Antiporters

反派者
  • 文章类型: Journal Article
    阴离子交换剂3(AE3)在调节整个兴奋组织的细胞内pH中起着关键作用,然而,与其他阴离子交换剂相比,其结构复杂性和功能动力学仍未得到充分探索。这项研究揭示了人类AE3的结构见解,包括AE3跨膜结构域(TMD)的低温电子显微镜结构以及将AE3N末端结构域(NTD)与AE2TMD(hAE3NTD2TMD)结合的嵌合体。我们的分析揭示了一个底物结合位点,NTD-TMD联锁机构,以及对面向外的构象的偏好。与AE2不同,AE2具有更强大的酸负载能力,AE3的结构,包括由于缺少关键的NTD-TMD相互作用而导致的不太稳定的面向内的构象,有助于其适度的pH调节活性和增加对抑制剂DIDS的敏感性。这些结构差异强调了AE3在特定组织中的独特功能作用,并强调了阴离子交换家族中结构动力学和功能特异性之间的复杂相互作用。增强我们对阴离子交换家族的生理和病理作用的理解。
    Anion exchanger 3 (AE3) is pivotal in regulating intracellular pH across excitable tissues, yet its structural intricacies and functional dynamics remain underexplored compared to other anion exchangers. This study unveils the structural insights into human AE3, including the cryo-electron microscopy structures for AE3 transmembrane domains (TMD) and a chimera combining AE3 N-terminal domain (NTD) with AE2 TMD (hAE3NTD2TMD). Our analyzes reveal a substrate binding site, an NTD-TMD interlock mechanism, and a preference for an outward-facing conformation. Unlike AE2, which has more robust acid-loading capabilities, AE3\'s structure, including a less stable inward-facing conformation due to missing key NTD-TMD interactions, contributes to its moderated pH-modulating activity and increased sensitivity to the inhibitor DIDS. These structural differences underline AE3\'s distinct functional roles in specific tissues and underscore the complex interplay between structural dynamics and functional specificity within the anion exchanger family, enhancing our understanding of the physiological and pathological roles of the anion exchanger family.
    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
    肠上皮被粘液覆盖,保护组织免受腔内容物的影响。研究表明,通过囊性纤维化电导调节因子(Cftr)分泌的阴离子调节小肠中的粘液形成。然而,调节结肠粘液形成的机制知之甚少。这项研究的目的是探索阴离子转运在稳态期间以及对氨甲酰基胆碱(CCh)和前列腺素E2(PGE2)的反应中对粘液形成的调节中的作用。广谱阴离子转运抑制剂4,4'-二异硫氰基二苯乙烯-2,2'-二磺酸盐(DIDS),CftrdF508(CF)小鼠,和slc26a3抑制剂SLC26A3-IN-2用于抑制阴离子转运。在远端结肠,SLC26A3-IN-2减少了CF小鼠的稳态粘液扩张和正常。PGE2通过slc26a3敏感机制在野生型(WT)和CF结肠中刺激粘液扩张而没有从头粘液释放,而CCh在WT中诱导从头粘液分泌,而在CF结肠中不诱导。然而,当同时添加时,CCh和PGE2通过DIDS敏感途径刺激CF结肠中的从头粘液分泌。在CF回肠中观察到类似的反应,对CCh和PGE2有DIDS敏感的从头粘液分泌反应。总之,这项研究表明,slc26a3调节结肠粘液扩张,而Cftr调节回肠和远端结肠隐窝的CCh诱导的从头粘液分泌。此外,这些发现表明,在没有功能性Cftr通道的情况下,CCh和PGE2的平行刺激激活了其他阴离子转运过程,有助于从肠杯状细胞释放粘液。
    The intestinal epithelium is covered by mucus that protects the tissue from the luminal content. Studies have shown that anion secretion via the cystic fibrosis conductance regulator (Cftr) regulates mucus formation in the small intestine. However, mechanisms regulating mucus formation in the colon are less understood. The aim of this study was to explore the role of anion transport in the regulation of mucus formation during steady state and in response to carbamylcholine (CCh) and prostaglandin E2 (PGE2). The broad-spectrum anion transport inhibitor 4,4\'-diisothiocyanatostilbene-2,2\'-disulfonate (DIDS), CftrdF508 (CF) mice, and the slc26a3 inhibitor SLC26A3-IN-2 were used to inhibit anion transport. In the distal colon, steady-state mucus expansion was reduced by SLC26A3-IN-2 and normal in CF mice. PGE2 stimulated mucus expansion without de novo mucus release in wild type (WT) and CF colon via slc26a3 sensitive mechanisms, while CCh induced de novo mucus secretion in WT but not in CF colon. However, when added simultaneously, CCh and PGE2 stimulated de novo mucus secretion in the CF colon via DIDS-sensitive pathways. A similar response was observed in CF ileum that responded to CCh and PGE2 with DIDS-sensitive de novo mucus secretion. In conclusion, this study suggests that slc26a3 regulates colonic mucus expansion, while Cftr regulates CCh-induced de novo mucus secretion from ileal and distal colon crypts. Furthermore, these findings demonstrate that in the absence of a functional Cftr channel, parallel stimulation with CCh and PGE2 activates additional anion transport processes that help release mucus from intestinal goblet cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    背景:在这项研究中,我们使用靶向下一代测序(NGS)研究了一名19岁突尼斯男性患有严重甲状腺功能减退症和甲状腺肿的先天性甲状腺功能减退症(CH)的遗传基础.
    方法:患者18岁时出现甲状腺肿。重要的是,他没有智力低下的迹象,他的成长是相称的。通过高氯酸盐诱导的碘化物放电测试检测到部分有机化缺陷。NGS在SLC26A7基因(P628Qfs*11)的第18外显子中发现了一个新的纯合突变,编码一种新的碘化物转运蛋白。预测该变体产生截短的蛋白质。值得注意的是,患者的甲状腺功能正常的兄弟是相同突变的杂合子。未发现肾脏酸碱异常,服用1mg碘未能纠正甲状腺功能减退。
    结论:我们描述了由于在青春期后期诊断的SLC26A7基因纯合突变导致的第一例甲状腺肿性CH。
    BACKGROUND: In this study, we used targeted next-generation sequencing (NGS) to investigate the genetic basis of congenital hypothyroidism (CH) in a 19-year-old Tunisian man who presented with severe hypothyroidism and goiter.
    METHODS: The propositus reported the appearance of goiter when he was 18. Importantly, he did not show signs of mental retardation, and his growth was proportionate. A partial organification defect was detected through the perchlorate-induced iodide discharge test. NGS identified a novel homozygous mutation in exon 18 of the SLC26A7 gene (P628Qfs*11), which encodes for a new iodide transporter. This variant is predicted to result in a truncated protein. Notably, the patient\'s euthyroid brother was heterozygous for the same mutation. No renal acid-base abnormalities were found and the administration of 1 mg of iodine failed to correct hypothyroidism.
    CONCLUSIONS: We described the first case of goitrous CH due to a homozygous mutation of the SLC26A7 gene diagnosed during late adolescence.
    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
    来自大肠杆菌的多药外排转运蛋白EmrE需要底物结合袋中的阴离子残基,以将药物转运与质子动力偶联。这里,我们展示了EmrE同二聚体中单个膜包埋的谷氨酸残基(Glu14)的质子化如何使用NMR光谱以变构方式调节结构和动力学。处于Glu14质子化状态的EmrE的结构显示出部分封闭的构象,该构象由于在结合袋中存在芳香族残基而无法进行药物结合。一个单体中单个Glu14残基的去质子化通过改变其侧链位置和附近色氨酸残基的位置来诱导向开放状态的平衡移动。这种结构变化促进开放构象,其通过构象选择机制促进药物结合,并使结合亲和力增加约2000倍。质子耦合交换在外排系统中的普遍性表明了一种可能在其他反转运蛋白中共享的机制,其中酸/碱化学调节药物进入底物结合袋。
    The multidrug efflux transporter EmrE from Escherichia coli requires anionic residues in the substrate binding pocket for coupling drug transport with the proton motive force. Here, we show how protonation of a single membrane embedded glutamate residue (Glu14) within the homodimer of EmrE modulates the structure and dynamics in an allosteric manner using NMR spectroscopy. The structure of EmrE in the Glu14 protonated state displays a partially occluded conformation that is inaccessible for drug binding by the presence of aromatic residues in the binding pocket. Deprotonation of a single Glu14 residue in one monomer induces an equilibrium shift toward the open state by altering its side chain position and that of a nearby tryptophan residue. This structural change promotes an open conformation that facilitates drug binding through a conformational selection mechanism and increases the binding affinity by approximately 2000-fold. The prevalence of proton-coupled exchange in efflux systems suggests a mechanism that may be shared in other antiporters where acid/base chemistry modulates access of drugs to the substrate binding pocket.
    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
    阴离子交换剂SLC26A9,由跨膜(TM)结构域和胞质STAS结构域组成,在调节氯化物跨细胞膜的运输中起着至关重要的作用。最近的研究表明,C末端螺旋阻断了假定的离子转运途径的入口。然而,STAS结构域和C端螺旋的精确功能,以及控制运输过程的潜在分子机制,仍然知之甚少。在这项研究中,我们对人类SLC26A9的三种不同模型进行了分子动力学模拟:全长(FL),STAS域移除(ΔSTAS),和C末端去除(ΔC),研究其构象动力学和离子结合特性。在这些模拟中,在ΔC模型中仅观察到离子与结合位点的稳定结合。比较FL和ΔC模拟,ΔC模型显示了STAS域的增强运动。此外,比较ΔSTAS和ΔC模拟,尽管两个系统中都没有C末端阻断离子传输途径,但ΔSTAS模拟未能显示出稳定的离子结合位点。这些结果表明,C末端的去除不仅可以阻断离子进入渗透途径,还可以触发STAS结构域运动。门控TM域以促进离子进入其结合位点。进一步的分析表明,STAS域的非对称运动导致TM域内离子渗透途径的扩展,导致离子结合位点附近的柔性TM12螺旋变硬。TM12螺旋的这种结构变化稳定了氯离子的结合,这对于SLC26A9的备用访问机制至关重要。总的来说,我们的研究为SLC26A9转运的分子机制提供了新的见解,并可能为开发与离子转运失调相关的疾病的新疗法铺平道路。
    The anion exchanger solute carrier family 26 (SLC26)A9, consisting of the transmembrane (TM) domain and the cytoplasmic STAS domain, plays an essential role in regulating chloride transport across cell membranes. Recent studies have indicated that C-terminal helices block the entrance of the putative ion transport pathway. However, the precise functions of the STAS domain and C-terminal helix, as well as the underlying molecular mechanisms governing the transport process, remain poorly understood. In this study, we performed molecular dynamics simulations of three distinct models of human SLC26A9, full-length, STAS domain removal (ΔSTAS), and C-terminus removal (ΔC), to investigate their conformational dynamics and ion-binding properties. Stable binding of ions to the binding sites was exclusively observed in the ΔC model in these simulations. Comparing the full-length and ΔC simulations, the ΔC model displayed enhanced motion of the STAS domain. Furthermore, comparing the ΔSTAS and ΔC simulations, the ΔSTAS simulation failed to exhibit stable ion bindings to the sites despite the absence of the C-terminus blocking the ion transmission pathway in both systems. These results suggest that the removal of the C-terminus not only unblocks the access of ions to the permeation pathway but also triggers STAS domain motion, gating the TM domain to promote ions\' entry into their binding site. Further analysis revealed that the asymmetric motion of the STAS domain leads to the expansion of the ion permeation pathway within the TM domain, resulting in the stiffening of the flexible TM12 helix near the ion-binding site. This structural change in the TM12 helix stabilizes chloride ion binding, which is essential for SLC26A9\'s alternate-access mechanism. Overall, our study provides new insights into the molecular mechanisms of SLC26A9 transport and may pave the way for the development of novel treatments for diseases associated with dysregulated ion transport.
    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
    背景:Triptans是抗偏头痛药物,具有潜在的中枢作用位点。然而,目前尚不清楚曲坦通过血脑屏障(BBB)的程度。因此,这项研究的目的是确定曲坦类药物是否通过脑毛细血管内皮,并研究可能的潜在机制,重点是推定的质子偶联有机阳离子(H/OC)反转运蛋白的参与。此外,我们评估了曲坦是否与外排转运蛋白相互作用,P-糖蛋白(P-gp)。
    方法:我们研究了原型H+/OC反转运底物的细胞摄取特征,吡喃胺和羟考酮,和人脑微血管内皮细胞系中的七种不同的曲坦类药物,hCMEC/D3。使用IPEC-J2MDR1细胞系研究了曲坦与P-gp的相互作用。最后,在野生型和mdr1a/1b基因敲除小鼠中进行了eletriptan未结合脑-血浆配置的体内神经药代动力学评估.
    结果:我们证明大多数曲坦类药物能够抑制H+/OC反转运底物的摄取,吡喃胺,其中eletriptan成为最强的抑制剂。Eletriptan,阿莫曲坦,和舒马曲坦表现出pH依赖性摄取到hCMEC/D3细胞中。Eletriptan表现出可饱和的摄取动力学,表观Km为89±38µM,Jmax为2.2±0.7nmol·min-1·mg蛋白-1(n=3)。跨IPEC-J2MDR1单层的双向运输实验表明,eletriptan通过P-gp运输,因此表明eletriptan是H/OC反转运蛋白和P-gp的底物。这在体内得到进一步证实,其中未结合的脑与未结合的血浆浓度比(Kp,uu)在野生型小鼠中为0.04,而mdr1a/1b敲除小鼠的比率上升至1.32。
    结论:我们已经证明了化合物的曲坦家族对H+/OC反转运蛋白具有亲和力,表明推定的H+/OC反转运蛋白在曲坦的BBB转运中起作用。尤其是依来曲坦.我们的体内研究表明,eletriptan同时进行脑摄取和外排,可能是由推定的H+/OC反转录因子和P-GP促进的,分别。我们的发现为曲坦类药物治疗偏头痛的潜在中枢作用位点提供了新的见解,并强调了潜在转运蛋白相关药物-药物相互作用的重要性。
    BACKGROUND: Triptans are anti-migraine drugs with a potential central site of action. However, it is not known to what extent triptans cross the blood-brain barrier (BBB). The aim of this study was therefore to determine if triptans pass the brain capillary endothelium and investigate the possible underlying mechanisms with focus on the involvement of the putative proton-coupled organic cation (H+/OC) antiporter. Additionally, we evaluated whether triptans interacted with the efflux transporter, P-glycoprotein (P-gp).
    METHODS: We investigated the cellular uptake characteristics of the prototypical H+/OC antiporter substrates, pyrilamine and oxycodone, and seven different triptans in the human brain microvascular endothelial cell line, hCMEC/D3. Triptan interactions with P-gp were studied using the IPEC-J2 MDR1 cell line. Lastly, in vivo neuropharmacokinetic assessment of the unbound brain-to-plasma disposition of eletriptan was conducted in wild type and mdr1a/1b knockout mice.
    RESULTS: We demonstrated that most triptans were able to inhibit uptake of the H+/OC antiporter substrate, pyrilamine, with eletriptan emerging as the strongest inhibitor. Eletriptan, almotriptan, and sumatriptan exhibited a pH-dependent uptake into hCMEC/D3 cells. Eletriptan demonstrated saturable uptake kinetics with an apparent Km of 89 ± 38 µM and a Jmax of 2.2 ± 0.7 nmol·min-1·mg protein-1 (n = 3). Bidirectional transport experiments across IPEC-J2 MDR1 monolayers showed that eletriptan is transported by P-gp, thus indicating that eletriptan is both a substrate of the H+/OC antiporter and P-gp. This was further confirmed in vivo, where the unbound brain-to-unbound plasma concentration ratio (Kp,uu) was 0.04 in wild type mice while the ratio rose to 1.32 in mdr1a/1b knockout mice.
    CONCLUSIONS: We have demonstrated that the triptan family of compounds possesses affinity for the H+/OC antiporter proposing that the putative H+/OC antiporter plays a role in the BBB transport of triptans, particularly eletriptan. Our in vivo studies indicate that eletriptan is subjected to simultaneous brain uptake and efflux, possibly facilitated by the putative H+/OC antiporter and P-gp, respectively. Our findings offer novel insights into the potential central site of action involved in migraine treatment with triptans and highlight the significance of potential transporter related drug-drug interactions.
    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
    糖原贮积病Ib型(GSD-Ib)是由SLC37A4突变引起的一种罕见的先天性糖原代谢错误。患有GSD-Ib的患者具有发生炎症性肠病(IBD)的高风险。我们评估了依帕列净的疗效,肾钠葡萄糖协同转运蛋白2(SGLT2)抑制剂,GSD相关性IBD患者结肠黏膜愈合的研究.一个潜在的,单臂,开放标签临床试验纳入了2022年7月1日至2023年12月31日中国广东省人民医院8例GSD相关IBD患者.招募了8名患者,平均年龄为10.34±2.61岁。四男四女。内镜特征包括深圆形和大圆形溃疡,炎性增生,阻塞和狭窄。与依帕列净之前相比,在第48周SES-CD评分显着降低。6例患者完成了48周的依帕列净治疗和内窥镜检查显示粘膜溃疡的显著改善或愈合,炎性增生,狭窄,和阻塞。一名患者出汗严重,需要补液,并出现尿路感染。无严重或危及生命的不良事件。本研究提示依帕列净可能促进结肠黏膜愈合,减少增生。狭窄,与GSD相关的IBD患儿的梗阻。
    Glycogen storage disease type Ib (GSD-Ib) is a rare inborn error of glycogen metabolism caused by mutations in SLC37A4. Patients with GSD-Ib are at high risk of developing inflammatory bowel disease (IBD). We evaluated the efficacy of empagliflozin, a renal sodium‒glucose cotransporter protein 2 (SGLT2) inhibitor, on colonic mucosal healing in patients with GSD-associated IBD. A prospective, single-arm, open-label clinical trial enrolled eight patients with GSD-associated IBD from Guangdong Provincial People\'s Hospital in China from July 1, 2022 through December 31, 2023. Eight patients were enrolled with a mean age of 10.34 ± 2.61 years. Four male and four female. The endoscopic features included deep and large circular ulcers, inflammatory hyperplasia, obstruction and stenosis. The SES-CD score significantly decreased at week 48 compared with before empagliflozin. Six patients completed 48 weeks of empagliflozin therapy and endoscopy showed significant improvement or healing of mucosal ulcers, inflammatory hyperplasia, stenosis, and obstruction. One patient had severe sweating that required rehydration and developed a urinary tract infection. No serious or life-threatening adverse events. This study suggested that empagliflozin may promote colonic mucosal healing and reduce hyperplasia, stenosis, and obstruction in children with GSD-associated IBD.
    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
    产气荚膜梭菌是一类广泛存在于人和动物肠道组织中的厌氧革兰氏阳性菌。产气荚膜梭菌的主要毒力因子是其外毒素。产气荚膜梭菌C型是家畜疾病的主要毒株,其外毒素可引起坏死性肠炎和肠毒血症,导致饲料转化率降低,严重影响育种生产性能。我们的研究发现,用外毒素治疗会降低人单核细胞白血病细胞(THP-1)细胞的细胞活力并触发细胞内活性氧(ROS)。通过转录组测序分析,我们发现,用外毒素治疗后,血红素加氧酶1(HO-1)和铁凋亡信号通路等相关蛋白的水平显著升高.为了研究巨噬细胞外毒素处理后是否发生铁凋亡,我们证实了抗氧化因子谷胱甘肽过氧化物酶4/铁凋亡抑制蛋白1/胱氨酸/谷氨酸反转运溶质载体家族7成员11(GPX4/FSP1/xCT)的蛋白表达水平,铁凋亡相关蛋白核受体共激活因子4/转铁蛋白/转铁蛋白受体(NCOA4/TF/TFR)/铁蛋白和脂质过氧化水平均有显著改变。基于以上结果,我们的研究表明,产气荚膜梭菌C型外毒素可通过氧化应激和铁凋亡诱导巨噬细胞损伤。
    Clostridium perfringens is a kind of anaerobic Gram-positive bacterium that widely exists in the intestinal tissue of humans and animals. And the main virulence factor in Clostridium perfringens is its exotoxins. Clostridium perfringens type C is the main strain of livestock disease, its exotoxins can induce necrotizing enteritis and enterotoxemia, which lead to the reduction in feed conversion, and a serious impact on breeding production performance. Our study found that treatment with exotoxins reduced cell viability and triggered intracellular reactive oxygen species (ROS) in human mononuclear leukemia cells (THP-1) cells. Through transcriptome sequencing analysis, we found that the levels of related proteins such as heme oxygenase 1 (HO-1) and ferroptosis signaling pathway increased significantly after treatment with exotoxins. To investigate whether ferroptosis occurred after exotoxin treatment in macrophages, we confirmed that the protein expression levels of antioxidant factors glutathione peroxidase 4/ferroptosis-suppressor-protein 1/the cystine/glutamate antiporter solute carrier family 7 member 11 (GPX4/FSP1/xCT), ferroptosis-related protein nuclear receptor coactivator 4/transferrin/transferrin receptor (NCOA4/TF/TFR)/ferritin and the level of lipid peroxidation were significantly changed. Based on the above results, our study suggested that Clostridium perfringens type C exotoxins can induce macrophage injury through oxidative stress and ferroptosis.
    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
    背景:糖原贮积病Ib型(GSDIb)是一种罕见的疾病,其特征是由SLC37A4基因突变引起的葡萄糖稳态受损。它是一种与低血糖相关的严重遗传性代谢疾病,高脂血症,乳酸性酸中毒,肝肿大,和中性粒细胞减少症.传统治疗包括饲喂生玉米淀粉,这可以帮助调节能量代谢,但对中性粒细胞减少症没有积极作用,这对这些患者来说是致命的。最近,已经发现GSDIb中性粒细胞功能障碍和中性粒细胞减少的病理生理机制,SGLT2抑制剂empaglifozin的治疗现在已经确立。2020年,SGLT2抑制剂empagliflozin开始在全球GSDIb患者的中性粒细胞中用作有前途的1,5AG6P的有效去除剂。然而,有必要考虑一种新型治疗方法的长期效用和安全性。
    结果:在这项研究中,我们回顾性地检查了临床表现,生化检查结果,基因型,自2009年以来在我们部门就诊的35名GSDIb儿童的长期结局和随访。自2020年以来,其中14名患者接受了empagliflozin治疗。这项研究是中国最大的儿童GSDIb患者队列,也是迄今为止在单个中心接受依帕列净治疗的最大的儿童GSDIb患者队列。该研究还讨论了小儿GSDIb患者的长期管理经验。
    结论:Empagliflozin治疗小儿GSDIb患者是有效和安全的。尿糖的增加是药效的信号,然而,建议注意尿路感染和低血糖。
    BACKGROUND: Glycogen storage disease type Ib (GSD Ib) is a rare disorder characterized by impaired glucose homeostasis caused by mutations in the SLC37A4 gene. It is a severe inherited metabolic disease associated with hypoglycemia, hyperlipidemia, lactic acidosis, hepatomegaly, and neutropenia. Traditional treatment consists of feeding raw cornstarch which can help to adjust energy metabolism but has no positive effect on neutropenia, which is fatal for these patients. Recently, the pathophysiologic mechanism of the neutrophil dysfunction and neutropenia in GSD Ib has been found, and the treatment with the SGLT2 inhibitor empaglifozin is now well established. In 2020, SGLT2 inhibitor empagliflozin started to be used as a promising efficient remover of 1,5AG6P in neutrophil of GSD Ib patients worldwide. However, it is necessary to consider long-term utility and safety of a novel treatment.
    RESULTS: In this study, we retrospectively examined the clinical manifestations, biochemical examination results, genotypes, long-term outcomes and follow-up of thirty-five GSD Ib children who visited our department since 2009. Fourteen patients among them underwent empagliflozin treatment since 2020. This study is the largest cohort of pediatric GSD Ib patients in China as well as the largest cohort of pediatric GSD Ib patients treated with empagliflozin in a single center to date. The study also discussed the experience of long-term management on pediatric GSD Ib patients.
    CONCLUSIONS: Empagliflozin treatment for pediatric GSD Ib patients is efficient and safe. Increase of urine glucose is a signal for pharmaceutical effect, however attention to urinary infection and hypoglycemia is suggested.
    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
    次级活性转运蛋白催化无数物质在所有细胞膜上的运动,通常针对相反的浓度梯度,并且不消耗任何ATP。要做到这一点,这些蛋白质采用一种有趣的结构机制,进化为只有在识别或释放运输物种时才被激活。我们使用心脏Na/Ca2交换剂的同系物作为模型系统来检查这种自我调节的机制。使用先进的计算机模拟,我们绘制出这个运输机的完整功能周期,包括我们根据现有实验数据验证的未知构象。计算出的自由能景观揭示了为什么这种运输机充当反运输机而不是符号运输机,为什么它专门交换Na+和Ca2+,以及为什么这种交换的化学计量是3:1。我们还合理化了为什么蛋白质不会将H交换为Ca2或Na,尽管能够结合H+及其与H+/Ca2+交换剂的高度相似性。有趣的是,这种转运蛋白的性质不能用它的主要结构状态来解释,被称为向内和向外开放的构象;相反,决定性因素是这些状态之间构象中间体的可行性,其中通向底物结合位点的通路同时从膜的两侧闭塞。这种分析提供了物理上连贯的,广泛转移的途径,以了解二级活性膜转运蛋白从结构中功能的出现。
    Secondary-active transporters catalyze the movement of myriad substances across all cellular membranes, typically against opposing concentration gradients, and without consuming any ATP. To do so, these proteins employ an intriguing structural mechanism evolved to be activated only upon recognition or release of the transported species. We examine this self-regulated mechanism using a homolog of the cardiac Na+/Ca2+ exchanger as a model system. Using advanced computer simulations, we map out the complete functional cycle of this transporter, including unknown conformations that we validate against existing experimental data. Calculated free-energy landscapes reveal why this transporter functions as an antiporter rather than a symporter, why it specifically exchanges Na+ and Ca2+, and why the stoichiometry of this exchange is exactly 3:1. We also rationalize why the protein does not exchange H+ for either Ca2+ or Na+, despite being able to bind H+ and its high similarity with H+/Ca2+ exchangers. Interestingly, the nature of this transporter is not explained by its primary structural states, known as inward- and outward-open conformations; instead, the defining factor is the feasibility of conformational intermediates between those states, wherein access pathways leading to the substrate binding sites become simultaneously occluded from both sides of the membrane. This analysis offers a physically coherent, broadly transferable route to understand the emergence of function from structure among secondary-active membrane transporters.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号