bile acid transporter

  • 文章类型: Journal Article
    据报道,肠肝循环在PFAS的生物积累中起着重要作用。在这项研究中,全氟辛烷磺酸及其替代品的组织分布和排泄,使用小鼠测定法研究了6:2和8:2氟调聚物磺酸(FTSA),重点是肠肝循环的作用。肝脏是全氟辛烷磺酸和8:2FTSA的主要积累器官(14天后吸收总剂量的33.4%和25.8%),而65%的6:2FTSA在24小时内通过尿液排出。胆囊中发现8:2FTSA和PFOS的峰值水平,提示肝肠循环在PFAS重吸收中的重要作用。通过8:2FTSA和PFOS与药物(即二甲双胍(MET)和熊去氧胆酸(UDCA))共同暴露,进一步评估了肠肝循环的作用。MET使肝脏中8:2FTSA和PFOS的积累减少了68.6%和65.8%,通过下调胆汁酸转运蛋白(Asbt)和增强粪便排泄。相反,与仅暴露于PFAS的浓度相比,UDCA将其浓度提高了21.9%和34.6%。在PFAS血清水平和Asbt表达之间鉴定出强的正相关。这项研究阐明了PFAS的生物积累机制,并提出了减轻暴露风险的潜在策略。
    Enterohepatic circulation has been reported to play a significant role in the bioaccumulation of PFASs. In this study, the tissue distribution and excretion of PFOS and its alternatives, namely 6:2 and 8:2 fluorotelomer sulfonic acid (FTSA) was investigated using a mouse assay with a focus on role of enterohepatic circulation. Liver was the primarily accumulating organ for PFOS and 8:2 FTSA (33.4 % and 25.8 % of total doses absorbed after 14 days), whereas 65 % of 6:2 FTSA was excreted via urine within 24 h. Peak levels of 8:2 FTSA and PFOS were found in the gallbladder, implying the important role of enterohepatic circulation in PFASs reabsorption. The role of enterohepatic circulation was further evaluated through co-exposure of 8:2 FTSA and PFOS with medicines (namely metformin (MET) and ursodeoxycholic acid (UDCA)). MET reduced accumulation of 8:2 FTSA and PFOS in the liver by 68.6 % and 65.8 %, through down-regulation of bile acid transporter (Asbt) and enhancement of fecal excretion. Conversely, UDCA raised their concentrations by 21.9 % and 34.6 % compared to that exposed solely to PFASs. A strong positive correlation was identified between PFASs serum levels and Asbt expression. This study illuminated PFAS bioaccumulation mechanisms and suggested potential strategies to mitigate the exposure risks.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:口服胰岛素是治疗糖尿病的潜在候选药物。然而,它被胃肠道屏障阻塞,导致口服生物利用度可忽略不计。
    方法:本研究提出了一种旨在应对这些挑战的新型纳米载体平台。在这方面,该过程涉及海藻酸钠被乙二胺胺化,然后与脱氧胆酸结合。
    结果:所得的DCA@Alg@INS纳米载体显示出63.6±1.03%的胰岛素负荷含量和87.6±3.84%的包封率,粒径为206nm,ζ电位为-3mV。体外研究显示胰岛素从纳米颗粒的持续和pH依赖性释放曲线。体外细胞研究,共聚焦激光扫描显微镜和流式细胞术分析证实了DCA@Alg@INS纳米颗粒在Caco-2细胞中的成功附着和内化。此外,相对于胰岛素溶液,DCA@Alg@INS表现出优异的细胞摄取能力和渗透系数,表现出六倍和4.94倍的增强,分别。根据摄取机制研究,结果表明,DCA@Alg@INS主要通过能量依赖性活性途径运输,因为在NaN3存在下,细胞对DCA@Alg@INS的摄取显着降低了〜92%,在4°C的低温下降低了〜94%。
    结论:鉴于口服胰岛素的重要性,脱氧胆酸修饰的藻酸盐纳米颗粒提供了一种可行的选择,可以克服胃肠道带来的各种障碍。
    OBJECTIVE: Oral administration of insulin is a potential candidate for managing diabetes. However, it is obstructed by the gastrointestinal tract barriers resulting in negligible oral bioavailability.
    METHODS: This investigation presents a novel nanocarrier platform designed to address these challenges. In this regard, the process involved amination of sodium alginate by ethylene diamine, followed by its conjugation with deoxycholic acid.
    RESULTS: The resulting DCA@Alg@INS nanocarrier revealed a significantly high insulin loading content of 63.6 ± 1.03% and encapsulation efficiency of 87.6 ± 3.84%, with a particle size of 206 nm and zeta potentials of -3 mV. In vitro studies showed sustained and pH-dependent release profiles of insulin from nanoparticles. In vitro cellular studies, confocal laser scanning microscopy and flow cytometry analysis confirmed the successful attachment and internalization of DCA@Alg@INS nanoparticles in Caco-2 cells. Furthermore, the DCA@Alg@INS demonstrated a superior capacity for cellular uptake and permeability coefficient relative to the insulin solution, exhibiting sixfold and 4.94-fold enhancement, respectively. According to the uptake mechanism studies, the results indicated that DCA@Alg@INS was mostly transported through an energy-dependent active pathway since the uptake of DCA@Alg@INS by cells was significantly reduced in the presence of NaN3 by ~ 92% and at a low temperature of 4°C by ~ 94%.
    CONCLUSIONS: Given the significance of administering insulin through oral route, deoxycholic acid-modified alginate nanoparticles present a viable option to surmount various obstacles presented by the gastrointestinal.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Preprint
    胆汁酸钠转运体(BASS)家族跨膜运输广泛的分子,包括人类的胆汁酸,和植物中的小代谢产物。这些运输机,其中许多是钠偶联的,已经显示出使用电梯运输机构,但是底物结合与钠离子结合和转运的确切方式尚不清楚。在这里,我们解决了来自脑膜炎奈瑟菌(ASBTNM)的转运体与泛解物的2.3µA的晶体结构,ASBTNM的潜在底物。BASS家族的特征在于两个螺旋在蛋白质的中心交叉排列,所述排列由两个钠离子错综复杂地保持在一起。我们观察到泛解酯结合,具体来说,在该交叉区域中两个相对螺旋的N末端之间。在分子动力学模拟过程中,当存在钠离子时,泛素酸保持在该位置,但在不存在钠离子时更具流动性。在存在和不存在泛素酸的情况下的结构比较表明,泛素酸引起交叉螺旋之一的构象变化。这修改了相对于彼此移动的两个域之间的界面以引出升降机构。这些结果有影响,不仅对于ASBTNM,而且对于整个BASS家族以及通过电梯机制工作的其他运输者。
    The Bile Acid Sodium Symporter (BASS) family transports a wide array of molecules across membranes, including bile acids in humans, and small metabolites in plants. These transporters, many of which are sodium-coupled, have been shown to use an elevator mechanism of transport, but exactly how substrate binding is coupled to sodium ion binding and transport is not clear. Here we solve the crystal structure at 2.3 Å of a transporter from Neisseria Meningitidis (ASBTNM) in complex with pantoate, a potential substrate of ASBTNM. The BASS family is characterised by two helices that cross-over in the centre of the protein in an arrangement that is intricately held together by two sodium ions. We observe that the pantoate binds, specifically, between the N-termini of two of the opposing helices in this cross-over region. During molecular dynamics simulations the pantoate remains in this position when sodium ions are present but is more mobile in their absence. Comparison of structures in the presence and absence of pantoate demonstrates that pantoate elicits a conformational change in one of the cross-over helices. This modifies the interface between the two domains that move relative to one another to elicit the elevator mechanism. These results have implications, not only for ASBTNM but for the BASS family as a whole and indeed other transporters that work through the elevator mechanism.
    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
    NAFLD和NASH的患病率在全球范围内不断增加,到目前为止还没有批准的医疗方法。有证据表明,干扰胆汁酸代谢可能会导致NASH的改善。在这项研究中,对28例胆汁淤积性肝功能检查(尤其是GGT)升高的患者进行胆汁酸基因多态性筛查,并接受UDCA治疗。所有患者在ABCB4或ABCB11中都存在胆汁酸基因多态性。用UDCA治疗12个月显著降低了所有患者的GGT和纯合患者的ALT。使用FIb-4,NFS,和瞬时弹性成像(TE)。PNPLA3和TM6SF2是最常见的NASH相关多态性,并且TM6SF2患者在施用UDCA后GGT和ALT显着降低。总之,GGT升高的NASH患者应筛查胆汁酸基因多态性,因为UDCA治疗可以改善肝功能检查。然而,临床结果没有差异,比如进展为肝硬化,已使用非侵入性测试(NIT)观察到。
    The prevalence of NAFLD and NASH is increasing worldwide, and there is no approved medical treatment until now. Evidence has emerged that interfering with bile acid metabolism may lead to improvement in NASH. In this study, 28 patients with elevated cholestatic liver function tests (especially GGT) were screened for bile acid gene polymorphisms and treated with UDCA. All patients had a bile acid gene polymorphism in ABCB4 or ABCB11. Treatment with UDCA for 12 months significantly reduced GGT in all patients and ALT in homozygous patients. No difference in fibrosis was observed using FIb-4, NFS, and transient elastography (TE). PNPLA3 and TM6SF2 were the most common NASH-associated polymorphisms, and patients with TM6SF2 showed a significant reduction in GGT and ALT with the administration of UDCA. In conclusion, NASH patients with elevated GGT should be screened for bile acid gene polymorphisms, as UDCA therapy may improve liver function tests. However, no difference in clinical outcomes, such as progression to cirrhosis, has been observed using non-invasive tests (NITs).
    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
    目的:OATP1B3/SLCO1B3是用于清除内源性化合物(例如胆汁酸/BA)和外源性物质的人类肝脏特异性转运蛋白。OATP1B3在人类中的功能作用尚未得到表征,因为SLCO1B3在没有小鼠直系同源物的物种中保守性差。
    方法:Slc10a1-敲除(Slc10a1-/-),Slc10a1hSLCO1B3(内源性小鼠Slc10a1启动子驱动的人SLCO1B3在Slc10a1-/-小鼠中的表达),和人SLCO1B3肝脏特异性转基因(hSLCO1B3-LTG)小鼠产生并用0.1%熊去氧胆酸(UDCA)攻击,1%胆酸(CA)饮食或胆管结扎(BDL)用于功能研究。原代肝细胞和肝癌-PLC/RPF/5细胞用于机理研究。
    结果:与WT小鼠相比,在有或没有0.1%UDCA喂养的情况下,Slc10a1-/-小鼠中的血清BA水平显著增加。这种增加在Slc10a1hSLCO1B3-小鼠中减弱,表明OATP1B3作为重要的肝脏BA摄取转运蛋白。使用来自WT的原代肝细胞的体外测定,Slc10a1-/-,Slc10a1hSLCO1B3-小鼠表明OATP1B3具有与Ntcp相似的摄取牛磺胆酸盐/TCA的能力。此外,TCA诱导的胆汁流量在Slc10a1-/-小鼠中明显受损,但在Slc10a1hSLC01B3小鼠中部分恢复,表明OATP1B3可以在体内部分补偿NTCP功能。OATP1B3的肝脏特异性过表达显着增加了1%CA喂养和BDL小鼠的肝结合BA和胆汁淤积性肝损伤的水平。机制研究表明,缀合的BAs刺激Ccl2和Cxcl2在肝细胞中增加肝中性粒细胞浸润和促炎细胞因子的产生(例如IL-6),通过与其启动子结合激活STAT3来抑制OATP1B3的表达。
    结论:人OATP1B3是一种重要的BA摄取转运蛋白,并且可以部分补偿Ntcp在小鼠中的共轭BA摄取。其在胆汁淤积中的下调是一种适应性保护性反应。
    OATP1B3/SLCO1B3 is a human liver-specific transporter for the clearance of endogenous compounds (eg, bile acid [BA]) and xenobiotics. The functional role of OATP1B3 in humans has not been characterized, as SLCO1B3 is poorly conserved among species without mouse orthologs.
    Slc10a1-knockout (Slc10a1-/-), Slc10a1hSLCO1B3 (endogenous mouse Slc10a1 promoter-driven human-SLCO1B3 expression in Slc10a1-/- mice), and human SLCO1B3 liver-specific transgenic (hSLCO1B3-LTG) mice were generated and challenged with 0.1% ursodeoxycholic-acid (UDCA), 1% cholic-acid (CA) diet, or bile duct ligation (BDL) for functional studies. Primary hepatocytes and hepatoma-PLC/RPF/5 cells were used for mechanistic studies.
    Serum BA levels in Slc10a1-/- mice were substantially increased with or without 0.1% UDCA feeding compared with wild-type (WT) mice. This increase was attenuated in Slc10a1hSLCO1B3-mice, indicating that OATP1B3 functions as a significant hepatic BA uptake transporter. In vitro assay using primary hepatocytes from WT, Slc10a1-/-, and Slc10a1hSLCO1B3-mice indicated that OATP1B3 has a similar capacity in taking up taurocholate/TCA as Ntcp. Furthermore, TCA-induced bile flow was significantly impaired in Slc10a1-/- mice but partially recovered in Slc10a1hSLC01B3-mice, indicating that OATP1B3 can partially compensate the NTCP function in vivo. Liver-specific overexpression of OATP1B3 markedly increased the level of hepatic conjugated BA and cholestatic liver injury in 1% CA-fed and BDL mice. Mechanistic studies revealed that conjugated BAs stimulated Ccl2 and Cxcl2 in hepatocytes to increase hepatic neutrophil infiltration and proinflammatory cytokine production (eg, IL-6), which activated STAT3 to repress OATP1B3 expression by binding to its promoter.
    Human OATP1B3 is a significant BA uptake transporter and can partially compensate Ntcp for conjugated BA uptake in mice. Its downregulation in cholestasis is an adaptive protective response.
    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
    目的:比较GIMMER患者和日本人群的患者特征和结局。
    方法:GLIMMER是一个多中心,双盲,随机化,安慰剂对照,2b期研究评估linerixibat对原发性胆汁性胆管炎(PBC)患者瘙痒的治疗。
    结果:在GLIMMER总体人群中随机分配了147例患者,其中38例患者包括日本人群。人口统计学和基线临床特征在治疗组之间和两个人群之间相似。从基线到第16周(主要终点),所有组的平均最差每日瘙痒评分均降低,对于日本和整个人群,linerixibat每天两次(BID;-2.92[95%CI:-5.07,-0.76]和-2.86[95%CI:-3.76,-1.95]观察到最大的减少,分别)。无论所应用的响应者定义如何,在两个群体中,响应者的比例最高通常在40mgBID组中。健康相关生活质量的改善在两个人群中通常是一致的。在日本和总人口中,安慰剂组中25%和19%的患者以及linerixibat组中0-86%和31-78%的患者报告了治疗中药物相关的不良事件,分别。与作用机制一致,最常见的事件是胃肠道事件.利奈西巴特对药效学生物标志物的影响有利于BID给药。
    结论:linerixibat的治疗反应和安全性在日本人和GIMMER的总体人群之间是一致的。Linerixibat可能为PBC患者的胆汁淤积性瘙痒提供有效的治疗选择。本文受版权保护。保留所有权利。
    OBJECTIVE: To compare patient characteristics and outcomes between the overall and Japanese populations of GLIMMER.
    METHODS: GLIMMER was a multicenter, double-blind, randomized, placebo-controlled, Phase IIb study evaluating linerixibat for the treatment of pruritus in patients with primary biliary cholangitis.
    RESULTS: In total, 147 patients were randomized in the GLIMMER overall population with 38 patients comprising the Japanese population. Demographics and baseline clinical characteristics were similar across treatment groups and between both populations. A reduction in mean worst daily itch score from baseline to week 16 (primary endpoint) was seen in all groups, with the largest reduction observed with linerixibat 40 mg twice daily (BID; -2.92 [95% confidence interval: -5.07, -0.76] and -2.86 [95% confidence interval: -3.76, -1.95] for Japanese and overall populations, respectively). The highest proportion of responders was generally in the 40 mg BID group in both populations regardless of the responder definition applied. Improvements in health-related quality of life were generally consistent in both populations. In the Japanese and overall populations, on-treatment drug-related adverse events were reported in 25% and 19% of patients in the placebo group and 0%-86% and 31%-78% of patients in the linerixibat groups, respectively. Consistent with the mechanism of action, the most common events were gastrointestinal in nature. The effects of linerixibat on pharmacodynamic biomarkers favored BID dosing.
    CONCLUSIONS: Therapeutic responses and safety of linerixibat were consistent between the Japanese and overall populations of GLIMMER. Linerixibat may provide an effective treatment option for cholestatic pruritus in patients with primary biliary cholangitis.
    BACKGROUND: NCT02966834.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Letter
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    由于缺乏基因动物模型,充分重建肝硬化的关键临床特征,肝硬化的分子发病机制尚未明确,治疗仍然有限。因此,我们的目的是使用一种新型小鼠模型更好地阐明肝硬化的病理机制。
    我们报道了第一个模拟由肝细胞特异性消除微珠蛋白1(MCRS1)诱导的人类肝硬化的小鼠遗传模型,非特异性致死(NSL)和INO80染色质修饰复合物的成员。使用这种遗传工具和其他小鼠模型,细胞培养和人体样本,结合定量蛋白质组学,细胞核/细胞RNA测序和染色质免疫沉淀测定,我们研究了肝硬化的机制。
    小鼠肝细胞中的MCRS1缺失调节胆汁酸(BA)转运体的表达-Na-牛磺胆酸共转运多肽(NTCP)的显着下调-浓缩窦中的BA,从而通过法尼醇X受体(FXR)激活肝星状细胞(HSC),其主要在人和小鼠HSC中表达。始终如一,NTCP在小鼠中的再表达减少肝硬化,HSC中FXR的遗传消融抑制小鼠和体外细胞培养中的纤维化标记。机械上,从MCRS1中删除假定的SANT结构域会从组蛋白H3锚定位点驱逐组蛋白脱乙酰酶1,增加BA转运蛋白基因的组蛋白乙酰化,调节它们的表达并扰乱BA流。因此,人肝硬化显示核MCRS1和NTCP表达降低。
    我们的数据揭示了MCRS1作为关键组蛋白乙酰化调节因子的先前未被识别的功能,维持基因表达和肝脏稳态。MCRS1缺失诱导BA转运蛋白基因乙酰化,BA流的扰动,因此,HSC中的FXR活化。该轴代表肝硬化中的中心和普遍信号事件,这对肝硬化治疗有重要意义。
    通过MCRS1在小鼠肝细胞中的遗传消融,我们产生了第一个能概括人类特征的肝硬化遗传小鼠模型。在这里,我们证明肝成纤维细胞中胆汁酸/FXR轴的激活是肝硬化发展的关键。
    Owing to the lack of genetic animal models that adequately recreate key clinical characteristics of cirrhosis, the molecular pathogenesis of cirrhosis has been poorly characterized, and treatments remain limited. Hence, we aimed to better elucidate the pathological mechanisms of cirrhosis using a novel murine model.
    We report on the first murine genetic model mimicking human cirrhosis induced by hepatocyte-specific elimination of microspherule protein 1 (MCRS1), a member of non-specific lethal (NSL) and INO80 chromatin-modifier complexes. Using this genetic tool with other mouse models, cell culture and human samples, combined with quantitative proteomics, single nuclei/cell RNA sequencing and chromatin immunoprecipitation assays, we investigated mechanisms of cirrhosis.
    MCRS1 loss in mouse hepatocytes modulates the expression of bile acid (BA) transporters - with a pronounced downregulation of Na+-taurocholate cotransporting polypeptide (NTCP) - concentrating BAs in sinusoids and thereby activating hepatic stellate cells (HSCs) via the farnesoid X receptor (FXR), which is predominantly expressed in human and mouse HSCs. Consistently, re-expression of NTCP in mice reduces cirrhosis, and genetic ablation of FXR in HSCs suppresses fibrotic marks in mice and in vitro cell culture. Mechanistically, deletion of a putative SANT domain from MCRS1 evicts histone deacetylase 1 from its histone H3 anchoring sites, increasing histone acetylation of BA transporter genes, modulating their expression and perturbing BA flow. Accordingly, human cirrhosis displays decreased nuclear MCRS1 and NTCP expression.
    Our data reveal a previously unrecognized function of MCRS1 as a critical histone acetylation regulator, maintaining gene expression and liver homeostasis. MCRS1 loss induces acetylation of BA transporter genes, perturbation of BA flow, and consequently, FXR activation in HSCs. This axis represents a central and universal signaling event in cirrhosis, which has significant implications for cirrhosis treatment.
    By genetic ablation of MCRS1 in mouse hepatocytes, we generate the first genetic mouse model of cirrhosis that recapitulates human features. Herein, we demonstrate that the activation of the bile acid/FXR axis in liver fibroblasts is key in cirrhosis development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Treatment of β-lactamase positive bacterial infections with a combination of amoxicillin (AMOX) and clavulanic acid (CLAV) causes idiosyncratic drug-induced liver injury (iDILI) in a relevant number of patients, often with features of intrahepatic cholestasis. This study aims to determine serum bile acid (BA) levels in amoxicillin/clavulanate (A+C)-iDILI patients and to investigate the mechanism of cholestasis by A+C in human in vitro hepatic models. In six A+C-iDILI patients, significant elevations of serum primary conjugated BA definitely demonstrated A+C-induced cholestasis. In cultured human Upcyte hepatocytes and HepG2 cells, CLAV was more cytotoxic than AMOX, and, at subcytotoxic concentrations, it altered the expression of more than 1,300 genes. CLAV, but not AMOX, downregulated the expression of key genes for BA transport (BSEP, NTCP, OSTα and MDR2) and synthesis (CYP7A1 and CYP8B1). CLAV also caused early oxidative stress, with reduced GSH/GSSG ratio, along with induction of antioxidant nuclear factor erythroid 2-related factor 2 (NRF2) target genes. Activation of NRF2 by sulforaphane also resulted in downregulation of NTCP, OSTα, ABCG5, CYP7A1 and CYP8B1. CLAV also inhibited the BA-sensor farnesoid X receptor (FXR), in agreement with the downregulation of FXR targets BSEP, OSTα and ABCG5. We conclude that CLAV, the culprit molecule in A+C, downregulates several key biliary transporters by modulating NRF2 and FXR signaling, thus likely promoting intrahepatic cholestasis. On top of that, increased ROS production and GSH depletion may aggravate the cholestatic injury by A+C.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Despite many ongoing and innovative approaches, there are still formidable challenges in the clinical translation of oral peptide drugs into marketable products due to their low absorption and poor bioavailability. Herein, a novel nanocarrier platform was developed that employs a hydrophobic ion-pairing (HIP) of model peptide (insulin) and the anionic bile salt (sodium glycodeoxycholate, SGDC), and markedly improves intestinal absorption via the bile acid pathway. The developed HIP-nanocomplexes (C1 and C2) were optimized, characterized, and in vitro and in vivo evaluation were performed to assess oral efficacy of these system. The optimal molar ratios of C1 and C2-nanocomplexes were 30:1 and 6:1 (SGDC:insulin), respectively. Compared to the insulin solution, the C1 and C2 nanocomplexes significantly enhanced the permeation of insulin across the Caco-2 cell monolayers, with 6.36- and 4.05-fold increases in apparent permeability, respectively. Uptake mechanism studies were conducted using different endocytosis inhibitors and apical sodium-dependent bile acid transporter (ASBT)-transfected MDCK cells, which demonstrated the involvement of the energy-dependent ASBT-mediated active transport. Furthermore, the intrajejunal administration of C1 and C2 resulted in their pharmacological availabilities (PA) being 6.44% and 0.10%, respectively, indicating increased potential for C1, when compared to C2. Similarly, the PA and the relative bioavailability with intrajejunal administration of the C1 were 17.89-fold and 16.82-fold greater than those with intracolonic administration, respectively, confirming better jejunal absorption of C1. Overall, these findings indicate that the HIP-nanocomplexes could be a prominent platform for the effective delivery of peptides with improved intestinal absorption.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号