15/10/02说明:此前论坛服务器频繁出错,现已更换服务器。今后论坛继续数据库备份,不备份上传附件。

肝胆相照论坛

 

 

肝胆相照论坛 论坛 学术讨论& HBV English 环孢菌素衍生物抑制乙型肝炎病毒进入而不干扰NTCP转运蛋 ...
查看: 886|回复: 6
go

环孢菌素衍生物抑制乙型肝炎病毒进入而不干扰NTCP转运蛋白 [复制链接]

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30437 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

1
发表于 2017-3-17 20:06 |只看该作者 |倒序浏览 |打印
Cyclosporin derivatives inhibit hepatitis B virus entry without interfering with NTCP transporter activity [url=]Satomi Shimura[/url]
,  [url=]Koichi Watashi[/url][url=]Correspondence information about the author  Koichi Watashi[/url]Email the author  Koichi Watashi
,  [url=]Kento Fukano[/url]
,  [url=]Michael Peel[/url]
,  [url=]Ann Sluder[/url]
,  [url=]Fumihiro Kawai[/url]
,  [url=]Masashi Iwamoto[/url]
,  [url=]Senko Tsukuda[/url]
,  [url=]Junko S. Takeuchi[/url]
,  [url=]Takeshi Miyake[/url]
,  [url=]Masaya Sugiyama[/url]
,  [url=]Yuki Ogasawara[/url]
,  [url=]Sam-Yong Park[/url]
,  [url=]Yasuhito Tanaka[/url]
,  [url=]Hiroyuki Kusuhara[/url]
,  [url=]Masashi Mizokami[/url]
,  [url=]Camille Sureau[/url]
,  [url=]Takaji Wakita[/url]

See Editorial, pages 677–679

DOI:            http://dx.doi.org/10.1016/j.jhep.2016.11.009 |

Article Info






Background & AimsThe sodium taurocholate co-transporting polypeptide (NTCP) is the main target of most hepatitis B virus (HBV) specific entry inhibitors. Unfortunately, these agents also block NTCP transport of bile acids into hepatocytes, and thus have the potential to cause adverse effects. We aimed to identify small molecules that inhibit HBV entry while maintaining NTCP transporter function.


MethodsWe characterized a series of cyclosporine (CsA) derivatives for their anti-HBV activity and NTCP binding specificity using HepG2 cells overexpressing NTCP and primary human hepatocytes. The four most potent derivatives were tested for their capacity to prevent HBV entry, but maintain NTCP transporter function. Their antiviral activity against different HBV genotypes was analysed.


ResultsWe identified several CsA derivatives that inhibited HBV infection with a sub-micromolar IC50. Among them, SCY446 and SCY450 showed low activity against calcineurin (CN) and cyclophilins (CyPs), two major CsA cellular targets. This suggested that instead, these compounds interacted directly with NTCP to inhibit viral attachment to host cells, and have no immunosuppressive function.

Importantly, we found that SCY450 and SCY995 did not impair the NTCP-dependent uptake of bile acids, and inhibited multiple HBV genotypes including a clinically relevant nucleoside analog-resistant HBV isolate.


ConclusionsThis is the first example of small molecule selective inhibition of HBV entry with no decrease in NTCP transporter activity. It suggests that the anti-HBV activity can be functionally separated from bile acid transport. These broadly active anti-HBV molecules are potential candidates for developing new drugs with fewer adverse effects.


Lay summaryIn this study, we identified new compounds that selectively inhibited hepatitis B virus (HBV) entry, and did not impair bile acid uptake. Our evidence offers a new strategy for developing anti-HBV drugs with fewer side effects.



Keywords:                HBV, Infection, NTCP, Membrane transport proteins, Cyclosporine, Antiviral, PreS1, Replication, Hepatitis B virus, Hepatitis D virus, Bile acids and salts, Cyclophilins

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30437 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

2
发表于 2017-3-17 20:06 |只看该作者
环孢菌素衍生物抑制乙型肝炎病毒进入而不干扰NTCP转运蛋白活性
Satomi志村
,Koichi Watashi'关于作者Koichi Watashi的通信信息电子邮件作者Koichi Watashi
,Kento Fukano
,迈克尔·皮尔
,Ann Sluder
,Fumihiro Kawai
,Masashi Iwamoto
,Tsenko Tsukuda
,Junko S.Buchuchi
,Takeshi Miyake
,Masaya Sugiyama
,小笠原
,三永公园
,Yasuhito Tanaka
,Kuzuhara Hiroyuki
,Masashi Mizokami
,Camille Sureau
,Takaji Wakita
见编辑,第677-679页
文章的得分为15
DOI:http://dx.doi.org/10.1016/j.jhep.2016.11.009 |
显示文章信息

    抽象
    全文
    图片
    参考文献
    补充材料

    图形抽象

图缩略图fx1
背景和目的

牛磺胆酸钠共转运多肽(NTCP)是大多数乙型肝炎病毒(HBV)特异性进入抑制剂的主要靶标。不幸的是,这些试剂还阻断NTCP将胆汁酸转运到肝细胞中,因此具有引起不良作用的可能性。我们的目标是识别抑制HBV进入同时保持NTCP转运蛋白功能的小分子。
方法

我们使用过表达NTCP和原代人肝细胞的HepG2细胞来表征一系列的环孢菌素(CsA)衍生物的抗HBV活性和NTCP结合特异性。测试了四种最有效的衍生物防止HBV进入,但保持NTCP转运蛋白功能的能力。分析了它们对不同HBV基因型的抗病毒活性。
结果

我们确定几个CsA衍生物抑制HBV感染与亚微摩尔IC50。其中,SCY446和SCY450显示对钙调神经磷酸(CN)和亲环素(CyP),两种主要的CsA细胞靶标的低活性。这表明,相反,这些化合物直接与NTCP相互作用以抑制病毒附着于宿主细胞,并且没有免疫抑制功能。

重要的是,我们发现SCY450和SCY995没有损害NTCP依赖性胆汁酸摄取,并抑制多种HBV基因型,包括临床相关的核苷类似物抗性HBV隔离。
结论

这是小分子选择性抑制HBV进入的第一个实例,没有NTCP转运蛋白活性的降低。这表明抗HBV活性可以在功能上与胆汁酸转运分离。这些广泛活性的抗HBV分子是开发具有较少副作用的新药物的潜在候选者。
放置摘要

在这项研究中,我们确定了选择性抑制乙型肝炎病毒(HBV)进入,并没有削弱胆汁酸摄取的新化合物。我们的证据提供了一种新的策略,用于开发抗HBV药物,副作用较少。
关键词:
HBV,感染,NTCP,膜转运蛋白,环孢霉素,抗病毒,PreS1,复制,乙型肝炎病毒,D型肝炎病毒,胆汁酸和盐,亲环蛋白

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30437 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

3
发表于 2017-3-17 20:07 |只看该作者
Advancing hepatitis B virus entry inhibitors
Eloi R. Verrier
, Catherine Schuster
, Thomas F. Baumert'Correspondence information about the author Thomas F. BaumertEmail the author Thomas F. Baumert
See Article, pages 685–692
Article has an altmetric score of 5
DOI: http://dx.doi.org/10.1016/j.jhep.2016.11.028 |
showArticle Info

    Abstract
    Full Text
    Images
    References

Article Outline

    Conflict of interest
    Authors’ contributions
    References

Despite the effective antivirals and vaccines, chronic hepatitis B virus (HBV) infection remains a major public health problem worldwide [[1], [2]]. Due to limited access, late or absent detection and vertical transmission, the prevalence of HBV infection is estimated to be 250–350 million infected individuals [[1], [3]]. HBV infection is a leading cause of hepatocellular carcinoma worldwide. While current therapies, based on pegylated-interferon and nucleos(t)ide analogs effectively decrease viral load, viral elimination is rare. Thus, there is an unmet medical need for novel therapeutic strategies effectively curing HBV infection [[1], [2], [4]]. HBV entry inhibitors have been introduced as a novel class of antivirals for viral control and cure [[1], [5], [6], [7]]. The discovery of the Na+-taurocholate cotransporting polypeptide (NTCP) as the first receptor of HBV opened the door to understand the molecular mechanisms of viral cell entry and to identify entry inhibitors (reviewed in [8]). In the liver, HBV virions bind to NTCP, a bile acid transporter expressed at the basolateral membrane of hepatocytes [9], triggering viral entry. The preS1 region of HBV envelope interacts with the bile acid pocket site of the transporter, corresponding to the amino acids 157 to 165 of the protein [8], therefore interfering with its transporter function. At the same time NTCP has been explored in great detail as a target for antiviral therapy (reviewed in [5]). Indeed, small molecules interacting with NTCP exhibit antiviral activity, including the immunosuppressive drug cyclosporin A (CsA) [5]. Moreover, a preS1 peptide derived from the HBV envelope and specifically binding to the receptor has been shown to exhibit marked antiviral activity in cell culture, animal models and patients (for reviews, see [[5], [6]]). However, as HBV and bile acids share the same interaction site with NTCP, the current HBV NTCP entry inhibitors are also capable of interfering with bile acid uptake. This may induce putative adverse effects by impairing Na+-taurocholate transport in hepatocytes. Indeed NTCP deficiency in patients leads to hypercholanemia for instance [10] and an increase in glycine-conjugated bile acid concentrations was also observed in some patients treated with myrcludex B, a preS1 peptide binding to NTCP and currently evaluated in a clinical trial [11]. Although bile acid related adverse effects appeared to be limited or absent [11], the discovery of HBV entry inhibitors not affecting the NTCP transporter function would represent an interesting conceptual advancement addressing a potential safety concern.

In this issue of Journal of Hepatology, Shimura and colleagues [19] addressed this issue, by investigating the ability of CsA derivatives to inhibit HBV infection without interfering with bile acid transport. To do so, they took advantage of an HBV infection model based on NTCP-overexpressing HepG2 cells, named HepG2-hNTCP-C4, which enables HBV infection [12]. The authors synthesized eleven CsA derivatives and characterized their inhibitory activity on virus infection. Observing that five of these drugs were able to inhibit HBV infection in a pan-genotypic manner, they demonstrated that four of them (named SCY806, SCY446, SCY450, and SCY995) have no immunosuppressive activity and were capable of suppressing HBV infection in primary human hepatocytes (PHH). To further characterize the antiviral activity of these compounds, they then tested their inhibitory effect at different stages of viral infection. Using non-susceptible HBV replicating cells, they demonstrated that CsA derivatives had no effect on HBV replication. In contrast, taking advantage of a fluorescent-labeled preS1 peptide, which specifically binds to NTCP [13], the authors showed that these drugs were able to prevent preS1-NTCP interaction, confirming the capacity of CsA derivatives to inhibit HBV entry by interfering with the binding of the virus to its receptor. They validated these findings using surface plasmon resonance analysis, showing that CsA derivatives were capable of interacting with recombinant NTCP protein, such as CsA itself. As CsA is known to target NTCP-mediated bile acid uptake as well, the authors set up a bile acid uptake assay based on HepG2-hNTCP-C4 cells to evaluate the putative effect of CsA derivatives on NTCP transporter function. Interestingly, while SCY806 and SCY446 do inhibit bile acid uptake similarly to CsA, the two other derivatives, SCY450 and SCY995, had no effect on NTCP-mediated bile acid transport in spite of a strong HBV inhibition, suggesting that the bile acid transporter and HBV receptor functions of NTCP can potentially be dissociated. This hypothesis was further confirmed by sulfobromophthalein, a substrate of NTCP interacting with the bile acid pocket site of the protein [14], which was shown to inhibit bile acid uptake without affecting HBV infection. Taken together, the authors conclude that it is possible to develop NTCP-targeting HBV entry inhibitors without affecting the bile acid transporter function of NTCP.

These results have implications for the understanding of HBV entry as well as the development of HBV entry inhibitors as antivirals. First, the results obtained by Shimiura et al. suggest that NTCP domains mediating HBV entry and bile acid transport may not be completely identical. The newly identified compounds could serve as a tool to further elucidate virus-NTCP interactions during HBV cell entry. Furthermore, their findings could advance the further development of HBV entry inhibitors: while host-targeting agents (HTAs) have been shown to be effective against HBV, HDV and HCV infections [[7], [15], [16]], target-specific adverse effects need to be carefully addressed [17]. Although it has been suggested that auxiliary transporters may be able to sustain the enterohepatic cycle in the absence of NTCP [10], it cannot be excluded that pharmacological long-term modulation of bile acid transport may result in previously undiscovered adverse effects. Shimura and colleagues addressed this issue by the identification of CsA derivatives which neither appeared to exhibit immunosuppressive activity, nor interfer with NTCP-mediated bile acid transport. Mechanistically, their study suggests that the SCY995 molecule may target an alternative site of NTCP (putatively overlapping with the bile acid pocket site) compared to native CsA, which may allow to inhibit viral entry without blocking bile acid uptake (Fig. 1). However, it is important to note while no effect was observed in the range of concentrations used in the study, it cannot be excluded that SCY995 interferes with NTCP bile acid transporter function at higher concentrations. Furthermore it would be of interest to investigate whether SCY995 interferes with the recently described antiviral innate immune responses mediated by the NTCP bile acid transporter function [18]. Collectively, the findings of Shimura et al. advance our knowledge by showing: (i) that it appears to be possible to dissect HBV entry and bile acid transporter function; and (ii) providing an opportunity to optimize entry inhibitors by potentially increasing their specificity and decreasing potential adverse effects. However, it would be important to assess whether the compounds exhibit other off-target effects such as interference with other transporters (e.g. the hepatic organic anion transporting polypeptide OATP). Detailed studies of efficacy and safety in animal models and ultimately in the HBV infected patients are needed to evaluate the positioning of the approach introduced by Shimura and colleagues compared to the entry inhibitor myrcludex B [11], other HTAs (for review see [7]) and clinically licensed HBV antivirals.
Opens large image
Fig. 1

Model of interaction of selected HBV entry inhibitors and Na+-taurocholate cotransporting polypeptide (NCTP) at the hepatocyte basolateral membrane. According to Shimura and colleagues, CSY995, a CsA derivative exhibiting no immunosuppressive activity, contrary to CsA, appears to bind to NTCP via an alternative site (putatively overlapping with the bile acid pocket site of the protein), allowing bile acid transport in spite of a strong anti-HBV activity. Myrcludex B (MyrB) is a myristoylated peptide derived from the HBV envelope specifically binding to NTCP, inhibiting both HBV entry and bile acid transport. CsA, cyclosporine A; HBV, hepatitis B virus.

View Large Image | View Hi-Res Image | Download PowerPoint Slide

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30437 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

4
发表于 2017-3-17 20:12 |只看该作者
推进乙型肝炎病毒进入抑制剂
Eloi R. Verrier
,凯瑟琳·舒斯特
,Thomas F. Baumert'Correspondence information about the author Thomas F. BaumertEmail the author Thomas F. Baumert
参见文章,第685-692页
文章的得分为5
DOI:http://dx.doi.org/10.1016/j.jhep.2016.11.028 |
显示文章信息


尽管有效的抗病毒和疫苗,慢性乙型肝炎病毒(HBV)感染仍然是全世界的主要公共卫生问题[[1],[2]]。由于访问受限,检测和垂直传播迟到或缺失,HBV感染的流行率估计为2.5-3.5亿感染者[[1],[3]]。 HBV感染是全世界肝细胞癌的主要原因。虽然目前的治疗,基于聚乙二醇化干扰素和核苷类似物有效减少病毒载量,病毒清除是罕见的。因此,对于有效治愈HBV感染的新的治疗策略存在未满足的医学需要[[1],[2],[4]]。已经引入HBV进入抑制剂作为用于病毒控制和治愈的新型抗病毒剂[[1],[5],[6],[7]]。作为HBV的第一受体的Na + - 交联胆酸盐共转运多肽(NTCP)的发现开启了了解病毒细胞进入的分子机制和识别进入抑制剂的门(参见[8])。在肝脏中,HBV病毒颗粒结合NTCP,一种在肝细胞基底外膜表达的胆汁酸转运蛋白[9],触发病毒进入。 HBV包膜的前S1区域与转运蛋白的胆汁酸口袋位点相互作用,对应于蛋白质的氨基酸157至165,因此干扰其转运蛋白功能。同时,NTCP作为抗病毒治疗的目标被详细研究(参见[5])。事实上,与NTCP相互作用的小分子表现出抗病毒活性,包括免疫抑制药物环孢菌素A(CsA)[5]。此外,来自HBV包膜并特异性结合受体的preS1肽已显示在细胞培养物,动物模型和患者中显示出显着的抗病毒活性(参见[[5],[6]]。然而,由于HBV和胆汁酸与NTCP共享相同的相互作用位点,所以目前的HBV NTCP进入抑制剂也能够干扰胆汁酸摄取。这可能通过损害肝细胞中Na + - 胆固醇的转运来诱导推定的不良反应。实际上,NTCP缺乏导致高胆固醇血症[10],并且在一些用myrcludex B(一种与NTCP结合的preS1肽)治疗的患者中观察到甘氨酸结合的胆汁酸浓度的增加,并且目前在临床试验中进行评估[11] 。虽然胆汁酸相关的不良反应似乎有限或不存在[11],发现不影响NTCP转运蛋白功能的HBV进入抑制剂将代表一个有趣的概念进步,解决潜在的安全问题。

在这期“肝脏病学杂志”上,志村及其同事[19]通过研究CsA衍生物抑制HBV感染而不干扰胆汁酸转运的能力来解决这个问题。为此,他们利用了基于过表达的NTCP过表达HepG2细胞的HBV感染模型,命名为HepG2-hNTCP-C4,这使HBV感染成为可能[12]。作者合成了十一种CsA衍生物,并表征了它们对病毒感染的抑制活性。观察到这些药物中的五种能够以泛基因型方式抑制HBV感染,他们证明其中四种(命名为SCY806,SCY446,SCY450和SCY995)没有免疫抑制活性,能够抑制原发性人类的HBV感染肝细胞(PHH)。为了进一步表征这些化合物的抗病毒活性,他们然后测试了它们在病毒感染的不同阶段的抑制作用。使用不敏感的HBV复制细胞,他们表明CsA衍生物对HBV复制没有影响。相比之下,利用特异性结合NTCP的荧光标记的preS1肽[13],作者显示这些药物能够预防preS1-NTCP相互作用,证实CsA衍生物通过干扰抑制HBV进入的能力病毒与其受体的结合。他们使用表面等离振子共振分析验证了这些发现,显示CsA衍生物能够与重组NTCP蛋白(如CsA本身)相互作用。由于已知CsA靶向NTCP介导的胆汁酸摄取,作者建立了基于HepG2-hNTCP-C4细胞的胆汁酸摄取测定以评价CsA衍生物对NTCP转运蛋白功能的推定效应。有趣的是,尽管SCY806和SCY446确实抑制胆汁酸吸收类似于CsA,但是两种其它衍生物SCY450和SCY995对于NTCP介导的胆汁酸转运没有影响,尽管HBV抑制作用强,这表明胆汁酸转运蛋白和HBV NTCP的受体功能可以潜在地解离。这一假说进一步由磺基溴菌素(NTCP与蛋白质的胆汁酸口袋位点相互作用的底物[14])证实,其显示抑制胆汁酸摄取而不影响HBV感染。总之,作者得出结论,可以开发靶向NTCP的HBV进入抑制剂而不影响NTCP的胆汁酸转运蛋白功能。

这些结果对于了解HBV进入以及作为抗病毒药的HBV进入抑制剂的发展具有影响。首先,Shimiura等人表明介导HBV进入和胆汁酸转运的NTCP结构域可能不完全相同。新鉴定的化合物可以作为进一步阐明HBV细胞进入期间病毒-NTCP相互作用的工具。此外,他们的发现可以推动HBV进入抑制剂的进一步发展:虽然主机靶向剂(HTAs)已被证明对HBV,HDV和HCV感染有效[[7],[15],[16]],靶 - 具体的不良反应需要仔细处理[17]。尽管已经提出辅助转运蛋白可以在不存在NTCP的情况下维持肠肝周期[10],但不能排除胆汁酸转运的药理学长期调节可能导致先前未发现的不良反应。 Shimura和同事通过鉴定既不表现出免疫抑制活性的CsA衍生物也不干扰NTCP介导的胆汁酸转运来解决这个问题。机械地,他们的研究表明,与天然CsA相比,SCY995分子可以靶向NTCP的替代位点(推定与胆汁酸口袋位点重叠),这可以允许抑制病毒进入而不阻断胆汁酸摄取(图1)。然而,重要的是注意,虽然在研究中使用的浓度范围没有观察到效果,但不能排除SCY995在较高浓度下干扰NTCP胆汁酸转运蛋白功能。此外,调查SCY995是否干扰最近描述的由NTCP胆汁酸转运蛋白功能介导的抗病毒先天免疫应答是有意义的[18]。总的来说,Shimura et al。通过显示:(i)似乎有可能解剖HBV进入和胆汁酸转运蛋白功能;和(ii)通过潜在地增加其特异性和减少潜在的不利影响提供优化进入抑制剂的机会。然而,重要的是评估化合物是否显示其它脱靶效应,例如干扰其它转运蛋白(例如肝有机阴离子转运多肽OATP)。需要详细研究动物模型和最终在HBV感染患者中的功效和安全性,以评估由Shimura和同事介绍的方法与进入抑制剂myrcludex B [11],其他HTAs(参见[7] )和临床许可的HBV抗病毒药。
打开大图像
图。 1

选择的HBV进入抑制剂和Na + - 牛磺胆酸酯共转运多肽(NCTP)在肝细胞基底外膜的相互作用模型。根据志村和同事,CSY995,显示没有免疫抑制活性,与CsA相反的CsA衍生物似乎通过替代位点(推定与蛋白质的胆汁酸口袋位点重叠)结合NTCP,允许胆汁酸运输,尽管具有很强的抗HBV活性。 Myrcludex B(MyrB)是衍生自特异性结合NTCP的HBV包膜的豆蔻酰化肽,其抑制HBV进入和胆汁酸转运。 CsA,环孢霉素A; HBV,乙型肝炎病毒。

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30437 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

5
发表于 2017-3-17 20:13 |只看该作者
已有 1 人评分现金 收起 理由
MP4 + 1

总评分: 现金 + 1   查看全部评分

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30437 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

6
发表于 2017-3-17 20:13 |只看该作者

Rank: 6Rank: 6

现金
309 元 
精华
帖子
295 
注册时间
2015-6-30 
最后登录
2020-5-31 
7
发表于 2017-3-17 21:39 |只看该作者
感谢分享!
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册

肝胆相照论坛

GMT+8, 2024-5-22 23:58 , Processed in 0.038746 second(s), 12 queries , Gzip On.

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.