Your Good Partner in Biology Research

TM4SF1:四次跨膜蛋白TM4SF家族成員,抗腫瘤血管新生潛力靶點(diǎn)?

日期:2023-11-23 14:54:30

近年來(lái),TM4SF1作為多種惡性腫瘤治療的新靶點(diǎn)被廣泛研究。TM4SF1屬于四次跨膜蛋白TM4SF家族成員,其家族蛋白可以與細(xì)胞內(nèi)外的多種分子相互作用,調(diào)控細(xì)胞信號(hào)轉(zhuǎn)導(dǎo)、細(xì)胞黏附和遷移過(guò)程,從而對(duì)腫瘤的發(fā)生和發(fā)展產(chǎn)生影響。最新研究報(bào)道,TM4SF1-CAR-T細(xì)胞在體外顯示出特異性細(xì)胞毒性,能夠殺死TM4SF1陽(yáng)性的腫瘤細(xì)胞,并釋放干擾素-γ(IFN-γ)和腫瘤壞死因子α(TNF-α)。TM4SF1-CAR-T細(xì)胞在體內(nèi)抑制了SKOV3來(lái)源的腫瘤生長(zhǎng),達(dá)到了90%的緩解率,包括低劑量和高劑量組 [1]。TM4SF1作為一種在多種腫瘤中潛在的生物標(biāo)志物,已被證實(shí)在多種腫瘤中高表達(dá)。越來(lái)越多的研究顯示TM4SF1在癌癥研究中具有潛在的應(yīng)用價(jià)值,并展示了基于TM4SF1蛋白的靶向潛力。因此,新靶點(diǎn)TM4SF1正成為許多研究學(xué)者關(guān)注的焦點(diǎn)!


1. 什么是TM4SF家族?

TM4SF(Transmembrane 4 Superfamily)是四次跨膜蛋白超家族,在多數(shù)哺乳動(dòng)物組織和白細(xì)胞中均有不同程度的表達(dá)。目前,在哺乳動(dòng)物中有約30多個(gè)公認(rèn)的四次跨膜蛋白超家族成員,包括CD9(MRP1)、CD82(KAI1)、CD63(LAMP-3)、CD81(TAPA1)、CD151(PETA3)、CD37、CD53、C0-029和Sm23等 [2-4]。其中,TM4SF1(L6)與TM4SF家族其它成員相比,雖具有相似的拓?fù)浣Y(jié)構(gòu),但缺乏四次跨膜蛋白整體序列的同源性-高度保守的CCG(Cys-Cys-GLy)序列,因此,將其從4次跨膜蛋白家族中細(xì)化出來(lái),又歸為4次跨膜蛋白L6超家族(L6 superfamily),這一家族成員還包括TM4SF4、TM4SF5、TM4SF18、TM4SF19、TM4SF20[5-6]。TM4SF家族有些成員對(duì)腫瘤細(xì)胞的生長(zhǎng)、移動(dòng)和轉(zhuǎn)移起著負(fù)性調(diào)節(jié)的作用,即可以抑制腫瘤細(xì)胞的遷移及侵襲,但也有些可以促進(jìn)惡性腫瘤的發(fā)生發(fā)展及轉(zhuǎn)移浸潤(rùn) [1, 2-4]。


2. 什么是TM4SF1?

2.1 TM4SF1的結(jié)構(gòu)

TM4SF1(Transmembrane-4-L-six-family-1)又稱(chēng)L6、TAL6。TM4SF1是近十幾年才發(fā)現(xiàn)的一種癌基因,因結(jié)構(gòu)和功能上都與TM4SF家族類(lèi)似,屬于四次跨膜蛋白家族(TM4SF)成員之一。TM4SF家族成員結(jié)構(gòu)相似,大多由200-350個(gè)氨基酸組成,具有四個(gè)跨膜區(qū)域。蛋白內(nèi)部有短的C末端和N末端,可能與細(xì)胞骨架和信號(hào)傳導(dǎo)相關(guān)。在細(xì)胞外部,它們與特定受體等空間蛋白相互作用 [7-8]。目前,TM4SF1的分子結(jié)構(gòu)尚未完全闡明,一項(xiàng)研究揭示TM4SF1蛋白是由202個(gè)氨基酸組成的一個(gè)富含半胱氨酸且呈現(xiàn)疏水性的多肽,該肽鏈有四個(gè)疏水結(jié)構(gòu)域并四次跨越細(xì)胞膜,其中三個(gè)疏水區(qū)靠近氨基端,第四個(gè)靠近羥基末端,疏水區(qū)域Ⅲ與Ⅳ之間被一個(gè)由29個(gè)氨基酸組成的親水結(jié)構(gòu)域分隔開(kāi),該親水結(jié)構(gòu)域含有兩個(gè)潛在的糖基化位點(diǎn),同時(shí)與類(lèi)似拓?fù)涿附Y(jié)構(gòu)的細(xì)胞表面蛋白相關(guān)聯(lián) [10]。

TM4SF1的結(jié)構(gòu)

圖1. TM4SF1的結(jié)構(gòu) [10]

2.2 TM4SF1的表達(dá)及功能研究

通常情況下,在正常組織中TM4SF1呈現(xiàn)低表達(dá)。然而,研究發(fā)現(xiàn)TM4SF1在胰腺癌、卵巢癌、肝癌、前列腺癌、結(jié)直腸癌以及乳腺癌等多種上皮性惡性腫瘤細(xì)胞中高表達(dá),參與腫瘤細(xì)胞的生長(zhǎng)、黏附、侵襲和轉(zhuǎn)移,以及調(diào)控血管內(nèi)皮細(xì)胞的功能及病理血管的生成 [11-12]。此外,研究發(fā)現(xiàn),TM4SF1抗體可能通過(guò)調(diào)控人類(lèi)單核細(xì)胞抗體依賴(lài)性細(xì)胞毒作用和人類(lèi)補(bǔ)充因子補(bǔ)體依賴(lài)性細(xì)胞毒作用來(lái)殺滅腫瘤細(xì)胞,并抑制它們代謝產(chǎn)物的生成 [13]。對(duì)乳腺癌、結(jié)腸癌、肺癌、卵巢癌等復(fù)發(fā)患者進(jìn)行I期臨床研究發(fā)現(xiàn),TM4SF1抗體的耐受性較好,且也有效地局限了腫瘤細(xì)胞,有復(fù)發(fā)的患者甚至得到了完全緩解。因此,TM4SF1抗體有望成為未來(lái)惡性腫瘤研究的新靶點(diǎn) [14-16]。


3. TM4SF1相關(guān)的抗腫瘤血管生成的調(diào)控機(jī)制

腫瘤生長(zhǎng)依賴(lài)腫瘤血管的形成,它是腫瘤代謝的關(guān)鍵途徑。TM4SF1通過(guò)與整合素家族成員形成跨膜復(fù)合體來(lái)參與細(xì)胞的多項(xiàng)生命活動(dòng)及影響腫瘤細(xì)胞的生長(zhǎng)、黏附、侵襲和轉(zhuǎn)移能力,并參與腫瘤血管生成。也有報(bào)道TM4SF1能夠激活Rho信號(hào)通路,從而介導(dǎo)上皮-間質(zhì)的轉(zhuǎn)化(EMT),進(jìn)而參與腫瘤細(xì)胞的浸潤(rùn)轉(zhuǎn)移,具體作用機(jī)制尚在研究中 [17]。

研究報(bào)道,TM4SF1基因敲除后可以阻止絲狀偽足的形成,抑制細(xì)胞遷移,阻止細(xì)胞分裂,促進(jìn)內(nèi)皮細(xì)胞衰老。整合素α5(Integrin α5)和整合素β1(Integrin beta 1/CD29)亞基,與TM4SF1結(jié)合后相互作用,且整合素只在血管內(nèi)皮生長(zhǎng)因子A(VEGF-A)或凝血酶刺激作用下才與TM4SF1相互結(jié)合。因此,TM4SF1敲除后能抑制血管生成內(nèi)皮生長(zhǎng)因子的成熟。同時(shí),TM4SF1存在于腫瘤細(xì)胞膜及細(xì)胞器(如溶酶體)中,可與LAMP-1LAMP-2或乳鐵蛋白結(jié)合蛋白(LTF)實(shí)現(xiàn)共定位后影響細(xì)胞功能;TM4SF1促進(jìn)血管的生成同時(shí),可通過(guò)募集到TEMs區(qū)域及相關(guān)的泛素化修飾影響細(xì)胞的遷移能力;還可通過(guò)與CD13相互作用以及調(diào)節(jié)跨膜蛋白CD63、CD82等的表達(dá),參與調(diào)節(jié)細(xì)胞的運(yùn)動(dòng)及侵襲能力。無(wú)論無(wú)何,TM4SF1對(duì)體外內(nèi)皮細(xì)胞和體內(nèi)病理性血管形成具有重要作用,有望作為潛在的抗腫瘤血管生成的靶點(diǎn) [17]

TM4SF1的抗腫瘤血管生成的調(diào)控機(jī)制

圖2. TM4SF1的抗腫瘤血管生成的調(diào)控機(jī)制 [17]


4. TM4SF1相關(guān)的腫瘤研究

4.1 TM4SF1和胰腺癌研究

TM4SF1在胰腺癌中的表達(dá)量明顯高于良性腫瘤和正常組織。研究表明,通過(guò)siRNA沉默TM4SF1后,胰腺癌細(xì)胞的侵襲力和遷移能力受到抑制,但腫瘤細(xì)胞的增值能力并未受到影響。這可能是因?yàn)門(mén)M4SF1僅在腫瘤細(xì)胞的遷移和轉(zhuǎn)移途徑中發(fā)揮作用,與腫瘤的發(fā)生并無(wú)顯著關(guān)系。siRNA沉默TM4SF1后,腫瘤細(xì)胞對(duì)化療藥物卡培他濱的敏感性增加,然而其具體機(jī)制尚不明確。有研究提示,TM4SF1通過(guò)調(diào)節(jié)MMP-2MMP-9的基因表達(dá)影響胰腺癌細(xì)胞的遷移和侵襲。事實(shí)上,TM4SF家族多種成員在胰腺癌組織中高表達(dá)。例如,CD151CD9、CD81等家族成員在胰腺導(dǎo)管腺癌的細(xì)胞膜以及胞漿中均呈彌漫性表達(dá) [18-21]

4.2 TM4SF1和乳腺癌研究

研究發(fā)現(xiàn),TM4SF1在乳腺癌組織中高表達(dá),可能與乳腺癌的發(fā)生發(fā)展及轉(zhuǎn)移密切相關(guān),能夠促進(jìn)腫瘤細(xì)胞的轉(zhuǎn)移。在HER-2高表達(dá)型乳腺癌中,TM4SF1蛋白的表達(dá)量最高,暗示兩者可能存在某種潛在聯(lián)系 [22]。另有研究發(fā)現(xiàn),與周?chē)H橄俳M織相比,乳腺腫瘤組織中高表達(dá)TM4SF1。其后,合成來(lái)源于抗原表位的衍生物HLA-A2,發(fā)現(xiàn)它能抑制腫瘤細(xì)胞生長(zhǎng)。這些研究結(jié)果的發(fā)現(xiàn)對(duì)未來(lái)的乳腺癌新藥研發(fā)具有重要意義 [23]。此外,有研究揭示,TM4SF1可能通過(guò)PI3K/AKT/mTOR信號(hào)通路在TNBC細(xì)胞的侵襲和凋亡中起重要作用 [24]。

4.3 TM4SF1和前列腺癌研究

TM4SF1是雄激素受體(Androgen receptor,AR)轉(zhuǎn)錄因子的直接靶基因。雄激素受體在前列腺癌內(nèi)環(huán)境中扮演關(guān)鍵角色,促進(jìn)了前列腺癌的發(fā)展。雄激素與其配體結(jié)合后,可能完全失去對(duì)其活性的控制,導(dǎo)致腫瘤細(xì)胞過(guò)度增殖。研究發(fā)現(xiàn),TM4SF1其啟動(dòng)子區(qū)含有雄激素反應(yīng)元件(ARE),并且受雄激素調(diào)節(jié)。在前列腺癌中,TM4SF1表達(dá)水平高于良性前列腺增生。進(jìn)一步的功能研究表明,TM4SF1參與調(diào)控腫瘤細(xì)胞的遷移,可能在腫瘤轉(zhuǎn)移中發(fā)揮作用。深入研究TM4SF1作為雄激素受體(AR)的靶點(diǎn),有望為前列腺癌研究開(kāi)辟新的治療途徑。 [25-27]。

4.4 TM4SF1和卵巢癌研究

應(yīng)用SEREX技術(shù)將TM4SF1從人卵巢癌腹水cDNA文庫(kù)中篩選出來(lái),并通過(guò)分離并純化相關(guān)抗原,對(duì)其進(jìn)行了進(jìn)一步的分析。研究發(fā)現(xiàn),TM4SF1基因在不同卵巢組織中的表達(dá),發(fā)現(xiàn)隨著卵巢癌的發(fā)生與發(fā)展,TM4SF1呈逐漸增高的趨勢(shì),其基因表達(dá)顯示出與卵巢癌發(fā)生發(fā)展相關(guān)的特征 [28]。另有報(bào)道,通過(guò)基因芯片數(shù)據(jù)庫(kù)分析和蛋白相互作用網(wǎng)絡(luò)分析顯示,TM4SF1和DDR1有直接相互作用,并且與細(xì)胞外基質(zhì)、膠原、整合素等信號(hào)通路相關(guān) [29]。

4.5 TM4SF1和其它癌癥研究

TM4SF1在多種惡性癌癥的遷移和侵襲中起重要作用,如研究發(fā)現(xiàn)TM4SF1通過(guò)調(diào)節(jié)caspase-3、caspase-9、MMP-2、MMP-9和VEG等相關(guān)基因的表達(dá)促進(jìn)肝癌細(xì)胞的增殖、侵襲和轉(zhuǎn)移 [30];根據(jù)基于芯片結(jié)果,篩選宮頸癌相關(guān)的目標(biāo)基因2個(gè):EFNB2和TM4SF1。EFNB2和TM4SF1在宮頸癌原代SP細(xì)胞中有較高表達(dá),且兩者與腫瘤的轉(zhuǎn)移、侵襲均有密切相關(guān)性 [28, 31];TM4SF1通過(guò)調(diào)控凋亡相關(guān)蛋白caspase-3等促進(jìn)胃癌細(xì)胞的增殖、遷移和侵襲;亦或通過(guò)調(diào)控PPARγ-SIRT1反饋環(huán)影響膀胱癌細(xì)胞凋亡、細(xì)胞周期 [32]。


5. TM4SF1臨床研究前景

目前,有四種針對(duì)TM4SF1的藥物在研發(fā)中,分別是Angiex公司的抗體偶聯(lián)藥物Anti-TM4SF1 ADC,上??破逅帢I(yè)科技有限公司的兩種CAR-T細(xì)胞療法CART-TM4SF1 cells和KQ-L6,以及上??漆t(yī)聯(lián)創(chuàng)生物科技有限公司的CAR-T細(xì)胞療法Anti-TM4SF1-CAR-T-cell therapy。這些藥物都通過(guò)不同的作用機(jī)制抑制或殺死表達(dá)TM4SF1的腫瘤細(xì)胞,主要適用于實(shí)體瘤以及消化系統(tǒng)腫瘤。其中,Anti-TM4SF1 ADC還處于臨床前階段,而其他三種藥物已進(jìn)入臨床I期階段。TM4SF1作為潛力巨大的腫瘤研究靶點(diǎn),其在臨床藥物研究方面具有廣闊前景。然而,要證明其有效性和優(yōu)勢(shì),需要進(jìn)一步收集臨床研究數(shù)據(jù)并進(jìn)行安全性評(píng)估。

為鼎力協(xié)助科研和藥企人員針對(duì)TM4SF1在腫瘤中的臨床應(yīng)用研究,CUSABIO推出TM4SF1活性蛋白(Code:CSB-MP013330HU)以及重組抗體(Code:CSB-RA023615MA1HU),助力您在TM4SF1機(jī)制方面的研究或其潛在臨床價(jià)值的探索。

華美 CUSABIO TM4SF1 蛋白

Recombinant Human Transmembrane 4 L6 family member 1(TM4SF1)-VLPs (Active) Code: CSB-MP023615HU

High Specifity Validated by SDS-PAGE
CSB-MP023615HU High Specifity Validated by SDS-PAGE

CSB-MP023615HU is detected by Mouse anti-6*His monoclonal antibody.The two bands respectively correspond to monomer, Homodimer.

Excellent Bioactivity Validated by Functional ELISA
High Purity Validated of CSB-MP023615HU

Immobilized Human TM4SF1 at 5μg/mL can bind Anti-TM4SF1 recombinant antibody(CSB-RA023615MA1HU). The EC50 is 4.079-4.472 ng/mL. VLPs (CSB-MP3838) is negative control.

Recombinant Macaca fascicularis Transmembrane 4 L6 family member 1 (TM4SF1)-VLPs (Active) Code: CSB-MP5031MOV

High Specifity Validated by SDS-PAGE
CSB-MP5031MOV High Specifity Validated by SDS-PAGE

CSB-MP5031MOV is detected by Mouse anti-6*His monoclonal antibody.The two bands respectively correspond to monomer, Homodimer.

Excellent Bioactivity Validated by Functional ELISA
High Purity Validated of CSB-MP5031MOV

Immobilized Cynomolgus TM4SF1 at 5μg/mL can bind Anti-TM4SF1 recombinant antibody(CSB-RA023615MA1HU). The EC50 is 4.480-4.930 ng/mL. VLPs (CSB-MP3838) is negative control.


參考文獻(xiàn):

[1] Shen, Yijie, et al. "Construction of CAR-T cells targeting TM4SF1 and its anti-tumor capacity in ovarian cancer." Immunology Letters 255 (2023): 1-9.

[2] Rahim, Nur Syafiqah, et al. "Three Members of Transmembrane-4-Superfamily, TM4SF1, TM4SF4, and TM4SF5, as Emerging Anticancer Molecular Targets against Cancer Phenotypes and Chemoresistance." Pharmaceuticals 16.1 (2023): 110.

[3] Qi, Yaoyue, et al. "Expression and function of transmembrane 4 superfamily proteins in digestive system cancers." Cancer Cell International 20.1 (2020): 1-12.

[4] Seldin, Michael F., et al. "Mapping of the genes for four members of the transmembrane 4 superfamily: mouse Cd9, Cd63, Cd81, and Cd82." Immunogenetics 42 (1995): 422-425.

[5] Fu, Fangmei, et al. "Role of transmembrane 4 L six family 1 in the development and progression of cancer." Frontiers in molecular biosciences 7 (2020): 202.

[6] Xu, Lijian, et al. "hsa-miR-141 downregulates TM4SF1 to inhibit pancreatic cancer cell invasion and migration." International journal of oncology 44.2 (2014): 459-466.

[7] Xian, Jinwen, et al. "Molecular Characterization of a Tetraspanin TSP11 Gene in Echinococcus granulosus and Evaluation Its Immunoprotection in Model Dogs." Frontiers in Veterinary Science 8 (2021): 759283.

[8] Murungi, Edwin K., et al. "Evolution and structural analyses of Glossina morsitans (Diptera; Glossinidae) tetraspanins." Insects 5.4 (2014): 885-908.

[9] Fu, Fangmei, et al. "Role of transmembrane 4 L six family 1 in the development and progression of cancer." Frontiers in molecular biosciences 7 (2020): 202.

[10] Shih, Shou-Ching, et al. "The L6 protein TM4SF1 is critical for endothelial cell function and tumor angiogenesis." Cancer research 69.8 (2009): 3272-3277.

[11] Visintin, Alberto, et al. "Novel anti-TM4SF1 antibody–drug conjugates with activity against tumor cells and tumor vasculature." Molecular Cancer Therapeutics 14.8 (2015): 1868-1876.

[12] Lin, Chi-Iou, et al. "TM4SF1: a new vascular therapeutic target in cancer." Angiogenesis 17 (2014): 897-907.

[13] Sciuto, Tracey E., et al. "Intracellular distribution of TM4SF1 and internalization of TM4SF1-antibody complex in vascular endothelial cells." Biochemical and biophysical research communications 465.3 (2015): 338-343.

[14] Chen, Guang, et al. "Targeting TM4SF1 exhibits therapeutic potential via inhibition of cancer stem cells." Signal Transduction and Targeted Therapy 7.1 (2022): 350.

[15] Fu, Fangmei, et al. "Role of transmembrane 4 L six family 1 in the development and progression of cancer." Frontiers in molecular biosciences 7 (2020): 202.

[16] Allioli, Nathalie, et al. "TM4SF1, a novel primary androgen receptor target gene over‐expressed in human prostate cancer and involved in cell migration." The Prostate 71.11 (2011): 1239-1250.

[17] Rahim, Nur Syafiqah, et al. "Three Members of Transmembrane-4-Superfamily, TM4SF1, TM4SF4, and TM4SF5, as Emerging Anticancer Molecular Targets against Cancer Phenotypes and Chemoresistance." Pharmaceuticals 16.1 (2023): 110.

[18] Visintin, Alberto, et al. "Novel anti-TM4SF1 antibody–drug conjugates with activity against tumor cells and tumor vasculature." Molecular Cancer Therapeutics 14.8 (2015): 1868-1876.

[19] Cao, Jia, et al. "TM4SF1 regulates pancreatic cancer migration and invasion in vitro and in vivo." Cellular Physiology and Biochemistry 39.2 (2016): 740-750.

[20] Cao, Jia, et al. "TM4SF1 promotes gemcitabine resistance of pancreatic cancer in vitro and in vivo." PloS one 10.12 (2015): e0144969.

[21] Zheng, Biao, et al. "TM4SF1 as a prognostic marker of pancreatic ductal adenocarcinoma is involved in migration and invasion of cancer cells." International journal of oncology 47.2 (2015): 490-498.

[22] Gao, Xinya, et al. "Expression of TM4SF1 in breast cancer tissue and its clinical significance." Journal of Jilin University (Medicine Edition) (2017): 1186-1192.

[23] Chen, Jie, et al. "Transmembrane 4 L Six Family Member 1 Suppresses Hormone Receptor-–Positive, HER2-Negative Breast Cancer Cell Proliferation." Frontiers in Pharmacology 13 (2022): 770993.

[24] Sun, Yonghong, et al. "Role of TM4SF1 in regulating breast cancer cell migration and apoptosis through PI3K/AKT/mTOR pathway." International journal of clinical and experimental pathology 8.8 (2015): 9081.

[25] Allioli, Nathalie, et al. "TM4SF1, a novel primary androgen receptor target gene over‐expressed in human prostate cancer and involved in cell migration." The Prostate 71.11 (2011): 1239-1250.

[26] Chen, Junyi, et al. "Over-expression of TM4SF1 improves cell metastasis and growth by activating ERK1/2 signaling pathway in human prostate cancer." J. BUON 24 (2019): 2531-2538.

[27] Cao, Jia, et al. "TM4SF1 regulates pancreatic cancer migration and invasion in vitro and in vivo." Cellular Physiology and Biochemistry 39.2 (2016): 740-750.

[28] Gao, Caiyun, et al. "TM4SF1 is a potential target for anti-invasion and metastasis in ovarian cancer." BMC cancer 19 (2019): 1-12.

[29] Huang, Zhijiong, Hongyu Yao, and Zhijun Yang. "Prognostic significance of TM4SF1 and DDR1 expression in epithelial ovarian cancer." Oncology Letters 26.4 (2023): 1-12.

[30] Huang, Yu-Kun, Xue-Gong Fan, and Fu Qiu. "TM4SF1 promotes proliferation, invasion, and metastasis in human liver cancer cells." International journal of molecular sciences 17.5 (2016): 661.

[31] Alam, Syed Mahfuzul, et al. "Coexpression of EphB4 and ephrinB2 in tumor advancement of uterine cervical cancers." Gynecologic oncology 114.1 (2009): 84-88.

[32] Cao, Rui, et al. "TM4SF1 regulates apoptosis, cell cycle and ROS metabolism via the PPARγ-SIRT1 feedback loop in human bladder cancer cells." Cancer letters 414 (2018): 278-293.