variant does not occur, the pre-mRNA variant is identical to the mRNA variant. Variants can be generated by using alternative signals to initiate or terminate transcription. The pre-mRNA and mRNA may have more than one start codon or stop codon. Variants derived from pre-mRNA or mRNA using alternative start codons are referred to as "alternative start variants" of the pre-mRNA or mRNA. Their transcripts using alternative stop codons are referred to as "alternative termination variants" of the pre-mRNA or mRNA. A particular type of alternative termination variant is a "polyA variant" in which multiple transcripts are produced by an alternative selection of a "polyA termination signal" by a transcriptional machinery, thereby generating a unique polyA site. Terminated transcripts are produced. In the context of the present invention, the variant types described herein are also examples of target nucleic acids. The position on the target nucleic acid that hybridizes to the antisense compound is defined as the portion of the target region that is targeted by the active antisense compound and that is at least 5 nucleotides in length. Although specific sequences of certain exemplary target segments are set forth herein, those skilled in the art will recognize that such sequences are used to illustrate and describe particular embodiments within the scope of the invention. Other target segments are readily identified by one of ordinary skill in the art in view of the present invention. A target segment of 5 to 100 nucleotides in length comprising an extension of at least five (5) contiguous nucleotides selected from the illustrative preferred target segments is considered to be also suitable for targeting. The target segment can comprise a DNA or RNA sequence comprising at least 5 contiguous nucleotides from the 5' end of an illustrative preferred target segment (the remaining nucleotides are consecutive extensions of the same DNA or RNA, which are immediately followed by the target The upstream of the 5' end of the segment begins and continues until the DNA or RNA contains from about 5 to about 100 nucleotides). A similar preferred target segment is represented by a DNA or RNA sequence comprising at least 5 contiguous nucleotides from the 3' end of an illustrative preferred target segment (the remaining nucleotides are consecutive extensions of the same DNA or RNA, Beginning immediately downstream of the 3' end of the target segment and continuing until the DNA or RNA contains from about 5 to about 100 nucleotides). Those skilled in the art having the subject areas described herein will be able to identify other preferred target segments without performing an experiment. Once one or more of the target regions, segments or sites have been identified, an antisense compound that is sufficiently complementary to the target (ie, sufficiently good to hybridize) and of sufficient specificity is selected to produce the desired effect. In an embodiment of the invention, the oligonucleotide is associated with an antisense strand of a particular target. Oligonucleotides are at least 5 nucleotides in length and can be synthesized such that each oligonucleotide targets an overlapping sequence such that the oligonucleotide is synthesized to encompass the entire length of the polynucleotide of interest. Targets also include coding and non-coding areas. In one embodiment, the specific nucleic acid is preferably targeted with an antisense oligonucleotide. Targeting an antisense compound to a particular nucleic acid molecule is a multi-step process. This process typically begins with the identification of the nucleic acid sequence whose function is to be modulated. This can be, for example, a cellular gene (or mRNA transcribed from a gene), or a non-coding polynucleotide, such as a non-coding RNA (ncRNA), that is associated with a particular disorder or disease state. RNA can be classified into (1) messenger RNA (mRNA), which is translated into protein, and (2) RNA that does not encode protein (ncRNA). ncRNAs contain microRNAs, antisense transcripts and other transcription units (TUs) that contain high-density stop codons and lack any extended "open reading frame". Many ncRNAs appear to start from the start site in the 3' untranslated region (3' UTR) of the locus of the encoded protein. ncRNAs are usually sparse and at least half of the ncRNAs that have been sequenced by the FANTOM Association do not appear to be polyadenylated. Most researchers have focused on polyadenylation mRNA that is processed and excreted into the cytoplasm for obvious reasons. Recently, the collection of non-polyadenylated nuclear RNAs has been shown to be extremely large, and many of these transcripts are produced by so-called intergenic regions. The mechanism by which ncRNAs can regulate gene expression is based on base pairing with a target transcript. RNA acting by base pairing can be grouped into (1) cis-encoding RNA, which is encoded at the same genetic position but on the opposite strand of the RNA to which it acts and thus exhibits complete complementarity with its target, and (2 Trans-encoding RNA, which encodes at a chromosomal location different from the RNA it acts on and typically does not exhibit full base pairing potential with its target. Without wishing to be bound by theory, the perturbation of the antisense oligonucleotides described herein by antisense oligonucleotides can alter the performance of the corresponding sense messenger RNA. However, this regulation may be inconsistent (antisense blocking gene expression results in elevated messenger RNA) or consensus (antisense blocking gene expression results in concomitant messenger RNA reduction). In such cases, the antisense oligonucleotide can be targeted to overlapping or non-overlapping portions of the antisense transcript, thereby causing it to block gene expression or sequestration. Both coding and non-coding antisense can be targeted in the same manner and any of the species can modulate the corresponding sense transcript in a consistent or inconsistent manner. The strategy for identifying novel oligonucleotides for use against a target can be based on blocking the gene expression of the antisense RNA transcript by antisense oligonucleotides or any other means of modulating the desired target.Strategy 1 : In the case of inconsistent regulation, blocking the expression of the antisense transcript gene will increase the performance of the conventional (sense) gene. If a known gene encodes a known or putative drug target, it is conceivable that the gene expression that blocks its antisense counterpart mimics the effect of a receptor agonist or enzyme stimulant.Strategy 2 : In the case of consistent regulation, gene expression of both antisense and sense transcripts can be blocked and thereby synergistically reduce conventional (sense) gene expression. If, for example, an antisense oligonucleotide is used to block gene expression, this strategy can be used to administer an antisense oligonucleotide that targets a sense transcript and another antisense oligo that targets the corresponding antisense transcript. Glycosidic acid, or a single energy symmetric antisense oligonucleotide that simultaneously targets overlapping sense and antisense transcripts. According to the invention, antisense compounds include antisense oligonucleotides, ribonucleases, external leader sequence (EGS) oligonucleotides, siRNA compounds, single or double stranded RNA interference (RNAi) compounds (such as siRNA compounds), And other oligomeric compounds that hybridize to at least a portion of the target nucleic acid and modulate their function. Thus, it can be DNA, RNA, DNA-like, RNA-like or a mixture thereof, or a mimetic of one or more of these. Such compounds may be single-stranded, double-stranded, circular or hairpin oligomeric compounds and may contain structural elements such as internal or terminal protrusions, mismatches or rings. The antisense compounds are prepared in a conventional manner in a linear form but may be linked or otherwise prepared to form a ring and/or a branch. An antisense compound can include a construct, such as a single strand that hybridizes to form a fully or partially double-stranded compound or has sufficient self-complementarity to allow hybridization and formation of a fully or partially double-stranded compound. The two strands may be internally connected to leave a free 3' or 5' end; or may be joined to form a continuous hairpin structure or loop. The hairpin structure may have protrusions on the 5' or 3' end to create an extension having a single strand characteristic. The double-stranded compound may optionally include protrusions on the ends. Other modifications may include a binding group attached to one end, a selected nucleotide position, a sugar position, or an internucleoside linkage. Alternatively, the two strands may be linked via a non-nucleic acid moiety or a linking group. When formed from only one strand, the dsRNA can take the form of a self-complementary hairpin-type molecule that folds in itself to form a double helix. Thus, the dsRNA can be fully or partially double stranded. Gene expression can be specifically regulated by stably expressing dsRNA hairpins in a transgenic cell line, however, in some embodiments, gene expression or function is upregulated. When formed from two strands, or a single strand in the form of a self-complementary hairpin type molecule that is folded in half to form a double helix, the two strands (or single stranded double helix forming regions) are taken in Waldorf-Kerry Complementary RNA strands that base pairing in gram mode. Once introduced into the system, the compounds of the invention may elicit one or more enzymes or structural proteins to effect cleavage or other modification of the target nucleic acid or may function via an occupancy-based mechanism. In general, nucleic acids (including oligonucleotides) can be described as "DNA-like" (ie, typically having one or more 2'-deoxy sugars and usually having a T base rather than a U base) or "RNA-like" (ie, typically having one or more 2'-hydroxy or 2'-modified sugars and typically having a U base rather than a T base). Nucleic acid helices can employ more than one type of structure, most commonly A and B. In general, an oligonucleotide having a B-like structure is "DNA-like" and an oligonucleotide having an A-like structure is "RNA-like". In some (chimeric) embodiments, an antisense compound can contain both an A-form region and a B-form region. In one embodiment, the desired oligonucleotide or antisense compound comprises at least one of: an antisense RNA, an antisense DNA, a chimeric antisense oligonucleotide, an antisense oligonucleotide comprising a modified bond , interfering RNA (RNAi), short interfering RNA (siRNA); micro interfering RNA (miRNA); small temporal RNA (small, temporal RNA) (stRNA); or short hairpin RNA (shRNA); small RNA-induced gene activation ( RNAa); small activating RNA (saRNA) or a combination thereof. dsRNA also activates gene expression, a mechanism known as "small RNA-induced gene activation" or RNAa. The dsRNA targeting the gene promoter induces strong transcriptional activation of related genes. RNAa is demonstrated in human cells using synthetic dsRNA called "small activating RNA" (saRNA). It is currently unknown whether RNAa is conserved in other organisms. Small double-stranded RNA (dsRNA) such as small interfering RNA (siRNA) and microRNA (miRNA) has been found to be a trigger for an evolutionarily conserved mechanism called RNA interference (RNAi). RNAi always results in gene quiescence by remodeling chromatin to thereby inhibit transcription, degrade complementary mRNA, or block protein translation. However, as detailed in the Examples section that follows, oligonucleotides are shown to increase the performance and/or function of voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotides and their encoded products. dsRNA can also act as a small activating RNA (saRNA). Without wishing to be bound by theory, by targeting a sequence in a gene promoter, the saRNA will induce expression of the target gene, a phenomenon known as dsRNA-induced transcriptional activation (RNAa). In another embodiment, the "better target segment" identified herein can be used in the screening of other compounds that modulate the performance of voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotides. A "modulator" is one that reduces or increases the performance of a nucleic acid molecule encoding a SCNA and comprises a compound that is complementary to at least a 5 nucleotide portion of the preferred target segment. The screening method comprises the steps of contacting a preferred target segment of a nucleic acid molecule encoding a sense or natural antisense polynucleotide of SCNA with one or more candidate modulators, and selecting one or more reducing or increasing encoding of the SCNA poly Candidate modulators of the expression of nucleic acid molecules of nucleotides, such as SEQ ID NOS: 29-94. Once the candidate modulator is shown to be capable of modulating (eg, reducing or increasing) the expression of the nucleic acid molecule encoding the SCNA polynucleotide, the modulator can then be used in other exploratory studies of the function of the SCNA polynucleotide, or as The research agent, diagnostic agent or therapeutic agent of the invention. Targeting the natural antisense sequence preferably modulates the function of the target gene. For example, SCNA genes (eg, accession numbers NM_001165963, NM_021007, NM_006922, NM_000334, NM_198056, NM_002976, NM_014191, NM_002977, NM_006514, NM_014139, AF109737). In one embodiment, the target is an antisense polynucleotide of the SCNA gene. In one embodiment, the antisense oligonucleotides are targeted to SCNA polynucleotides (eg, accession numbers NM_001165963, NM_021007, NM_006922, NM_000334, NM_198056, NM_002976, NM_014191, NM_002977, NM_006514, NM_014139, AF109737) and/or Or a natural antisense sequence, variants thereof, dual genes, isoforms, homologs, mutants, derivatives, fragments and complementary sequences. Preferably, the oligonucleotide is an antisense molecule and the target comprises an encoding and non-coding region of an antisense and/or sense SCNA polynucleotide. Preferred target segments of the invention may also be combined with the respective complementary antisense compounds of the invention to form stabilized double stranded (double helix) oligonucleotides. In the art, these double-stranded oligonucleotide moieties have been shown to modulate target expression via antisense mechanisms and regulate translation and RNA processing. In addition, the double-stranded portion can undergo chemical modification. For example, such double-stranded portions have been shown to undergo classical cross-linking with the target by the antisense strand of the double helix, thereby triggering enzymatic degradation of the target to inhibit the target. In one embodiment, the antisense oligonucleotide targets a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide (eg, accession numbers NM_001165963, NM_021007, NM_006922, NM_000334, NM_198056, NM_002976, NM_014191, NM_002977, NM_006514, NM_014139, AF109737), variants thereof, dual genes, isoforms, homologs, mutants, derivatives, fragments and complementary sequences. Preferably, the oligonucleotide is an antisense molecule. According to an embodiment of the invention, the target nucleic acid molecule is not limited to SCNA but extends to any of the isoforms, receptors, homologs and analogs thereof of the SCNA molecule. In one embodiment, the oligonucleotide targets a natural antisense sequence of a SCNA polynucleotide (eg, the polynucleotides set forth in SEQ ID NOs: 12 to 28) and any variants thereof, dual genes, homologs , mutants, derivatives, fragments and complementary sequences. Examples of antisense oligonucleotides are set forth in SEQ ID NOs: 29-94. In one embodiment, the oligonucleotide is complementary to or binds to a nucleic acid sequence of SCNA antisense, including, without limitation, non-coding sense and/or antisense sequences associated with a SCNA polynucleotide, and modulating the performance of the SCNA molecule And / or function. In one embodiment, the oligonucleotide is complementary to or binds to the nucleic acid sequence of the SCNA natural antisense as set forth in SEQ ID NOs: 12 to 28, and modulates the expression and/or function of the SCNA molecule. In one embodiment, the oligonucleotide comprises a sequence of at least 5 contiguous nucleotides of SEQ ID NOs: 29 to 94 and modulates the expression and/or function of the SCNA molecule. Polynucleotide targets include SCNAs, including family members, variants of SCNA; mutants of SCNA, including SNPs; non-coding sequences of SCNA; dual genes of SCNA; species variants, fragments and analogs thereof. Preferably, the oligonucleotide is an antisense molecule. In one embodiment, the oligonucleotide that targets the SCNA polynucleotide comprises: antisense RNA, interfering RNA (RNAi), short interfering RNA (siRNA); micro interfering RNA (miRNA); small temporal RNA (stRNA) Or short hairpin RNA (shRNA); small RNA-induced gene activation (RNAa); or small activating RNA (saRNA). In one embodiment, targeting voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotides, such as SEQ ID NOS: 1 to 11, modulate the performance or function of such targets. In one embodiment, the performance or function is up-regulated compared to the control. In one embodiment, the performance or function is down-regulated compared to the control. In another embodiment, targeting a natural antisense transcript (eg, SEQ ID NOs: 12 to 28) and any other target NAT of such target polynucleotides results in upregulation of the target mRNA and corresponding protein. In one embodiment, the antisense compound comprises the sequences set forth in SEQ ID NOs: 29-94. Such oligonucleotides may comprise one or more modified nucleotides, shorter or longer fragments, modified linkages, and the like. In one embodiment, SEQ ID NOs: 29 to 94 comprise one or more LNA nucleotides. Table 1 shows exemplary antisense oligonucleotides suitable for use in the methods of the invention. table 1 : * indicates a phosphorothioate linkage, + indicates LNA, "r" indicates RNA and "m" indicates a methyl group on the 2' oxygen atom on the designated sugar moiety of the oligonucleotide. To avoid ambiguity, this LNA has the following formula:Wherein B is a specific designated base. table 2 : Relative expression of SCN1A mRNA in cells treated with antisense oligonucleotides targeting SCN1A-specific natural antisense transcripts. The mean fold difference in the performance of SCN1A compared to the control transfected with Avg-; the Std-standard deviation, the P-treated sample is no different from the probability of simulating the control. The total number of N-parallel measurements. table 2 : The desired target nucleic acid can be modulated in a number of ways known in the art. For example, antisense oligonucleotides, siRNA, and the like. Enzymatic nucleic acid molecules (e.g., ribonucleases) are those capable of catalyzing one or more of a variety of reactions, including nucleic acid molecules capable of repeatedly cleaving other individual nucleic acid molecules in a nucleotide base sequence specific manner. Such enzymatic nucleic acid molecules can be used, for example, to target virtually any RNA transcript. Trans-lytic enzymatic nucleic acid molecules are promising as therapeutic agents for human diseases due to their sequence specificity. Enzymatic nucleic acid molecules can be designed to cleave specific RNA targets within the context of cellular RNA. This cleavage event renders the mRNA non-functional and abolishes the expression of the protein from the other RNA. In this way, the synthesis of proteins associated with disease states can be selectively inhibited. In general, an enzymatic nucleic acid having an RNA cleavage activity functions by first binding to a target RNA. This binding occurs via a target binding moiety of the enzymatic nucleic acid that remains in close proximity to the enzymatic portion of the molecule that functions to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes the target RNA and then binds to the target RNA via complementary base pairing, and once bound to the correct site, acts enzymatically to cleave the target RNA. The key cleavage of this target RNA will destroy its ability to direct synthesis of the encoded protein. After the enzymatic nucleic acid has bound and cleaves its RNA target, it is released from the other RNA to search for another target and can repeatedly bind and cleave the new target. Several methods such as the In Vitro Selection (Evolution) Strategy (Orgel, (1979) Proc. R. Soc. London, B 205, 435) have been used to develop a variety of reactions, such as cleavage and linkage of phosphodiester bonds and guanamines. A novel nucleic acid catalyst for the bond. The development of a catalytically active ribonuclease will significantly contribute to the use of RNA cleavage ribonuclease to achieve any strategy for the regulation of gene expression. The hammerhead ribonuclease acts, for example, at a catalytic rate (kcat) of about 1 min-1 in the presence of a saturated (10 mM) concentration of Mg2+ cofactor. Artificial "RNA ligase" ribonucleases have been shown to catalyze the corresponding self-modification reactions at a rate of about 100 min-1. Furthermore, it is known that certain modified hammerhead ribonucleases having a binding arm composed of DNA catalyze RNA cleavage at multiple conversion rates of approximately 100 min-1. Finally, replacement of a particular residue within the catalytic core of the hammerhead with certain nucleotide analogs results in a modified ribonuclease that exhibits up to a 10-fold improvement in catalytic rate. These findings demonstrate that ribonuclease can significantly promote chemical conversion at a catalytic rate that is significantly greater than the catalytic rate exhibited by most native self-cleaving ribonucleases in vitro. It is then possible that the structure of certain self-cleaving ribonucleases can be optimized to produce maximum catalytic activity, or completely novel RNA motifs that exhibit significantly faster RNA phosphodiester cleavage rates can be prepared. The intermolecular cleavage of RNA catalysts for RNA conformation was first shown in 1987 (Uhlenbeck, O. C. (1987) Nature, 328: 596-600). The RNA catalyst is recovered and reacted with a plurality of RNA molecules to prove that it does catalyze. Catalytic RNA designed based on the "hammerhead" motif has been used to cleave specific target sequences by making appropriate base changes in the catalytic RNA to maintain the necessary base pairing with the target sequence. This has allowed the use of catalytic RNA to cleave specific target sequences and indicates that catalytic RNA designed according to the "hammerhead" model may cleave specific receptor RNA in vivo. RNA interference (RNAi) has become a powerful tool for regulating gene expression in mammalian and mammalian cells. This method requires the transmission of small interfering RNA (siRNA) in the form of self RNA or in the form of DNA using a coding sequence that expresses a plastid or virus and a small hairpin RNA that is processed into siRNA. This system is capable of efficiently transporting pre-siRNA to the cytoplasm where the pro-siRNA is active and allows the use of a regulated and tissue-specific promoter to achieve gene expression. In one embodiment, the oligonucleotide or antisense compound comprises an oligo or polymer of ribonucleic acid (RNA) and/or deoxyribonucleic acid (DNA), or a mimetic, chimera, analog or the like Source. The term includes oligonucleotides consisting of naturally occurring nucleotides, sugars, and covalent internucleoside (backbone) linkages, as well as oligonucleotides having portions that are not naturally occurring and functioning in a similar manner. Such modified or substituted oligonucleotides are often required to exceed the native form due to desirable properties such as enhanced cellular uptake, increased affinity for the target nucleic acid, and increased stability in the presence of nucleases. According to the invention, an oligonucleotide or "antisense compound" includes an antisense oligonucleotide (eg, an RNA, DNA, mimetic, chimera, analog or homolog thereof), a ribonuclease, an external leader sequence ( EGS) Oligonucleotides, siRNA compounds, single or double stranded RNA interference (RNAi) compounds (such as siRNA compounds), saRNA, aRNA, and other oligomeric compounds that hybridize to at least a portion of the target nucleic acid and modulate its function. Thus, it can be DNA, RNA, DNA-like, RNA-like or a mixture thereof, or a mimetic of one or more of these. Such compounds may be single-stranded, double-stranded, circular or hairpin oligomeric compounds and may contain structural elements such as internal or terminal protrusions, mismatches or rings. The antisense compounds are prepared in a conventional manner in a linear form but may be linked or otherwise prepared to form a ring and/or a branch. An antisense compound can include a construct, such as a single strand that hybridizes to form a fully or partially double-stranded compound or has sufficient self-complementarity to allow hybridization and formation of a fully or partially double-stranded compound. The two strands may be internally connected to leave a free 3' or 5' end; or may be joined to form a continuous hairpin structure or loop. The hairpin structure may have protrusions on the 5' or 3' end to create an extension having a single strand characteristic. The double-stranded compound may optionally include protrusions on the ends. Other modifications may include a binding group attached to one end, a selected nucleotide position, a sugar position, or an internucleoside linkage. Alternatively, the two strands may be linked via a non-nucleic acid moiety or a linking group. When formed from only one strand, the dsRNA can take the form of a self-complementary hairpin-type molecule that folds in itself to form a double helix. Thus, dsRNAs can be fully or partially double stranded. Gene expression can be specifically regulated by stably expressing dsRNA hairpins in transgenic cell lines. When formed from two strands, or a single strand in the form of self-complementary hairpin-type molecules that are folded in half to form a double helix, the two strands (or single-stranded double helix forming regions) are in the form of Waldorf-German Complementary RNA strands for base pairing in Rick mode. Once introduced into the system, the compounds of the invention may elicit the action of one or more enzymes or structural proteins to effect cleavage or other modification of the target nucleic acid or may act via a mechanism based on occupancy. In general, nucleic acids (including oligonucleotides) can be described as "DNA-like" (ie, typically having one or more 2'-deoxy sugars and usually having a T base rather than a U base) or "RNA-like" (ie, typically having one or more 2'-hydroxy or 2'-modified sugars and typically having a U base rather than a T base). Nucleic acid helices can be used in more than one type of structure, most commonly in the A form and the B form. In general, an oligonucleotide having a B-like structure is "DNA-like" and an oligonucleotide having an A-like structure is "RNA-like". In some (chimeric) embodiments, an antisense compound can contain both an A-form region and a B-form region. Antisense compounds of the invention may comprise an antisense portion of from about 5 to about 80 nucleotides in length (i.e., from about 5 to about 80 linked nucleosides). This refers to the length of the antisense strand or portion of the antisense compound. In other words, the single antisense compound of the invention comprises from 5 to about 80 nucleotides, and the double antisense compound of the invention (such as dsRNA) comprises sense and antisense of from 5 to about 80 nucleotides in length. Share or part. One of ordinary skill will appreciate that this includes lengths of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 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, 49, 50 , 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75 , 76, 77, 78, 79 or 80 nucleotides or any range of antisense portions therebetween. In one embodiment, an antisense compound of the invention has an antisense portion of 10 to 50 nucleotides in length. The average technician should understand that this includes the antisense portion lengths of 10, 11, 12, 13, 14, 15, 16, 17, 18, 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, 49 or 50 nucleotides or any Range of oligonucleotides. In some embodiments, the oligonucleotide is 15 nucleotides in length. In one embodiment, an antisense or oligonucleotide compound of the invention has an antisense portion of 12 or 13 to 30 nucleotides in length. One of ordinary skill will appreciate that this includes antisense portions of length 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 Nucleotide or any range of antisense compounds therebetween. In one embodiment, the oligomeric compounds of the invention also include variants in which different bases are present at one or more nucleotide positions in the compound. For example, if the initial nucleotide is adenosine, a variant containing thymidine, guanosine or cytidine at this position can be produced. This can be done at any position of the antisense or dsRNA compound. These compounds are then tested using the methods described herein to determine their ability to inhibit the performance of the target nucleic acid. In some embodiments, the homology, sequence identity, or complementarity between the antisense compound and the target is from about 40% to about 60%. In some embodiments, the homology, sequence identity, or complementarity is from about 60% to about 70%. In some embodiments, the homology, sequence identity, or complementarity is from about 70% to about 80%. In some embodiments, the homology, sequence identity, or complementarity is from about 80% to about 90%. In some embodiments, the homology, sequence identity, or complementarity is about 90%, about 92%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%. In one embodiment, antisense oligonucleotides such as the nucleic acid molecules set forth in SEQ ID NOs: 29-94 comprise one or more substitutions or modifications. In one embodiment, the nucleotide is substituted with a locked nucleic acid (LNA). In one embodiment, the oligonucleotide targets one or more of the sequence of the nucleic acid molecule encoding and/or non-coding sequence associated with the SCNA, and/or antisense, and the sequences set forth in SEQ ID NOs: 1 to 28. region. Oligonucleotides are also targeted to the overlapping regions of SEQ ID NOS: 1 to 28. Certain preferred oligonucleotides of the invention are chimeric oligonucleotides. In the context of the present invention, a "chimeric oligonucleotide" or "chimera" is an oligonucleotide comprising two or more chemically distinct regions each consisting of at least one nucleotide. Such oligonucleotides typically contain at least one region of the modified nucleotide that confers one or more beneficial properties, such as increased resistance to nucleases, increased uptake into cells, increased binding affinity to the target, and as capable of cleavage. RNA: A region of the DNA or RNA: RNA hybrid that is responsible for the enzyme. For example, RNase H is a cellular endonuclease that cleaves RNA strands of RNA:DNA duplexes. Thus, activation of RNase H results in cleavage of the RNA target, thereby greatly enhancing the efficiency of antisense regulation of gene expression. Thus, similar results obtained with shorter oligonucleotides are typically obtained when chimeric oligonucleotides are used as compared to hybridization of phosphorothioate deoxyoligonucleotides to the same target region. Cleavage of the RNA target can be detected in a conventional manner by gel electrophoresis and, if desired, related nucleic acid hybridization techniques known in the art. In one embodiment, the chimeric oligonucleotide comprises at least one region modified to increase the binding affinity of the target and a region that normally acts as a substrate for RNAse H. The affinity of an oligonucleotide for its target (in this case, the nucleic acid encoding ras) is determined in a conventional manner by measuring the Tm of the oligonucleotide/target pair, and Tm is the temperature at which the oligonucleotide dissociates from the target. Dissociation is detected by spectrophotometry. The higher the Tm, the greater the affinity of the oligonucleotide for the target. The chimeric antisense compounds of the invention can be formed into a composite structure of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds are also referred to in the art as hybrids or gapmers. Representative US patents for the preparation of such hybrid structures include, but are not limited to, U.S. Patent Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350 , 5, 652, 355; 5, 652, 355; 5, 652, 356; and 5,700, 922, each incorporated herein by reference. In one embodiment, the region of the modified oligonucleotide comprises at least one nucleotide modified at the 2' position of the sugar, most preferably 2'-O alkyl, 2'-O-alkyl-O - an alkyl or 2'-fluoro-modified nucleotide. In another embodiment, the RNA modification comprises a 2'-fluoro, 2'-amino and 2' O-methyl modification on the ribose of the pyrimidine, an abasic residue or an inverted base at the 3' end of the RNA. Such modifications are incorporated into the oligonucleotides in a conventional manner, and such oligonucleotides have been shown to have a higher Tm (i.e., higher target binding affinity) than the 2'-deoxyoligonucleotide for a given target. The effect of this increased affinity is to greatly enhance RNAi oligonucleotide inhibition gene expression. RNAse H is a cellular endonuclease that cleaves RNA:DNA double helix RNA strands; therefore, this enzyme activation results in cleavage of the RNA target and thus greatly enhances the efficiency of RNAi inhibition. Cleavage of the RNA target can be demonstrated by gel electrophoresis in a conventional manner. In one embodiment, the chimeric oligonucleotide is also modified to enhance nuclease resistance. The cell contains a plurality of exonuclease and endonuclease which degrade the nucleic acid. Many nucleotide and nucleoside modifications have been shown to make the incorporated oligonucleotides more resistant to nuclease digestion than natural oligodeoxynucleotides. Nuclease resistance is measured in a conventional manner by culturing the oligonucleotide with a cell extract or an isolated nuclease solution and measuring the extent to which the intact oligonucleotide remains over time, typically by gel electrophoresis. Oligonucleotides that have been modified to enhance their nuclease resistance are more fully present than unmodified oligonucleotides. A variety of oligonucleotide modifications have been shown to enhance or confer nuclease resistance. Oligonucleotides currently containing at least one phosphorothioate modification are preferred. In some cases, oligonucleotide modifications that enhance target binding affinity are also independently capable of enhancing nuclease resistance. Specific examples of some preferred oligonucleotides contemplated by the present invention include such modified backbones, such as phosphorothioates, phosphotriesters, methylphosphonates, short chain alkyl or cycloalkyl sugar linkages. A bond between a short or a short chain hetero atom or a heterocyclic sugar. Preferably, it is an oligonucleotide having a phosphorothioate skeleton, and has a hetero atom skeleton, particularly CH2--NH--O--CH2, CH, -N(CH3)--O--CH2 [called Is a methylene (methylimido) or MMI skeleton], CH2--O--N(CH3)--CH2, CH2-N(CH3)--N(CH3)--CH2 and O--N (CH3)--CH2--CH2 skeleton in which the natural phosphodiester skeleton is represented by O--P--O--CH. De Mesmaeker et al. (1995) Acc. Chem. Res. 28: 366-374 discloses that the indoleamine backbone is also preferred. Oligonucleotides having a morpholino skeleton structure (Summerton and Weller, U.S. Patent No. 5,034,506) are also preferred. In another embodiment, a peptide nucleic acid (PNA) backbone, such as an oligonucleotide, and a phosphodiester backbone are replaced by a polyamine backbone, wherein the nucleotide is bonded directly or indirectly to the aza nitrogen atom of the polyamine backbone. The oligonucleotide may also comprise one or more substituted sugar moieties. Preferred oligonucleotides comprise one of the following at the 2' position: OH, SH, SCH3, F, OCN, OCH3 OCH3, OCH3 O(CH2)n CH3, O(CH2)n NH2 or O(CH2)n CH3 Wherein n is from 1 to about 10; C1 to C10 lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF3; OCF3; O--, S-- or N-alkyl; O--, S-- or N-alkenyl; SOCH3; SO2 CH3; ONO2; NO2; N3; NH2; heterocycloalkyl; heterocycloalkylaryl; Aminoalkylamino; polyalkylamino; substituted decyl; RNA cleavage group; reporter group; intercalator; group for improving the pharmacokinetic properties of the oligonucleotide; or modified oligonucleotide Groups of pharmacodynamic properties and other substituents of similar nature. Preferred modifications include 2'-methoxyethoxy [2'-O-CH2 CH2 OCH3, also known as 2'-O-(2-methoxyethyl)]. Other preferred modifications include 2'-methoxy (2'-O--CH3), 2'-propoxy (2'-OCH2 CH2CH3) and 2'-fluoro (2'-F). Similar modifications can also be made at other positions on the oligonucleotide, at the 3' position of the sugar at the 3' end nucleotide and at the 5' position of the 5' end nucleotide. Oligonucleotides may also have a sugar mimetic, such as a cyclobutyl group in place of a pentofuranosyl group. Oligonucleotides may also or alternatively include modifications or substitutions of nucleobases (often referred to in the art as "bases"). As used herein, "unmodified" or "natural" nucleotides include adenine (A), guanine (G), thymine (T), cytosine (C), and uracil (U). Modified nucleotides include nucleotides that are only occasionally or transiently found in natural nucleic acids, such as hypoxanthine, 6-methyladenine, 5-Mepyrimidine, specifically 5-methylcytosine (also known as 5-methylcytosine) 5-methyl-2'deoxycytosine and is often referred to in the art as 5-Me-C), 5-hydroxymethylcytosine (HMC), glycosyl HMC, and gentobiosyl HMC And synthetic nucleotides such as 2-aminoadenine, 2-(methylamino)adenine, 2-(imidazolylalkyl)adenine, 2-(aminoalkylamino) adenine or other miscellaneous Substituted alkyl adenine, 2-thiouracil, 2-thiothymidine, 5-bromouracil, 5-hydroxymethyluracil, 8-azaguanine, 7-deazaguanine, N6 (6- Aminohexyl) adenine and 2,6-diaminopurine. "General" bases known in the art, such as inosine, can be included. The 5-Me-C substitution has been shown to increase the stability of the nucleic acid duplex by 0.6-1.2 ° C and is currently a preferred base substitution. Another modification of the oligonucleotides of the invention involves chemically linking one or more portions or conjugates of the enhanced oligonucleotide to the oligonucleotide. Such moieties include, but are not limited to, lipid moieties (such as cholesterol moieties, cholesteryl moieties), aliphatic chains (such as dodecanediol or eleven base residues), polyamines or polyethylene glycol chains, or diamonds Adamantane acetic acid. Oligonucleotides comprising a lipophilic moiety and methods of preparing such oligonucleotides are known in the art, for example, in U.S. Patent Nos. 5,138,045, 5,218,105, and 5,459,255. Not all positions in a given oligonucleotide must be uniformly modified, and indeed more than one of the above mentioned modifications can be incorporated into a single oligonucleotide or even within a single nucleoside within an oligonucleotide. The invention also includes oligonucleotides that are chimeric oligonucleotides as defined above. In another embodiment, a nucleic acid molecule of the invention is conjugated to another moiety, including but not limited to, abasic nucleotides, polyethers, polyamines, polyamines, peptides, carbohydrates, lipids or Polyhydrocarbon compound. Those skilled in the art will recognize that such molecules can be linked to one or more of any of the nucleotides comprising the nucleic acid molecule at several positions on the sugar, base or phosphate group. Oligonucleotides for use in accordance with the present invention can be conveniently and routinely prepared via well known solid phase synthesis techniques. The equipment used for this synthesis is sold by several suppliers including Applied Biosystems. Any other means for this synthesis can also be employed; the actual synthesis of the oligonucleotides is well within the skill of the average skilled artisan. It is also well known to use similar techniques to prepare other oligonucleotides, such as phosphorothioates and alkylated derivatives. It is also well known to use similar techniques and commercially available modified amidites and controlled-pore glass (CPG) products, such as biotin, luciferin, acridine or psoralen. (psoralen) modified amino acid esters and/or CPG (available from Glen Research, Sterling VA) to synthesize fluorescently labeled oligonucleotides, biotinylated oligonucleotides or other modified oligos Nucleotides, such as cholesterol-modified oligonucleotides. In accordance with the present invention, modifications such as the use of LNA monomers to enhance the potency, specificity and duration of action and broaden the route of administration of oligonucleotides comprising current chemical compositions (such as MOE, ANA, FANA, PS, etc.) are used. This can be achieved by replacing some of the monomers in the current oligonucleotide with LNA monomers. The LNA modified oligonucleotide may have a size similar to the parent compound or may be larger or preferably smaller. Preferably, such LNA-modified oligonucleotides contain less than about 70%, more preferably less than about 60%, optimally less than about 50% of the LNA monomer and are between about 5 and 25 nucleotides in size. More preferably, between about 12 and 20 nucleotides. Preferred modified oligonucleotide backbones include, but are not limited to, phosphorothioates, palmitic phosphorothioates, dithiophosphates, phosphotriesters, aminoalkyl phosphates, methylphosphines Acid esters and other alkyl phosphonates (including 3' phosphonic acid alkyl esters and palmitic phosphonates), phosphonites, amino phosphates (including 3'-amino amino phosphates and amine groups) Alkylamino phosphate), thiocarbonylamino phosphate, thiocarbonylalkylphosphonate, thiocarbonylalkylphosphoric acid triester, and boranophosphate having a normal 3'-5' bond, a 2'-5' linked analog of borane phosphate, and a borane phosphate having a reverse polarity, wherein the adjacent nucleoside unit pair is 3'-5' to 5'-3' or 2'-5' Connect to 5'-2'. Also included are various salts, mixed salts and free acid forms. Representative US patents for the preparation of the above phosphorus-containing bonds include, but are not limited to, U.S. Patent Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019 , No. 5, 286, No. 5, No. 5, 286, No. 5, No. 5, 351, No. 5, 563, 253; Preferred modified oligonucleotide backbones which do not include a phosphorus atom have a short chain alkyl or cycloalkyl internucleoside linkage, a mixed heteroatom and an alkyl or cycloalkyl internucleoside linkage, or one or more A skeleton formed by a short chain hetero atom or a heterocyclic nucleoside bond. Such oligonucleotide backbones comprise an oligonucleotide backbone having the following: a morpholino linkage (partially formed from a sugar moiety of a nucleoside); a oxoxane skeleton; a sulfide, an anthracene and an anthracene skeleton; Formacetyl and thioformacetyl skeleton; methylene methyl ethyl and methylene thiomethyl sulfonyl skeleton; olefin-containing skeleton; amino sulfonate skeleton; methylene imino group And a methylene fluorenyl skeleton; a sulfonate and a sulfonamide skeleton; a guanamine skeleton; and other skeletons having a mixture of N, O, S and CH2 components. Representative US patents for the preparation of the above oligonucleosides include, but are not limited to, U.S. Patent Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564 , 5, 405, 938; 5, 434, 257; 5, 466, 677; 5, 470, 967; 5, 489, 677; 5, 541, 307; 5, 561, 225; 5, 596, 086; 5, 602, 240; 5, 610, 289; 5, 602, 240; 5, 608, 046; U.S. Patent Nos. 5,610, 399; 5, 618, 704; 5, 623, 070; 5, 663, 312; 5, 633, 360; 5, 677, 437; and 5, 677, 439 each incorporated herein by reference. In other preferred oligonucleotide mimetics, both the sugar and the internucleoside linkage (i.e., the backbone) of the nucleotide unit are replaced by a novel group. Base units are retained for hybridization to the appropriate nucleic acid target compound. One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as peptide nucleic acid (PNA). In the PNA compound, the sugar-backbone of the oligonucleotide is replaced by a guanamine-containing backbone, specifically an amine ethylglycine skeleton. The nucleobase is retained and directly or indirectly bound to the aza nitrogen atom of the backbone guanamine moiety. Representative US patents for the preparation of PNA compounds include, but are not limited to, U.S. Patent Nos. 5,539,082; 5,714,331; and 5,719,262 each incorporated herein by reference. Other teachings of PNA compounds can be found in Nielsen et al. (1991) Science 254, 1497-1500. In one embodiment of the invention, the oligonucleotide has a phosphorothioate backbone and the oligonucleoside has a heteroatom backbone, and in particular CH2-NH-O-CH2-; referred to as methylene (methyl Amino) or MMI backbone -CH2-N(CH3)-O-CH2-;-CH2-ON(CH3)-CH2-;-CH2N(CH3)-N(CH3)CH2- and -ON(CH3)- CH2-CH2-, wherein the natural phosphodiester backbone is represented by the above-mentioned U.S. Patent No. 5,489,677 -OPO-CH2-; and the above-cited U.S. Patent No. 5,602,240. Oligonucleotides having the morpholino skeleton structure of U.S. Patent No. 5,034,506, which is incorporated by reference above, are also preferred. The modified oligonucleotide may also contain one or more substituted sugar moieties. Preferred oligonucleotides comprise one of the following at the 2' position: OH; F; O-, S- or N-alkyl; O-, S- or N-alkenyl; O-, S- or N- Alkynyl; or Oalkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl groups may be substituted or unsubstituted C to CO alkyl or C2 to CO alkenyl and alkynyl. Particularly preferred are O(CH2)n OmCH3, O(CH2)n, OCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2 and O(CH2nON(CH2)nCH3)2, wherein n and m can be from 1 to about 10. Other preferred oligonucleotides comprise one of the following at the 2' position: C to CO, lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkylaryl or O- Aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkylaryl, aminoalkylamine, poly An alkylamino group, a substituted alkylene group, an RNA cleavage group, a reporter group, an intercalator, a group that modifies the pharmacokinetic properties of the oligonucleotide, or a group that improves the pharmacodynamic properties of the oligonucleotide And other substituents having similar properties. A preferred modification comprises 2'-methoxyethoxy (2'-O-CH2CH2OCH3, also known as 2'-O-(2-methoxyethyl) or 2'-MOE), ie alkoxy Alkoxy group. Another preferred modification comprises a 2'-dimethylaminooxyethoxy group, that is, an O(CH2)2ON(CH3)2 group, also known as 2'-DMAOE, as described in the Examples herein below; 2'-Dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAEOE), ie 2'-O- CH2-O-CH2-N (CH2)2. Other preferred modifications include 2'-methoxy (2'-O CH3), 2'-aminopropoxy (2'-O CH2CH2CH2NH2) and 2'-fluoro (2'-F). It can also be located at other positions on the oligonucleotide, specifically at the 3' end nucleotide or in the 2'-5' linked oligonucleotide, at the 3' position of the sugar and at the 5' end nucleotide 5 Similar modifications were made at the location. The oligonucleotide may also have a sugar mimetic, such as a cyclobutyl moiety in place of the pentofuranosyl sugar. Representative US patents for the preparation of such modified sugar structures include, but are not limited to, U.S. Patent Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; Nos. 5, 514, 785; 5, 519, 134; 5, 567, 811; 5, 576, 427; 5, 591, 722; 5, 597, 909; 5, 610, 300; 5, 627, 053; 5, 639, 873; 5, 646, 265; 5, 658, 873; 5, 670, 633; No. 5,700,920, each of which is incorporated herein by reference. Oligonucleotides may also contain modifications or substitutions of nucleobases (often referred to in the art as "bases"). As used herein, "unmodified" or "natural" nucleotides include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil. (U). The modified nucleotide comprises other synthetic and natural nucleotides, such as 5-methylcytosine (5-me-C); 5-hydroxymethylcytosine; xanthine; hypoxanthine; 2-aminoglycan嘌呤; 6-methyl and other alkyl derivatives of adenine and guanine; 2-propyl and other alkyl derivatives of adenine and guanine; 2-thiouracil; 2-thiothymidine and 2- Thiocytosine; 5-halouracil and 5-halocytosine; 5-propynyl uracil and 5-propynyl cytosine; 6-azouracil, 6-azocytosine and 6-azo Thymine; 5-uracil (pseudouracil); 4-thiouracil; 8-halo, 8-amino, 8-thiol, 8-sulfanyl, 8-hydroxy and other 8-substituted adenines and birds嘌呤; 5-halo (specifically 5-bromine), 5-trifluoromethyl and other 5-substituted uracils and cytosines; 7-methylquanine and 7-methyladenine; 8-Azaguanine and 8-azadenine; 7-deazaguanine and 7-deaza adenine, 3-deazaguanine and 3-deaza adenine. In addition, the nucleotides include the nucleotides disclosed in U.S. Patent No. 3,687,808, "The Concise Encyclopedia of Polymer Science And Engineering", pp. 858-859, Kroschwitz, JI, John Wiley & Sons, 1990, Englisch, et al. People, "Angewandle Chemie, International Edition", 1991, 30, p. 613, and Sanghvi, YS, Chapter 15, "Antisense Research and Applications", pp. 289-302, Crooke, ST and Lebleu, B. CRC Press, 1993. Certain of these nucleotides are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted indole, including 2-aminopropyl adenine, 5-propynyl uracil and 5-propynyl Pyrimidine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2 ° C (Sanghvi, YS, Crooke, ST and Lebleu, B. ed., "Antisense Research and Applications", CRC Press, Boca Raton, 1993 , pp. 276-278) and presently preferred base substitutions, even more particularly preferred when combined with 2'-O methoxyethyl sugar modifications. Representative US patents that teach the preparation of the above-described modified nucleotides and other modified nucleotides include, but are not limited to, U.S. Patent Nos. 3,687,808 and 4,845,205; 5,130,302; 5,134,066; 5,175,273 No. 5, 367, 266; 5, 432, 272; 5, 457, 187; 5, 459, 255; 5, 484, 908; 5, 502, 177; 5, 525, 711, 5, 552, 540; 5, 587, 469; 5, 596, 091; 5, 614, 617; 5, 750, 692; No. 5,681,941, each of which is incorporated herein by reference. Another modification of the oligonucleotides of the invention involves chemically linking one or more portions or conjugates of the enhanced oligonucleotide, cell distribution or cellular uptake to the oligonucleotide. Such moieties include, but are not limited to, lipid moieties (such as cholesterol moieties), cholic acid, thioethers (such as hexyl-S-trityl mercaptan), thiocholesterol, aliphatic chains (eg, dodecanediol) Or eleven base residues), phospholipids (eg, di-hexadecyl-racemic-glycerol or 1,2-di-O-hexadecyl-racemic-glyceryl-3-H-phosphonic acid triethyl) Alkyl ammonium), polyamine or polyethylene glycol chain, or adamantane acetic acid, palmityl moiety, or octadecylamine or hexylamino-carbonyl-t oxycholesterol moiety. Representative US patents for the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Patent Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538 [5,578,717; 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4, 605, 044; 4, 605, 735; 4, 667, 025; 4, 762, 779; 4, 789, 737; 4, 824, 941; 4, 835, 263; 4, 876, 335; 4, 904, 582; 4, 958, 013; 5, 082, 830; 5, 112, 963; 5, 214, 136 , 5, 082, 830; 5, 112, 963; 5, 214, 136; 5, 245, 022; 5, 254, 469; 5, 258, 506; 5, 262, 536; 5, 272, 250; 5, 292, 873; 5, 317, 098; 5, 371, 241; 5, 391, 723; 5, 416, 203; 5, 451, 463; 5, 510, 475; 5, 512, 667; 5, 514, 785; 5, 565, 552; 567,810; 5, 574, 142; 5, 585, 481; 5, 587, 371; 5, 595, 726; 5, 597, 696; 5, 599, 923; 5, 599, 928 and 5, 688, 941, each of which is incorporated herein by reference.Drug discovery: The compounds of the invention are also useful in the field of drug discovery and target validation. The present invention includes efforts to clarify the relationship between voltage-gated sodium channel alpha subunit (SCNA) polynucleotides and disease states, phenotypes or conditions using the compounds identified herein and preferred target segments. The drug. Such methods comprise detecting or modulating a SCNA polynucleotide comprising contacting a sample, tissue, cell or organism with a compound of the invention, and measuring the nucleic acid or protein content and/or correlation of the SCNA polynucleotide at a time after treatment. Phenotype or chemical endpoint, and compare measurements to untreated samples or samples treated with another compound of the invention, as appropriate. Such methods can also be performed in parallel or in combination with other assays to determine the function of an unknown gene for a target validation method or to determine the effectiveness of a particular gene product as a target for treating or preventing a particular disease, condition or phenotype.Assess gene expression up-regulation or inhibition: The transfer of an exogenous nucleic acid into a host cell or organism can be assessed by directly detecting the presence of the nucleic acid in the cell or organism. This detection can be achieved by several methods well known in the art. For example, the presence of an exogenous nucleic acid can be detected by Southern blot analysis or by polymerase chain reaction (PCR) techniques using primers that specifically amplify nucleic acid-related nucleotide sequences. The performance of exogenous nucleic acids can also be measured using conventional methods including gene expression analysis. For example, mRNA produced by exogenous nucleic acids can be detected and quantified using Northern blot analysis and reverse transcription PCR (RT-PCR). The RNA expressed by the exogenous nucleic acid can also be detected by measuring the activity of the enzyme or reporting the activity of the protein. For example, antisense modulating activity can be indirectly measured as a decrease or increase in the performance of a target nucleic acid, which indicates that the exogenous nucleic acid is producing an effector RNA. Based on sequence conservation, primers can be designed and used to amplify coding regions of a target gene. Initially, the most highly expressed coding region for each gene can be used to construct a model control gene, but any coding or non-coding region can be used. Each control gene is assembled by inserting each coding region between the reporter region and its poly (A) signal. These plastids will produce mRNA with a reporter gene in the upstream portion of the gene and a potential RNAi target in the 3' non-coding region. The utility of individual antisense oligonucleotides will be assayed by modulation of the reporter gene. Reporter genes suitable for use in the methods of the invention include acetamyl hydroxyacid synthase (AHAS), alkaline phosphatase (AP), beta galactosidase (LacZ), beta glucuronidase (GUS), and chloramphenicol Chloramphenicol acetyltransferase (CAT), green fluorescent protein (GFP), red fluorescent protein (RFP), yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), horseradish Horseradish peroxidase (HRP), luciferase (Luc), nopaline synthase (NOS), octopine synthase (OCS) and its derivatives. A variety of anesthetic (ampicillin), bleomycin, chloramphenicol, gentamycin, hygromycin, kanamycin, lincomycin Selectable markers for resistance to methotrexate, phosphinothricin, puromycin, and tetracycline are available. Methods for determining modulation of a reporter gene are well known in the art and include, but are not limited to, fluorescence assays (e.g., fluorescence spectrometry, fluorescence activated cell sorting (FACS), fluorescent microscopy). (Technology), antibiotic resistance is determined. Target nucleic acid segments can also be detected in cell-based assays. Experiments were performed to detect Scn1a natural antisense BG724147 in HepG2, primary human fibroblasts with Dravet syndrome-related mutations, and human testis. For HepG2 and primary human fibroblasts with Dravid syndrome-associated mutations, cells are grown and RNA is extracted. For human testicles, RNA is isolated and purchased using polyA. This experiment was called RACE (rapid amplification of the cDNA ends) and specific primers for the BG724147 RNA transcript were used. A very similar PCR product was detected in RNA isolated from polyA isolated from HepG2 and polyA isolated from primary human fibroblasts with Dravid syndrome-related mutations but this product was not isolated from polyA from human testis Detected in RNA. Furthermore, the PCR product was not detected (or at a very low level) in total RNA from HepG2 cells and total RNA from primary human fibroblasts with Dravid syndrome-associated mutations. The results indicate that the natural antisense of Scn1a, designated BG724147, is present in HepG2 cells and primary human fibroblasts with mutations associated with Dravid syndrome but not in human testis. SCNA protein and mRNA expression can be assayed using methods known to those skilled in the art and described elsewhere herein. For example, an immunoassay such as ELISA can be used to measure protein content. The SCNA ELISA assay kit is commercially available, for example, from R&D Systems (Minneapolis, MN). In an embodiment, SCNA expression (eg, mRNA or protein) in a sample (eg, an in vivo or ex vivo cell or tissue) treated with an antisense oligonucleotide of the invention is performed by SCNA expression in a control sample. Compare to evaluate. For example, the performance of a protein or nucleic acid can be compared to the performance of a protein or nucleic acid in a simulated or untreated sample using methods known to those skilled in the art. Alternatively, it may be compared to a sample treated with a control antisense oligonucleotide (eg, an antisense oligonucleotide having altered or different sequences) depending on the desired information. In another embodiment, the difference in performance of the SCNA protein or nucleic acid in the treated sample relative to the untreated sample may be relative to the untreated sample, different nucleic acids in the treated sample (including any deemed appropriate by the researcher) Differences in performance of standards, such as housekeeping genes, were compared. The observed differences can be expressed as needed, for example in the form of ratios or fractions, for comparison with controls. In an embodiment, the amount of SCNA mRNA or protein in the sample treated with the antisense oligonucleotide of the invention is increased or decreased from about 1.25 fold to about 10 fold or 10 relative to the untreated sample or the control nucleic acid treated sample. More than double. In embodiments, the amount of SCNA mRNA or protein is increased or decreased by at least about 1.25 fold, at least about 1.3 fold, at least about 1.4 fold, at least about 1.5 fold, at least about 1.6 fold, at least about 1.7 fold, at least about 1.8 fold, at least About 2 times, at least about 2.5 times, at least about 3 times, at least about 3.5 times, at least about 4 times, at least about 4.5 times, at least about 5 times, at least about 5.5 times, at least about 6 times, at least about 6.5 times, at least About 7 times, at least about 7.5 times, at least about 8 times, at least about 8.5 times, at least about 9 times, at least about 9.5 times, or at least about 10 times or more.Kits, research reagents, diagnostics, and therapeutics The compounds of the invention are useful in the diagnosis, treatment, and prevention, and can be used as components of research reagents and kits. Furthermore, antisense oligonucleotides capable of inhibiting gene expression with strong specificity are often used by the general practitioner to elucidate the function of a particular gene or to distinguish the function of each member of the biological pathway. For use in kits and diagnostics and various biological systems, the compounds of the invention, alone or in combination with other compounds or therapeutic agents, are useful as tools in differential analysis and/or combinatorial analysis to elucidate expression in cells and tissues. The expression pattern of one part of the gene or the entire complementary sequence. As used herein, the term "biological system" or "system" is defined to mean any organism, cell, cell culture or tissue that exhibits or enables the ability to express a voltage-gated sodium ion channel alpha subunit (SCNA) gene product. These include, but are not limited to, humans, transgenic animals, cells, cell cultures, tissues, xenografts, grafts, and combinations thereof. As a non-limiting example, a pattern of expression in a cell or tissue treated with one or more antisense compounds is compared to a control cell or tissue that has not been treated with an antisense compound and the resulting pattern is analyzed to obtain a difference in gene expression, Because it relates to, for example, the disease correlation, signal transduction pathway, cellular localization, amount of expression, size, structure or function of the gene being examined. Such analysis can be performed on stimulated or unstimulated cells in the presence or absence of other compounds that affect the performance profile. Examples of gene expression analysis methods known in the art include DNA arrays or microarrays, serial analysis of gene expression (SAGE), and restriction enzyme amplification of digested cDNAs (restriction enzyme amplification of digested cDNAs). ;READS), total gene expression analysis (TOGA), protein array and proteomic research, expressed sequence tag (EST) sequencing, subtractive RNA fingerprinting (SuRF), Differential display, differential display (DD), comparative genomic hybridization, fluorescence in situ hybridization (FISH) and mass spectrometry. The compounds of the invention are useful in research and diagnostics because such compounds hybridize to nucleic acids encoding voltage-gated sodium ion channel alpha subunits (SCNA). For example, oligonucleotides that hybridize as effective SCNA modulators under such conditions as disclosed herein are effective primers or probes, respectively, under conditions conducive to gene amplification or detection. Such primers and probes are useful in methods that require specific detection of nucleic acid molecules encoding SCNA and in other studies for amplifying such nucleic acid molecules to detect SCNA or for SCNA. Hybridization of an antisense oligonucleotide of the invention, in particular a primer and probe, to a nucleic acid encoding SCNA can be detected by means known in the art. Such means may include binding the enzyme to the oligonucleotide, radiolabeling the oligonucleotide or any other suitable means of detection. Kits using such detection means for detecting the amount of SCNA in a sample can also be prepared. The specificity and sensitivity of antisense are also utilized by those skilled in the art to achieve therapeutic use. Antisense compounds have been used as therapeutic moieties in the treatment of disease states including animals in humans. Antisense oligonucleotide drugs have been safely and effectively administered to humans and numerous clinical trials are currently underway. It is thus determined that the antisense compound can be a suitable therapeutic modality that can be configured to be useful in the treatment of cells, tissues, and animals, particularly humans. For treatment, an animal suspected of having a disease or condition treatable by modulating the performance of a SCNA polynucleotide is preferred for administration by administering an antisense compound of the invention. For example, in one non-limiting embodiment, the method comprises the step of administering to a subject in need of treatment a therapeutically effective amount of a SCNA modulator. The SCNA modulators of the invention are effective in modulating the activity of SCNA or modulating the performance of SCNA proteins. In one embodiment, the activity or performance of SCNA in an animal is inhibited by about 10% compared to a control. Preferably, the activity or performance of SCNA in an animal is inhibited by about 30%. More preferably, SCNA inhibits activity or performance in animals by 50% or more. Thus, the oligomeric compound modulates the performance of the voltage-gated sodium ion channel alpha subunit (SCNA) mRNA by at least 10%, at least 50%, at least 25%, at least 30%, at least 40%, at least 50%, compared to the control, At least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99% or 100%. In one embodiment, the activity or performance of a voltage-gated sodium ion channel alpha subunit (SCNA) in an animal is increased by about 10% compared to a control. Preferably, the activity or performance of SCNA in an animal is increased by about 30%. More preferably, the activity or performance of SCNA in animals is increased by 50% or more. Thus, the oligomeric compound modulates the performance of SCNA mRNA by at least 10%, at least 50%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75% compared to the control. At least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99% or 100%. For example, a decrease in the performance of a voltage-gated sodium ion channel alpha subunit (SCNA) in serum, blood, adipose tissue, liver, or any other body fluid, tissue, or organ of an animal can be measured. Preferably, the cells contained in the fluid, tissue or organ analyzed comprise a nucleic acid molecule encoding the SCNA peptide and/or the SCNA protein itself. The compounds of the present invention can be utilized in pharmaceutical compositions by the addition of an effective amount of the compound to a pharmaceutically acceptable diluent or carrier. The use of the compounds and methods of the invention may also be applied prophylactically.Conjugate Another modification of the oligonucleotides of the invention involves chemically linking one or more portions or conjugates of the enhanced oligonucleotide, cell distribution or cellular uptake to the oligonucleotide. Such moieties or combinations may include a binding group covalently bonded to a functional group such as a primary or secondary hydroxyl group. The binding groups of the present invention include intercalating agents, reporter molecules, polyamines, polyamines, polyethylene glycols, polyethers, groups which enhance the pharmacodynamic properties of oligomers and enhance the pharmacokinetic properties of oligomers. The group. Typical binding groups include cholesterol, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, luciferin, rhodamine, coumarin, and dyes. In the context of the present invention, groups that enhance pharmacodynamic properties include groups that improve uptake, enhance resistance to degradation, and/or enhance sequence-specific hybridization to a target nucleic acid. In the context of the present invention, groups which enhance the pharmacokinetic properties include groups which improve the uptake, distribution, metabolism or excretion of the compounds of the invention. A representative binding group is disclosed in International Patent Application No. PCT/US92/09196, filed on Oct. 23, 1992, and U.S. Pat. Binding moieties include, but are not limited to, lipid moieties (such as cholesterol moieties), cholic acid, thioethers (such as hexyl-5-trityl mercaptan), thiocholesterol, aliphatic chains (such as dodecanediol or Eleven base residues), phospholipids (eg, di-hexadecyl-racemic-glycerol or 1,2-di-O-hexadecyl-racemic-glyceryl-3-Hphosphonic acid triethylammonium) a polyamine or polyethylene glycol chain, or adamantane acetic acid, a palmitoyl moiety, or an octadecylamine or hexylamino-carbonyl-hydroxycholesterol moiety. The oligonucleotide of the present invention may also be combined with an active drug substance such as aspirin, warfarin, phenylbutazone, ibuprofen, Suprofen, fenbufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansyl sarcosine ( Dansylsarcosine), 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, benzothiadiazide, chlorothiazide, diazepine (diazepine), indomethicin, barbiturate, cephalosporin, sulfa drugs, antidiabetic agents, antibacterial agents or antibiotics. Representative US patents for the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Patent Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538 [5,578,717; 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4, 605, 044; 4, 605, 735; 4, 667, 025; 4, 762, 779; 4, 789, 737; 4, 824, 941; 4, 835, 263; 4, 876, 335; 4, 904, 582; 4, 958, 013; 5, 082, 830; 5, 112, 963; 5, 214, 136 , No. 5, 082, 830; 5, 317, 098; 5, 371, 241; 5, 391, 723; 5, 416, 203; 5, 451, 463; 5, 510, 475; Nos. 5, 514, 785; 5, 565, 552; 5, 574, 142; 5, 585, 481; 5, 587, 371;Formulation The compounds of the invention may also be incorporated, encapsulated, with other molecules, molecular structures or mixtures of compounds, such as liposomes, receptor targeting molecules, oral formulations, rectal formulations, topical formulations or other formulations. Binding or otherwise association to aid ingestion, distribution, and/or absorption. Representative US patents for the preparation of such formulations which facilitate ingestion, distribution and/or absorption include, but are not limited to, U.S. Patent Nos. 5,108,921; 5,354,844; 5,416,016; 5,459,127; 5,521,291; No. 5, 543, pp; Nos. 5,416,016; 5,417,978; 5,462,854; 5,469,854; 5,512,295; 5,527,528; 5,534,259; 5,543,152; 5,556,948; 5,580,575; and 5,595,756, each patent The manner of reference is incorporated herein. Although antisense oligonucleotides need not be administered in the context of a vector to modulate target expression and/or function, embodiments of the invention pertain to expression vector constructs for use in expressing antisense oligonucleotides, including The promoter, hybridization initiation gene sequence and has strong constitutive promoter activity or promoter activity that can be induced if desired. In one embodiment, the practice of the invention involves administering at least one of the foregoing antisense oligonucleotides in a suitable nucleic acid delivery system. In one embodiment, the system includes a non-viral vector operably linked to a polynucleotide. Examples of such non-viral vectors include oligonucleotides alone (e.g., any one or more of SEQ ID NOS: 29-94) or oligonucleotides in combination with suitable protein, polysaccharide or lipid formulations. Further suitable nucleic acid delivery systems include viral vectors, the sequences of which are typically derived from adenovirus, adeno-associated virus (AAV), helper virus-dependent adenovirus, retrovirus or hemagglutinatin virus of Japan-liposome (hemagglutinatin virus of Japan-liposome, At least one of the HVJ) complexes. Preferably, the viral vector comprises a strong eukaryotic promoter, such as a cytomegalovirus (CMV) promoter operably linked to a polynucleotide. Further preferred vectors include viral vectors, fusion proteins, and chemical conjugates. Retroviral vectors include Moloney murine leukemia viruses and HIV-based viruses. A preferred HIV-based viral vector comprises at least two vectors, wherein the gag and pol genes are derived from the HIV genome and the env gene is derived from another virus. A DNA viral vector is preferred. Such vectors include vaccinia vectors, such as acne or avian pox vectors; herpesvirus vectors, such as herpes simplex I virus (HSV) vectors; adenoviral vectors and adeno-associated viral vectors. The antisense compound of the present invention encompasses any pharmaceutically acceptable salt, ester or salt of such an ester, or is capable of providing (directly or indirectly) a biologically active metabolite or residue thereof when administered to an animal including humans. Any other compound. The term "pharmaceutically acceptable salt" refers to a physiologically and pharmaceutically acceptable salt of a compound of the invention: that is, a salt which retains the desired biological activity of the parent compound and which does not impart an undesirable toxicological effect. For oligonucleotides, preferred examples of pharmaceutically acceptable salts and their use are further described in U.S. Patent No. 6,287,860, incorporated herein by reference. The invention also includes pharmaceutical compositions and formulations comprising the antisense compounds of the invention. The pharmaceutical compositions of the present invention can be administered in a number of ways, depending on the local or systemic treatment desired and the area to be treated. Administration may be topical (including ophthalmic and mucosal administration, including vaginal and rectal delivery); pulmonary administration (eg, by inhalation or insufflation of a powder or aerosol, including the use of a nebulizer; intratracheal administration; intranasal administration; Transdermal administration and transdermal administration); oral administration or parenteral administration. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, such as intrathecal or intraventricular administration. For the treatment of tissues in the central nervous system, administration can be carried out by, for example, injection or infusion into the cerebrospinal fluid. The administration of antisense RNA to the cerebrospinal fluid is described, for example, in U.S. Patent Application Publication No. 2007/0117772, the entire disclosure of which is incorporated herein by reference. When the antisense oligonucleotide of the present invention is intended to be administered to cells in the central nervous system, it can be administered with one or more agents capable of promoting the penetration of the target antisense oligonucleotide across the blood-brain barrier. Injection can be performed, for example, in an entorhinal cortex or hippocampus. The delivery of a neurotrophic factor by administering an adenoviral vector to a motor neuron in muscle tissue is described in, for example, U.S. Patent No. 6,632,427, "Adenoviral-vector-mediated gene transfer into medullary motor neurons", which is incorporated by reference. Incorporated herein. Direct delivery to the brain, such as the striatum, thalamus, hippocampus or substantia nigra, is known in the art and is described, for example, in U.S. Patent No. 6,756,523, "Adenovirus vectors For the transfer of foreign genes into cells of the central nervous system particularly in brain, the patent is incorporated herein by reference. When administered by injection, the administration can be rapid, or when the formulation is slowly infused or administered by slow infusion, the administration can take a period of time. The subject antisense oligonucleotide can also be linked or associated with an agent that will provide the desired pharmaceutical or pharmacodynamic properties. For example, an antisense oligonucleotide can be coupled to any substance known in the art to facilitate penetration or transport across the blood-brain barrier, such as an antibody to a transferrin receptor, and by intravenous The injection is administered. An antisense compound can be linked to, for example, a viral vector that renders the antisense compound more effective and/or increases the transport of the antisense compound across the blood-brain barrier. Infiltration of blood-brain barriers can also be accomplished by, for example, infusion of sugars including, but not limited to, meso erythritol, xylitol, D(+) galactose, D(+) lactose. , D(+) xylose, dulcitol, myo-inositol, L(-) fructose, D(-) mannitol, D(+) glucose, D(+) arabinose ( D(+) arabinose), D(-)arabinose, cellobiose, D(+) maltose, D(+) raffinose (D(+) raffinose), L(+) rhamnose (L(+) ) rhamnose), D(+) melibiose (D(+) melibiose), D(-) ribose, adonitol, D(+) arabitol, L(-) arabitol, D(+ ) trehalose (D(+) fucose), L(-) trehalose, D(-) to threose (D(-) lyxose), L(+) to threose and L(-) to threose; or Amino acids including, but not limited to, glutamic acid, lysine, arginine, aspartame, aspartic acid, cysteine, glutamic acid, glycine, histidine, It is achieved by leucine, methionine, phenylalanine, valine, serine, threonine, tyrosine, proline and taurine. Methods and materials for enhancing blood-brain barrier penetration are described in, for example, U.S. Patent No. 4,866,042, "Method for the delivery of genetic material across the blood brain barrier", No. 6,294,520, "Material for passage through the blood-brain barrier", And U.S. Patent No. 6,936, 589, the entire disclosure of each of which is incorporated herein by reference. The subject antisense compound can be incorporated, encapsulated, with other molecules, molecular structures or mixtures of compounds, such as liposomes, receptor targeting molecules, oral formulations, rectal formulations, topical formulations or other formulations. Binding or otherwise association to aid ingestion, distribution, and/or absorption. For example, cationic lipids can be included in the formulation to facilitate oligonucleotide uptake. One such composition which is shown to promote uptake is LIPOFECTIN (available from GIBCO-BRL, Bethesda, MD). Oligonucleotides having at least one 2'-O-methoxyethyl modification are particularly suitable for oral administration. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids, and powders. Conventional pharmaceutical carriers, aqueous bases, powder bases or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be suitable. The pharmaceutical formulations of the present invention, which are preferably provided in unit dosage form, can be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing the active ingredient into association with a pharmaceutical carrier or excipient. In general, formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and, if necessary, shaping the product. The compositions of the present invention may be formulated into any of a wide variety of possible dosage forms such as, but not limited to, lozenges, capsules, gel capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media. The aqueous suspensions may additionally contain materials which increase the viscosity of the suspension, including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers. Pharmaceutical compositions of the invention include, but are not limited to, solutions, emulsions, foams, and liposome-containing formulations. The pharmaceutical compositions and formulations of the present invention may comprise one or more penetration enhancers, carriers, excipients or other active or inactive ingredients. The emulsion is typically a heterogeneous system in the form of droplets of a liquid diameter typically exceeding 0.1 μm dispersed in another liquid. The emulsion may contain other components in addition to the dispersed phase and the solution which may be present in the aqueous phase, the oil phase, or the active drug itself present in the separate phase. Microemulsions are included as an embodiment of the invention. Emulsions and their use are well known in the art and are further described in U.S. Patent No. 6,287,860. Formulations of the invention include liposome formulations. As used in the present invention, the term "liposome" means a vesicle composed of two pherophilic lipids arranged in a spherical bilayer. Liposomes are single or multi-layered vesicles having a film formed from a lipophilic material and an aqueous interior containing the composition to be delivered. Cationic liposomes are positively charged liposomes that interact with negatively charged DNA molecules to form stable complexes. Salt-sensitive or negatively charged liposomes enticle DNA rather than complex with it. Both cationic liposomes and non-cationic liposomes have been used to deliver DNA to cells. Liposomes also include "sterically stabilized" liposomes, as the term is used herein to refer to liposomes comprising one or more specialized lipids. When incorporated into liposomes, such specialized lipids produce liposomes with increased cycle life relative to liposomes lacking such specialized lipids. Examples of sterically stabilized liposomes are liposomes: a portion of the liposome-forming lipid portion of the liposome comprising one or more glycolipids or partially derivatized with one or more hydrophilic polymers, such as polyethylene glycol (PEG) . Liposomes and their use are further described in U.S. Patent No. 6,287,860. The pharmaceutical formulations and compositions of the present invention may also include a surfactant. The use of surfactants in pharmaceuticals, formulations and emulsions is well known in the art. Surfactants and their use are further described in U.S. Patent No. 6,287,860, incorporated herein by reference. In one embodiment, the invention employs various permeation enhancers to efficiently deliver nucleic acids, in particular oligonucleotides. In addition to helping non-lipophilic drugs to diffuse across the cell membrane, the penetration enhancer also enhances the permeability of the lipophilic drug. Penetration enhancers can be classified into one of five categories: surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants. Permeation enhancers and their use are further described in U.S. Patent No. 6,287,860, the disclosure of which is incorporated herein by reference. Those skilled in the art will recognize that the formulation is designed in a conventional manner in accordance with its intended use, i.e., the route of administration. Preferred formulations for topical administration include formulations in which the oligonucleotides of the invention are admixed with topical delivery agents such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents, and surfactants. Preferred lipids and liposomes include neutral (eg, dioleoyl-phospholipid oxime ethanolamine DOPE, dimyristoyl phosphatidylcholine DMPC, distearyl phosphatidylcholine), negatively charged (e.g., dimyristylphospholipid glycerol DMPG) and cationic (e.g., dinonyltetramethylammonium propyl DOTAP and dioleyl-phosphothioguanylethanolamine DOTMA). For topical or other administration, the oligonucleotides of the invention may be encapsulated within a liposome or may form a complex with a liposome, in particular with a cationic liposome. Alternatively, the oligonucleotide may be complexed with a lipid, in particular with a cationic lipid. Preferred fatty acids and esters, pharmaceutically acceptable salts thereof, and their use are further described in U.S. Patent No. 6,287,860. Compositions and formulations for oral administration include powders or granules, microparticles, nanoparticles, suspensions or solutions in water or non-aqueous medium, capsules, gel capsules, sachets, lozenges Or a small lozenge. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable. Preferred oral formulations are formulations of the oligonucleotides of the invention administered with one or more penetration enhancers, surfactants, and chelating agents. Preferred surfactants include fatty acids and/or esters or salts thereof, bile acids and/or salts thereof. Preferred bile acids/salts and fatty acids and their use are further described in U.S. Patent No. 6,287,860, incorporated herein by reference. Combinations of penetration enhancers, such as fatty acid/salts and bile acids/salts, are also preferred. A particularly preferred combination is the sodium salt of lauric acid, citric acid and UDCA. Other penetration enhancers include polyoxyethylene-9-lauryl ether, polyoxyethylene-20-hexadecyl ether. The oligonucleotides of the invention may be delivered orally in particulate form (including spray dried particles) or by complex formation of microparticles or nanoparticles. Oligonucleotide complexes and their use are further described in U.S. Patent No. 6,287,860, incorporated herein by reference. Compositions and formulations for parenteral, intrathecal or intrapharmaceutical administration may also include buffers, diluents, and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds, and other pharmaceutically acceptable agents. A sterile aqueous solution of the carrier or excipient is accepted. Certain embodiments of the invention provide pharmaceutical compositions comprising one or more oligomeric compounds and one or more other chemotherapeutic agents that act by a non-antisense mechanism. Examples of such chemotherapeutic agents include, but are not limited to, cancer chemotherapeutic drugs such as daunorubicin, daunomycin, dactinomycin, cranberry (doxorubicin), Epirubicin, idarubicin, esorubicin, bleomycin, mafosfamide, ifosfamide, arsenic Cytosine arabinoside, bischloroethyl-nitrosurea, busulfan sulfate, mitomycin C, actinomycin D, light god Mithramycin, prednisone, hydroxyprogesterone, testosterone, tamoxifen, dacarbazine, procarbazine, hexamethyl Hexamethylmelamine, pentamethyl melamine, mitoxantrone, amsacrine, chlorambucil, methylcyclohexyl nitrosourea, nitrogen mustard, beauty Melphalan, cyclophosphamide, 6-mercaptopurine, 6-thio bird呤, cytarabine, 5-azacytidine, hydroxyurea, deoxycoformycin, 4-hydroxyperoxyguanamine, 5-fluorouracil (5-FU), 5- Fluorodeoxyuridine (5-FUdR), methotrexate (MTX), colchicine, taxol, vincristine, vinblastine, etoposide (VP-) 16), trimetrexate, irinotecan, topotecan, gemcitabine, teniposide, cisplatin and diethylstilbestrol (DES) . When used with a compound of the invention, such chemotherapeutic agents can be used individually (eg, 5-FU and oligonucleotides), sequentially (eg, 5-FU and oligonucleotides for a period of time, followed by MTX and oligos) Nucleotides) or in combination with one or more other such chemotherapeutic agents (eg, 5-FU, MTX and oligonucleotides, or 5-FU, radiation therapy, and oligonucleotides). Anti-inflammatory drugs may also be combined in the compositions of the present invention, including but not limited to non-steroidal anti-inflammatory drugs and corticosteroids; and antiviral drugs including, but not limited to, ribivirin, vidarabine, Acyclovir and ganciclovir. Combinations of antisense compounds with other non-antisense drugs are also within the scope of the invention. Compounds of two or more combinations may be used together or sequentially. In another related embodiment, the compositions of the invention may contain one or more antisense compounds that target the first nucleic acid, in particular, oligonucleotides; and one or more other antisense compounds that target the second nucleic acid target . For example, the first target can be a specific antisense sequence of a voltage-gated sodium ion channel alpha subunit (SCNA), and the second target can be a region from another nucleotide sequence. Alternatively, the compositions of the invention may contain two or more antisense compounds that target different regions of the same voltage-gated sodium ion channel alpha subunit (SCNA) nucleic acid target. Numerous examples of antisense compounds are described herein and other examples can be selected from suitable compounds known in the art. Compounds of two or more combinations may be used together or sequentially.Dosing: The formulation of the Xianxin therapeutic composition and its subsequent administration (administration) are within the skill of the relevant personnel in the art. Administration depends on the severity and responsiveness of the condition being treated, wherein the course of treatment lasts from several days to several months, or until the condition is cured or attenuated. The optimal dosing schedule can be calculated from the cumulative measurement of the drug in the patient. The average dosage, method of administration, and repetition rate can be readily determined by one of ordinary skill. The optimal dose will vary depending on the relative potency of the individual oligonucleotides and can generally be estimated based on the EC50 considered to be effective in both in vitro and in vivo animal models. Generally, the dosage is from 0.01 μg to 100 mg per kilogram of body weight and may be administered once or more daily, weekly, monthly or yearly, or even every 2 to 20 years. One of ordinary skill can readily estimate the repetition rate for administration based on measuring the residence time and concentration of the drug in body fluids or tissues. After successful treatment, it may be necessary to subject the patient to maintenance therapy to prevent recurrence of the disease state, with a maintenance dose ranging from 0.01 μg to 100 mg per kilogram of body weight, once or more once a day to once every 20 years. Nucleotide. In an embodiment, the patient is treated with a dose of at least about 1 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 6 mg, at least about 7 mg per kilogram of body weight, At least about 8 mg, at least about 9 mg, at least about 10 mg, at least about 15 mg, at least about 20 mg, at least about 25 mg, at least about 30 mg, at least about 35 mg, at least about 40 mg, at least about 45 mg, At least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, or at least about 100 mg. Some of the injectable doses of the antisense oligonucleotides are described, for example, in U.S. Patent No. 7,563,884, the disclosure of which is incorporated herein by reference. While the various embodiments of the invention have been described, Numerous variations of the disclosed embodiments can be made without departing from the spirit and scope of the invention. Therefore, the breadth and scope of the invention should not be limited by any of the embodiments described above. All documents mentioned herein are hereby incorporated by reference. All publications and patent documents cited in this application are hereby incorporated by reference in their entirety in their entirety in the extent of the disclosures in To the extent that the various references are cited in this document, the Applicant does not recognize that any particular reference is the "pre-technical" of the invention. Examples of the compositions and methods of the present invention are illustrated in the following examples.Instance The following non-limiting examples are illustrative of selected embodiments of the invention. It is to be understood that the changes in the proportions and the alternatives to the elements of the invention are obvious to those skilled in the art and are within the scope of the embodiments of the invention.Instance 1 : Designed for voltage-gated sodium ion channels α Subunit (SCNA) Antisense nucleic acid molecule and / or SCNA The sense strand of the polynucleotide has a specific antisense oligonucleotide As indicated above, the term "oligonucleotide specific for" or "oligonucleotide targeting" refers to having (i) capable of forming a stable complex with a portion of the targeted gene, or (ii) capable of An oligonucleotide that forms a stable duplex sequence with a portion of the mRNA transcript of the targeted gene. Promote the selection of appropriate oligonucleotides by using a computer program (eg IDT AntiSense Design, IDT OligoAnalyzer) that automatically identifies the desired sequence with the desired melting temperature (usually 50-60 ° C) and the target nucleus The nucleotide sequence forms a hybrid and will not form a 19-25 nucleotide subsequence of the self-dimer or other complex secondary structure. The selection of appropriate oligonucleotides is further facilitated by the use of computer programs that automatically align nucleic acid sequences and indicate regions of homology or homology. Such programs are used to compare the resulting nucleic acid sequences, for example, by searching a library such as GenBank or by sequencing the PCR products. Comparing nucleic acid sequences from a range of genes and intergenic regions of a given genome allows selection of nucleic acid sequences that exhibit an appropriate degree of specificity for the relevant genes. Such procedures allow for the selection of oligonucleotides that exhibit a high degree of complementarity to a target nucleic acid sequence and a lower degree of complementarity to other nucleic acid sequences in a given genome. Those skilled in the art will recognize that there is a great deal of freedom in selecting the appropriate region for the genes used in the present invention. When the antisense compound binds to the target nucleic acid, it interferes with the normal function of the target nucleic acid to regulate the function and/or activity, and there is a sufficient degree of complementarity to avoid the specific binding of the antisense compound to the non-target nucleic acid sequence. The antisense compound is administered under conditions (ie, in the case of in vivo assay or therapeutic treatment, under physiological conditions, and in the case of in vitro assays, under conditions of assay), the antisense compound is "Specially hybridizable." The hybridization properties of the oligonucleotides described herein can be determined by one or more in vitro assays known in the art. For example, the properties of the oligonucleotides described herein can be obtained by measuring the binding strength between a target natural antisense and a potential drug molecule using a melting curve assay. The strength of binding between the target natural antisense and the potential drug molecule (molecule) can be estimated using any established method of measuring the strength of the intermolecular interaction, such as a melting curve assay. The melting curve assay determines the temperature at which a natural antisense/molecular complex rapidly transitions from a double-strand configuration to a single-strand configuration. This temperature is widely accepted as a reliable measure of the strength of the interaction between two molecules. The melting curve assay can be performed using a cDNA copy of the actual native antisense RNA molecule or a synthetic DNA or RNA nucleotide corresponding to the binding site of the molecule. Multiple kits are available that contain all the reagents necessary to perform this assay (eg, Applied Biosystems Inc. MeltDoctor kit). Such kits include suitable buffer solutions containing a double-stranded DNA (dsDNA) binding dye such as ABI HRM dye, SYBR Green, SYTO, and the like. The nature of the dsDNA dye is that it emits almost no fluorescence in free form, but is highly fluorescent when bound to dsDNA. For the assay, the cDNA or corresponding oligonucleotide is mixed with the molecule at a concentration determined by a particular manufacturer's protocol. The mixture was heated to 95 ° C to dissociate all preformed dsDNA complexes, then slowly cooled to room temperature or other lower temperatures as determined by the kit manufacturer to allow DNA molecules to adhere. The newly formed composite was then slowly heated to 95 ° C while continuously collecting data on the amount of fluorescence generated by the reaction. The intensity of the fluorescence is inversely proportional to the amount of dsDNA present in the reaction. Data can be collected using a real-time PCR instrument compatible with the kit (eg, ABI's StepOne Plus Instant PCR System or lightTyper instrument, Roche Diagnostics, Lewes, UK). Construct by using a suitable software (such as lightTyper (Roche) or SDS Dissociation Curve, ABI) to plot the negative derivative of fluorescence relative to temperature (on the y-axis (descent) / dT) relative to temperature (x-axis) Melting peak. The data was analyzed to identify the temperature at which the dsDNA complex rapidly transitions to a single molecule. This temperature is called Tm and is proportional to the strength of the interaction between the two molecules. Typically, the Tm will exceed 40 °C.Instance 2 : adjustment SCNA Polynucleotide Treatment with antisense oligonucleotides HepG2 cell At 37 ° C and 5% CO2 Next, make HepG2 cells from ATCC (catalog number HB-8065) in growth medium (MEM/EBSS (Hyclone catalog number SH30024, or Mediatech catalog number MT-10-010-CV) + 10% FBS (Mediatech catalog number MT35- 011-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI)) was grown. One day before the experiment, the cells were 1.5×105 The density of /ml was re-inoculated in a 6-well plate at 37 ° C and 5% CO2 Under cultivation. On the day of the experiment, the medium in the 6-well plate was changed to fresh growth medium. All antisense oligonucleotides were diluted to a concentration of 20 μM. 2 μl of this solution was incubated with 400 μl of Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μl of Lipofectamine 2000 (Invitrogen Cat. No. 11668019) for 20 minutes at room temperature and HepG2 cells were applied to each well of a 6-well plate. . A similar mixture comprising 2 μl of water instead of oligonucleotide solution was used to simulate the transfection control. At 37 ° C and 5% CO2 After 3-18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and the Promega SV Total RNA Isolation System (Cat. No. Z3105) or the RNeasy Total RNA Isolation Kit from Qiagen (Cat. No. 74181) was followed from the cells according to the manufacturer's instructions. Extract RNA. 600 ng of RNA was added to the reverse transcription reaction using the Thermo Scientific cDNA kit (Cat. No. AB1453B) or the high capacity cDNA reverse transcription kit (Cat. No. 4368813) as described in the manufacturer's protocol. The cDNA from this reverse transcription reaction was used to monitor gene expression by using ABI Taqman Gene Expression Mixture (Cat. No. 4369510) and primers/probes designed by ABI for real-time PCR (according to Applied Biosystems Inc., Foster City CA Applied) Biosystems Taqman Gene Performance Assay: Hs00374696_m1, Hs00897350_m1 or Hs00897341_m1 for human SCNA). The following PCR cycles were used: 50 °C for 2 minutes; 95 °C for 10 minutes; 40 cycles (95 °C for 15 seconds, 60 °C for 1 minute) using a StepOne Plus real-time PCR machine (Applied Biosystems). The fold change in gene expression after treatment with the antisense oligonucleotide was calculated based on the difference in the 18S corrected dCt value between the treated sample and the mock transfected sample.result: The real-time PCR results showed that the content of SCN1A mRNA in HepG2 cells was significantly increased 48 hours after treatment with SCN1A antisense BG724147 antisense oligonucleotide (Fig. 1, 4). Other oligonucleotides designed for SCN1A antisense BG724147 and Hs.662210 did not increase SCN1A content (Figures 2, 3).Instance 3 : by targeting SCNA Antisense oligonucleotide treatment of specific natural antisense transcripts to upregulate different cell lines SCNA mRNA In Example 3, antisense oligonucleotides of different chemical compositions targeting SCN1A-specific natural antisense transcripts were screened at a final concentration of 20 nM in a panel of various cell lines. The cell lines used are derived from different organs and different animal species. The following data demonstrate that up-regulation of SCN1A mRNA/protein via a function of modulating the SCN1A-specific natural antisense transcript is not limited to a single oligonucleotide, tissue or species and thus represents a general phenomenon.Materials and methods Primary human fibroblasts with mutations associated with Dravid syndrome. At 37 ° C and 5% CO2 Next, primary human skin fibroblasts with Dravid syndrome-associated mutation E1099X introduced into culture by Dr. N. Kenyon (University of Miami) were obtained from MEM (Gibco, catalog number: 12561-056) + 10 % FBS (Mediatech, catalog number: 35-015 CV) was grown in growth medium consisting of +1% antifungal antibiotic (Gibco, catalog number: 15240-062). Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in growth medium at approximately 2 x 10 days prior to the experiment.5 /The density of the wells was re-inoculated in a 6-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 6-well plate was changed to fresh growth medium (1.5 ml/well) and the cells were administered with antisense oligonucleotides. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of all oligonucleotides are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. For administration to 1 well, 2 μl of this solution was incubated with 400 μl of Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μl of Lipofectamine 2000 (Invitrogen Cat. No. 11668019) for 20 minutes at room temperature and One well of the 6-well plate of the cells was applied dropwise. A similar mixture comprising 2 μl of water instead of oligonucleotide solution was used to simulate the transfection control. In addition, the inactive oligonucleotide CUR-1462 at the same concentration was used as a control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and RNA was extracted from the cells using Promega's SV Total RNA Isolation System (catalog number Z3105) following the manufacturer's instructions. 600 ng of purified total RNA was added to the reverse transcription reaction using Invitrogen's SuperScript VILO cDNA Synthesis Kit (catalog number 11754-250) as described in the manufacturer's protocol. The cDNA from this reverse transcription reaction was used to monitor gene expression by using ABI Taqman gene expression mixture (catalog number 4369510) and primers/probes designed by ABI for real-time PCR (for Hs00374696_m1, Hs00897350_m1 or Hs00897341_m1 for human SCN1A) . The results obtained using all 3 tests are very similar. The following PCR cycles were used: 50 °C for 2 minutes; 95 °C for 10 minutes; 40 cycles (95 °C for 15 seconds, 60 °C for 1 minute) using the StepOne Plus Real-Time PCR System (Applied Biosystems). The 18S test is manufactured by ABI (catalog number 4319413E). The fold change in gene expression after treatment with the antisense oligonucleotide was calculated based on the difference in the 18S corrected dCt value between the treated sample and the mock transfected sample. For the alternative same day method, all procedures were performed similarly, but cells were dosed with antisense oligonucleotides on the first day after they were dispensed into 6-well plates.SK-N-AS Cell line . At 37 ° C and 5% CO2 Next, make SK-N-AS human neuroblastoma cells from ATCC (Catalog No. CRL-2137) in growth medium (DMEM (Mediatech catalog number 10-013-CV) + 10% FBS (Mediatech catalog number MT35-011) -CV) + Penicillin/Streptomycin (Mediatech Cat. No. MT30-002-CI) + Non-essential Amino Acid (NEAA) (HyClone SH30238.01)). Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in growth medium at approximately 3 x 10 days prior to the experiment.5 /The density of the wells was re-inoculated in a 6-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 6-well plate was changed to fresh growth medium (1.5 ml/well) and the cells were administered with antisense oligonucleotides. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of all oligonucleotides are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. For administration to 1 well, 2 μl of this solution was incubated with 400 μl of Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μl of Lipofectamine 2000 (Invitrogen Cat. No. 11668019) for 20 minutes at room temperature and One well of the 6-well plate of the cells was applied dropwise. A similar mixture comprising 2 μl of water instead of oligonucleotide solution was used to simulate the transfection control. In addition, the inactive oligonucleotide CUR-1462 at the same concentration was used as a control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and RNA was extracted from the cells using Promega's SV Total RNA Isolation System (catalog number Z3105) following the manufacturer's instructions. 600 ng of purified total RNA was added to the reverse transcription reaction using Invitrogen's SuperScript VILO cDNA Synthesis Kit (catalog number 11754-250) as described in the manufacturer's protocol. The cDNA from this reverse transcription reaction was used to monitor gene expression by using ABI Taqman gene expression mixture (catalog number 4369510) and primers/probes designed by ABI for real-time PCR (for Hs00374696_m1, Hs00897350_m1 or Hs00897341_m1 for human SCN1A) . The results obtained using all 3 tests are very similar. The following PCR cycles were used: 50 °C for 2 minutes; 95 °C for 10 minutes; 40 cycles (95 °C for 15 seconds, 60 °C for 1 minute) using the StepOne Plus Real-Time PCR System (Applied Biosystems). The 18S test is manufactured by ABI (catalog number 4319413E). The fold change in gene expression after treatment with the antisense oligonucleotide was calculated based on the difference in the 18S corrected dCt value between the treated sample and the mock transfected sample. For the alternative same day method, all procedures were performed similarly, but cells were dosed with antisense oligonucleotides on the first day after they were dispensed into 6-well plates.CHP-212 Cell line. At 37 ° C and 5% CO2 Next, make CHP-212 human neuroblastoma cells from ATCC (Catalog No. CRL-2273) in growth medium (1: MEM and F12 (ATCC catalog number 30-2003 and Mediatech catalog number 10-080-CV, respectively) The mixture was grown in +10% FBS (Mediatech catalog number MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI)). Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in growth medium at approximately 2 x 10 days prior to the experiment.5 /The density of the wells was re-inoculated in a 6-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 6-well plate was changed to fresh growth medium (1.5 ml/well) and the cells were administered with antisense oligonucleotides. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of all oligonucleotides are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. For administration to 1 well, 2 μl of this solution was incubated with 400 μl of Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μl of Lipofectamine 2000 (Invitrogen Cat. No. 11668019) for 20 minutes at room temperature and One well of the 6-well plate of the cells was applied dropwise. A similar mixture comprising 2 μl of water instead of oligonucleotide solution was used to simulate the transfection control. In addition, the inactive oligonucleotide CUR-1462 at the same concentration was used as a control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and RNA was extracted from the cells using Promega's SV Total RNA Isolation System (catalog number Z3105) following the manufacturer's instructions. 600 ng of purified total RNA was added to the reverse transcription reaction using Invitrogen's SuperScript VILO cDNA Synthesis Kit (catalog number 11754-250) as described in the manufacturer's protocol. The cDNA from this reverse transcription reaction was used to monitor gene expression by using ABI Taqman gene expression mixture (catalog number 4369510) and primers/probes designed by ABI for real-time PCR (for Hs00374696_m1, Hs00897350_m1 or Hs00897341_m1 for human SCN1A) . The results obtained using all 3 tests are very similar. The following PCR cycles were used: 50 °C for 2 minutes; 95 °C for 10 minutes; 40 cycles (95 °C for 15 seconds, 60 °C for 1 minute) using the StepOne Plus Real-Time PCR System (Applied Biosystems). The 18S test is manufactured by ABI (catalog number 4319413E). The fold change in gene expression after treatment with the antisense oligonucleotide was calculated based on the difference in the 18S corrected dCt value between the treated sample and the mock transfected sample. For the alternative same day method, all procedures were performed similarly, but cells were dosed with antisense oligonucleotides on the first day after they were dispensed into 6-well plates.Vero76 Cell line. At 37 ° C and 5% CO2 Next, make Vero76 African green monkey embryo kidney cells from ATCC (Catalog No. CRL-1587) in growth medium (Dulbecco's Modified Eagle's Medium (Cellgrow 10-013-CV) + 5 % FBS (Mediatech catalog number MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI)) was grown. Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in the growth medium at approximately 1 day prior to the experiment.5 /The density of the wells was re-inoculated in a 6-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 6-well plate was changed to fresh growth medium (1.5 ml/well) and the cells were administered with antisense oligonucleotides. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of all oligonucleotides are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. For administration to 1 well, 2 μl of this solution was incubated with 400 μl of Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μl of Lipofectamine 2000 (Invitrogen Cat. No. 11668019) for 20 minutes at room temperature and One well of the 6-well plate of the cells was applied dropwise. A similar mixture comprising 2 μl of water instead of oligonucleotide solution was used to simulate the transfection control. In addition, the inactive oligonucleotide CUR-1462 at the same concentration was used as a control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and RNA was extracted from the cells using Promega's SV Total RNA Isolation System (catalog number Z3105) following the manufacturer's instructions. 600 ng of purified total RNA was added to the reverse transcription reaction using Invitrogen's SuperScript VILO cDNA Synthesis Kit (catalog number 11754-250) as described in the manufacturer's protocol. The cDNA from this reverse transcription reaction was used to monitor gene expression by using ABI Taqman gene expression mixture (catalog number 4369510) and primers/probes designed by ABI for real-time PCR (for Hs00374696_m1, Hs00897350_m1 or Hs00897341_m1 for human SCN1A) . The following PCR cycles were used: 50 °C for 2 minutes; 95 °C for 10 minutes; 40 cycles (95 °C for 15 seconds, 60 °C for 1 minute) using the StepOne Plus Real-Time PCR System (Applied Biosystems). The 18S test is manufactured by ABI (catalog number 4319413E). The fold change in gene expression after treatment with the antisense oligonucleotide was calculated based on the difference in the 18S corrected dCt value between the treated sample and the mock transfected sample. For the alternative same day method, all procedures were performed similarly, but cells were dosed with antisense oligonucleotides on the first day after they were dispensed into 6-well plates.3T3 Cell line. At 37 ° C and 5% CO2 Next, let 3T3 mouse embryonic fibroblasts from ATCC (Catalog No. CRL-1658) in growth medium (Dellbeck modified Eagle's medium (Cellgrow 10-013-CV) + 10% fetal bovine serum (Cellgrow) Growth in 35-22-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI)). Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in the growth medium at approximately 1 day prior to the experiment.5 /The density of the wells was re-inoculated in a 6-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 6-well plate was changed to fresh growth medium (1.5 ml/well) and the cells were administered with antisense oligonucleotides. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of all oligonucleotides are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. For administration to 1 well, 2 μl of this solution was incubated with 400 μl of Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μl of Lipofectamine 2000 (Invitrogen Cat. No. 11668019) for 20 minutes at room temperature and One well of the 6-well plate of the cells was applied dropwise. A similar mixture comprising 2 μl of water instead of oligonucleotide solution was used to simulate the transfection control. In addition, the inactive oligonucleotide CUR-1462 at the same concentration was used as a control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and RNA was extracted from the cells using Promega's SV Total RNA Isolation System (catalog number Z3105) following the manufacturer's instructions. 600 ng of purified total RNA was added to the reverse transcription reaction using Invitrogen's SuperScript VILO cDNA Synthesis Kit (catalog number 11754-250) as described in the manufacturer's protocol. The cDNA from this reverse transcription reaction was used to monitor gene expression by using ABI Taqman gene expression mixture (catalog number 4369510) and primers/probes designed by ABI for real-time PCR (for Hs00374696_m1, Hs00897350_m1 or Hs00897341_m1 for human SCN1A) . The results obtained using all 3 tests are very similar. The following PCR cycles were used: 50 °C for 2 minutes; 95 °C for 10 minutes; 40 cycles (95 °C for 15 seconds, 60 °C for 1 minute) using the StepOne Plus Real-Time PCR System (Applied Biosystems). The 18S test is manufactured by ABI (catalog number 4319413E). The fold change in gene expression after treatment with the antisense oligonucleotide was calculated based on the difference in the 18S corrected dCt value between the treated sample and the mock transfected sample. For the alternative same day method, all procedures were performed similarly, but cells were dosed with antisense oligonucleotides on the first day after they were dispensed into 6-well plates.HepG2 Cell line. HepG2 human hepatocellular carcinoma cells from ATCC (catalog number HB-8065) in growth medium (MEM/EBSS (Hyclone catalog number SH30024, or Mediatech catalog number MT-10-010-CV) + 10% FBS (Mediatech catalog number MT35) -011-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI)) at 37 ° C and 5% CO2 Growing. Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in growth medium at approximately 3 x 10 days prior to the experiment.5 Density of holes/wells inoculated in 6-well plates at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 6-well plate was changed to fresh growth medium (1.5 ml/well) and the cell antisense oligonucleotide was administered. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of all oligonucleotides are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. For 1 well, 2 μl of this solution was incubated with 400 μl of Opti-MEM medium (Gibco Cat. No. 31985-070) and 4 μl of Lipofectamine 2000 (Invitrogen Cat. No. 11668019) for 20 minutes at room temperature and to 6 wells with cells. One hole of the disk was applied dropwise. A similar mixture including 2 μl of water replacement oligonucleotide solution was used to simulate the transfection control. In addition, the same concentration of the inactive oligonucleotide CUR-1462 was used as a control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and RNA was extracted from the cells using Promega's SV Total RNA Isolation System (catalog number Z3105) following the manufacturer's instructions. 600 ng of purified total RNA was added to the reverse transcription reaction using Invitrogen's SuperScript VILO cDNA Synthesis Kit (Cat. No. 11754-250) as described in the manufacturer's protocol. The gene expression (Hs00374696_m1, Hs00897350_m1 or Hs00897341_m1 for human SCN1A) was monitored by real-time PCR using the ABI Taqman gene expression mixture (catalog number 4369510) and the ABI-designed primer/probe using cDNA from this reverse transcription reaction. The results obtained using all three assays are very similar. The following PCR cycles were used: 50 °C for 2 minutes, 95 °C for 10 minutes, 40 cycles (95 °C for 15 seconds, 60 °C for 1 minute) using the StepOne Plus Real-Time PCR System (Applied Biosystems). The 18S test is manufactured by ABI (catalog number 4319413E). The fold change in gene expression after treatment with antisense oligonucleotides was calculated based on the difference in 18S corrected dCt values between the treated samples and the mock transfected samples. For the alternative same day method, all procedures were performed similarly, but the antisense oligonucleotides were administered immediately after the cells were dispensed to the 6-well plate on the first day.result. The amount of SCN1A mRNA in different cell lines after treatment with 20 nM antisense oligonucleotides is shown in Table 2 compared to mock-transfected controls. As can be seen from the literature, some oligonucleotides are highly active in up-regulating SCN1A mRNA levels when applied at 20 nM and are derived from several species (human, African green monkeys and mice), from different organs/cell types Cell lines of (liver, kidney, brain, embryonic fibroblasts) and primary skin fibroblasts with SCN1A mutations consistently showed up-regulation. Up-regulation of the SCN1A protein in cells with a Dewitt mutation supports the suitability of a method for treating a disease associated with a mutation in the SCN1A gene. Some oligonucleotides designed for natural antisense sequences do not affect or only slightly affect the SCN1A mRNA content in all or some of the test cell lines. These differences are consistent with the literature indicating that the binding of the oligonucleotides may depend on the secondary and tertiary structure of the target sequence of the oligonucleotide. It is noteworthy that the SCN1A content in cells treated with an oligonucleotide that is not homologous to the SCN1A natural antisense sequence but has a similar chemical composition (CUR-1462) is not significantly different from the mock-transfected control, which confirms targeting The effect of oligonucleotides does not depend on the non-specific toxicity of such molecules.Instance 4 : by targeting SCNA Antisense oligonucleotide treatment of specific natural antisense transcripts to upregulate different cell lines SCNA mRNA Dose dependence In Example 4, antisense oligonucleotides of different chemical compositions targeting SCNA-specific natural antisense transcripts were screened at a final concentration ranging from 5 to 80 nM in a panel of various cell lines. The cell lines used are derived from different organs and different animal species. The following data demonstrate that the degree of up-regulation of SCNA mRNA by modulating the function of SCNA-specific natural antisense transcripts can be altered by applying different amounts of active oligonucleotides.Materials and methods. SK-N-AS, Vero 76 and primary human fibroblasts with a Dravid mutation were treated with antisense oligonucleotides as described in Example 2 with the exception of oligonucleosides for treatment of each well. Acid and Lipofectamine 2000 concentrations are different. Adjust the concentration of oligonucleotide and Lipofectamine 2000 to ensure a final oligonucleotide concentration of 5, 10, 20, 40 and 80 nM and a 2:1 (v:v) Lipofectamine 2000 and 20 μM oligonucleotide stock solution ratio.result. Results of Dose Response Experiments Antisense oligonucleotides targeting SCN1A-specific natural antisense RNA have been shown to induce dose-dependent up-regulation of SCN1A mRNA (Figures 1-3). In some cases, at higher doses, this up-regulation is extremely strong (up to 60 times) (Figure 1-3). The degree of up-regulation induced by the same nucleotide in different cell lines appears to be different, for example 10-40 fold in primary fibroblasts at 40 nM, and by the same oligonucleotide at the same concentration The overshoot was achieved in Vero 76 cells by 2-6 fold (Figure 1 vs. Figure 3). These differences can be attributed to different transfection efficiencies of different cell lines and/or various feedback pathways manifested by them. The effect of most oligonucleotides reached a plateau at approximately 40 nM, with the exception of CUR-1764 and CUR-1770 in SCN1A fibroblasts and all oligonucleotides tested in Vero 76 cells, in the above case The plateau was not reached at the highest test concentration (Figure 1-3).Instance 5 : by targeting SCNA Up-regulation of antisense oligonucleotides specific for natural antisense transcripts SCNA mRNA Sequence specificity In Example 5, the targeted SCN1A-specific natural antisense transcript was tested in an assay designed to demonstrate that SCN1A up-regulation by oligonucleotides is independent of non-specific toxicity associated with the chemical composition of the oligonucleotide used. Antisense oligonucleotides. The following data demonstrate that the extent to which SCN1A mRNA is up-regulated by modulating the function of the SCN1A-specific natural antisense transcript depends only on the amount of active oligonucleotide, rather than the total amount of molecules of similar chemical composition.Materials and methods : Vero 76 and primary human fibroblasts with a Dravid mutation were treated with antisense oligonucleotides as described in Example 2 with the exception of different concentrations of oligonucleotides used to treat each well. The active oligonucleotide is co-administered with an inactive oligonucleotide (CUR-1462) that is similar in chemical composition but has no known target in the human genome and has no effect on the performance of the multiple genes tested (data not shown). The total amount of oligonucleotides and the amount of Lipofectamine 2000 remained constant while changing the ratio of active oligonucleotides in the mixture. Oligonucleotide concentrations were adjusted to ensure a final active oligonucleotide concentration of 5, 10, 20 and 40 nM and a total oligonucleotide concentration of 40 nM (active + inactive). As seen from the data (Figure 7), the dose-dependent effects of oligonucleotides targeting SCN1A natural antisense are not produced by non-specific toxicity potentially associated with such molecules. The SCN1A mRNA content is dependent on the dose of active oligonucleotide used to treat it (Figure 7).Instance 6 : by targeting SCNA Up-regulation of antisense oligonucleotides specific for natural antisense transcripts SCNA mRNA Target specificity In Example 6, antisense oligonucleotides that target the SCN1A-specific natural antisense transcript were tested in experiments designed to confirm the specificity of their target (ie, SCN1A). The following data demonstrate that up-regulation of SCN1A mRNA is restricted to SCN1A mRNA by modulating the function of the SCN1A-specific natural antisense transcript and does not affect the relevant sodium channels SCN9A, SCN8A, SCN7A, SCN3A, and SCN2A.Materials and methods. Vero 76 and primary human fibroblasts with a Dravid mutation were treated with antisense oligonucleotides as described in Example 3. After treatment, the isolated RNA was analyzed as described in Example 2 with the exception of using Taqman gene expression assay to detect mRNA of SCN9A, SCN8A, SCN7A, SCN3A and SCN2A channels by real-time PCR. The assays for the alpha subunits of the human SCN9A, SCN8A, SCN7A, SCN3A and SCN2A channels were obtained from ABI Inc. (catalog numbers Hs00161567_m1, Hs00274075_m1, Hs00161546_m1, Hs00366902_m1 and Hs00221379_m1, respectively).result . As shown in Figure 8, treatment with oligonucleotides CUR-1916 and CUR-1770 did not significantly affect the performance of SCN8A and SCN9A channels in human fibroblasts with a de lavert mutation. The performance of the SCN7A, SCN3A and SCN2A channels was not detectable in these cells before or after treatment (data not shown). The data demonstrates the specificity of gene expression regulation using oligonucleotides directed against the natural antisense RNA of a given gene.Instance 7 :Targeting SCNA Stability of antisense oligonucleotides specific for natural antisense transcripts In Example 7, two batches of antisense oligonucleotides targeting SCN1A-specific natural antisense transcripts were tested in an assay designed to check their stability after storage in diluted (1 mM) aqueous solution at 4 °C. Test it. The following data shows that oligonucleotides can be stable for at least 6 months in these conditions without significant loss of activity.Materials and methods. Vero 76 cells were treated with two different sets of antisense oligonucleotides as described in Example 2. The batches were synthesized in August 2010 and March 2011. The oligonucleotide synthesized in August 2010 was stored as a 1 mM aqueous solution at 4 °C. Oligonucleotides synthesized in March 2011 were shipped in lyophilized form within 3 days after synthesis and tested as soon as they arrived.result: As shown in Figure 9, there was no significant loss of biological activity after the oligonucleotide was stored in aqueous solution for 6 months at 4 °C.Instance 8 : targeting SCNA Specific antisense oligos specific for natural antisense transcripts in primary human fibroblasts with Dravid syndrome-associated mutations SCNA Protein upregulation The aim of this experiment was to classify the SCNA protein by its ability to up-regulate the performance of the SCNA protein in fibroblasts with Dravid syndrome-associated mutations based on antisense oligonucleotides CUR-1740, CUR-1770 and CUR-1916.Materials and methods. At 37 ° C and 5% CO2 Next, fibroblasts with Dravid syndrome-associated mutations introduced into culture by Dr. N. Kenyon (University of Miami) were used in MEM (Gibco, catalog number: 12561-056) + 10% FBS (Mediatech , Catalog No.: 35-015 CV) Growth in a growth medium consisting of +1% antifungal antibiotic (Gibco, catalog number: 15240-062). Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in growth medium at approximately 4 x 10 days prior to the experiment.4 /The density of the wells was re-inoculated in a 24-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 24-well plate was changed to fresh growth medium (1 ml/well) and the cells were administered with antisense oligonucleotides CUR-1740, CUR-1770 and CUR-1916. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of oligonucleotides CUR-1740, CUR-1770 and CUR-1916 are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. To administer 1 well at a final concentration of 20 nM, 1 μl of 20 μM oligonucleotide stock solution and 200 μl Opti-MEM medium (Gibco Cat. No. 31985-070) and 2 μl Lipofectamine 2000 (at room temperature) Invitrogen catalog number 11668019) was incubated for 20 minutes and applied dropwise to one well of a 24-well plate with cells. To achieve final concentrations of 5, 10, 40 and 80 nM, the volume of the 20 μM oligonucleotide stock solution used was adjusted accordingly. The ratio of 20 μM oligonucleotide stock solution to Lipofectamine 2000 was 1:2 (v:v). A similar mixture of 8 μl of water replacement oligonucleotide solution and corresponding volume of Lipofectamine 2000 was used to simulate the transfection control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and the cells were washed 3 times with calcium and magnesium-free Dulbecco's phosphate buffered saline (PBS) (Mediatech Cat. No. 21-031-CV). The PBS was then discarded and the cells were fixed in a 24-well plate for 15 minutes at -20 °C using 300 μl of 100% methanol. After removing methanol and washing with PBS, the cells were incubated with 3% hydrogen peroxide (Fisher Chemical Cat. No. H325-100) for 5 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each, followed by incubation with 300 μl of PBS containing 0.1% bovine serum albumin (BSA) (Sigma Cat. No. A-9647) for 30 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each, followed by incubation with 300 μl of avidin solution (Vector Laboratories Cat. No. SP-2001) for 30 minutes at 21 °C. The cells were briefly washed 3 times with PBS, followed by incubation with biotin solution (Vector Laboratories Cat. No. SP-2001) for 30 minutes at 21 °C. The cells were washed 3 times with PBS and then incubated overnight at 4 °C with 300 μl of rabbit antibody (Abcam catalog number ab24820) against human SCN1A diluted 250-fold in PBS/0.1% BSA overnight. After equilibrating the plate for 5 minutes at 21 ° C, the cells were washed 3 times with PBS for 5 minutes each, followed by incubation with goat anti-rabbit antibody diluted 200-fold in PBS/0.1% BSA for 30 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each and then incubated with 300 μl of Vectastain Elite ABC Reagent A+B solution (Vector Laboratories Cat. No. PK-6101) for 30 minutes; Vectastain Elite ABC Reagent A+B solution at 21 °C Prepare 30 minutes before incubation with the cells by sequentially adding and mixing 2 drops of Reagent A and 2 drops of Reagent B to 5 ml of PBS. The cells were washed 3 times with PBS for 5 minutes at 21 ° C, and then incubated with diaminobenzidine (DAB) peroxidase substrate (Vector Laboratories catalog number SK-4105) until cell staining; DAB over The oxidase substrate was reconstituted by mixing 1 ml of ImmPACTTM DAB dilution with 30 μl of ImmPACTTM DAB chromogen concentrate prior to addition to the cells. At this time, the cells were briefly washed 3 times with PBS and 300 μl of PBS was left in each well. Cell staining was analyzed directly on the inside of each well of a 24-well plate using a reverse Nikon Eclipse TS100 microscope equipped with a Nikon DS-Ri1 camera coupled to a Nikon digital observation device on a Dell Latitude D630 laptop screen. The photographs of the individual holes were taken using a software NIS-Elements D 3.0 with a Nikon camera.result. All tested antisense oligonucleotides up-regulated SCN1A protein efficiently, with CUR-1770 and CUR-1916 being the two best antisense oligonucleotides (Figure 10).Instance 9 : targeting SCNA Antisense oligonucleotide treatment of specific natural antisense transcripts SK-N-AS In the cell SCNA Protein upregulation The purpose of this experiment was to classify antisense oligonucleotides CUR-1740, CUR-1764, CUR-1770 and CUR-1916 by upregulating the ability of the SCN1A protein to be expressed in SK-N-AS cells. SK-N-AS is a human neuroblastoma cell line.Materials and methods : at 37 ° C and 5% CO2 SK-N-AS human neuroblastoma cells from ATCC (Catalog No. CRL-2137) were grown in growth medium (DMEM (Mediatech catalog number 10-013-CV) + 10% FBS (Mediatech catalog number MT35-011- CV) + penicillin/streptomycin (Mediatech Cat. No. MT30-002-CI) + non-essential amino acid (NEAA) (HyClone SH30238.01)). Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in the growth medium at approximately 5 x 10 days prior to the experiment.4 /The density of the wells was re-inoculated in a 24-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 24-well plate was changed to fresh growth medium (1 ml/well) and the cells were administered with antisense oligonucleotides CUR-1740, CUR-1764, CUR-1770 and CUR-1916. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of oligonucleotides CUR-1740, CUR-1764, CUR-1770 and CUR-1916 are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. To administer 1 well at a final concentration of 20 nM, 1 μl of 20 μM oligonucleotide stock solution and 200 μl Opti-MEM medium (Gibco Cat. No. 31985-070) and 2 μl Lipofectamine 2000 (at room temperature) Invitrogen catalog number 11668019) was incubated for 20 minutes and applied dropwise to one well of a 24-well plate with cells. To achieve final concentrations of 5, 10, 40 and 80 nM, the volume of the 20 μM oligonucleotide stock solution used was adjusted accordingly. The ratio of 20 μM oligonucleotide stock solution to Lipofectamine 2000 was 1:2 (v:v). A similar mixture of 8 μl of water replacement oligonucleotide solution and corresponding volume of Lipofectamine 2000 was used to simulate the transfection control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and the cells were washed 3 times with calcium and magnesium-free Dulbecco's phosphate buffered saline (PBS) (Mediatech Cat. No. 21-031-CV). The PBS was then discarded and the cells were fixed in a 24-well plate for 15 minutes at -20 °C using 300 μl of 100% methanol. After removing methanol and washing with PBS, the cells were incubated with 3% hydrogen peroxide (Fisher Chemical Cat. No. H325-100) for 5 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each, followed by incubation with 300 μl of PBS containing 0.1% bovine serum albumin (BSA) (Sigma Cat. No. A-9647) for 30 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each, followed by incubation with 300 μl of antibiotic solution (Vector Laboratories Cat. No. SP-2001) for 30 minutes at 21 °C. The cells were briefly washed 3 times with PBS, followed by incubation with biotin solution (Vector Laboratories Cat. No. SP-2001) for 30 minutes at 21 °C. The cells were washed 3 times with PBS and then incubated overnight at 4 °C with 300 μl of rabbit antibody (Abcam catalog number ab24820) against human SCN1A diluted 250-fold in PBS/0.1% BSA overnight. After equilibrating the plate for 5 minutes at 21 ° C, the cells were washed 3 times with PBS for 5 minutes each, followed by incubation with goat anti-rabbit antibody diluted 200-fold in PBS/0.1% BSA for 30 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each and then incubated with 300 μl of Vectastain Elite ABC Reagent A+B solution (Vector Laboratories Cat. No. PK-6101) for 30 minutes; Vectastain Elite ABC Reagent A+B solution at 21 °C Prepare 30 minutes before incubation with the cells by sequentially adding and mixing 2 drops of Reagent A and 2 drops of Reagent B to 5 ml of PBS. The cells were washed 3 times with PBS for 5 minutes at 21 ° C, and then incubated with diaminobenzidine (DAB) peroxidase substrate (Vector Laboratories catalog number SK-4105) until cell staining; DAB peroxidation The enzyme substrate was reconstituted by mixing 1 ml of ImmPACTTM DAB dilution with 30 μl of ImmPACTTM DAB chromogen concentrate prior to addition to the cells. At this time, the cells were briefly washed 3 times with PBS and 300 μl of PBS was left in each well. Cell staining was analyzed directly on the inside of each well of a 24-well plate using a reverse Nikon Eclipse TS100 microscope equipped with a Nikon DS-Ri1 camera coupled to a Nikon digital observation device on a Dell Latitude D630 laptop screen. The photographs of the individual holes were taken using a software NIS-Elements D 3.0 with a Nikon camera.result: All tested antisense oligonucleotides up-regulated the SCN1A protein, with CUR-1764 and CUR-1770 being the two best antisense oligonucleotides (Figure 11).Instance 10 : targeting SCNA Antisense oligonucleotide treatment of specific natural antisense transcripts Vero 76 In the cell SCNA Protein upregulation The purpose of this experiment was to classify antisense oligonucleotides CUR-1740, CUR-1770, CUR-1916, CUR-1924 and CUR-1945 by upregulating the ability of the SCN1A protein to behave in Vero 76 cells. Vero76 is a prairie monkey (Cercopithecus aethiops (Green vervet or African green monkey) kidney cell line.Materials and methods: At 37 ° C and 5% CO2 Next, make Vero76 African green monkey embryo kidney cells from ATCC (Catalog No. CRL-1587) in growth medium (Dellbeck modified Eagle's medium (Cellgrow 10-013-CV) + 5% FBS (Mediatech catalog number) Growth in MT35-011-CV) + penicillin/streptomycin (Mediatech catalog number MT30-002-CI)). Cells were treated with antisense oligonucleotides using one of the following methods. For the next day method, cells were grown in growth medium at approximately 4 x 10 days prior to the experiment.4 /The density of the wells was re-inoculated in a 24-well plate at 37 ° C and 5% CO2 Cultivate overnight. The next day, the medium in the 24-well plate was changed to fresh growth medium (1 ml/well) and the cells were administered with antisense oligonucleotides CUR-1740, CUR-1770 and CUR-1916. All antisense oligonucleotides were manufactured by IDT Inc. (Coralville, IA) or Exiqon (Vedbaek, Denmark). The sequences of oligonucleotides CUR-1740, CUR-1770, CUR-1916, CUR-1924 and CUR-1945 are listed in Table 1. The stock solution of the oligonucleotide was diluted to a concentration of 20 μM in sterile water without DNAse/RNAse. To administer 1 well at a final concentration of 20 nM, 1 μl of 20 μM oligonucleotide solution and 200 μl Opti-MEM medium (Gibco Cat. No. 31985-070) and 2 μl Lipofectamine 2000 (Invitrogen) at room temperature Catalog No. 11668019) was incubated for 20 minutes and applied dropwise to one well of a 24-well plate with cells. To achieve final concentrations of 5, 10, 40 and 80 nM, the volume of the 20 μM oligonucleotide stock solution used was adjusted accordingly. The ratio of 20 μM oligonucleotide stock solution to Lipofectamine 2000 was 1:2 (v:v). A similar mixture of 8 μl of water replacement oligonucleotide solution and corresponding volume of Lipofectamine 2000 was used to simulate the transfection control. At 37 ° C and 5% CO2 After about 18 hours of incubation, the medium was changed to fresh growth medium. 48 hours after the addition of the antisense oligonucleotide, the medium was removed and the cells were washed 3 times with calcium and magnesium-free Dulbecco's phosphate buffered saline (PBS) (Mediatech Cat. No. 21-031-CV). PBS was discarded and Vero 76 cells were fixed in a 24-well plate for 15 minutes at -20 °C using 300 μl of 100% methanol. After removing the methanol and washing the cells with PBS, the cells were incubated with 3% hydrogen peroxide (Fisher Chemical Cat. No. H325-100) for 5 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each, followed by incubation with 300 μl of PBS containing 0.1% bovine serum albumin (BSA) (Sigma Cat. No. A-9647) for 30 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each, followed by incubation with 300 μl of antibiotic solution (Vector Laboratories Cat. No. SP-2001) for 30 minutes at 21 °C. The cells were briefly washed 3 times with PBS, followed by incubation with biotin solution (Vector Laboratories Cat. No. SP-2001) for 30 minutes at 21 °C. The cells were washed 3 times with PBS and then incubated overnight at 4 °C with 300 μl of rabbit antibody (Abcam catalog number ab24820) against human SCN1A diluted 250-fold in PBS/0.1% BSA overnight. After equilibrating the plate for 5 minutes at 21 ° C, the cells were washed 3 times with PBS for 5 minutes each, followed by incubation with goat anti-rabbit antibody diluted 200-fold in PBS/0.1% BSA for 30 minutes at 21 °C. The cells were washed 3 times with PBS for 5 minutes each and then incubated with 300 μl of Vectastain Elite ABC Reagent A+B solution (Vector Laboratories Cat. No. PK-6101) for 30 minutes; Vectastain Elite ABC Reagent A+B solution at 21 °C Prepare 30 minutes before incubation with the cells by sequentially adding and mixing 2 drops of Reagent A and 2 drops of Reagent B to 5 ml of PBS. The cells were washed 3 times with PBS for 5 minutes at 21 ° C, and then incubated with diaminobenzidine (DAB) peroxidase substrate (Vector Laboratories catalog number SK-4105) until cell staining; DAB peroxidation The enzyme substrate was reconstituted by mixing 1 ml of ImmPACTTM DAB dilution with 30 μl of ImmPACTTM DAB chromogen concentrate prior to addition to the cells. At this time, the cells were briefly washed 3 times with PBS and 300 μl of PBS was left in each well. Cell staining was analyzed directly on the inside of each well of a 24-well plate using a reverse Nikon Eclipse TS100 microscope equipped with a Nikon DS-Ri1 camera coupled to a Nikon digital observation device on a Dell Latitude D630 laptop screen. The photographs of the individual holes were taken using a software NIS-Elements D 3.0 with a Nikon camera.result. All tested antisense oligonucleotides up-regulated the SCN1A protein, with CUR-1764 and CUR-1770 producing the highest upregulation (Figure 12).Instance 11 : Powerful up SCNA mRNA Targeting SCNA Oligonucleotides specific for natural antisense transcripts are not upregulated Vero76 Actin in cells (actin) mRNA The purpose of this experiment was to examine whether antisense oligonucleotides (CUR-1924, CUR-1740, CUR-1838) targeting SCN1A-specific natural antisense transcripts that upregulate SCN1A mRNA and protein can regulate Vero76 African green mRNA of other non-related genes (such as actin) in monkey embryonic kidney cells.Materials and methods. The Vero76 African green monkey embryo kidney cell line (Catalog No. CRL-1587) from ATCC was administered under the same conditions as described in Example 2. Actin mRNA was quantified by real-time PCR as described in Example 2 with the exception that the ABI-designed primer/probe was specific for actin (catalog number Hs99999903_m1). The data is presented in Figure 13.result. As shown in Figure 13, oligonucleotides (CUR-1924, CUR-1740, which target SCN1A-specific natural antisense transcripts that up-regulate SCN1A mRNA and protein in Vero76 cells are shown in Examples 3 and 10. CUR-1838) Effect on actin mRNA expression in Vero 76 cells. The data in Figure 13 demonstrates that oligonucleotides targeting SCN1A-specific natural antisense transcripts do not upregulate non-related genes, such as actin. Thus, such oligonucleotides are specific for up-regulating SCN1A.Instance 12 : Display will be raised SCNA mRNA And protein targeting SCNA Oligonucleotides specific for the natural antisense transcript do not upregulate actin in primary fibroblasts with Dravid-related mutations mRNA The purpose of this experiment was to examine whether antisense oligonucleotides (CUR-1916, CUR-1945), which display SCN1A-specific mRNA and protein-targeting SCN1A-specific natural antisense transcripts, can be regulated with Dravid syndrome Related mRNA of other non-related genes (such as actin) in primary human skin fibroblasts of mutant E1099X.Materials and methods. Primary human skin fibroblasts bearing the Delavid syndrome-associated mutation E1099X introduced into the culture by Dr. N. Kenyon (University of Miami) were administered under the same conditions as described in Example 3. Actin mRNA was quantified by real-time PCR as described in Example 3 with the exception that the primer/probe designed by ABI was specific for actin (catalog number Hs99999903_m1). The data is presented in Figure 14.result : As shown in Figure 14, oligonucleotides that target the SCN1A-specific natural antisense transcript do not upregulate non-related genes, such as actin. Thus, such oligonucleotides are specific for up-regulating SCN1A.Instance 13 : Display will be raised SCNA mRNA And protein targeting SCNA Oligonucleotides specific for natural antisense transcripts are not upregulated SK-N-AS Actin in cells mRNA The purpose of this experiment was to examine antisense oligonucleotides (CUR-1740, CUR-1764, CUR-1770, CUR-1838, CUR-) that display SCN1A-specific natural antisense transcripts that up-regulate SCN1A mRNA and protein. 1916) Whether it is capable of regulating mRNA of other non-related genes (such as actin) in SK-N-AS human neuroblastoma cells.Materials and methods. SK-N-AS human neuroblastoma cells (Catalog No. CRL-2137) from ATCC were administered under the same conditions as described in Example 2. Actin mRNA was quantified by real-time PCR as described in Example 2 with the exception that the ABI-designed primer/probe was specific for actin (catalog number Hs99999903_m1). The data is presented in Figure 15.result. As shown in Figure 15, oligonucleotides that target the SCN1A-specific natural antisense transcript do not upregulate non-related genes, such as actin. Thus, such oligonucleotides are specific for up-regulating SCN1A.Instance 14 : Actin is targeted SCNA Antisense oligonucleotide treatment of specific natural antisense transcripts SK-N-AS Not upregulated in cells The purpose of this experiment was to determine whether oligonucleotides (CUR-1740, CUR-1764, CUR-1770, and CUR-1916) that target the SCN1A-specific natural antisense transcript and are capable of upregulating the SCN1A protein can also regulate SK-N. - The expression of non-related proteins (such as actin) in AS cells. SK-N-AS is a human neuroblastoma cell line.Materials and methods: SK-N-AS human neuroblastoma cells (Catalog No. CRL-2137) from ATCC were grown in the same conditions as described in Example 9. The cells were precisely fixed and stained under the same conditions as described in Example 8, except that the first antibody was rabbit anti-actin (Abcam catalog number ab1801) diluted 500-fold. The staining of the cells was directly analyzed inside each well of a 24-well plate using the same method as described in Example 9.result: As shown in Figure 16, none of the antisense oligonucleotides tested up-regulated actin. Thus, such oligonucleotides are specific for up-regulating the SCN1A protein.Instance 15 : Actin is targeted SCNA Antisense oligonucleotide treatment of specific natural antisense transcripts Vero 76 Not upregulated in cells The purpose of this experiment was to identify specific antisense oligonucleotides that target the SCN1A-specific natural antisense transcript and are capable of up-regulating the SCN1A protein (CUR-1740, CUR-1770, CUR-1916, CUR-1924, and CUR-1945). Whether it is also capable of regulating the expression of proteins (such as actin) of non-related genes in Vero76 cells. Vero76 is a rhesus monkey (green monkey or African green monkey) kidney cell line.Materials and methods. The Vero76 African green monkey embryo kidney cell line (catalog number CRL-1587) from ATCC was grown in the same conditions as described in Example 10. The cells were precisely fixed and stained under the same conditions as described in Example 10 except that the first antibody was rabbit anti-actin (Abcam catalog number ab1801) diluted 500-fold. The staining of the cells was directly analyzed inside each well of a 24-well plate using the same method as described in Example 10.result. As shown in Figure 17, none of the antisense oligonucleotides tested up-regulated actin. Thus, such oligonucleotides are specific for up-regulating the SCN1A protein.Instance 16 : Actin is targeted SCNA Anti-sense oligonucleotides specific for natural antisense transcripts are not upregulated in primary human fibroblasts with Dravid syndrome-associated mutations The purpose of this experiment was to determine whether oligonucleotides (CUR-1740, CUR-1764, CUR-1770, CUR-1838, and CUR-1916) that target the SCN1A-specific natural antisense transcript and are capable of upregulating the SCNA protein are also capable of Proteins (such as actin) that regulate non-related genes in primary human fibroblasts with Dravid syndrome-associated mutations.Materials and methods. Fibroblasts with a Dehwat syndrome-associated mutation introduced into the culture by Dr. N. Kenyon (University of Miami) were grown under the same conditions as described in Example 8. The cells were precisely fixed and stained under the same conditions as described in Example 8, except that the first antibody was rabbit anti-actin (Abcam catalog number ab1801) diluted 500-fold. The staining of the cells was directly analyzed inside each well of a 24-well plate using the same method as described in Example 8.result: As shown in Figure 18, none of the antisense oligonucleotides tested up-regulated actin. Thus, such oligonucleotides are specific for up-regulating the SCN1A protein.Instance 17 :use ELISA Targeting SCNA Oligonucleotide-specific natural antisense transcripts in primary human fibroblasts with Dravid syndrome-associated mutations SCNA Protein quantification The purpose of this experiment was to quantify the affluent with Dravid syndrome due to treatment with oligonucleotides targeting SCN1A-specific natural antisense transcripts (CUR-1740, CUR-1770, and CUR-1916) using ELISA. The degree of up-regulation of SCN1A protein in primary human fibroblasts with related mutations.Materials and methods: Fibroblasts with Dravid syndrome-associated mutations introduced into culture by Dr. N. Kenyon (University of Miami) were grown under the same conditions as described in Example 8, but only at concentrations of 0 and 80 nM Oligonucleotides are used for administration. Cells were then counted and re-seeded in 96 well plates. After 24 hours, the cells were precisely fixed in the same conditions as described in Examples 8 and 16, with the exception that all 300 μl volumes were reduced to 100 μl. Parallel assay wells were stained with actin and SCN1A antibodies as described in Example 8, with the exception that all reactions were performed in a volume of 100 μl. The anti-actin antibody dilution was 1:500, the anti-SCN1A dilution was 1:250 and the anti-mouse dilution was 1:250. Further, in place of the diaminobenzidine (DAB) peroxidase receptor solution, a tetramethylbenzidine (TMB) peroxidase receptor solution (Thermo Scientific catalog No. N301) was used. After the supernatant turned blue, it was transferred to a new 96-well plate (Greiner bio one catalog number 65121) and 1 M sulfuric acid was added. Absorbance was read at 450 nm using a Multiskan Spectrum spectrophotometer (Thermo Scientific). Background signals were subtracted from all SCN1A and actin readings (read in wells used as anti-mouse stains for primary antibodies). The SCN1A signal for each condition is then corrected relative to the actin signal.result: Figure 19 shows that all of the tested antisense oligonucleotides (CUR-1740, CUR-1770 and CUR-1916) up-regulated SCN1A protein by up to 40%.Instance 18 :use ELISA Targeting SCNA Oligonucleotide treatment of specific natural antisense transcripts Vero76 In the cell SCNA Protein quantification The purpose of this experiment was to quantify the use of ELISA to quantify the oligonucleotides (CUR-1740, CUR-1770, CUR-1916, CUR-1924, CUR-1945) that target the SCN1A-specific natural antisense transcript. The extent of SCN1A protein up-regulation in primary human fibroblasts with Dravid syndrome-associated mutations.Materials and methods: Vero76 African green monkey embryo kidney cells were grown in the same conditions as described in Example 10, but only oligonucleotides at concentrations of 0 and 80 nM were used for administration. Cells were then counted and re-seeded in 96 well plates. After 24 hours, the cells were precisely fixed in the same conditions as described in Example 8, with the exception that all 300 μl volumes were reduced to 100 μl. Parallel assay wells were stained with actin and SCN1A antibodies as described in Examples 10 and 15, with the exception that all reactions were performed at 100 μl, the anti-actin antibody dilution was 1:500, and the anti-SCN1A dilution was 1:250 and the anti-mouse dilution was 1:250. Further, instead of using a diaminobenzidine (DAB) peroxidase receptor solution, a tetramethylbenzidine (TMB) peroxidase receptor solution (Thermo Scientific catalog No. N301) was used. After the supernatant turned blue, it was transferred to a new 96-well plate (Greiner bio one catalog number 651201) and 1 M sulfuric acid was added. Absorbance was read at 450 nm using a Multiskan Spectrum spectrophotometer (Thermo Scientific). Background signals were subtracted from all SCN1A and actin readings (read in wells used as anti-mouse stains for primary antibodies). The SCN1A signal for each condition is then corrected relative to the actin signal.result: Figure 20 shows that all of the tested antisense oligonucleotides (CUR-1740, CUR-1770, CUR-1916, CUR-1924, CUR-1945) efficiently upregulated the SCN1A protein by up to 300%.Instance 19 :use ELISA Targeting SCNA Oligonucleotide treatment of specific natural antisense transcripts SK-N-AS In the cell SCNA Protein quantification The purpose of this experiment was to quantify SK-N-AS due to treatment with oligonucleotides (CUR-1740, CUR-1770, CUR-1924 and CUR-1945) that target the SCN1A-specific natural antisense transcript. The degree of up-regulation of SCN1A protein in cells.Materials and methods: SK-N-AS human neuroblastoma cells from ATCC (Catalog No. CRL-2137) were grown under the same conditions as described in Example 10, but only oligonucleotides at concentrations of 0 and 20 nM were used to give medicine. Cells were then counted and re-seeded in 96 well plates. After 24 hours, the cells were precisely fixed in the same conditions as described in Example 9, with the exception that all 300 μl volumes were reduced to 100 μl. Parallel assay wells were stained with actin and SCN1A antibodies as described in Examples 9 and 13, with the exception that all reactions were performed at 100 μl, the anti-actin antibody dilution was 1:500, and the anti-SCN1A dilution was 1:250 and the anti-mouse dilution was 1:250. Further, in place of the diaminobenzidine (DAB) peroxidase receptor solution, a tetramethylbenzidine (TMB) peroxidase receptor solution (Thermo Scientific catalog No. N301) was used. After the supernatant turned blue, it was transferred to a new 96-well plate (Greiner bio one catalog number 651201) and 1 M sulfuric acid was added. Absorbance was read at 450 nm using a Multiskan Spectrum (Thermo Scientific). Background signals were subtracted from all SCN1A and actin readings (read in wells used as anti-mouse stains for primary antibodies). The SCN1A signal for each condition is then corrected relative to the actin signal.result: Figure 21 shows that all of the tested antisense oligonucleotides (CUR-1740, CUR-1770, CUR-1924, and CUR-1945) efficiently up-regulated SCN1A protein in SK-N-AS cells by up to 500%.Instance 20 :Detection HepG2 Natural antisense in cells and primary human fibroblasts with mutations related to Dravid syndrome BG724147 . The purpose of this experiment was to determine whether natural antisense BG724147 is present in human hepatocellular carcinoma HepG2 cell lines and primary human fibroblasts with mutations associated with Dravid syndrome. To achieve this, two different RNAs (poly A RNA and total RNA) isolated from each cell type were used. The PCR product was obtained after two consecutive rounds of PCR using two cell types, which were analyzed on a gel. Amplification of a similarly sized band obtained using the BG724147-specific primer confirmed the presence of BG724147 in both cell types.Materials and methods Separation total RNA . At 75 cm2 The 80% confluent HepG2 cells grown in the culture flask or the primary human fibroblasts with the Dravid syndrome-associated mutation were washed twice with PBS AccuGENE 1× (Lonza Rockeland Inc., Rockeland, ME). After discarding the PBS, 5 ml of RLT buffer containing b-mercaptoethanol (QIAGEN Inc.-USA, Valencia, CA) was added to the cells and the cell lysate was stored in a 1 ml aliquot at -80 °C. The total RNA was isolated in a microcentrifuge tube. Total RNA was isolated from these cells using the RNeasy Medium Kit (QIAGEN Inc. - USA, Valencia, CA) following the manufacturer's protocol. Briefly, cell lysates were centrifuged at 3000 xg for 5 minutes to clarify the lysate and discard any centrifugation. The clarified cell lysate was centrifuged through a QIAshredder column (inside a 2 ml microcentrifuge tube) at 14800 xg and the resulting homogenized lysate was mixed with an equal volume of 70% ethanol. The cell lysate mixed with ethanol was applied to an RNeasy medium column (inside a 15 ml conical tube) and centrifuged at 3000 x g for 5 minutes. The column was washed once with 4 ml of RW1 buffer and then subjected to DNase digestion on a column with 140 μl of RDD buffer containing RNase-free DNase for 15 minutes. The DNase digestion was terminated by the addition of 4 ml RW1 buffer and the column was centrifuged at 3000 xg. The column was washed twice with RPE buffer and the total RNA bound to the filter was dissolved in 150 μl of water without DNase and RNAse. Total RNA was stored at -80 °C until the next step.from HepG2 Total cell RNA Separation poly-A RNA . Separation of total RNA from HepG2 cells and primary human fibroblasts with Dravid syndrome-associated mutations using the kit for separation of poly-A from Ambion (Applied Biosystems/ Ambion, Austin, TX) following the manufacturer's protocol Poly A RNA. Basically, 100 μg of total RNA was resuspended so that the final concentration in water without DNase/RNAse was 600 μg/ml and an equal volume of 2× binding solution was added. During this time, 10 μl of Oligo (dT) magnetic beads were placed in a microcentrifuge tube, which was captured by placing the tube on a magnetic stand and discarding the storage buffer. 50 μl of Wash Solution 1 was added to the beads and the tube was removed from the magnetic bench and the wash solution was discarded. At this time, 1× binding buffer containing total RNA from HepG2 cells was mixed with magnetic beads and heated at 70 ° C for 5 minutes, followed by incubation at room temperature for 60 minutes with gentle agitation. The poly A RNA combined with the magnetic beads was captured by using a magnetic stand for 5 minutes. Discard the supernatant. The Oligo (dT) magnetic beads were washed twice with Wash Solution 1 and once with Wash Solution 2 to remove non-specifically bound RNA. The magnetic beads were captured with a magnetic stand and 200 μl of warm RNA storage solution (preheated at 70 ° C for 5 minutes) was added to the beads. The magnetic beads are captured by a magnetic gantry and the supernatant (the first solution of poly A RNA) is stored. A second 200 μl of warm RNA storage solution (preheated at 70 ° C for 5 minutes) was then added to the beads. A second eluate of polyA RNA is added to the first eluate. At this time, the eluted RNA was precipitated overnight at -20 ° C using 5 M ammonium acetate, glycogen and 100% ethanol. The poly RNA was centrifuged at 14800 x g for 30 minutes at 4 °C. The supernatant was discarded and the RNA pellet was washed 3 times with 1 ml of 70% ethanol, and the RNA pellet was recovered each time by centrifugation at 4 ° C for 10 minutes. Finally, the poly A RNA pellet was resuspended in an RNA storage solution heated to 70 ° C to dissolve the RNA. Poly A RNA was stored at -80 °C. Adenosine was added to the 3' end of the RNA transcript. Total RNA (40 μg) from HepG2 cells or primary human fibroblasts with Dravid syndrome-associated mutations was mixed with 2 units of RNA Poly (A) polymerase to a final reaction volume of 100 μl (Ambion, Applied Biosystems, St. Austin TX). The ATP used in the polyadenylation reaction was from Invitrogen. After polyadenylation, RNA was purified using phenol/chloroform techniques followed by hepatic glucose/sodium acetate precipitation. This RNA was resuspended in 40 μl of DNAse/RNAse-free water and used in the 3' RACE reaction (from the FirstChoice RLM-RACE kit from Ambion, Applied Biosystems, St. Austin TX). 3' extension of BCN724147 natural antisense transcript of SCN1A. The 3' rapid amplification (RACE) reaction of the two different cDNA ends was performed using the FirstChoice RLM-RACE kit from Ambion, Applied Biosystems (St. Austin, TX). One group used poly A RNA and the other group used total RNA supplemented with 1 adenosine, and the poly A RNA and total RNA were derived from HepG2 cells or primary human fibroblasts with mutations associated with Dravid syndrome. Two consecutive rounds of PCR were performed. The first PCR was performed using the 3' external primer supplied in the kit and the 5' primer (5' GATTCTCCTACA GCAATTGGTA 3') specific for BG724147 designed by OPKO CURNA. The second round of PCR was performed using the 3' external primer supplied in the kit and the different 5' primer (5' GACATGTAATCACTTTCATCAA 3') specific for BG724147 designed by OPKO CURNA. The product of the second PCR reaction was run on a 1% agarose-1 x TAE gel.result : Figure 22 shows the use of poly A RNA from HepG2 cells and total RNA supplemented with adenosine and poly A RNA from primary human fibroblasts with Dravid syndrome-associated mutations and total RNA supplemented with adenosine The product of the second round of PCR reaction of the 3' RACE experiment was performed. One identical bright band was observed in poly A RNA from HepG2 cells and primary human fibroblasts with Dravid syndrome-associated mutations.in conclusion: PCR amplification using a primer specific for the BG724147 natural antisense transcript of SCN1A produces a common PCR bright in two different cells (HepG2 cells and primary human fibroblasts with mutations associated with Dravid syndrome) band. Furthermore, antisense oligonucleotides targeting SCN1A natural antisense BG724147 have been shown to upregulate SCN1A mRNA and protein in such cells as shown in Examples 2, 7 and 16. This data indicates that BG724147 is actually present in both cells (HepG2 cells and primary human fibroblasts with mutations associated with Dravid syndrome).Instance twenty one : SCNA Natural antisense sequence BG724147 Extension The purpose of this experiment was to extend its known sequence by sequencing all sequences of SCN1A natural antisense BG724147. The original BG724147 RNA transcript was obtained from the human test pellet obtained by Miklos Palkovits. A cDNA library prepared in pBluescriptR vector by Michael J. Brownstein (at NHGRI), Shiraki Toshiyuki, and Piero Carninci (at RIKEN). The cDNA libraries were arranged by the I.M.A.G.E. Association (or LLNL) and purely sequenced by Incyte Genomics, Inc. in May 2001. BG724147 pure line is available from Open Biosystems (Open Biosystems Products, Huntsville, AL). In 2001, the cDNA inserted into the BG724147 pure line was not completely sequenced. OPKO-CURNA obtained the BG724147 pure line and sequenced all inserts. To achieve this, a bacterial strain containing a plastid having the BG724147 insert was obtained from Open Biosystems and plated in a Luria Bertani (LB) agar plate containing ampicillin to isolate individual colonies. The colonies were then expanded in 5 ml of LB medium. The plastid containing the BG724147 insert was then isolated from the bacteria and delivered to Davis Sequencing (Davis, CA) for sequencing. materialAnd method: Separation and sequencing SCNA Natural antisense BG724147 It cDNA The plastid. A suspension of frozen bacteria containing BG724147 plastids (Open Biosystems Products, Cat. No. 4829512) was purchased from Open Biosystems, diluted 10, 100, 1000, 10,000, 100000 times, followed by inoculation with 100 μg/ml ampicillin (Calbiochem, catalogue) No. 171254) Luria Bertani (LB) (BD, Cat. No. 244520) agar plate (Falcon, Cat. No. 351055). After 15 hours, 20 individual bacterial colonies were isolated from the 100,000-fold dilution plate and individually grown in 5 ml LB medium (Fisher Scientific, Cat. No. BP1426-2) for 15 hours to 24 hours. At this point, the bacteria were pooled and the plastids (cDNA containing the BG724147 RNA transcript) were isolated using Promega's PureYieldTM plastid small scale purification system kit (Promega, Cat. No. A1222) following the manufacturer's protocol. The isolated DNA was diluted to 200 ng/ml and 12 μl of plastids from each community were delivered to Davis sequencing (Davis, CA) for sequencing.result: The sequence obtained from Davis sequencing provides an extended BG724147 (SEQ ID NO: 12).in conclusion: The BG724147 sequence is known to successfully extend 403 nucleotides to serve as the basis for designing antisense oligonucleotides against the SCN1A natural antisense transcript BG724147. While the invention has been illustrated and described with reference to the embodiments In addition, although a particular feature of the invention may have been disclosed in connection with only one of several embodiments, this feature can be combined with one of the other embodiments as may be required for any given or specific application and for any given or particular application. Or a combination of multiple other features. The Abstract of the Invention will allow the reader to quickly ascertain the nature of the technical disclosure. It should be understood that it does not apply to the scope or meaning of the following claims.

圖1為即時PCR結果之圖,其展示相較於對照,在用使用Lipofectamine 2000引入之硫代磷酸酯寡核苷酸處理HepG2細胞之後SCN1A mRNA之倍數變化+標準偏差。即時PCR結果顯示HepG2細胞中之SCN1A mRNA之含量在用SCN1A反義BG724147之1種反義寡核苷酸處理之後48小時顯著增加。以CUR-1624至CUR-1627表示之柱條分別對應於用SEQ ID NO: 30至33處理之樣品。 圖2為即時PCR結果之圖,其展示相較於對照,在用使用Lipofectamine 2000引入之硫代磷酸酯寡核苷酸處理HepG2細胞之後SCN1A mRNA之倍數變化+標準偏差。以CUR-1628至CUR-1631表示之柱條分別對應於用SEQ ID NO: 34至37處理之樣品。 圖3為即時PCR結果之圖,其展示相較於對照,在用使用Lipofectamine 2000引入之硫代磷酸酯寡核苷酸處理HepG2細胞之後SCN1A mRNA之倍數變化+標準偏差。以CUR-1632至CUR-1636表示之柱條分別對應於用SEQ ID NO: 38至42處理之樣品。 圖4顯示帶有德拉威特症候群相關突變之初級人類皮膚纖維母細胞中的SCN1A mRNA之劑量依賴性上調。CUR-1916、CUR-1740、CUR-1764及CUR-1770分別對應於用SEQ ID NO: 70、45、52及57處理之樣品。 圖5顯示SK-N-AS細胞中之SCN1A mRNA的劑量依賴性上調。CUR-1916、CUR-1740、CUR-1764及CUR-1770分別對應於用SEQ ID NO: 70、45、52及57處理之樣品。 圖6顯示Vero76細胞中之SCN1A mRNA的劑量依賴性上調。CUR-1916、CUR-1740、CUR-1764及CUR-1770分別對應於用SEQ ID NO: 70、45、52及57處理之樣品。 圖7顯示SCN1A mRNA之上調並非由反義寡核苷酸之非特異性毒性引起。A-由CUR-1916引起之上調;B-由CUR-1770引起之上調。CUR-1462為化學組成類似之非活性對照寡核苷酸。 圖8顯示帶有德拉威特相關突變之人類纖維母細胞中的SCN8A及SCN9A通道之表現並不因用靶向SCN1A天然反義轉錄物之反義寡核苷酸進行處理而受顯著影響。A-用CUR-1770處理;B-用CUR-1916處理。 圖9展示靶向SCN1A特異性天然反義轉錄物之反義寡核苷酸之穩定性。Vero 76細胞如實例2中所述用在2010年8月及2011年3月合成之兩批不同CUR-1916處理。2010年8月合成之寡核苷酸在4℃下以1 mM水溶液形式儲存。2011年3月合成之寡核苷酸以凍乾形式運送且在到達後即刻加以測試。 圖10展示用互補於SCN1A天然反義之反義寡核苷酸處理之帶有德拉威特症候群突變的纖維母細胞中之SCN1A蛋白之上調。使纖維母細胞在24孔盤中生長且用互補於SCN1A天然反義之反義寡核苷酸以20 nM(圖c:CUR-1740;d:CUR-1770;e:CUR-1916)及0 nM(b)加以處理。細胞係使用抗SCN1A抗體(Abcam目錄號ab24820)及用抗生蛋白/生物素方法(Vector Laboratories目錄號SP-2001;Vector Laboratories目錄號PK-6101;Vector Laboratories目錄號SK-4105)進行之二次抗體染色/擴增,藉由間接免疫組織化學針對SCN1A(b-e)進行染色;圖a-陰性對照,兔抗小鼠抗體用作一次抗體,隨後進行與圖b-e中相同之染色程序。 圖11展示用互補於SCN1A天然反義之反義寡核苷酸處理之SK-N-AS細胞中的SCN1A蛋白之上調。使SK-N-AS細胞在24孔盤中生長且用寡核苷酸以20 nM(c:CUR-1740;d:CUR-1764;e:CUR-1770;f:CUR-1916)及(b:0 nM)加以處理。SK-N-AS細胞係使用抗SCN1A抗體(Abcam目錄號ab24820)及使用抗生蛋白/生物素方法(Vector Laboratories目錄號SP-2001;Vector Laboratories目錄號PK-6101;Vector Laboratories目錄號SK-4105)進行之二次抗體染色/擴增,藉由間接免疫組織化學針對SCN1A(b-f)進行染色;作為陰性對照,兔抗小鼠抗體用作一次抗體,隨後進行與圖b-f中相同之染色程序(圖a)。 圖12展示用互補於SCN1A天然反義之反義寡核苷酸處理之Vero 76細胞中的SCN1A蛋白之上調。使Vero 76細胞在24孔盤中生長且用互補於SCN1A天然反義之反義寡核苷酸以20 nM(c: CUR-1740;d: CUR-1945;e: CUR-1770;f: CUR-1916;g: CUR-1924)及0 nM(b)加以處理。Vero 76細胞係使用抗SCN1A抗體(Abcam目錄號ab24820)及用抗生蛋白/生物素方法(Vector Laboratories目錄號SP-2001;Vector Laboratories目錄號PK-6101;Vector Laboratories目錄號SK-4105)進行之二次抗體染色/擴增,藉由間接免疫組織化學針對SCN1A(b-f)進行染色;圖a-作為陰性對照,兔抗小鼠抗體用作一次抗體,隨後進行與圖b-g中相同之染色程序。 圖13顯示會上調SCN1A mRNA之靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調Vero76細胞中之肌動蛋白。測試在實例5及12中顯示會上調SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物的相同反義寡核苷酸(CUR-1740、CUR-1838、CUR-1924)對Vero 76細胞中之β-肌動蛋白mRNA表現的影響。資料證實靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調非相關基因,諸如肌動蛋白。以CUR-1740、CUR-1838及CUR-1924表示之柱條分別對應於用SEQ ID NO: 45、62及78處理之樣品。 圖14顯示顯示會上調SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調帶有德拉威特相關突變之纖維母細胞中的肌動蛋白。測試在實例2及7中顯示會上調SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物之寡核苷酸(CUR-1916、CUR-1945)對帶有德拉威特相關突變之纖維母細胞中的肌動蛋白mRNA表現之影響。以下資料證實靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調非相關基因,諸如肌動蛋白。以CUR-1916及CUR-1945表示之柱條分別對應於用SEQ ID NO: 70及93處理之樣品。 圖15顯示顯示會上調SCN1A mRNA及蛋白質之靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調SK-N-AS細胞中的肌動蛋白。測試在實例中顯示會上調SCN1A mRNA及蛋白質之相同反義寡核苷酸(CUR-1740、CUR-1770、CUR-1916、CUR-1764、CUR-1838)對SK-N-AS細胞中之肌動蛋白mRNA表現的影響。資料證實靶向SCN1A特異性天然反義轉錄物之寡核苷酸不上調非相關基因,諸如肌動蛋白。以CUR-1740、CUR-1770、CUR-1916、CUR-1764、CUR-1838表示之柱條分別對應於用SEQ ID NO: 45、57、70、52及62處理之樣品。 圖16展示用互補於SCN1A天然反義之反義寡核苷酸處理之SK-N-AS細胞中的肌動蛋白之染色。使SK-N-AS細胞在24孔盤中生長且用寡核苷酸以20 nM(b:CUR-1740;c:CUR-1764;d:CUR-1770;e:CUR-1916)及0 nM(a)加以處理。SK-N-AS細胞係使用抗肌動蛋白抗體(Abcam目錄號ab1801)及使用抗生蛋白/生物素方法(Vector Laboratories目錄號SP-2001;Vector Laboratories目錄號PK-6101;Vector Laboratories目錄號SK-4105)進行之二次抗體染色/擴增,藉由間接免疫組織化學針對肌動蛋白(a-e)進行染色。 圖17展示用互補於SCN1A天然反義之反義寡核苷酸處理之Vero 76細胞中的肌動蛋白之染色。使Vero 76細胞在24孔盤中生長且用互補於SCN1A天然反義之反義寡核苷酸以20 nM(b:CUR-1740;c:CUR-1770;d:CUR-1916;e:CUR-1924;f:CUR-1945)及0 nM(a)加以處理。Vero 76細胞係使用抗肌動蛋白抗體(Abcam目錄號ab1801)及用抗生蛋白/生物素方法(Vector Laboratories目錄號SP-2001;Vector Laboratories目錄號PK-6101;Vector Laboratories目錄號SK-4105)進行之二次抗體染色/擴增,藉由間接免疫組織化學針對肌動蛋白(b-f)進行染色;圖a-陰性對照,兔抗小鼠抗體用作一次抗體,隨後進行與圖b-g中相同之染色程序。 圖18展示用互補於SCN1A天然反義之反義寡核苷酸處理之帶有德拉威特症候群突變的纖維母細胞中之肌動蛋白之上調。使纖維母細胞在24孔盤中生長且用互補於SCN1A天然反義之反義寡核苷酸以20 nM(圖b:CUR-1740;c:CUR-1764;d:CUR-1770;e:CUR-1838及f:CUR-1916)及0 nM(a)加以處理。細胞係使用抗肌動蛋白抗體(Abcam目錄號ab1801)及用抗生蛋白/生物素方法(Vector Laboratories目錄號SP-2001;Vector Laboratories目錄號PK-6101;Vector Laboratories目錄號SK-4105)進行之二次抗體染色/擴增,藉由間接免疫組織化學針對肌動蛋白(a-f)進行染色。 圖19展示用互補於SCN1A天然反義之反義寡核苷酸處理之帶有德拉威特症候群突變的纖維母細胞中之SCN1A蛋白的上調,使用ELISA定量。用互補於SCN1A天然反義之寡核苷酸以0或80 nM處理纖維母細胞。在48小時之後,細胞轉移至96孔盤中,持續24小時,隨後經固定且用於SCN1A及肌動蛋白ELISA。SCN1A信號之OD讀數相對於相同實驗條件之肌動蛋白信號進行校正。用0 nM寡核苷酸給藥之細胞中之經校正的SCN1A信號用作基線(100%)。以CUR-1740、CUR-1770及CUR-1916表示之柱條分別對應於用SEQ ID NO: 45、57及70處理之樣品。 圖20展示用互補於SCN1A天然反義之反義寡核苷酸處理之Vero76細胞中的SCN1A蛋白的上調,使用ELISA定量。用互補於SCN1A天然反義之反義寡核苷酸以0或80 nM處理Vero76細胞。在48小時之後,細胞轉移至96孔盤中,持續24小時,隨後經固定且用於SCN1A及肌動蛋白ELISA。SCN1A信號之OD讀數相對於相同實驗條件之肌動蛋白信號進行校正。用0 nM寡核苷酸給藥之細胞中之經校正的SCN1A信號用作基線(100%)。以CUR-1740、CUR-1770、CUR-1916、CUR-1924、CUR-1945表示之柱條分別對應於用SEQ ID NO: 45、57、70、78及93處理之樣品。 圖21展示用互補於SCN1A天然反義之寡核苷酸處理之SK-N-AS細胞中的SCN1A蛋白的上調,使用ELISA定量。用互補於SCN1A天然反義之反義寡核苷酸以0或20 nM處理SK-N-AS細胞。在48小時之後,細胞轉移至96孔盤中,持續24小時,隨後經固定且用於SCN1A及肌動蛋白ELISA。SCN1A信號之OD讀數相對於相同實驗條件之肌動蛋白信號進行校正。用0 nM寡核苷酸給藥之細胞中之經校正的SCN1A信號用作基線(100%)。以CUR-1740、CUR-1770、CUR-1924、CUR-1945表示之柱條分別對應於用SEQ ID NO: 45、57、78及93處理之樣品。 圖22展示SCN1A天然反義轉錄物BG724147之3'RACE之第二輪PCR的產物。對以下進行3'RACE:a)來自HepG2細胞之添加有腺苷之總RNA;b-自HepG2細胞分離之poly A RNA;c-來自帶有德拉威特症候群相關突變之初級人類纖維母細胞的添加有腺苷之總RNA;d-自帶有德拉威特症候群相關突變之初級人類纖維母細胞分離的poly A RNA。圖示用GelRed(GenScript,目錄號M00120)染色之1%瓊脂糖凝膠/1×TAE之負片(negative)。箭頭展示為HepG2細胞及帶有德拉威特症候群相關突變之初級人類纖維母細胞所共有的亮帶,從而證明此等細胞中存在BG724147天然反義轉錄物。序列表描述 - SEQ ID NO: 1:智人電壓門控鈉離子通道第I型α次單元(SCN1A),轉錄變異體1,mRNA(NCBI寄存編號:NM_001165963);SEQ ID NO: 2:智人電壓門控鈉離子通道第II型α次單元(SCN2A),轉錄變異體1,mRNA(NCBI寄存編號:NM_021007);SEQ ID NO: 3:智人電壓門控鈉離子通道第III型α次單元(SCN3A),轉錄變異體1,mRNA(NCBI寄存編號:NM_006922);SEQ ID NO: 4:智人電壓門控鈉離子通道第IV型α次單元(SCN4A),mRNA(NCBI寄存編號:NM_000334);SEQ ID NO: 5:智人電壓門控鈉離子通道第V型α次單元(SCN5A),轉錄變異體1,mRNA(NCBI寄存編號:NM_198056);SEQ ID NO:6:智人電壓門控鈉離子通道第VII型α(SCN7A),mRNA(NCBI寄存編號:NM_002976);SEQ ID NO: 7:智人電壓門控鈉離子通道第VIII型α次單元(SCN8A),轉錄變異體1,mRNA(NCBI寄存編號:NM_014191);SEQ ID NO: 8:智人電壓門控鈉離子通道第IX型α次單元(SCN9A),mRNA(NCBI寄存編號:NM_002977);SEQ ID NO: 9:智人電壓門控鈉離子通道第X型α次單元(SCN10A),mRNA(NCBI寄存編號:NM_006514);SEQ ID NO: 10:智人電壓門控鈉離子通道第XI型α次單元(SCN11A),mRNA(NCBI寄存編號:NM_014139);SEQ ID NO: 11:智人電壓門控鈉離子通道α次單元SCN12A(SCN12A)mRNA,完全cds(NCBI寄存編號:AF109737);SEQ ID NO: 12:天然SCN1A反義序列(延伸之BG724147);SEQ ID NO: 13:天然SCN1A反義序列(Hs.662210);SEQ ID NO: 14:天然SCN1A反義序列(AA383040);SEQ ID NO: 15:天然SCN1A反義序列(BC029452);SEQ ID NO: 16:天然SCN1A反義序列(AA630035);SEQ ID NO: 17:天然SCN1A反義序列(BE566126);SEQ ID NO: 18:天然SCN1A反義序列(BF673100);SEQ ID NO: 19:天然SCN1A反義序列(BG181807);SEQ ID NO: 20:天然SCN1A反義序列(BG183871);SEQ ID NO: 21:天然SCN1A反義序列(BG215777);SEQ ID NO: 22:天然SCN1A反義序列(BG227970);SEQ ID NO: 23:天然SCN1A反義序列(BM905527);SEQ ID NO: 24:天然SCN1A反義序列(BU180772);SEQ ID NO: 25:小鼠天然SCN1A反義序列(BG724147 ExtMouse);SEQ ID NO: 26:小鼠天然SCN1A反義序列(Hs.662210小鼠AS1);SEQ ID NO: 27:小鼠天然SCN1A反義序列(Hs.662210小鼠AS2);SEQ ID NO: 28:小鼠天然SCN1A反義序列(Hs.662210小鼠AS3);SEQ ID NO: 29至94:反義寡核苷酸。SEQ ID NO: 95及96分別為反義寡核苷酸SEQ ID NO: 58及59之反向互補序列。*指示硫代磷酸酯鍵,+指示LNA,「r」指示RNA且「m」指示寡核苷酸之指定糖部分上之2'氧原子上的甲基。Figure 1 is a graph of real-time PCR results showing fold change + standard deviation of SCN1A mRNA after treatment of HepG2 cells with phosphorothioate oligonucleotides introduced using Lipofectamine 2000 compared to controls. The real-time PCR results showed that the content of SCN1A mRNA in HepG2 cells was significantly increased 48 hours after treatment with one antisense oligonucleotide of SCN1A antisense BG724147. The bars indicated by CUR-1624 to CUR-1627 correspond to the samples treated with SEQ ID NOS: 30 to 33, respectively. Figure 2 is a graph of real-time PCR results showing fold change + standard deviation of SCN1A mRNA after treatment of HepG2 cells with phosphorothioate oligonucleotides introduced using Lipofectamine 2000 compared to controls. The bars indicated by CUR-1628 to CUR-1631 correspond to the samples treated with SEQ ID NOS: 34 to 37, respectively. Figure 3 is a graph of real-time PCR results showing fold change + standard deviation of SCN1A mRNA after treatment of HepG2 cells with phosphorothioate oligonucleotides introduced using Lipofectamine 2000 compared to controls. The bars indicated by CUR-1632 to CUR-1636 correspond to the samples treated with SEQ ID NOS: 38 to 42, respectively. Figure 4 shows dose-dependent up-regulation of SCN1A mRNA in primary human skin fibroblasts with Dravid syndrome-associated mutations. CUR-1916, CUR-1740, CUR-1764 and CUR-1770 correspond to the samples treated with SEQ ID NOS: 70, 45, 52 and 57, respectively. Figure 5 shows a dose-dependent up-regulation of SCN1A mRNA in SK-N-AS cells. CUR-1916, CUR-1740, CUR-1764 and CUR-1770 correspond to the samples treated with SEQ ID NOS: 70, 45, 52 and 57, respectively. Figure 6 shows a dose-dependent up-regulation of SCN1A mRNA in Vero76 cells. CUR-1916, CUR-1740, CUR-1764 and CUR-1770 correspond to the samples treated with SEQ ID NOS: 70, 45, 52 and 57, respectively. Figure 7 shows that SCN1A mRNA upregulation is not caused by non-specific toxicity of antisense oligonucleotides. A- is caused by CUR-1916; B- is caused by CUR-1770. CUR-1462 is an inactive control oligonucleotide of similar chemical composition. Figure 8 shows that the expression of SCN8A and SCN9A channels in human fibroblasts with Dravid-related mutations was not significantly affected by treatment with antisense oligonucleotides targeting the SCN1A natural antisense transcript. A-treated with CUR-1770; B- treated with CUR-1916. Figure 9 shows the stability of antisense oligonucleotides that target SCN1A-specific natural antisense transcripts. Vero 76 cells were treated with two different batches of CUR-1916 synthesized in August 2010 and March 2011 as described in Example 2. The oligonucleotide synthesized in August 2010 was stored as a 1 mM aqueous solution at 4 °C. Oligonucleotides synthesized in March 2011 were shipped in lyophilized form and tested as soon as they arrived. Figure 10 shows the up-regulation of SCN1A protein in fibroblasts with Dravid syndrome mutations treated with antisense oligonucleotides complementary to SCN1A natural antisense. Fibroblasts were grown in 24-well plates and treated with antisense oligonucleotides complementary to SCN1A natural antisense at 20 nM (panel c: CUR-1740; d: CUR-1770; e: CUR-1916) and 0 nM (b) deal with it. Cell lines were treated with anti-SCN1A antibody (Abeam catalog number ab24820) and anti-protein/biotin method (Vector Laboratories catalog number SP-2001; Vector Laboratories catalog number PK-6101; Vector Laboratories catalog number SK-4105). Staining/amplification, staining for SCN1A (be) by indirect immunohistochemistry; Figure a - Negative control, rabbit anti-mouse antibody was used as primary antibody, followed by the same staining procedure as in Figure be. Figure 11 shows up-regulation of SCN1A protein in SK-N-AS cells treated with antisense oligonucleotides complementary to SCN1A natural antisense. SK-N-AS cells were grown in 24-well plates and oligonucleotides were used at 20 nM (c: CUR-1740; d: CUR-1764; e: CUR-1770; f: CUR-1916) and (b :0 nM) to be processed. The SK-N-AS cell line used anti-SCN1A antibody (Abeam catalog number ab24820) and the antibiotic/biotin method (Vector Laboratories catalog number SP-2001; Vector Laboratories catalog number PK-6101; Vector Laboratories catalog number SK-4105) Secondary antibody staining/amplification was performed, and SCN1A (bf) was stained by indirect immunohistochemistry; as a negative control, a rabbit anti-mouse antibody was used as a primary antibody, followed by the same staining procedure as in Figure bf (Fig. a). Figure 12 shows up-regulation of SCN1A protein in Vero 76 cells treated with antisense oligonucleotides complementary to SCN1A natural antisense. Vero 76 cells were grown in 24-well plates and treated with antisense oligonucleotides complementary to SCN1A natural antisense at 20 nM (c: CUR-1740; d: CUR-1945; e: CUR-1770; f: CUR- 1916; g: CUR-1924) and 0 nM (b) are processed. The Vero 76 cell line was treated with anti-SCN1A antibody (Abeam catalog number ab24820) and antibiotic/biotin method (Vector Laboratories catalog number SP-2001; Vector Laboratories catalog number PK-6101; Vector Laboratories catalog number SK-4105). Sub-body staining/amplification, staining for SCN1A (bf) by indirect immunohistochemistry; Figure a - As a negative control, a rabbit anti-mouse antibody was used as a primary antibody, followed by the same staining procedure as in Figure bg. Figure 13 shows that oligonucleotides that target SCN1A mRNA targeting SCN1A-specific natural antisense transcripts do not upregulate actin in Vero76 cells. The same antisense oligonucleotides (CUR-1740, CUR-1838, CUR-1924) targeting SCN1A-specific natural antisense transcripts that up-regulate SCN1A mRNA and protein were shown in Examples 5 and 12 for Vero 76 cells. The effect of β-actin mRNA expression. The data demonstrate that oligonucleotides that target the SCN1A-specific natural antisense transcript do not upregulate non-related genes, such as actin. The bars indicated by CUR-1740, CUR-1838 and CUR-1924 correspond to the samples treated with SEQ ID NOS: 45, 62 and 78, respectively. Figure 14 shows that oligonucleotides targeting SCN1A-specific natural antisense transcripts that up-regulate SCN1A mRNA and protein do not upregulate actin in fibroblasts with Dravid-related mutations. The oligonucleotides (CUR-1916, CUR-1945) targeting SCN1A-specific natural antisense transcripts that up-regulate SCN1A mRNA and protein are shown in Examples 2 and 7 for fibers with Dravid-related mutations. The effect of actin mRNA expression in mother cells. The following data demonstrate that oligonucleotides targeting SCN1A-specific natural antisense transcripts do not upregulate non-related genes, such as actin. The bars indicated by CUR-1916 and CUR-1945 correspond to the samples treated with SEQ ID NOS: 70 and 93, respectively. Figure 15 shows that oligonucleotides targeting SCN1A-specific natural antisense transcripts that up-regulate SCN1A mRNA and protein do not upregulate actin in SK-N-AS cells. The test showed in the examples that the same antisense oligonucleotides (CUR-1740, CUR-1770, CUR-1916, CUR-1764, CUR-1838) that upregulate SCN1A mRNA and protein are involved in the muscles of SK-N-AS cells. The effect of kinesin mRNA expression. The data demonstrate that oligonucleotides that target the SCN1A-specific natural antisense transcript do not upregulate non-related genes, such as actin. The bars indicated by CUR-1740, CUR-1770, CUR-1916, CUR-1764, CUR-1838 correspond to the samples treated with SEQ ID NOS: 45, 57, 70, 52 and 62, respectively. Figure 16 shows the staining of actin in SK-N-AS cells treated with antisense oligonucleotides complementary to SCN1A natural antisense. SK-N-AS cells were grown in 24-well plates with oligonucleotides at 20 nM (b: CUR-1740; c: CUR-1764; d: CUR-1770; e: CUR-1916) and 0 nM (a) deal with it. SK-N-AS cell line uses anti-actin antibody (Abeam catalog number ab1801) and antibiotic/biotin method (Vector Laboratories catalog number SP-2001; Vector Laboratories catalog number PK-6101; Vector Laboratories catalog number SK- 4105) Secondary antibody staining/amplification performed by indirect immunohistochemistry for actin (ae) staining. Figure 17 shows staining of actin in Vero 76 cells treated with antisense oligonucleotides complementary to SCN1A natural antisense. Vero 76 cells were grown in 24-well dishes and treated with antisense oligonucleotides complementary to SCN1A natural antisense at 20 nM (b: CUR-1740; c: CUR-1770; d: CUR-1916; e: CUR- 1924; f: CUR-1945) and 0 nM (a) are processed. The Vero 76 cell line was performed using an anti-actin antibody (Abeam catalog number ab1801) and using the antibiotic/biotin method (Vector Laboratories Cat. No. SP-2001; Vector Laboratories Cat. No. PK-6101; Vector Laboratories Cat. No. SK-4105). Secondary antibody staining/amplification, staining for actin (bf) by indirect immunohistochemistry; Figure a - negative control, rabbit anti-mouse antibody used as primary antibody, followed by staining as in Figure bg program. Figure 18 shows actin upregulation in fibroblasts with Dravid syndrome mutations treated with antisense oligonucleotides complementary to SCN1A natural antisense. Fibroblasts were grown in 24-well plates and treated with antisense oligonucleotides complementary to SCN1A natural antisense at 20 nM (panel b: CUR-1740; c: CUR-1764; d: CUR-1770; e: CUR -1838 and f:CUR-1916) and 0 nM(a) are processed. Cell lines were performed using anti-actin antibody (Abeam catalog number ab1801) and anti-protein/biotin method (Vector Laboratories catalog number SP-2001; Vector Laboratories catalog number PK-6101; Vector Laboratories catalog number SK-4105) Sub-body staining/amplification, staining for actin (af) by indirect immunohistochemistry. Figure 19 shows up-regulation of SCN1A protein in fibroblasts with Dravid syndrome mutations treated with antisense oligonucleotides complementary to SCN1A natural antisense, quantified using ELISA. Fibroblasts were treated with oligonucleotides complementary to the natural antisense of SCN1A at 0 or 80 nM. After 48 hours, the cells were transferred to 96-well plates for 24 hours, then fixed and used for SCN1A and actin ELISA. The OD reading of the SCN1A signal was corrected relative to the actin signal of the same experimental conditions. The corrected SCN1A signal in cells dosed with 0 nM oligonucleotide was used as the baseline (100%). The bars indicated by CUR-1740, CUR-1770 and CUR-1916 correspond to the samples treated with SEQ ID NOS: 45, 57 and 70, respectively. Figure 20 shows up-regulation of SCN1A protein in Vero76 cells treated with antisense oligonucleotides complementary to SCN1A natural antisense, quantified using ELISA. Vero76 cells were treated with antisense oligonucleotides complementary to SCN1A natural antisense at 0 or 80 nM. After 48 hours, the cells were transferred to 96-well plates for 24 hours, then fixed and used for SCN1A and actin ELISA. The OD reading of the SCN1A signal was corrected relative to the actin signal of the same experimental conditions. The corrected SCN1A signal in cells dosed with 0 nM oligonucleotide was used as the baseline (100%). The bars indicated by CUR-1740, CUR-1770, CUR-1916, CUR-1924, CUR-1945 correspond to the samples treated with SEQ ID NOS: 45, 57, 70, 78 and 93, respectively. Figure 21 shows up-regulation of SCN1A protein in SK-N-AS cells treated with oligonucleotides complementary to SCN1A natural antisense, quantified using ELISA. SK-N-AS cells were treated with antisense oligonucleotides complementary to SCN1A natural antisense at 0 or 20 nM. After 48 hours, the cells were transferred to 96-well plates for 24 hours, then fixed and used for SCN1A and actin ELISA. The OD reading of the SCN1A signal was corrected relative to the actin signal of the same experimental conditions. The corrected SCN1A signal in cells dosed with 0 nM oligonucleotide was used as the baseline (100%). The bars indicated by CUR-1740, CUR-1770, CUR-1924, CUR-1945 correspond to the samples treated with SEQ ID NOS: 45, 57, 78 and 93, respectively. Figure 22 shows the product of the second round of PCR of 3' RACE of SCN1A natural antisense transcript BG724147. 3' RACE for: a) total RNA with adenosine from HepG2 cells; b-poly A RNA isolated from HepG2 cells; c-primary human fibroblasts with mutations associated with Dravid syndrome Total RNA with adenosine added; d-poly A RNA isolated from primary human fibroblasts with Dravid syndrome-associated mutations. A 1% agarose gel/1 x TAE negative stained with GelRed (GenScript, catalog number M00120) is shown. The arrows show a bright band shared by HepG2 cells and primary human fibroblasts with Dravid syndrome-associated mutations, demonstrating the presence of BG724147 natural antisense transcripts in these cells. Sequence Listing - SEQ ID NO: 1: Homo sapiens voltage-gated sodium channel type I alpha subunit (SCN1A), transcript variant 1, mRNA (NCBI accession number: NM_001165963); SEQ ID NO: 2: Homo sapiens Voltage-gated sodium channel type II alpha subunit (SCN2A), transcript variant 1, mRNA (NCBI accession number: NM_021007); SEQ ID NO: 3: Homo sapiens voltage-gated sodium ion channel type III alpha subunit (SCN3A), transcript variant 1, mRNA (NCBI accession number: NM_006922); SEQ ID NO: 4: Homo sapiens voltage-gated sodium channel type IV subunit (SCN4A), mRNA (NCBI accession number: NM_000334) SEQ ID NO: 5: Homo sapiens voltage-gated sodium channel V-alpha subunit (SCN5A), transcript variant 1, mRNA (NCBI accession number: NM_198056); SEQ ID NO: 6: Homo sapiens voltage gating Sodium channel type VII alpha (SCN7A), mRNA (NCBI accession number: NM_002976); SEQ ID NO: 7: Homo sapiens voltage-gated sodium channel VIII type alpha subunit (SCN8A), transcript variant 1, mRNA (NCBI accession number: NM_014191); SEQ ID NO: 8: Homo sapiens voltage-gated sodium ion channel type IX alpha subunit (SCN9A), mRNA (NCBI registry No.: NM_002977); SEQ ID NO: 9: Homo sapiens voltage-gated sodium channel X-type alpha subunit (SCN10A), mRNA (NCBI accession number: NM_006514); SEQ ID NO: 10: Homo sapiens voltage-gated sodium Ion channel type XI alpha subunit (SCN11A), mRNA (NCBI accession number: NM_014139); SEQ ID NO: 11: Homo sapiens voltage-gated sodium channel alpha subunit SCN12A (SCN12A) mRNA, complete cds (NCBI registration number :AF109737); SEQ ID NO: 12: native SCN1A antisense sequence (extended BG724147); SEQ ID NO: 13: native SCN1A antisense sequence (Hs. 662210); SEQ ID NO: 14: native SCN1A antisense sequence ( AA383040); SEQ ID NO: 15: native SCN1A antisense sequence (BC029452); SEQ ID NO: 16: native SCN1A antisense sequence (AA630035); SEQ ID NO: 17: native SCN1A antisense sequence (BE566126); SEQ ID NO: 18: native SCN1A antisense sequence (BF673100); SEQ ID NO: 19: native SCN1A antisense sequence (BG181807); SEQ ID NO: 20: native SCN1A antisense sequence (BG183871); SEQ ID NO: 21: natural SCN1A antisense sequence (BG215777); SEQ ID NO: 22: natural SCN1A antisense sequence (BG227970); SEQ ID NO: 23: natural SCN1A antisense sequence (BM90552 7); SEQ ID NO: 24: native SCN1A antisense sequence (BU180772); SEQ ID NO: 25: mouse native SCN1A antisense sequence (BG724147 ExtMouse); SEQ ID NO: 26: mouse native SCN1A antisense sequence ( Hs.662210 mouse AS1); SEQ ID NO: 27: mouse native SCN1A antisense sequence (Hs.662210 mouse AS2); SEQ ID NO: 28: mouse native SCN1A antisense sequence (Hs.662210 mouse AS3) SEQ ID NOs: 29 to 94: Antisense oligonucleotides. SEQ ID NOS: 95 and 96 are the reverse complement of the antisense oligonucleotides SEQ ID NOS: 58 and 59, respectively. * indicates a phosphorothioate linkage, + indicates LNA, "r" indicates RNA and "m" indicates a methyl group on the 2' oxygen atom on the designated sugar moiety of the oligonucleotide.

Claims (56)

一種活體外調節電壓門控鈉離子通道α次單元(SCNA)聚核苷酸在生物系統中之功能及/或表現之方法,其包含:使該系統與至少一種長度5至30個核苷酸之反義寡核苷酸接觸,其中該至少一種寡核苷酸與電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之天然反義之反向互補序列具有至少50%序列一致性;藉此調節該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之功能及/或表現。A method for the in vitro regulation of the function and/or expression of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide in a biological system comprising: rendering the system with at least one nucleotide of 5 to 30 nucleotides in length An antisense oligonucleotide contact, wherein the at least one oligonucleotide has at least 50% sequence identity to a reverse complement of a natural antisense of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide; Thereby the function and/or performance of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide is modulated. 如請求項1之調節電壓門控鈉離子通道α次單元SCN1A聚核苷酸在生物系統中之功能及/或表現的方法,其包含:使該生物系統與至少一種長度5至30個核苷酸之反義寡核苷酸接觸,其中該至少一種寡核苷酸與包含以下天然反義轉錄物核苷酸內5至30個連續核苷酸之聚核苷酸的反向互補序列具有至少50%序列一致性:SEQ ID NO: 12之1至1123及SEQ ID NO: 13之1至2352、SEQ ID NO: 14之1至267、SEQ ID NO: 15之1至1080、SEQ ID NO: 16之1至173、SEQ ID NO: 17之1至618、SEQ ID NO: 18之1至871、SEQ ID NO: 19之1至304、SEQ ID NO: 20之1至293、SEQ ID NO: 21之1至892、SEQ ID NO: 22之1至260、SEQ ID NO: 23之1至982、SEQ ID NO: 24之1至906、SEQ ID NO: 25之1至476、SEQ ID NO: 26之1至185、SEQ ID NO: 27之1至162及SEQ ID NO: 28之1至94;藉此調節該電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之功能及/或表現。A method of regulating the function and/or performance of a voltage-gated sodium ion channel alpha subunit SCN1A polynucleotide in a biological system according to claim 1, comprising: bringing the biological system to at least one 5 to 30 nucleosides in length An acid antisense oligonucleotide contact, wherein the at least one oligonucleotide has at least a reverse complement sequence comprising a polynucleotide of 5 to 30 contiguous nucleotides within the nucleotide of the natural antisense transcript below 50% sequence identity: SEQ ID NO: 12 to 1123 and SEQ ID NO: 13 to 1 to 2352, SEQ ID NO: 14 to 1 to 267, SEQ ID NO: 15 to 1 to 1080, SEQ ID NO: 16 to 1 173, 1 to 618 of SEQ ID NO: 17, 1 to 871 of SEQ ID NO: 18, 1 to 304 of SEQ ID NO: 19, 1 to 293 of SEQ ID NO: 20, SEQ ID NO: 21 to 1 892, 1 to 260 of SEQ ID NO: 22, 1 to 982 of SEQ ID NO: 23, 1 to 906 of SEQ ID NO: 24, 1 to 476 of SEQ ID NO: 25, SEQ ID NO: 26 to 1 185, SEQ ID NO: 27 to 1 to 162, and SEQ ID NO: 28 to 1 to 94; thereby modulating the voltage-gated sodium ion channel type I alpha subunit (SCN1A) polynucleotide Function and / or performance. 一種活體外調節電壓門控鈉離子通道α次單元(SCNA)聚核苷酸在患者細胞或組織中之功能及/或表現之方法,其包含:使該等細胞或組織與至少一種長度5至30個核苷酸之反義寡核苷酸接觸,其中該寡核苷酸與該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義寡核苷酸具有至少50%序列一致性;藉此調節該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸在患者細胞或組織中或在活體外之功能及/或表現。A method for the in vitro regulation of the function and/or expression of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide in a patient cell or tissue, comprising: bringing the cells or tissues to at least one length 5 to a 30 nucleotide antisense oligonucleotide contact, wherein the oligonucleotide has at least 50% sequence to the antisense oligonucleotide of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide Consistency; thereby modulating the function and/or performance of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide in a patient cell or tissue or in vitro. 如請求項3之調節電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸在患者細胞或組織中之功能及/或表現的方法,其包含:使該生物系統與至少一種長度5至30個核苷酸之反義寡核苷酸接觸,其中該至少一種寡核苷酸與包含以下天然反義轉錄物核苷酸內5至30個連續核苷酸之聚核苷酸的反向互補序列具有至少50%序列一致性:SEQ ID NO: 12之1至1123及SEQ ID NO: 13之1至2352、SEQ ID NO: 14之1至267、SEQ ID NO: 15之1至1080、SEQ ID NO: 16之1至173、SEQ ID NO: 17之1至618、SEQ ID NO: 18之1至871、SEQ ID NO: 19之1至304、SEQ ID NO: 20之1至293、SEQ ID NO: 21之1至892、SEQ ID NO: 22之1至260、SEQ ID NO: 23之1至982、SEQ ID NO: 24之1至906、SEQ ID NO: 25之1至476、SEQ ID NO: 26之1至185、SEQ ID NO: 27之1至162及SEQ ID NO: 28之1至94;藉此調節該電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之功能及/或表現。A method of regulating the function and/or expression of a voltage-gated sodium ion channel type I alpha subunit (SCN1A) polynucleotide in a patient cell or tissue of claim 3, comprising: causing the biological system to be at least one An antisense oligonucleotide of 5 to 30 nucleotides in length, wherein the at least one oligonucleotide and a polynucleotide comprising 5 to 30 contiguous nucleotides within the nucleotide of the natural antisense transcript below The reverse complement sequence has at least 50% sequence identity: SEQ ID NO: 12 to 1123 and SEQ ID NO: 13 to 1 to 2352, SEQ ID NO: 14 to 1 to 267, and SEQ ID NO: 15 Up to 1080, SEQ ID NO: 16 to 1 to 173, SEQ ID NO: 17 to 1 to 618, SEQ ID NO: 18 to 1 to 871, SEQ ID NO: 19 to 1 to 304, and SEQ ID NO: 20 To 293, 1 to 892 of SEQ ID NO: 21, 1 to 260 of SEQ ID NO: 22, 1 to 982 of SEQ ID NO: 23, 1 to 906 of SEQ ID NO: 24, 1 of SEQ ID NO: 25 To 476, SEQ ID NO: 26 to 1 to 185, SEQ ID NO: 27 to 1 to 162, and SEQ ID NO: 28 to 1 to 94; thereby modulating the voltage-gated sodium ion channel type I alpha subunit ( SCN1A) The function and/or performance of a polynucleotide. 一種活體外調節電壓門控鈉離子通道α次單元(SCNA)聚核苷酸在生物系統中之功能及/或表現之方法,其包含:使該系統與至少一種靶向該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之天然反義寡核苷酸之一區域的反義寡核苷酸接觸;藉此調節該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之功能及/或表現。A method for the in vitro regulation of the function and/or performance of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide in a biological system, comprising: targeting the system with at least one of the voltage-gated sodium ions Antisense oligonucleotide contact of one of the natural antisense oligonucleotides of the channel alpha subunit (SCNA) polynucleotide; thereby modulating the voltage-gated sodium channel alpha subunit (SCNA) polynucleoside The function and / or performance of acid. 如請求項5之方法,其中相對於對照,該電壓門控鈉離子通道α次單元(SCNA)之功能及/或表現在活體外增加。The method of claim 5, wherein the function and/or performance of the voltage-gated sodium ion channel alpha subunit (SCNA) is increased in vitro relative to the control. 如請求項5之方法,其中該至少一種反義寡核苷酸靶向電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之天然反義序列。The method of claim 5, wherein the at least one antisense oligonucleotide targets a natural antisense sequence of a voltage-gated sodium ion channel type I alpha unit (SCN1A) polynucleotide. 如請求項5之方法,其中該至少一種反義寡核苷酸靶向包含電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之編碼及/或非編碼核酸序列的核酸序列。The method of claim 5, wherein the at least one antisense oligonucleotide targets a nucleic acid comprising a voltage-gated sodium ion channel type I alpha subunit (SCN1A) polynucleotide encoding and/or non-coding nucleic acid sequence sequence. 如請求項5之方法,其中該至少一種反義寡核苷酸靶向電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之重疊及/或不重疊序列。The method of claim 5, wherein the at least one antisense oligonucleotide targets an overlapping and/or non-overlapping sequence of a voltage-gated sodium ion channel type I alpha unit (SCN1A) polynucleotide. 如請求項5之方法,其中該至少一種反義寡核苷酸包含一或多個選自以下之修飾:至少一個經修飾之糖部分、至少一個經修飾之核苷間鍵聯、至少一個經修飾之核苷酸及其組合。The method of claim 5, wherein the at least one antisense oligonucleotide comprises one or more modifications selected from the group consisting of at least one modified sugar moiety, at least one modified internucleoside linkage, at least one Modified nucleotides and combinations thereof. 如請求項10之方法,其中該一或多個修飾包含至少一個選自以下之經修飾之糖部分:2'-O-甲氧基乙基修飾之糖部分、2'-甲氧基修飾之糖部分、2'-O-烷基修飾之糖部分、雙環糖部分及其組合。The method of claim 10, wherein the one or more modifications comprise at least one modified sugar moiety selected from the group consisting of 2'-O-methoxyethyl modified sugar moieties, 2'-methoxy modification A sugar moiety, a 2'-O-alkyl modified sugar moiety, a bicyclic sugar moiety, and combinations thereof. 如請求項10之方法,其中該一或多個修飾包含至少一個選自以下之經修飾之核苷間鍵聯:硫代磷酸酯、2'-O甲氧基乙基(MOE)、2'-氟、烷基膦酸酯、二硫代磷酸酯、硫代烷基膦酸酯、胺基磷酸酯(phosphoramidate)、胺基甲酸酯、碳酸酯、磷酸三酯、乙醯胺酸酯(acetami-date)、羧甲基酯及其組合。The method of claim 10, wherein the one or more modifications comprise at least one modified internucleoside linkage selected from the group consisting of phosphorothioate, 2'-Omethoxyethyl (MOE), 2' - fluorine, alkyl phosphonates, dithiophosphates, thioalkyl phosphonates, phosphoramidates, urethanes, carbonates, phosphates, acetates ( Acetami-date), carboxymethyl esters, and combinations thereof. 如請求項10之方法,其中該一或多個修飾包含至少一個選自以下之經修飾之核苷酸:肽核酸(PNA)、鎖核酸(LNA)、阿糖核酸(FANA),其類似物、衍生物及組合。The method of claim 10, wherein the one or more modifications comprise at least one modified nucleotide selected from the group consisting of a peptide nucleic acid (PNA), a locked nucleic acid (LNA), an arabinic acid (FANA), an analog thereof , derivatives and combinations. 如請求項1之方法,其中該至少一種寡核苷酸包含至少一種如SEQ ID NO: 29至94闡述之寡核苷酸序列。The method of claim 1, wherein the at least one oligonucleotide comprises at least one oligonucleotide sequence as set forth in SEQ ID NOs: 29-94. 一種活體外調節電壓門控鈉離子通道α次單元(SCNA)基因在哺乳動物細胞或組織中之功能及/或表現之方法,其包含:在活體外使該等細胞或組織與至少一種長度5至30個核苷酸之短干擾RNA(siRNA)寡核苷酸接觸,該至少一種siRNA寡核苷酸對電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義聚核苷酸具有特異性,其中該至少一種siRNA寡核苷酸與該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義及/或有義核酸分子之至少約5個連續核酸的互補序列具有至少50%序列一致性;及調節電壓門控鈉離子通道α次單元(SCNA)在哺乳動物細胞或組織中之功能及/或表現。A method for the in vitro regulation of the function and/or expression of a voltage-gated sodium channel alpha subunit (SCNA) gene in a mammalian cell or tissue, comprising: cultivating the cell or tissue with at least one length 5 in vitro Up to 30 nucleotide short interfering RNA (siRNA) oligonucleotide contacts, antisense polynucleosides of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide The acid is specific, wherein the at least one siRNA oligonucleotide is at least about 5 contiguous nucleic acids of the antisense and/or sense nucleic acid molecule of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide The complementary sequence has at least 50% sequence identity; and modulates the function and/or expression of a voltage-gated sodium ion channel alpha subunit (SCNA) in a mammalian cell or tissue. 如請求項15之方法,其中該寡核苷酸與該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義及/或有義核酸分子互補之至少約5個連續核酸的序列具有至少80%序列一致性。The method of claim 15, wherein the oligonucleotide is at least about 5 contiguous nucleic acids complementary to an antisense and/or sense nucleic acid molecule of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide. The sequence has at least 80% sequence identity. 一種活體外調節電壓門控鈉離子通道α次單元(SCNA)在哺乳動物細胞或組織中之功能及/或表現之方法,其包含:在活體外使該等細胞或組織與至少一種長度約5至30個核苷酸之對電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之有義及/或天然反義股的非編碼及/或編碼序列具有特異性之反義寡核苷酸接觸,其中該至少一種反義寡核苷酸與至少一種如SEQ ID NO: 1至28闡述之核酸序列具有至少50%序列一致性;及調節該電壓門控鈉離子通道α次單元(SCNA)在哺乳動物細胞或組織中之功能及/或表現。A method for the in vitro regulation of the function and/or expression of a voltage-gated sodium ion channel alpha subunit (SCNA) in a mammalian cell or tissue, comprising: subjecting the cell or tissue to at least one length of about 5 in vitro Antisense oligonucleotides specific to a non-coding and/or coding sequence of a 30-nucleotide pair of voltage-gated sodium channel alpha subunit (SCNA) polynucleotides and/or natural antisense strands Glucuronide contact, wherein the at least one antisense oligonucleotide has at least 50% sequence identity to at least one of the nucleic acid sequences set forth in SEQ ID NOS: 1 to 28; and modulating the voltage-gated sodium ion channel alpha subunit ( SCNA) Function and/or performance in mammalian cells or tissues. 一種至少一種長度5至30個核苷酸之反義寡核苷酸之用途,其係用於製造調節電壓門控鈉離子通道α次單元(SCNA)聚核苷酸在生物系統中之功能及/或表現;其中該至少一種寡核苷酸與電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之天然反義之反向互補序列具有至少50%序列一致性。Use of at least one antisense oligonucleotide of 5 to 30 nucleotides in length for the manufacture of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide in a biological system and And/or performance; wherein the at least one oligonucleotide has at least 50% sequence identity to a natural antisense reverse complement of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide. 如請求項18之用途,其中該寡核苷酸與包含以下天然反義轉錄物核苷酸內5至30個連續核苷酸之聚核苷酸的反向互補序列具有至少50%序列一致性:SEQ ID NO: 12之1至1123及SEQ ID NO: 13之1至2352、SEQ ID NO: 14之1至267、SEQ ID NO: 15之1至1080、SEQ ID NO: 16之1至173、SEQ ID NO: 17之1至618、SEQ ID NO: 18之1至871、SEQ ID NO: 19之1至304、SEQ ID NO: 20之1至293、SEQ ID NO: 21之1至892、SEQ ID NO: 22之1至260、SEQ ID NO: 23之1至982、SEQ ID NO: 24之1至906、SEQ ID NO: 25之1至476、SEQ ID NO: 26之1至185、SEQ ID NO: 27之1至162及SEQ ID NO: 28之1至94。The use of claim 18, wherein the oligonucleotide has at least 50% sequence identity to a reverse complement comprising a polynucleotide of 5 to 30 contiguous nucleotides within the nucleotide of the natural antisense transcript: SEQ ID NO: 12 to 1123 and SEQ ID NO: 13 to 1 to 2352, SEQ ID NO: 14 to 1 to 267, SEQ ID NO: 15 to 1 to 1080, and SEQ ID NO: 16 to 1 to 173 SEQ ID NO: 17 to 1 to 618, SEQ ID NO: 18 to 1 to 871, SEQ ID NO: 19 to 1 to 304, SEQ ID NO: 20 to 1 to 293, and SEQ ID NO: 21 to 1 to 892 1 to 260 of SEQ ID NO: 22, 1 to 982 of SEQ ID NO: 23, 1 to 906 of SEQ ID NO: 24, 1 to 476 of SEQ ID NO: 25, 1 to 185 of SEQ ID NO: 26. SEQ ID NO: 27 to 1 to 162 and SEQ ID NO: 28 to 1 to 94. 一種至少一種長度5至30個核苷酸之反義寡核苷酸之用途,其係用於製造供調節電壓門控鈉離子通道α次單元(SCNA)聚核苷酸在患者細胞或組織中之功能及/或表現之藥劑,其中該寡核苷酸與該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義寡核苷酸具有至少50%序列一致性。Use of at least one antisense oligonucleotide of 5 to 30 nucleotides in length for the manufacture of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide in a patient cell or tissue The agent and/or agent of the agent, wherein the oligonucleotide has at least 50% sequence identity to the antisense oligonucleotide of the voltage-gated sodium channel alpha subunit (SCNA) polynucleotide. 如請求項20之用途,其中該寡核苷酸與包含以下天然反義轉錄物核苷酸內5至30個連續核苷酸之聚核苷酸的反向互補序列具有至少50%序列一致性:SEQ ID NO: 12之1至1123及SEQ ID NO: 13之1至2352、SEQ ID NO: 14之1至267、SEQ ID NO: 15之1至1080、SEQ ID NO: 16之1至173、SEQ ID NO: 17之1至618、SEQ ID NO: 18之1至871、SEQ ID NO: 19之1至304、SEQ ID NO: 20之1至293、SEQ ID NO: 21之1至892、SEQ ID NO: 22之1至260、SEQ ID NO: 23之1至982、SEQ ID NO: 24之1至906、SEQ ID NO: 25之1至476、SEQ ID NO: 26之1至185、SEQ ID NO: 27之1至162及SEQ ID NO: 28之1至94。The use of claim 20, wherein the oligonucleotide has at least 50% sequence identity to a reverse complement comprising a polynucleotide of 5 to 30 contiguous nucleotides within the nucleotide of the natural antisense transcript: SEQ ID NO: 12 to 1123 and SEQ ID NO: 13 to 1 to 2352, SEQ ID NO: 14 to 1 to 267, SEQ ID NO: 15 to 1 to 1080, and SEQ ID NO: 16 to 1 to 173 SEQ ID NO: 17 to 1 to 618, SEQ ID NO: 18 to 1 to 871, SEQ ID NO: 19 to 1 to 304, SEQ ID NO: 20 to 1 to 293, and SEQ ID NO: 21 to 1 to 892 1 to 260 of SEQ ID NO: 22, 1 to 982 of SEQ ID NO: 23, 1 to 906 of SEQ ID NO: 24, 1 to 476 of SEQ ID NO: 25, 1 to 185 of SEQ ID NO: 26. SEQ ID NO: 27 to 1 to 162 and SEQ ID NO: 28 to 1 to 94. 一種靶向電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之天然反義寡核苷酸之一區域的反義寡核苷酸之用途,其係用於製造供調節電壓門控鈉離子通道α次單元(SCNA)聚核苷酸在生物系統中之功能及/或表現之藥劑。Use of an antisense oligonucleotide targeting a region of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide in the region of a natural antisense oligonucleotide, which is used in the manufacture of regulated voltage gating An agent that functions and/or expresses a sodium ion channel alpha subunit (SCNA) polynucleotide in a biological system. 如請求項22之用途,其中相對於對照,該電壓門控鈉離子通道α次單元(SCNA)之功能及/或表現在活體內增加。The use of claim 22, wherein the function and/or performance of the voltage-gated sodium ion channel alpha subunit (SCNA) is increased in vivo relative to a control. 如請求項22之用途,其中該反義寡核苷酸靶向電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之天然反義序列。The use of claim 22, wherein the antisense oligonucleotide targets a natural antisense sequence of a voltage-gated sodium ion channel type I alpha subunit (SCN1A) polynucleotide. 如請求項22之用途,其中該反義寡核苷酸靶向包含電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之編碼及/或非編碼核酸序列之核酸序列。The use of claim 22, wherein the antisense oligonucleotide targets a nucleic acid sequence comprising a voltage-gated sodium ion channel type I alpha subunit (SCN1A) polynucleotide encoding and/or non-coding nucleic acid sequence. 如請求項22之用途,其中該至少一種反義寡核苷酸靶向電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之重疊及/或不重疊序列。The use of claim 22, wherein the at least one antisense oligonucleotide targets an overlapping and/or non-overlapping sequence of a voltage-gated sodium ion channel type I alpha unit (SCN1A) polynucleotide. 如請求項22之用途,其中該反義寡核苷酸包含一或多個選自以下之修飾:至少一個經修飾之糖部分、至少一個經修飾之核苷間鍵聯、至少一個經修飾之核苷酸及其組合。The use of claim 22, wherein the antisense oligonucleotide comprises one or more modifications selected from the group consisting of at least one modified sugar moiety, at least one modified internucleoside linkage, at least one modified Nucleotides and combinations thereof. 如請求項27之用途,其中該一或多個修飾包含至少一個選自以下之經修飾之糖部分:2'-O-甲氧基乙基修飾之糖部分、2'-甲氧基修飾之糖部分、2'-O-烷基修飾之糖部分、雙環糖部分及其組合。The use of claim 27, wherein the one or more modifications comprise at least one modified sugar moiety selected from the group consisting of 2'-O-methoxyethyl modified sugar moieties, 2'-methoxy modification A sugar moiety, a 2'-O-alkyl modified sugar moiety, a bicyclic sugar moiety, and combinations thereof. 如請求項27之用途,其中該一或多個修飾包含至少一個選自以下之經修飾之核苷間鍵聯:硫代磷酸酯、2'-O甲氧基乙基(MOE)、2'-氟、烷基膦酸酯、二硫代磷酸酯、硫代烷基膦酸酯、胺基磷酸酯、胺基甲酸酯、碳酸酯、磷酸三酯、乙醯胺酸酯、羧甲基酯及其組合。The use of claim 27, wherein the one or more modifications comprise at least one modified internucleoside linkage selected from the group consisting of phosphorothioate, 2'-Omethoxyethyl (MOE), 2' -Fluorine, alkylphosphonate, dithiophosphate, thioalkylphosphonate, amino phosphate, urethane, carbonate, phosphotriester, acetate, carboxymethyl Esters and combinations thereof. 如請求項27之用途,其中該一或多個修飾包含至少一個選自以下之經修飾之核苷酸:肽核酸(PNA)、鎖核酸(LNA)、阿糖核酸(FANA),其類似物、衍生物及組合。The use of claim 27, wherein the one or more modifications comprise at least one modified nucleotide selected from the group consisting of peptide nucleic acid (PNA), locked nucleic acid (LNA), arabinic acid (FANA), analogs thereof , derivatives and combinations. 如請求項18之用途,其中該寡核苷酸包含至少一種如SEQ ID NO: 29至94闡述之寡核苷酸序列。The use of claim 18, wherein the oligonucleotide comprises at least one oligonucleotide sequence as set forth in SEQ ID NOs: 29-94. 一種至少一種長度5至30個核苷酸之短干擾RNA(siRNA)寡核苷酸之用途,其係用於製造供調節電壓門控鈉離子通道α次單元(SCNA)基因在哺乳動物細胞或組織中之功能及/或表現之藥劑;其中該至少一種siRNA寡核苷酸對電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義聚核苷酸具有特異性,其中該至少一種siRNA寡核苷酸與該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義及/或有義核酸分子之至少約5個連續核酸的互補序列具有至少50%序列一致性。Use of at least one short interfering RNA (siRNA) oligonucleotide of 5 to 30 nucleotides in length for the production of a voltage-gated sodium ion channel alpha subunit (SCNA) gene in a mammalian cell or An agent for function and/or performance in a tissue; wherein the at least one siRNA oligonucleotide is specific for an antisense polynucleotide of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide, wherein At least one siRNA oligonucleotide having at least 50% sequence to the antisense of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide and/or the complement of at least about 5 contiguous nucleic acids of the sense nucleic acid molecule consistency. 如請求項32之用途,其中該寡核苷酸與該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義及/或有義核酸分子互補之至少約5個連續核酸的序列具有至少80%序列一致性。The use of claim 32, wherein the oligonucleotide is at least about 5 contiguous nucleic acids complementary to an antisense and/or sense nucleic acid molecule of the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide. The sequence has at least 80% sequence identity. 一種長度約5至30個核苷酸之對於電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之有義及/或天然反義股的非編碼及/或編碼序列具有特異性之反義寡核苷酸之用途,其係用於製造供調節電壓門控鈉離子通道α次單元(SCNA)在哺乳動物細胞或組織中之功能及/或表現之藥劑,其中該反義寡核苷酸與至少一種如SEQ ID NO: 1至28闡述之核酸序列具有至少50%序列一致性。A non-coding and/or coding sequence of about 5 to 30 nucleotides in length for a sense-activated and/or natural antisense strand of a voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide Use of an antisense oligonucleotide for the manufacture of an agent for regulating the function and/or expression of a voltage-gated sodium ion channel alpha subunit (SCNA) in a mammalian cell or tissue, wherein the antisense oligonucleotide The nucleotide has at least 50% sequence identity to at least one of the nucleic acid sequences set forth in SEQ ID NOS: 1 to 28. 一種包含至少一個修飾之合成經修飾的寡核苷酸,其中該至少一個修飾係選自:至少一個經修飾之糖部分;至少一個經修飾之核苷酸間鍵聯;至少一個經修飾之核苷酸及其組合;其中該寡核苷酸為反義化合物,相較於正常對照,活體內或活體外與電壓門控鈉離子通道α次單元(SCNA)基因雜交且調節其功能及/或表現,且其中該寡核苷酸與該電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之反義及/或有義核酸分子及其對偶基因、同源物、同功異型物(isoforms)、變異體、衍生物、突變體、片段或組合互補之至少約5個連續核酸的序列具有至少50%序列一致性。A synthetic modified oligonucleotide comprising at least one modification, wherein the at least one modification is selected from the group consisting of: at least one modified sugar moiety; at least one modified internucleotide linkage; at least one modified core And a combination thereof; wherein the oligonucleotide is an antisense compound that hybridizes in vivo or in vitro to a voltage-gated sodium ion channel alpha subunit (SCNA) gene and modulates its function and/or Characterization, and wherein the oligonucleotide and the voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide antisense and/or sense nucleic acid molecule and its dual gene, homologue, isoform Sequences of at least about 5 contiguous nucleic acids that are complementary to isoforms, variants, derivatives, mutants, fragments or combinations have at least 50% sequence identity. 如請求項35之寡核苷酸,其中該寡核苷酸之長度為5至30個核苷酸且與該SCNA基因之天然反義轉錄物內之5-30個連續核苷酸的反向互補序列具有至少50%序列一致性。The oligonucleotide of claim 35, wherein the oligonucleotide is 5 to 30 nucleotides in length and is inverted from 5-30 contiguous nucleotides within the native antisense transcript of the SCNA gene The complementary sequence has at least 50% sequence identity. 如請求項36之寡核苷酸,其中該至少一個修飾包含選自由以下組成之群之核苷酸間鍵聯:硫代磷酸酯、烷基膦酸酯、二硫代磷酸酯、硫代烷基膦酸酯、胺基磷酸酯、胺基甲酸酯、碳酸酯、磷酸三酯、乙醯胺酸酯、羧甲基酯及其組合。The oligonucleotide of claim 36, wherein the at least one modification comprises an internucleotide linkage selected from the group consisting of phosphorothioate, alkyl phosphonate, dithiophosphate, thioalkyl Phosphonates, aminophosphates, urethanes, carbonates, phosphates, acetates, carboxymethyl esters, and combinations thereof. 如請求項36之寡核苷酸,其中該寡核苷酸包含至少一個硫代磷酸酯核苷酸間鍵聯。The oligonucleotide of claim 36, wherein the oligonucleotide comprises at least one phosphorothioate internucleotide linkage. 如請求項36之寡核苷酸,其中該寡核苷酸包含硫代磷酸酯核苷酸間鍵聯之骨架。The oligonucleotide of claim 36, wherein the oligonucleotide comprises a backbone of phosphorothioate internucleotide linkages. 如請求項36之寡核苷酸,其中該寡核苷酸包含至少一個經修飾之核苷酸,該經修飾之核苷酸選自:肽核酸、鎖核酸(LNA),其類似物、衍生物及組合。The oligonucleotide of claim 36, wherein the oligonucleotide comprises at least one modified nucleotide selected from the group consisting of: a peptide nucleic acid, a locked nucleic acid (LNA), an analog thereof, and a derivative. Things and combinations. 如請求項36之寡核苷酸,其中該寡核苷酸包含多個修飾,其中該等修飾包含選自以下之經修飾之核苷酸:硫代磷酸酯、烷基膦酸酯、二硫代磷酸酯、硫代烷基膦酸酯、胺基磷酸酯、胺基甲酸酯、碳酸酯、磷酸三酯、乙醯胺酸酯、羧甲基酯及其組合。The oligonucleotide of claim 36, wherein the oligonucleotide comprises a plurality of modifications, wherein the modifications comprise modified nucleotides selected from the group consisting of phosphorothioates, alkyl phosphonates, disulfides Phosphate, thioalkylphosphonate, amino phosphate, urethane, carbonate, phosphotriester, acetate, carboxymethyl ester, and combinations thereof. 如請求項36之寡核苷酸,其中該寡核苷酸包含多個修飾,其中該等修飾包含選自以下之經修飾之核苷酸:肽核酸、鎖核酸(LNA),其類似物、衍生物及組合。The oligonucleotide of claim 36, wherein the oligonucleotide comprises a plurality of modifications, wherein the modifications comprise modified nucleotides selected from the group consisting of peptide nucleic acids, locked nucleic acids (LNA), analogs thereof, Derivatives and combinations. 如請求項36之寡核苷酸,其中該寡核苷酸包含至少一個選自以下之經修飾之糖部分:2'-O-甲氧基乙基修飾之糖部分、2'-甲氧基修飾之糖部分、2'-O-烷基修飾之糖部分、雙環糖部分及其組合。The oligonucleotide of claim 36, wherein the oligonucleotide comprises at least one modified sugar moiety selected from the group consisting of a 2'-O-methoxyethyl modified sugar moiety, 2'-methoxy Modified sugar moieties, 2'-O-alkyl modified sugar moieties, bicyclic sugar moieties, and combinations thereof. 如請求項36之寡核苷酸,其中該寡核苷酸包含多個修飾,其中該等修飾包含選自以下之經修飾之糖部分:2'-O-甲氧基乙基修飾之糖部分、2'-甲氧基修飾之糖部分、2'-O-烷基修飾之糖部分、雙環糖部分及其組合。The oligonucleotide of claim 36, wherein the oligonucleotide comprises a plurality of modifications, wherein the modifications comprise a modified sugar moiety selected from the group consisting of 2'-O-methoxyethyl modified sugar moieties 2'-methoxy modified sugar moiety, 2'-O-alkyl modified sugar moiety, bicyclic sugar moiety, and combinations thereof. 如請求項36之寡核苷酸,其中該寡核苷酸具有至少約5至30個核苷酸之長度且與電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之反義及/或有義股雜交,其中該寡核苷酸與該電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之反義及/或有義編碼及/或非編碼核酸序列之至少約5個連續核酸的互補序列具有至少約60%序列一致性。The oligonucleotide of claim 36, wherein the oligonucleotide has a length of at least about 5 to 30 nucleotides and is associated with a voltage-gated sodium ion channel type I alpha unit (SCN1A) polynucleotide Antisense and/or sense strand hybridization, wherein the oligonucleotide is antisense and/or sense encoded and/or non-insensitive to the voltage-gated sodium channel type I alpha unit (SCN1A) polynucleotide A complementary sequence of at least about 5 contiguous nucleic acids encoding a nucleic acid sequence has at least about 60% sequence identity. 如請求項36之寡核苷酸,其中該寡核苷酸與該電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸之反義及/或有義編碼及/或非編碼核酸序列之至少約5個連續核酸的互補序列具有至少約80%序列一致性。An oligonucleotide according to claim 36, wherein the oligonucleotide is antisense and/or sense-coded and/or non-insensitive to the voltage-gated sodium ion channel type I alpha unit (SCN1A) polynucleotide A complementary sequence of at least about 5 contiguous nucleic acids encoding a nucleic acid sequence has at least about 80% sequence identity. 如請求項36之寡核苷酸,其中該寡核苷酸相較於正常對照在活體內或活體外與至少一種電壓門控鈉離子通道第I型α次單元(SCN1A)聚核苷酸雜交且調節其表現及/或功能。An oligonucleotide according to claim 36, wherein the oligonucleotide hybridizes to at least one voltage-gated sodium channel type I alpha unit (SCN1A) polynucleotide in vivo or in vitro compared to a normal control. And adjust its performance and / or function. 如請求項36之寡核苷酸,其中該寡核苷酸包含如SEQ ID NO: 29至94闡述之序列。The oligonucleotide of claim 36, wherein the oligonucleotide comprises the sequence set forth in SEQ ID NOs: 29-94. 一種醫藥組合物,其包含一或多種對於一或多種電壓門控鈉離子通道α次單元(SCNA)聚核苷酸具有特異性之如請求項35之寡核苷酸及醫藥學上可接受之賦形劑。A pharmaceutical composition comprising one or more oligonucleotides as claimed in claim 35 which are specific for one or more voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotides and pharmaceutically acceptable excipient. 如請求項49之組合物,其中該等寡核苷酸與SEQ ID NO: 29至94闡述之任一核苷酸序列具有至少約40%序列一致性。The composition of claim 49, wherein the oligonucleotides have at least about 40% sequence identity to any of the nucleotide sequences set forth in SEQ ID NOs: 29-94. 如請求項49之組合物,其中該等寡核苷酸包含如SEQ ID NO: 29至94闡述之核苷酸序列。The composition of claim 49, wherein the oligonucleotides comprise the nucleotide sequences set forth in SEQ ID NOs: 29-94. 如請求項51之組合物,其中如SEQ ID NO: 29至94闡述之寡核苷酸包含一或多個修飾或取代。The composition of claim 51, wherein the oligonucleotides set forth in SEQ ID NOs: 29 to 94 comprise one or more modifications or substitutions. 如請求項52之組合物,其中該一或多個修飾係選自:硫代磷酸酯、甲基膦酸酯、肽核酸、鎖核酸(LNA)分子及其組合。The composition of claim 52, wherein the one or more modifications are selected from the group consisting of: phosphorothioates, methylphosphonates, peptide nucleic acids, locked nucleic acid (LNA) molecules, and combinations thereof. 一種與至少一種電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之天然反義序列結合且調節該至少一種電壓門控鈉離子通道α次單元(SCNA)聚核苷酸之表現的反義寡核苷酸之用途,其係用於製造供預防或治療與至少一種電壓門控鈉離子通道α次單元(SCNA)聚核苷酸及/或其至少一種編碼產物相關之疾病的藥劑。a method of binding to a natural antisense sequence of at least one voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide and modulating the performance of the at least one voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide Use of an antisense oligonucleotide for the manufacture of a medicament for preventing or treating a disease associated with at least one voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide and/or at least one of its encoded products . 如請求項54之用途,其中與該至少一種電壓門控鈉離子通道α次單元(SCNA)聚核苷酸相關之疾病係選自:與SCNA之功能及/或表現異常相關之疾病或病症、神經疾病或病症、抽搐、疼痛(包括慢性疼痛)、涉及鈉離子通道功能障礙之受損電興奮性、與鈉離子通道功能障礙相關之疾病或病症、與電壓門控鈉離子通道α次單元活性之錯誤調控相關之疾病或病症(例如麻痺、高血鉀性週期性麻痺、先天性肌剛痙病、鉀惡化之肌強直、長Q-T症候群3、運動終板疾病(motor endplate disease)、共濟失調等)、歸因於腸神經系統之功能障礙之胃腸道疾病(例如結腸炎、迴腸炎、發炎性腸症候群等)、心血管疾病或病症(例如高血壓、充血性心臟衰竭等);涉及交感神經及副交感神經神經分佈之泌尿生殖道的疾病或病症(例如良性前列腺增生(prostate hyperplasia)、陽痿);與神經肌肉系統相關之疾病或病症(例如肌肉萎縮症、多發性硬化症、癲癇症、自閉症(autism)、偏頭痛(例如偶發性及家族性偏癱性偏頭痛等)、嬰兒期重度肌痙攣癲癇症(SMEI)、伴有熱性發作附加症(febrile seizure plus)之全身性癲癇症(GEFS+ )等)及SCNA相關之發作(seizure)病症。The use of claim 54, wherein the disease associated with the at least one voltage-gated sodium ion channel alpha subunit (SCNA) polynucleotide is selected from the group consisting of: a disease or condition associated with abnormal function and/or performance of SCNA, Neurological diseases or conditions, convulsions, pain (including chronic pain), impaired electrical excitability involving sodium ion channel dysfunction, diseases or conditions associated with sodium ion channel dysfunction, and voltage-gated sodium ion channel alpha subunit activity Diseases or conditions associated with misregulation (eg paralysis, hyperkalemia, periodic paralysis, congenital myocardium, muscle weakness of potassium deterioration, long QT syndrome 3, motor endplate disease, mutual aid) Disorders, etc., gastrointestinal diseases (such as colitis, ileitis, inflammatory bowel syndrome, etc.), cardiovascular diseases or conditions (such as hypertension, congestive heart failure, etc.) attributed to dysfunction of the enteric nervous system; Sympathetic and parasympathetic nerve distribution of the genitourinary tract diseases or conditions (eg, prostate hyperplasia, impotence); diseases associated with the neuromuscular system Disease or condition (eg, muscular dystrophy, multiple sclerosis, epilepsy, autism, migraine (eg, sporadic and familial hemiplegic migraine), severe infantile epilepsy (SMEI) , systemic epilepsy ( GEFS+ ), etc. with febrile seizure plus, and SCNA-related seizure disorders. 一種鑑別及選擇至少一種對SCNA基因之天然反義轉錄物所選目標聚核苷酸具有選擇性的寡核苷酸用於活體內投藥之方法,其包含鑑別至少一種包含至少部分互補於該所選目標聚核苷酸之反義聚核苷酸的至少5個連續核苷酸之寡核苷酸;測量在嚴格雜交條件下反義寡核苷酸與該目標聚核苷酸或該所選目標聚核苷酸反義之聚核苷酸的雜交體(hybrid)之熱熔點;及基於所獲得之資訊選擇至少一種寡核苷酸用於活體內投藥。An method for identifying and selecting at least one oligonucleotide that is selective for a selected target polynucleotide of a natural antisense transcript of a SCNA gene for in vivo administration, comprising identifying at least one comprising at least partially complementary to the An oligonucleotide of at least 5 contiguous nucleotides of an antisense polynucleotide of a polynucleotide of interest; measuring the antisense oligonucleotide under the stringent hybridization conditions with the target polynucleotide or the selected The thermal melting point of the hybrid of the polynucleotide of the target polynucleotide antisense; and based on the information obtained, at least one oligonucleotide is selected for in vivo administration.
TW107132956A 2011-06-24 2011-06-24 Antisense oligonucleotides that upregulate the expression and/or function of sodium channel, voltage-gated, type i, alpha subunit (scn1a), and use thereof TWI734935B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW107132956A TWI734935B (en) 2011-06-24 2011-06-24 Antisense oligonucleotides that upregulate the expression and/or function of sodium channel, voltage-gated, type i, alpha subunit (scn1a), and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW107132956A TWI734935B (en) 2011-06-24 2011-06-24 Antisense oligonucleotides that upregulate the expression and/or function of sodium channel, voltage-gated, type i, alpha subunit (scn1a), and use thereof

Publications (2)

Publication Number Publication Date
TW201916883A true TW201916883A (en) 2019-05-01
TWI734935B TWI734935B (en) 2021-08-01

Family

ID=67347667

Family Applications (1)

Application Number Title Priority Date Filing Date
TW107132956A TWI734935B (en) 2011-06-24 2011-06-24 Antisense oligonucleotides that upregulate the expression and/or function of sodium channel, voltage-gated, type i, alpha subunit (scn1a), and use thereof

Country Status (1)

Country Link
TW (1) TWI734935B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113274400A (en) * 2021-06-07 2021-08-20 四川大学 Use of tetrahedral framework nucleic acids in medicaments for the treatment of multiple sclerosis

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1904630A4 (en) * 2005-06-16 2009-10-21 Bionomics Ltd Methods of treatment, and diagnosis of epilepsy by detecting mutations in the scn1a gene

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113274400A (en) * 2021-06-07 2021-08-20 四川大学 Use of tetrahedral framework nucleic acids in medicaments for the treatment of multiple sclerosis
CN113274400B (en) * 2021-06-07 2022-07-12 四川大学 Use of tetrahedral framework nucleic acids in medicaments for the treatment of multiple sclerosis

Also Published As

Publication number Publication date
TWI734935B (en) 2021-08-01

Similar Documents

Publication Publication Date Title
US10793857B2 (en) Treatment of sodium channel, voltage-gated, alpha subunit (SCNA) related diseases by inhibition of natural antisense transcript to SCNA
US10253320B2 (en) Treatment of atonal homolog 1 (ATOH1) related diseases by inhibition of natural antisense transcript to ATOH1
TWI678375B (en) Treatment of frataxin (fxn) related diseases by inhibition of natural antisense transcript to fxn
TWI665301B (en) Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
CA2786535C (en) Treatment of sex hormone binding globulin (shbg) related diseases by inhibition of natural antisense transcript to shbg
US9902958B2 (en) Treatment of discs large homolog (DLG) related diseases by inhibition of natural antisense transcript to DLG
EP2519633B1 (en) Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
US10221413B2 (en) Treatment of uncoupling protein 2 (UCP2) related diseases by inhibition of natural antisense transcript to UCP2
CA2771228C (en) Treatment of &#39;c terminus of hsp70-interacting protein&#39; (chip) related diseases by inhibition of natural antisense transcript to chip
TWI675100B (en) Treatment of interferon-related developmental regulator 1 (ifrd1) related diseases by inhibition of natural antisense transcript to ifrd1
CA2765509C (en) Treatment of paraoxonase 1 (pon1) related diseases by inhibition of natural antisense transcript to pon1
CA2798401C (en) Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
US9234199B2 (en) Treatment of insulin gene (INS) related diseases by inhibition of natural antisense transcript to an insulin gene (INS)
EP2446036A2 (en) Treatment of tumor necrosis factor receptor 2 (tnfr2) related diseases by inhibition of natural antisense transcript to tnfr2
TWI642438B (en) Treatment of sodium channel, voltage-gated, alpha subunit (scna) related diseases by inhibition of natural antisense transcript to scna
TWI734935B (en) Antisense oligonucleotides that upregulate the expression and/or function of sodium channel, voltage-gated, type i, alpha subunit (scn1a), and use thereof