matriptase

间质蛋白酶
  • 文章类型: Journal Article
    抑制肝细胞生长因子(HGF)和巨噬细胞刺激蛋白(MSP)的蛋白水解加工是一种有吸引力的方法,用于发现预防肿瘤进展和转移的新型抗癌治疗剂。这里,我们利用了一种称为HyCoSuL的底物组合文库(PS-SCL)技术的改进和扩展版本来优化HGF/MSP激活丝氨酸蛋白酶的拟肽抑制剂,HGFA,间质蛋白酶,和hepsin。这些抑制剂具有亲电子酮丝氨酸捕获弹头,因此与蛋白酶形成可逆的共价键。我们证明,根据从HyCoSuL学习的蛋白酶底物偏好,通过使用非天然氨基酸改变抑制剂的P2,P3和P4位置,我们可以预测地改变抑制剂的效力和选择性。我们鉴定了四肽JH-1144(8)作为HGFA的单位数nM抑制剂,matriptase和hepsin对因子Xa和凝血酶具有优异的选择性。这些非天然肽相对于类似结构的天然肽具有增加的代谢稳定性。三肽抑制剂PK-1-89(2)在24小时内具有良好的化合物暴露的小鼠中具有优异的药代动力学。此外,我们获得了与matriptase结合的抑制剂MM1132(15)的X射线结构,揭示了一种可用于未来抑制剂设计的有趣的结合构象.
    Inhibition of the proteolytic processing of hepatocyte growth factor (HGF) and macrophage stimulating protein (MSP) is an attractive approach for the drug discovery of novel anticancer therapeutics which prevent tumor progression and metastasis. Here, we utilized an improved and expanded version of positional scanning of substrate combinatorial libraries (PS-SCL) technique called HyCoSuL to optimize peptidomimetic inhibitors of the HGF/MSP activating serine proteases, HGFA, matriptase, and hepsin. These inhibitors have an electrophilic ketone serine trapping warhead and thus form a reversible covalent bond to the protease. We demonstrate that by varying the P2, P3, and P4 positions of the inhibitor with unnatural amino acids based on the protease substrate preferences learned from HyCoSuL, we can predictably modify the potency and selectivity of the inhibitor. We identified the tetrapeptide JH-1144 (8) as a single digit nM inhibitor of HGFA, matriptase and hepsin with excellent selectivity over Factor Xa and thrombin. These unnatural peptides have increased metabolic stability relative to natural peptides of similar structure. The tripeptide inhibitor PK-1-89 (2) has excellent pharmacokinetics in mice with good compound exposure out to 24 h. In addition, we obtained an X-ray structure of the inhibitor MM1132 (15) bound to matriptase revealing an interesting binding conformation useful for future inhibitor design.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    膜膨胀整合了多种力以调节精确的管生长和网络形成。缺陷导致变形,如在多囊肾疾病等疾病中发现的,主动脉瘤,狭窄,和弯曲。我们确定了一种感知和响应气管导管膜驱动扩张的机制。顶端膜锚定到顶端细胞外基质(aECM),并引起伸长气管导管的膨胀力。aECM提供机械张力,平衡所产生的膨胀力,Dumpy是一种弹性分子,可在气管导管扩张过程中调节基质上的机械应力。我们在果蝇中显示透明带(ZP)结构域蛋白Piopio与气管细胞上的ZP蛋白Dumpy相互作用并合作。为了抵抗在管膨胀过程中产生的剪切应力,Piopio通过Matriptase同源物Notocribal经历了胞外域脱落,在膜和细胞外基质之间释放Piopio-Dumpy介导的连接。此过程的失败导致根尖膜的变形,撕裂顶端基质,并损害管状网络功能。我们还显示了人TGFβIII型受体的保守的胞外域脱落,为深入分析由细胞和管形状变化引起的疾病提供了新的发现。
    Membrane expansion integrates multiple forces to mediate precise tube growth and network formation. Defects lead to deformations, as found in diseases such as polycystic kidney diseases, aortic aneurysms, stenosis, and tortuosity. We identified a mechanism of sensing and responding to the membrane-driven expansion of tracheal tubes. The apical membrane is anchored to the apical extracellular matrix (aECM) and causes expansion forces that elongate the tracheal tubes. The aECM provides a mechanical tension that balances the resulting expansion forces, with Dumpy being an elastic molecule that modulates the mechanical stress on the matrix during tracheal tube expansion. We show in Drosophila that the zona pellucida (ZP) domain protein Piopio interacts and cooperates with the ZP protein Dumpy at tracheal cells. To resist shear stresses which arise during tube expansion, Piopio undergoes ectodomain shedding by the Matriptase homolog Notopleural, which releases Piopio-Dumpy-mediated linkages between membranes and extracellular matrix. Failure of this process leads to deformations of the apical membrane, tears the apical matrix, and impairs tubular network function. We also show conserved ectodomain shedding of the human TGFβ type III receptor by Notopleural and the human Matriptase, providing novel findings for in-depth analysis of diseases caused by cell and tube shape changes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    就其表面抗原表达而言,癌性B细胞与它们的非恶性对应物几乎无法区分。因此,面临的挑战在于消除恶性B细胞群,同时维持功能性适应性免疫系统.这里,我们提出了一种IgM特异性抗体-药物偶联物,该偶联物被携带表位的IgM恒定结构域融合所掩盖.抗体掩蔽与可溶性五聚体以及细胞表面表达的IgM分子的相互作用受损,使得抗体细胞毒性失活。抗IgM抗体药物缀合物的结合能力在条件性蛋白酶介导的去掩蔽后恢复,这因此使得靶依赖性抗体内化和随后在恶性B细胞中诱导凋亡。这种易于适应的方法可能为非霍奇金淋巴瘤治疗提供了一种新的克隆性B细胞淋巴瘤根除机制。
    Cancerous B cells are almost indistinguishable from their non-malignant counterparts regarding their surface antigen expression. Accordingly, the challenge to be faced consists in elimination of the malignant B cell population while maintaining a functional adaptive immune system. Here, we present an IgM-specific antibody-drug conjugate masked by fusion of the epitope-bearing IgM constant domain. Antibody masking impaired interaction with soluble pentameric as well as cell surface-expressed IgM molecules rendering the antibody cytotoxically inactive. Binding capacity of the anti-IgM antibody drug conjugate was restored upon conditional protease-mediated demasking which consequently enabled target-dependent antibody internalization and subsequent induction of apoptosis in malignant B cells. This easily adaptable approach potentially provides a novel mechanism of clonal B cell lymphoma eradication to the arsenal available for non-Hodgkin\'s lymphoma treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    前列腺素和间质蛋白酶是在实体上皮肿瘤中具有相反作用的细胞外膜丝氨酸蛋白酶。间质蛋白酶是一种促进肿瘤发生和进展的癌蛋白,prostasin是一种抑制肿瘤侵袭和转移的肿瘤抑制剂。先前的研究表明,伯基特淋巴瘤的一个亚组具有高水平的异位基质蛋白酶表达,但没有前列腺蛋白。通过B淋巴瘤细胞中的小干扰RNA降低间质蛋白酶水平阻碍了小鼠的肿瘤异种移植物生长。这里,我们报道了一种新的方法,通过前列腺蛋白通过外泌体启动前列腺蛋白-间质蛋白酶激活级联反应来调节B癌细胞中的间质蛋白酶。通过测量其数量和条件培养基中胰蛋白酶样丝氨酸蛋白酶活性来监测间质蛋白酶的活化和脱落。通过前列腺素的稳定表达实现了细胞中蛋白酶级联的持续激活。前列腺素表达的B癌细胞呈现与其肿瘤抑制作用一致的表型,如生长减少和细胞凋亡增加。通过在动物模型中的进一步研究,可以开发前列腺素外泌体作为启动前列腺素-间质蛋白酶级联反应的药物来治疗B淋巴瘤。
    Prostasin and matriptase are extracellular membrane serine proteases with opposing effects in solid epithelial tumors. Matriptase is an oncoprotein that promotes tumor initiation and progression, and prostasin is a tumor suppressor that reduces tumor invasion and metastasis. Previous studies have shown that a subgroup of Burkitt lymphoma have high levels of ectopic matriptase expression but no prostasin. Reducing the matriptase level via small interfering RNAs in B lymphoma cells impeded tumor xenograft growth in mice. Here, we report a novel approach to matriptase regulation in B cancer cells by prostasin via exosomes to initiate a prostasin-matriptase protease activation cascade. The activation and shedding of matriptase were monitored by measuring its quantity and trypsin-like serine protease activity in conditioned media. Sustained activation of the protease cascade in the cells was achieved by the stable expression of prostasin. The B cancer cells with prostasin expression presented phenotypes consistent with its tumor suppressor role, such as reduced growth and increased apoptosis. Prostasin exosomes could be developed as an agent to initiate the prostasin-matriptase cascade for treating B lymphoma with further studies in animal models.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    自从70多年前提出蛋白水解酶的促侵入活性以来,已经确定了蛋白酶在癌症进展中的几种作用.大约一半的473活性人类蛋白酶在前列腺中表达,并且该酶家族中许多最明确的成员受雄激素调节,前列腺癌发展所必需的激素。最值得注意的是,几种激肽释放酶相关肽酶,包括KLK3(前列腺特异性抗原,PSA),最著名的前列腺癌标志物,和II型跨膜丝氨酸蛋白酶,如TMPRSS2和间质蛋白酶,已被广泛研究,并发现促进前列腺癌的进展。最近的发现还表明,蛋白酶在晚期和侵袭性去势抵抗前列腺癌(CRPC)的发展中起着关键作用。也许最有趣的证据来自研究表明蛋白酶激活的跨膜蛋白,Notch和CDCP1与CRPC的发展有关。这里,我们综述了蛋白酶在前列腺癌中的作用,特别关注雄激素对它们的调节。
    Since the proposition of the pro-invasive activity of proteolytic enzymes over 70 years ago, several roles for proteases in cancer progression have been established. About half of the 473 active human proteases are expressed in the prostate and many of the most well-characterized members of this enzyme family are regulated by androgens, hormones essential for development of prostate cancer. Most notably, several kallikrein-related peptidases, including KLK3 (prostate-specific antigen, PSA), the most well-known prostate cancer marker, and type II transmembrane serine proteases, such as TMPRSS2 and matriptase, have been extensively studied and found to promote prostate cancer progression. Recent findings also suggest a critical role for proteases in the development of advanced and aggressive castration-resistant prostate cancer (CRPC). Perhaps the most intriguing evidence for this role comes from studies showing that the protease-activated transmembrane proteins, Notch and CDCP1, are associated with the development of CRPC. Here, we review the roles of proteases in prostate cancer, with a special focus on their regulation by androgens.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    整体膜,Kunitz型丝氨酸蛋白酶抑制剂HAI-1和HAI-2可以抑制2型跨膜丝氨酸蛋白酶间质蛋白酶的蛋白水解活性,具有高特异性和效力。高水平的细胞外间质蛋白酶蛋白水解活性,然而,在一些肿瘤B细胞中观察到具有高水平的内源性HAI-2,表明HAI-2在细胞水平上可能是无效的间质蛋白酶抑制剂。此处检查了HAI在控制细胞外间质蛋白酶蛋白水解活性中的不同有效性。在HAITetonDaudiBurkitt淋巴瘤细胞中诱导间质蛋白酶酶原激活后,天然表达蛋白裂解酶的HAI-2含量非常低,而没有HAI-1,新生的活性蛋白裂解酶被迅速抑制或作为酶活性酶脱落。随着HAI-1表达的增加,细胞间质蛋白酶-HAI-1复合物增加,细胞外活性蛋白裂解酶成比例下降。增加HAI-2表达,然而,导致细胞间质蛋白酶-HAI-2复合物水平达到平台,而细胞外活性蛋白裂解酶仍然很高。与这种差异效应相反,HAI-1和HAI-2,即使在非常低的水平,被证明可促进内源性间质蛋白酶的表达和细胞表面易位。与HAI-2的主要细胞内定位相比,两种密切相关的丝氨酸蛋白酶抑制剂对细胞外活性蛋白蛋白酶抑制的差异可能是由于HAI-1的主要细胞表面表达所致。HAIs,因此,在促进间质蛋白酶表达和表面易位方面彼此相似,但在控制细胞外间质蛋白酶酶活性方面的有效性不同。
    The integral membrane, Kunitz-type serine protease inhibitors HAI-1 and HAI-2, can suppress the proteolytic activity of the type 2 transmembrane serine protease matriptase with high specificity and potency. High levels of extracellular matriptase proteolytic activity have, however, been observed in some neoplastic B-cells with high levels of endogenous HAI-2, indicating that HAI-2 may be an ineffective matriptase inhibitor at the cellular level. The different effectiveness of the HAIs in the control of extracellular matriptase proteolytic activity is examined here. Upon inducing matriptase zymogen activation in the HAI Teton Daudi Burkitt lymphoma cells, which naturally express matriptase with very low levels of HAI-2 and no HAI-1, nascent active matriptase was rapidly inhibited or shed as an enzymatically active enzyme. With increasing HAI-1 expression, cellular matriptase-HAI-1 complex increased, and extracellular active matriptase decreased proportionally. Increasing HAI-2 expression, however, resulted in cellular matriptase-HAI-2 complex levels reaching a plateau, while extracellular active matriptase remained high. In contrast to this differential effect, both HAI-1 and HAI-2, even at very low levels, were shown to promote the expression and cell-surface translocation of endogenous matriptase. The difference in the suppression of extracellular active matriptase by the two closely related serine protease inhibitors could result from the primarily cell surface expression of HAI-1 compared to the mainly intracellular localization of HAI-2. The HAIs, therefore, resemble one another with respect to promoting matriptase expression and surface translocation but differ in their effectiveness in the control of extracellular matriptase enzymatic activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Matriptases, members of the type II transmembrane serine protease family, are cell surface proteolytic enzymes that mediate tumor invasion and metastasis. Matriptase is highly expressed in breast cancer and is associated with poor patient outcome. However, the cellular mechanism by which matriptase mediates breast cancer invasion remains unknown. The present study aimed to determine the role of matriptase in the protein kinase C (PKC)‑mediated metastasis of MCF‑7 human breast cancer cells. Matriptase small interfering RNA‑mediated knockdown significantly attenuated the 12‑O‑tetradecanoylphorbol‑13‑acetate (TPA)‑induced invasiveness and migration of MCF‑7 cells, and inhibited the activation of phospholipase C γ2 (PLCγ2)/PKC/MAPK signaling pathways. Matriptase‑knockdown also suppressed the expression of MMP‑9 and inhibited the activation of NF‑κB/activator protein‑1 in MCF‑7 cells. Additionally, GB83 [an inhibitor of protease‑activated receptor‑2 (PAR‑2)] inhibited PKC‑mediated MMP‑9 expression and metastatic ability in MCF‑7 cells. Furthermore, downregulation of matriptase suppressed TPA‑induced MMP‑9 expression and invasiveness via PAR‑2/PLCγ2/PKC/MAPK activation. These findings shed light on the mechanism underlying the role of matriptase in MCF‑7 cell invasion and migration ability, and suggest that matriptase modulation could be a promising therapeutic strategy for preventing breast cancer metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Head and neck squamous cell carcinoma remains challenging to treat with no improvement in survival rates over the past 50 years. Thus, there is an urgent need to discover more reliable therapeutic targets and biomarkers for HNSCC. Matriptase, a type-II transmembrane serine protease, induces malignant transformation in epithelial stem cells through proteolytic activation of pro-HGF and PAR-2, triggering PI3K-AKT-mTOR and NFKB signaling. The serine protease inhibitor lympho-epithelial Kazal-type-related inhibitor (LEKTI) inhibits the matriptase-driven proteolytic pathway, directly blocking kallikreins in epithelial differentiation. Hence, we hypothesized LEKTI could inhibit matriptase-dependent squamous cell carcinogenesis, thus implicating kallikreins in this process. Double-transgenic mice with simultaneous expression of matriptase and LEKTI under the keratin-5 promoter showed a prominent rescue of K5-Matriptase+/0 premalignant phenotype. Notably, in DMBA-induced SCC, heterotopic co-expression of LEKTI and matriptase delayed matriptase-driven tumor incidence and progression. Co-expression of LEKTI reverted altered Kallikrein-5 expression observed in the skin of K5-Matriptase+/0 mice, indicating that matriptase-dependent proteolytic pathway inhibition by LEKTI occurs through kallikreins. Moreover, we showed that Kallikrein-5 is necessary for PAR-2-mediated IL-8 release, YAP1-TAZ/TEAD activation, and matriptase-mediated oral squamous cell carcinoma migration. Collectively, our data identify a third signaling pathway for matriptase-dependent carcinogenesis in vivo. These findings are critical for the identification of more reliable biomarkers and effective therapeutic targets in Head and Neck cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    BACKGROUND: Genomic profiles of specific gene sets have been established to guide personalized treatment and prognosis for patients with breast cancer (BC). However, epigenomic information has not yet been applied in a clinical setting. ST14 encodes matriptase, a proteinase that is widely expressed in BC with reported prognostic value.
    METHODS: In this present study, we evaluated the effect of ST14 DNA methylation (DNAm) on overall survival (OS) of patients with BC as a representative example to promote the use of the epigenome in clinical decisions. We analyzed publicly available genomic and epigenomic data from 1361 BC patients. Methylation was characterized by the β-value from CpG probes based on sequencing with the Illumina Human 450 K platform.
    RESULTS: A high mean DNAm (β > 0.6779) across 34 CpG probes for ST14, as the gene-associated methylation (GAM) pattern, was associated with a longer OS after adjusting age, stage, histology and molecular features in Cox model (p value < 0.001). A high GAM status was also associated with a higher XBP1 expression level and higher proportion of hormone-positive BC (p value < 0.001). Pathway analysis revealed that altered GAM was related to matrisome-associated pathway.
    CONCLUSIONS: Here we show the potential role of ST14 DNAm in BC prognosis and warrant further study.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    上皮组织通过激活上皮细胞运动性和快速炎症而对损伤作出反应。这种反应也在过表达膜结合蛋白酶时引起,间质蛋白酶,或其抑制剂的突变,Hai1.不受限制的基质蛋白酶活性也易患癌症。基质蛋白酶如何导致这些细胞结果是未知的。我们证明斑马鱼hai1a突变体显示增加的H2O2,NFκB信号,和IP3R介导的钙闪烁,这些促进炎症,但不产生上皮细胞运动。相比之下,在hai1a突变体中Gq亚基的抑制挽救了炎症和上皮表型,后者由DAG类似物概括,PMA。我们证明hai1a具有升高的MAPK通路活性,抑制其挽救表皮缺陷。最后,我们将RSK激酶鉴定为MAPK靶向破坏hai1a突变体中的粘附连接。我们的工作绘制了新的信号级联,介导了基质蛋白酶对上皮的有效作用,与组织损伤反应和癌症进展有关。
    当细胞中的正常过程被破坏或不受调节时,就会发生癌症。许多蛋白质可以做出贡献,包括一种叫做基质蛋白酶的酶,它可以在特定部位切割其他蛋白质。基质蛋白酶活性由一种叫做Hai1的蛋白质严格控制。在老鼠和斑马鱼中,当Hai1不能充分控制基质蛋白酶活性时,侵袭性癌症与严重的炎症发展。然而,目前尚不清楚未调节的基质蛋白酶如何导致炎症和癌症侵袭。基质蛋白酶活性的一个结果是从细胞表面去除称为钙黏着蛋白的蛋白质。这些蛋白质在细胞粘附中起作用:它们像胶水一样将细胞粘在一起。没有他们,细胞可以从组织中分离并离开,癌细胞侵入其他器官的关键步骤。然而,目前尚不清楚基质蛋白酶是如何触发钙黏着蛋白从细胞表面去除以促进侵袭的。以前的工作表明,间质蛋白酶在称为蛋白酶激活受体2或简称Par2的受体上切换,已知可以激活许多酶,包括一种叫做磷脂酶C的酶。这种酶向细胞释放两种信号:一种叫做肌醇三磷酸的糖,IP3;和一种叫做二酰基甘油的脂质或脂肪,DAG.这两种信号可能在基质蛋白酶如何从细胞表面去除钙黏着蛋白中起作用。为了找出答案,Maetal.绘制了缺乏Hai1蛋白的斑马鱼中基质蛋白酶的作用。这表明基质蛋白酶会增加IP3和DAG水平,引发炎症和入侵。IP3通过打开细胞内的促炎信号如化学过氧化氢来促进炎症。同时,DAG通过激活称为MAPK的众所周知的癌症信号通路来促进细胞侵袭。该途径激活称为RSK的蛋白质。Maetal.表明这种蛋白质是从细胞表面去除钙黏着蛋白所必需的,从而将基质蛋白酶对磷脂酶C的激活与其在破坏细胞粘附中的作用联系起来。在许多癌症中存在基质蛋白酶与HAI-1(Hai1的人类等效物)的比率的增加。出于这个原因,Ma等人描述的信号级联。可能对开发这些癌症的治疗方法感兴趣。了解这些信号如何协同工作可能会导致未来更直接的靶向抗癌方法。
    Epithelial tissues are primed to respond to insults by activating epithelial cell motility and rapid inflammation. Such responses are also elicited upon overexpression of the membrane-bound protease, Matriptase, or mutation of its inhibitor, Hai1. Unrestricted Matriptase activity also predisposes to carcinoma. How Matriptase leads to these cellular outcomes is unknown. We demonstrate that zebrafish hai1a mutants show increased H2O2, NfκB signalling, and IP3R -mediated calcium flashes, and that these promote inflammation, but do not generate epithelial cell motility. In contrast, inhibition of the Gq subunit in hai1a mutants rescues both the inflammation and epithelial phenotypes, with the latter recapitulated by the DAG analogue, PMA. We demonstrate that hai1a has elevated MAPK pathway activity, inhibition of which rescues the epidermal defects. Finally, we identify RSK kinases as MAPK targets disrupting adherens junctions in hai1a mutants. Our work maps novel signalling cascades mediating the potent effects of Matriptase on epithelia, with implications for tissue damage response and carcinoma progression.
    Cancer occurs when normal processes in the cell become corrupted or unregulated. Many proteins can contribute, including one enzyme called Matriptase that cuts other proteins at specific sites. Matriptase activity is tightly controlled by a protein called Hai1. In mice and zebrafish, when Hai1 cannot adequately control Matriptase activity, invasive cancers with severe inflammation develop. However, it is unclear how unregulated Matriptase leads to both inflammation and cancer invasion. One outcome of Matriptase activity is removal of proteins called Cadherins from the cell surface. These proteins have a role in cell adhesion: they act like glue to stick cells together. Without them, cells can dissociate from a tissue and move away, a critical step in cancer cells invading other organs. However, it is unknown exactly how Matriptase triggers the removal of Cadherins from the cell surface to promote invasion. Previous work has shown that Matriptase switches on a receptor called Proteinase-activated receptor 2, or Par2 for short, which is known to activate many enzymes, including one called phospholipase C. When activated, this enzyme releases two signals into the cell: a sugar called inositol triphosphate, IP3; and a lipid or fat called diacylglycerol, DAG. It is possible that these two signals have a role to play in how Matriptase removes Cadherins from the cell surface. To find out, Ma et al. mapped the effects of Matriptase in zebrafish lacking the Hai1 protein. This revealed that Matriptase increases IP3 and DAG levels, which initiate both inflammation and invasion. IP3 promotes inflammation by switching on pro-inflammatory signals inside the cell such as the chemical hydrogen peroxide. At the same time, DAG promotes cell invasion by activating a well-known cancer signalling pathway called MAPK. This pathway activates a protein called RSK. Ma et al. show that this protein is required to remove Cadherins from the surface of cells, thus connecting Matriptase’s activation of phospholipase C with its role in disrupting cell adhesion. An increase in the ratio of Matriptase to HAI-1 (the human equivalent of Hai1) is present in many cancers. For this reason, the signal cascades described by Ma et al. may be of interest in developing treatments for these cancers. Understanding how these signals work together could lead to more direct targeted anti-cancer approaches in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号