• 
    

    
    

      99热精品在线国产_美女午夜性视频免费_国产精品国产高清国产av_av欧美777_自拍偷自拍亚洲精品老妇_亚洲熟女精品中文字幕_www日本黄色视频网_国产精品野战在线观看 ?

      樺木酸對(duì)人胃癌SGC-7901細(xì)胞自噬的影響

      2022-07-07 07:00:34郭旭何孟奇張偉偉沈洋黃鑫孟令雪邵淑麗
      高師理科學(xué)刊 2022年6期
      關(guān)鍵詞:樺木梯度胃癌

      郭旭,何孟奇,張偉偉,沈洋,黃鑫,孟令雪,邵淑麗

      樺木酸對(duì)人胃癌SGC-7901細(xì)胞自噬的影響

      郭旭1,何孟奇1,張偉偉1,2,沈洋1,黃鑫1,孟令雪1,邵淑麗1,2

      (齊齊哈爾大學(xué) 1. 生命科學(xué)與農(nóng)林學(xué)院,2. 抗性基因工程與寒地生物多樣性保護(hù)黑龍江省重點(diǎn)實(shí)驗(yàn)室,黑龍江 齊齊哈爾 161006)

      通過不同梯度濃度的樺木酸處理SGC-7901細(xì)胞,研究樺木酸對(duì)細(xì)胞自噬相關(guān)基因表達(dá)的影響,從而確定樺木酸在胃癌細(xì)胞內(nèi)與自噬的關(guān)聯(lián).將人胃癌SGC-7091細(xì)胞分為5組,其中3組設(shè)置樺木酸的濃度梯度分別為10,20,30 mg/L,使用經(jīng)典抗癌藥物氟尿嘧啶(5-Fu)作為陽性對(duì)照組,使用0 mmol/L樺木酸為陰性對(duì)照組,每組3個(gè)復(fù)孔,處理細(xì)胞48 h,用qRT-PCR和Western blot方法檢測樺木酸對(duì)人胃癌SGC-7091細(xì)胞自噬相關(guān)基因mRNA和蛋白表達(dá)的影響,使用免疫熒光法定位并檢測SGC-7091細(xì)胞內(nèi)的蛋白.與陰性對(duì)照組相比,梯度樺木酸處理組的細(xì)胞中-Ⅱ,mRNA和蛋白的表達(dá)均顯著升高(<0.01),-Ⅰ蛋白的表達(dá)明顯降低(<0.05).同時(shí)激光共聚焦的結(jié)果顯示高濃度樺木酸誘導(dǎo)蛋白在細(xì)胞質(zhì)內(nèi)的聚集現(xiàn)象.在設(shè)置的梯度濃度范圍內(nèi),樺木酸能誘導(dǎo)人胃癌SGC-7091細(xì)胞發(fā)生自噬,且呈劑量依賴性,濃度升高誘導(dǎo)效果也升高.

      樺木酸;人胃癌SGC-7901細(xì)胞;細(xì)胞自噬

      2020年全球新增癌癥病例約1 930萬例,癌癥死亡近1 000萬例(均不包括非黑素瘤皮膚癌),其中胃癌分別占新增和死亡病例數(shù)的5.6%,7.7%[1],其誘發(fā)因素多,復(fù)發(fā)率高[2],早期診斷極為復(fù)雜,絕大部分患者在胃癌早期無癥狀,出現(xiàn)腹痛等癥狀時(shí)已是晚期.外科手術(shù)及藥物輔助治療仍是早期胃癌最主要的治療方式.由于大多數(shù)胃癌病例被診斷時(shí)已處于局部晚期甚至轉(zhuǎn)移階段,限制了根治性手術(shù)的適用性,且多半患者預(yù)后差,需化療予以緩解[3].而目前的幾種主流化療藥物(如氟尿嘧啶、順鉑等)毒副作用較大,在治療腫瘤的同時(shí)對(duì)患者自身正常組織也造成傷害,且礙于胃癌的獨(dú)特性,其對(duì)化學(xué)療法普遍不敏感.因此研發(fā)特異性強(qiáng)、副作用小的化療藥物刻不容緩.

      自噬是一種細(xì)胞營養(yǎng)匱乏導(dǎo)致的應(yīng)激所誘導(dǎo)的分解代謝降解過程,大多數(shù)情況是一種細(xì)胞保護(hù)措施[4],指細(xì)胞在特定因素(如饑餓)刺激下,調(diào)動(dòng)自噬相關(guān)蛋白分解自身物質(zhì),抵抗外來干擾,以維持內(nèi)環(huán)境穩(wěn)定的樞紐.但如果自噬過程調(diào)用頻繁,破壞原本細(xì)胞的生理結(jié)構(gòu),則也會(huì)引發(fā)細(xì)胞凋亡.通過引發(fā)癌細(xì)胞自噬進(jìn)而殺滅癌細(xì)胞是化療藥物的一種作用方式.研究表明,多種藥物可通過誘導(dǎo)自噬引發(fā)細(xì)胞凋亡[5-7].

      樺木酸(Betulinic acid,BA)是一種五環(huán)三萜類化合物,可從白樺樹、三葉樹和棗樹中提取[8].樺木酸具有廣泛的生物活性[9-13],在誘導(dǎo)細(xì)胞凋亡時(shí)也伴隨著自噬.樺木酸會(huì)通過抑制人宮頸癌細(xì)胞[14]、口腔鱗狀細(xì)胞癌[15]、乳腺癌[16]、白血病細(xì)胞[17]等的G0/G1期來抑制細(xì)胞增殖和誘導(dǎo)細(xì)胞凋亡,也會(huì)在骨髓瘤細(xì)胞[18]、大腸癌細(xì)胞[19]、肝癌細(xì)胞[20]中通過誘導(dǎo)自噬來介導(dǎo)細(xì)胞凋亡.

      但目前樺木酸對(duì)SGC-7901細(xì)胞自噬方面的研究還未見報(bào)道.本研究在體外添加樺木酸后對(duì)自噬相關(guān)指標(biāo)進(jìn)行檢測,完善樺木酸在胃癌自噬方面的研究,并為后續(xù)實(shí)驗(yàn)提供理論基礎(chǔ).

      1 材料與方法

      1.1 材料與試劑

      人胃癌SGC-7901細(xì)胞株(中國醫(yī)學(xué)科學(xué)院腫瘤研究所).

      RPMI-1640培養(yǎng)基干粉(Gibico);胎牛血清(Biological Iudustries);樺木酸(北京漢博生物有限公司);Trizol(上海生工生物工程股份有限公司);反轉(zhuǎn)錄及RT-PCR Mixture(寶生物工程(大連)有限公司);,-Ⅰ,-Ⅱ一抗,兔二抗(北京博奧森生物技術(shù)有限公司).

      1.2 細(xì)胞培養(yǎng)及藥物處理

      用含10%胎牛血清的RPMI-1640培養(yǎng)基在標(biāo)準(zhǔn)細(xì)胞培養(yǎng)箱內(nèi)(37 ℃,5%CO2)培養(yǎng)人胃癌SGC-7901細(xì)胞,每1~2 d傳代1次.細(xì)胞培養(yǎng)至對(duì)數(shù)生長期后,將細(xì)胞分為5組,設(shè)置3個(gè)梯度濃度,分別為10,20,30 mg/L,每組3個(gè)復(fù)孔,0 mg/L處理組作為陰性對(duì)照,70 μm/L的5-FU處理組作為陽性對(duì)照,處理48 h后收集細(xì)胞進(jìn)行相應(yīng)檢測.

      1.3 qRT-PCR檢測自噬相關(guān)基因mRNA的表達(dá)

      收集各組細(xì)胞,通過Trizol法提取RNA并進(jìn)行反轉(zhuǎn)錄,隨后使用cDNA作為模板,進(jìn)行qRT-PCR檢測,結(jié)果見表1.所有結(jié)果以2-△△Ct方法計(jì)算.

      表1 熒光定量引物

      1.4 Western blot檢測自噬相關(guān)基因的蛋白表達(dá)

      分別培養(yǎng)6瓶細(xì)胞,根據(jù)梯度濃度分別處理48 h后,收集細(xì)胞并提總蛋白,SDS-PAGE電泳后進(jìn)行轉(zhuǎn)膜,使用5%的脫脂奶封閉1 h,再按照蛋白大小進(jìn)行剪膜,隨后分別使用,-Ⅰ,-Ⅱ一抗(均按1∶500進(jìn)行稀釋)孵育對(duì)應(yīng)的PVDF膜,4 ℃下孵育過夜;第2天回收所有一抗,TBST洗膜3次,棄凈TBST后使用兔二抗(1∶1 000稀釋)室溫?fù)u床下避光孵育1 h,再使用TBST洗膜3次,最后通過奧德賽掃膜儀掃描條帶并分析處理.

      1.5 免疫熒光檢測人胃癌SGC-7901細(xì)胞自噬

      分別培養(yǎng)6皿細(xì)胞,待細(xì)胞數(shù)合適時(shí),根據(jù)梯度濃度設(shè)置處理細(xì)胞48 h,處理結(jié)束后使用甲醇進(jìn)行固定處理.固定結(jié)束后清洗甲醇,標(biāo)準(zhǔn)封閉液封閉1 h.一抗4 ℃過夜,TBST洗3次;二抗避光孵育1 h,TBST洗3次.隨后使用20 μL的DAPI孵育3~8 min,再使用PBST洗3次.清洗完畢后,滴加一滴抗熒光淬滅封片液,激光共聚焦顯微鏡下拍照.

      1.6 統(tǒng)計(jì)學(xué)處理

      2 結(jié)果

      2.1 qRT-PCR檢測樺木酸對(duì)自噬相關(guān)基因mRNA表達(dá)的影響

      和的mRNA表達(dá)量見表2.由表2可見,與陰性對(duì)照組相比,隨著樺木酸的質(zhì)量濃度梯度升高,和的mRNA表達(dá)逐步升高,當(dāng)樺木酸濃度為30 mg/L時(shí),和的mRNA表達(dá)量最高,5-FU陽性對(duì)照處理組與10 mg/L處理組結(jié)果相似.結(jié)果表明,樺木酸上調(diào)了人胃癌SGC-7901細(xì)胞中的,基因mRNA的表達(dá).

      表2 LC3和Beclin 1的mRNA表達(dá)量

      2.2 Western blot檢測樺木酸對(duì)自噬相關(guān)基因蛋白表達(dá)的影響

      不同濃度樺木酸對(duì)人胃癌SGC-7901細(xì)胞自噬相關(guān),蛋白表達(dá)影響見表3和圖1.由表3和圖1可見,在3組梯度濃度處理組中-Ⅱ,蛋白表達(dá)水平梯度遞增(<0.01),而-Ⅰ蛋白表達(dá)水平則隨濃度梯度變化遞減(<0.01),-Ⅱ/-Ⅰ比值隨濃度梯度變化遞增(<0.01).10 mg/mL樺木酸處理的效果與陽性對(duì)照組的效果相似.結(jié)果表明,樺木酸通過調(diào)控-Ⅱ,蛋白表達(dá)促進(jìn)胃癌SGC-7901細(xì)胞自噬.

      表3 樺木酸對(duì)Beclin 1,LC3-Ⅰ,LC3-Ⅱ蛋白水平的影響

      圖1 樺木酸對(duì)Beclin 1,LC3-Ⅰ,LC3-Ⅱ蛋白水平的影響(n=3)

      2.3 樺木酸對(duì)人胃癌SGC-7901細(xì)胞中LC3蛋白定位的影響

      通過激光共聚焦顯微鏡觀察蛋白在胃癌SGC-7901細(xì)胞內(nèi)的定位及表達(dá)變化(見圖2).鑒于30 mg/L的樺木酸處理細(xì)胞后細(xì)胞死亡過于明顯,不利于鏡下觀察和染色,因此選擇20 mg/L的質(zhì)量濃度進(jìn)行處理,48 h后蛋白在細(xì)胞質(zhì)內(nèi)的聚集現(xiàn)象顯著,5-FU陽性對(duì)照組與20 mg/L處理組有相似結(jié)果.結(jié)果表明,樺木酸的處理誘導(dǎo)胃癌SGC-7901細(xì)胞發(fā)生自噬.

      圖2 樺木酸處理細(xì)胞48 h,LC3的細(xì)胞內(nèi)定位(bar=100 μm,n=3)

      3 討論

      細(xì)胞保護(hù)性自噬是應(yīng)對(duì)化療藥物和放射治療的重要反應(yīng).在大多數(shù)情況下,抗癌治療中,自噬作用會(huì)支持癌細(xì)胞的存活;然而在某些情況下,它也會(huì)促進(jìn)細(xì)胞死亡.因此,調(diào)節(jié)自噬有可能成為提高化療和放療療效的策略之一.此外,自噬調(diào)節(jié)劑和傳統(tǒng)治療方法的結(jié)合可能使癌細(xì)胞對(duì)癌癥治療敏感.有越來越多的臨床前證據(jù)表明,靶向誘導(dǎo)自噬,利用其細(xì)胞自身功能瓦解腫瘤,其毒副作用相對(duì)較小,在聯(lián)合治療中的作用也較為明顯[21-22].如褪黑素對(duì)胃腸癌細(xì)胞的自噬具有調(diào)節(jié)作用,可通過聯(lián)合用藥增加胃腸癌細(xì)胞對(duì)藥物的敏感性[23];脫落酸在膠質(zhì)母細(xì)胞瘤細(xì)胞中可通過誘導(dǎo)腫瘤細(xì)胞自噬來抑制腫瘤的生長[24].樺木酸作為一種天然產(chǎn)物藥物,已有研究表明其對(duì)人類肺癌、乳腺癌、結(jié)腸癌等均具有明顯的細(xì)胞毒性[25-29].本實(shí)驗(yàn)研究證明,樺木酸可誘導(dǎo)人胃癌SGC-7901細(xì)胞自噬.

      自噬的誘導(dǎo)途徑有很多,其中自噬相關(guān)基因和是自噬過程的主要調(diào)節(jié)劑,許多自噬調(diào)控蛋白通過與的不同結(jié)構(gòu)域或氨基酸發(fā)生直接或間接結(jié)合,形成蛋白復(fù)合體,進(jìn)而調(diào)控自噬水平[30].而的功能主要參與了自噬小體的形成,饑餓時(shí),核會(huì)被SIRT1(sirtuin 1)脫乙?;?,并與TP53INP2一起重新分布到細(xì)胞質(zhì)中,以引發(fā)自噬[31].在自噬過程中,樺木酸可能通過觸發(fā)細(xì)胞內(nèi)的上游自噬信號(hào)(如PI3K/AKT/mTOR信號(hào)通路)來介導(dǎo)和的表達(dá)變化,同時(shí)樺木酸也可能通過對(duì)細(xì)胞內(nèi)原本自噬信號(hào)的破壞,導(dǎo)致細(xì)胞自噬失調(diào),進(jìn)而引發(fā)腫瘤細(xì)胞的過度自噬行為,導(dǎo)致細(xì)胞死亡.

      本實(shí)驗(yàn)5-FU為陽性對(duì)照組,證明隨著樺木酸濃度的增加,自噬標(biāo)志基因、mRNA和蛋白的表達(dá)量增加,但是-Ⅰ蛋白表達(dá)量減少.通過激光共聚焦顯微鏡觀察蛋白在胃癌SGC-7901細(xì)胞質(zhì)內(nèi)形成點(diǎn)狀聚集增多,證明樺木酸的處理誘導(dǎo)了胃癌SGC-7901細(xì)胞發(fā)生自噬.研究結(jié)果為樺木酸的開發(fā)利用提供了實(shí)驗(yàn)依據(jù).綜上所述,樺木酸通過對(duì)自噬標(biāo)志基因和的調(diào)節(jié),調(diào)控胃癌細(xì)胞SGC-7901自噬的發(fā)生.

      [1] SUNG H,F(xiàn)ERLAY J,SIEGEL R L,et al.Global cancer statistics 2020:GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J].CA Cancer J Clin,2021,7(31):209-249.

      [2] CHENG X J,LIN J C,TU S P.Etiology and Prevention of Gastric Cancer[J].Gastrointest Tumors,2016,3(1):25-36.

      [3] FUJITANI K,YANG H K,MIZUSAWA J,et al.Gastrectomy plus chemotherapy versus chemotherapy alone for advanced gastric cancer with a single non-curable factor(REGATTA):a phase 3,randomised controlled trial[J].Lancet Oncol,2016,17(3):309-318.

      [4] LEVINE B,KROEMER G.Biological Functions of Autophagy Genes:A Disease Perspective[J].Cell,2019,176(1-2):11-42.

      [5] DU Y,SHAO S L,JIAO K H,et al.Effects of hedyotis diffusa on mitochondrial membrane potential and expressions of apoptosis-related genes in human gastric cancer cell line MNK-45[J].,2020,36(2):171-175.

      [6] XU J,PAN Y,LIU Y,et al.A review of anti-tumour effects of ginsenoside in gastrointestinal cancer[J].J Pharm Pharmacol,2021,72(10):1292-1301.

      [7] JIN X,SUN P P,HONG Y,et al.Puerarin induces apoptosis in A549 cells[J].,2017,33(5):466-469.

      [8] DUBEY K K,GOEL N.Evaluation and optimization of downstream process parameters for extraction of betulinic acid from the bark of Ziziphus jujubae L[J].Scientific World Journal,2013(1):e469674.

      [9] COSTA J F,BARBOSA-FILHO J M,MAIA G L,et al.Potent anti-inflammatory activity of betulinic acid treatment in a model of lethal endotoxemia[J].Int Immunopharmacol,2014,23(2):469-474.

      [10] HONG E H,SONG J H,KANG K B,et al.Anti-Influenza Activity of Betulinic Acid from Zizyphus jujuba on Influenza A/PR/8 Virus[J].Biomol Ther(Seoul),2015,23(4):345-349.

      [11] LUO C,HUANG C,ZHU L,et al.Betulinic Acid Ameliorates the T-2 Toxin-Triggered Intestinal Impairment in Mice by Inhibiting Inflammation and Mucosal Barrier Dysfunction through the NF-kappaB Signaling Pathway[J].Toxins(Basel),2020,12(12):794.

      [12] QIAN K,YU D,CHEN C H,et al.Anti-AIDS agents.78.Design,synthesis,metabolic stability assessment,and antiviral evaluation of novel betulinic acid derivatives as potent anti-human immunodeficiency virus(HIV)agents[J].J Med Chem,2009,52(10):3248-3258.

      [13] QUAN H Y,KIM D Y,KIM S J,et al.Betulinic acid alleviates non-alcoholic fatty liver by inhibiting SREBP1 activity via the AMPK-mTOR-SREBP signaling pathway[J].Biochem Pharmacol,2013,85(9):1330-1340.

      [14] XU T,PANG Q,WANG Y,et al.Betulinic acid induces apoptosis by regulating PI3K/Akt signaling and mitochondrial pathways in human cervical cancer cells[J].Int J Mol Med,2017,40(6):1669-1678.

      [15] SHEN H,LIU L,YANG Y,et al.Betulinic Acid Inhibits Cell Proliferation in Human Oral Squamous Cell Carcinoma via Modulating ROS-Regulated p53 Signaling[J].Oncol Res,2017,25(7):1141-1152.

      [16] FOO J B,SAIFUL YAZAN L,TOR Y S,et al.Induction of cell cycle arrest and apoptosis by betulinic acid-rich fraction from Dillenia suffruticosa root in MCF-7 cells involved p53/p21 and mitochondrial signalling pathway[J].J Ethnopharmacol,2015,166:270-278.

      [17] CHEN Z,WU Q,CHEN Y,et al.Effects of betulinic acid on proliferation and apoptosis in Jurkat cells and its in vitro mechanism[J].J Huazhong Univ Sci Technolog Med Sci,2008,28(6):634-638.

      [18] YANG L J,CHEN Y,HE J,et al.Betulinic acid inhibits autophagic flux and induces apoptosis in human multiple myeloma cells in vitro[J].Acta Pharmacol Sin,2012,33(12):1542-1548.

      [19] WANG S,WANG K,ZHANG C,et al.Overaccumulation of p53-mediated autophagy protects against betulinic acid-induced apoptotic cell death in colorectal cancer cells[J].Cell Death Dis,2017,8(10):e3087.

      [20] LIU W P,LI S L,QU Z L,et al.Betulinic acid induces autophagy-mediated apoptosis through suppression of the PI3K/AKT/mTOR signaling pathway and inhibits hepatocellular carcinoma[J].Am J Transl Res,2019(11):6952-6964.

      [21] MOKARRAM P,ALBOKASHY M,ZARGHOONI M,et al.New frontiers in the treatment of colorectal cancer:Autophagy and the unfolded protein response as promising targets[J].Autophagy,2017,13(5):781-819.

      [22] DJAVAHERI-MERGNY M,GIURIATO S,TSCHAN M P,et al.Therapeutic Modulation of Autophagy in Leukaemia and Lymphoma[J].Cells,2019,8(2):103.

      [23] POURHANIFEH M H,MEHRZADI S,KAMALI M,et al.Melatonin and gastrointestinal cancers:Current evidence based on underlying signaling pathways[J].Eur J Pharmacol,2020,886:e173471.

      [24] ZHOU N,WEI Z,QI Z,et al.Abscisic Acid-Induced Autophagy Selectively via MAPK/JNK Signalling Pathway in Glioblastoma[J].Cell Mol Neurobiol,2021,41(4):813-826.

      [25] CHINTHARLAPALLI S,PAPINENI S,LEI P,et al.Betulinic acid inhibits colon cancer cell and tumor growth and induces proteasome-dependent and -independent downregulation of specificity proteins(Sp)transcription factors[J].BMC Cancer,2011,11:371.

      [26] CHINTHARLAPALLI S,PAPINENI S,RAMAIAH S K,et al.Betulinic acid inhibits prostate cancer growth through inhibition of specificity protein transcription factors[J].Cancer Res,2007,67(6):2816-2823.

      [27] HSU T I,WANG M C,CHEN S Y,et al.Betulinic acid decreases specificity protein 1(Sp1)level via increasing the sumoylation of sp1 to inhibit lung cancer growth[J].Mol Pharmacol,2012,82(6):1115-1128.

      [28] LI L,DU Y,KONG X,et al.Lamin B1 is a novel therapeutic target of betulinic acid in pancreatic cancer[J].Clin Cancer Res,2013,19(17):4651-4661.

      [29] REINER T,PARRONDO R,DE LAS POZAS A,et al.Betulinic acid selectively increases protein degradation and enhances prostate cancer-specific apoptosis:possible role for inhibition of deubiquitinase activity[J].PLoS One,2013,8(2):e56234.

      [30] KANG R,ZEH H J,LOTZE M T,et al.The Beclin 1 network regulates autophagy and apoptosis[J].Cell Death Differ,2011,18(4):571-580.

      [31] SHIM M S,NETTESHEIM A,HIRT J,et al.The autophagic proteintranslocates to the nucleus and localizes in the nucleolus associated to NUFIP1 in response to cyclic mechanical stress[J].Autophagy,2020,16(7):1248-1261.

      Effects of betulinic acid on autophagy of human gastric cancer SGC-7901 cells

      GUO Xu1,HE Mengqi1,ZHANG Weiwei1,2,SHEN Yang1,HUANG Xin1,MENG Lingxue1,SHAO Shuli1,2

      (1. School of Life Sciences,Agriculture and Forestry,2. Heilongjiang Provincial Key Laboratory of Resistance Gene Engineering and Protection of Biodiversity in Cold Areas,Qiqihar University,Qiqihar 161006,China)

      SGC-7901 cells were treated with betulinic acid at different concentrations to study the effect of betulinic acid on the expression of autophagy related genes,so as to determine the association between betulinic acid and autophagy in gastric cancer cells.Human gastric cancer SGC-7091 cells were divided into 5 groups.The concentration gradients of betulinic acid in the three groups were 10,20,30 mg/L,respectively.The classic drug 5-fluorouracil(5-Fu)was used as the positive control group,and 0 mmol/L betulinic acid was used as the negative control group.Each group had three replicates and the cells were treated for 48 h,then qRT-PCR and Western blot were used to detect the effects of betulinic acid on mRNA and protein expression of autophagy related genes in human gastric cancer SGC-7091 cells.protein in SGC-7091 cells was localized by immune of luorescence.Compared with the negative control group,the mRNA and protein expressions of-Ⅱ andwere significantly increased(<0.01),and the protein expression of-Ⅰwas significantly decreased(<0.05).Confocal laser microscopy showed that high concentration of betulinic acid inducedprotein aggregation in cytoplasm.In the gradient concentration range,betulinic acid can induce autophagy in human gastric cancer SGC-7091 cells in a dose-dependent manner,and the induction effect increased with the increase of concentration.

      betulinic acid;human gastric cancer SGC-7901 cells;autophagy

      Q2

      A

      10.3969/j.issn.1007-9831.2022.06.013

      1007-9831(2022)06-0075-05

      2022-02-26

      齊齊哈爾大學(xué)黑龍江省教育廳基本業(yè)務(wù)專項(xiàng)重點(diǎn)項(xiàng)目(135109104);黑龍江省高教強(qiáng)省優(yōu)勢(shì)特色學(xué)科——玉米“糧頭食尾”重點(diǎn)項(xiàng)目(LTSW201737);黑龍江省省屬高等學(xué)?;究蒲袠I(yè)務(wù)費(fèi)科研項(xiàng)目(YSTSXK201809)——植物性食品加工技術(shù)特色學(xué)科專項(xiàng);2020年齊齊哈爾大學(xué)研究生創(chuàng)新科研項(xiàng)目(YJSCX2020046)

      郭旭(1997-),男,黑龍江哈爾濱人,在讀碩士研究生,從事腫瘤細(xì)胞基因表達(dá)調(diào)控研究.E-mail:757011379@qq.com

      邵淑麗(1962-),女,黑龍江齊齊哈爾人,教授,博士,從事基因表達(dá)調(diào)控研究.E-mail:shshl32@163.com

      猜你喜歡
      樺木梯度胃癌
      一個(gè)改進(jìn)的WYL型三項(xiàng)共軛梯度法
      樺木酸提取、合成及藥理作用的研究進(jìn)展*
      一種自適應(yīng)Dai-Liao共軛梯度法
      一類扭積形式的梯度近Ricci孤立子
      P53及Ki67在胃癌中的表達(dá)及其臨床意義
      樺木醇對(duì)人結(jié)腸癌SW480細(xì)胞增殖和凋亡的影響
      黃龍山白樺樺木醇與樺木酸含量研究
      胃癌組織中LKB1和VEGF-C的表達(dá)及其意義
      胃癌組織中VEGF和ILK的表達(dá)及意義
      中醫(yī)辨證結(jié)合化療治療中晚期胃癌50例
      石城县| 新津县| 慈溪市| 拉孜县| 呼和浩特市| 舞阳县| 涿鹿县| 凤城市| 桓台县| 大渡口区| 高陵县| 区。| 陆丰市| 彝良县| 彭阳县| 甘肃省| 平罗县| 台前县| 开封县| 浏阳市| 蓬莱市| 广德县| 新沂市| 扎兰屯市| 精河县| 双柏县| 防城港市| 汝城县| 桐庐县| 南皮县| 江口县| 漳平市| 油尖旺区| 贵定县| 许昌市| 留坝县| 分宜县| 仁布县| 精河县| 固安县| 高青县|