CoQ10

辅酶 Q10
  • 文章类型: Journal Article
    补充辅酶Q10(CoQ10)似乎与较低的血压有关。然而,目前尚不清楚食物来源的CoQ10是否会影响一般成人的新发高血压.这项研究调查了一般人群中膳食辅酶Q10摄入量与新发高血压之间的关系。纳入了中国健康与营养调查(CHNS)前瞻性队列研究中基线无高血压的参与者(n=11,428)。通过经过验证的饮食召回和食物称重方法收集饮食中的辅酶Q10摄入量。使用多变量Cox比例风险模型和有限的三次样条分析了饮食中辅酶Q10摄入量与新发高血压之间的线性和非线性关系。在随访期间(中位数:6年),记录了4006例新发高血压病例。与非消费者相比,风险比(HR)和95%置信区间(CI)从五分之一2到4总膳食CoQ10为0.83(0.76,0.91),0.86(0.78,0.94)和1.01(0.92,1.11);总植物源性辅酶Q10为0.80(0.73,0.88),1.00(0.91,1.09)和1.10(1.00,1.20);动物源性辅酶Q10为0.65(0.59,0.71),0.58(0.53,0.64)和0.68(0.62,0.75)。在适度摄入时风险最低,呈非线性关系(P非线性<0.05)。此外,在不饮酒或低脂饮食的个体中,总体负相关更强.适度的长期饮食摄入辅酶Q10可能对新发高血压具有保护作用。然而,呈非线性关系,过量摄入可能会增加中国人群新发高血压的风险.
    Coenzyme Q10 (CoQ10) supplementation appears to be associated with a lower blood pressure. Nevertheless, it remains unclear whether food-sourced CoQ10 will affect new-onset hypertension in general adults. This study investigated the relationship between dietary CoQ10 intake and new-onset hypertension among the general population. Participants without hypertension at baseline from the China Health and Nutrition Survey (CHNS) prospective cohort study were included (n = 11,428). Dietary CoQ10 intake was collected by validated dietary recalls and the food weighing method. Linear and non-linear relationships between dietary CoQ10 intake and new-onset hypertension were analyzed using multivariable Cox proportional hazards models and restricted cubic splines. During follow-up (median: 6 years), 4006 new-onset hypertension cases were documented. Compared with non-consumers, the hazard ratio (HR) and 95% confidence interval (CI) from quintile 2 to 4 total dietary CoQ10 were 0.83 (0.76, 0.91), 0.86 (0.78, 0.94) and 1.01 (0.92, 1.11); total plant-derived CoQ10 were 0.80 (0.73, 0.88), 1.00 (0.91, 1.09) and 1.10 (1.00, 1.20); and animal-derived CoQ10 were 0.65 (0.59, 0.71), 0.58 (0.53, 0.64) and 0.68 (0.62, 0.75). The lowest risk was found at moderate intake, with a non-linear relationship (P nonlinearity < 0.05). Furthermore, the overall inverse association was stronger among individuals without alcohol consumption or eating a low-fat diet. Moderate long-term dietary CoQ10 intake might be protective against new-onset hypertension. However, it follows a non-linear relationship and excessive intake may increase the risk of new-onset hypertension in the Chinese population.
    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
    七叶皂苷钠(SA),在马栗种子中发现的一种活性化合物,广泛应用于临床。最近,SA引起的不良事件的发生率,特别是肾功能损害,增加了。我们以前的工作表明,SA通过肾细胞铁性凋亡引起严重的肾毒性;然而,潜在的机制仍有待充分阐明。在目前的研究中,我们研究了与SA诱导的肾毒性有关的其他分子途径.我们的结果表明,SA抑制细胞活力,破坏细胞膜的完整性,和增强的活性氧(ROS),亚铁(Fe2+),丙二醛(MDA)水平,以及大鼠近端肾小管上皮细胞系(NRK-52E)细胞的脂质过氧化。SA还耗尽了辅酶Q10(CoQ10,泛醌)和烟酰胺腺嘌呤二核苷酸(NADH)以及降低的铁凋亡抑制蛋白1(FSP1)和聚异戊二烯基转移酶(辅酶Q2,COQ2)活性,在小鼠肾脏和NRK-52E细胞中触发脂质过氧化和ROS积累。补充COQ2,FSP1或CoQ10(泛醌)的过表达有效地减弱了SA诱导的铁细胞凋亡,而iFSP1或4-甲酰苯甲酸(4-CBA)预处理会加剧SA诱导的肾毒性。此外,SA降低了核因子-红系-2相关因子2(Nrf2)水平,并抑制了Nrf2与FSP1启动子中-1170/-1180bpARE位点的结合,导致FSP1抑制。过表达Nrf2或其激动剂富马酸二甲酯(DMF)促进FSP1表达,从而提高细胞抗氧化能力并减轻SA诱导的铁凋亡。这些结果表明,SA通过氧化应激和铁凋亡引发肾损伤,由Nrf2/FSP1/CoQ10轴的抑制驱动。
    Sodium aescinate (SA), an active compound found in horse chestnut seeds, is widely used in clinical practice. Recently, the incidence of SA-induced adverse events, particularly renal impairment, has increased. Our previous work demonstrated that SA causes severe nephrotoxicity via nephrocyte ferroptosis; however, the underlying mechanism remains to be fully elucidated. In the current study, we investigated additional molecular pathways involved in SA-induced nephrotoxicity. Our results showed that SA inhibited cell viability, disrupted cellular membrane integrity, and enhanced reactive oxygen species (ROS), ferrous iron (Fe2+), and malondialdehyde (MDA) levels, as well as lipid peroxidation in rat proximal renal tubular epithelial cell line (NRK-52E) cells. SA also depleted coenzyme Q10 (CoQ10, ubiquinone) and nicotinamide adenine dinucleotide (NADH) and reduced ferroptosis suppressor protein 1 (FSP1) and polyprenyltransferase (coenzyme Q2, COQ2) activity, triggering lipid peroxidation and ROS accumulation in mouse kidneys and NRK-52E cells. The overexpression of COQ2, FSP1, or CoQ10 (ubiquinone) supplementation effectively attenuated SA-induced ferroptosis, whereas iFSP1 or 4-formylbenzoic acid (4-CBA) pretreatment exacerbated SA-induced nephrotoxicity. Additionally, SA decreased nuclear factor-erythroid-2-related factor 2 (Nrf2) levels and inhibited Nrf2 binding to the -1170/-1180 bp ARE site in FSP1 promoter, resulting in FSP1 suppression. Overexpression of Nrf2 or its agonist dimethyl fumarate (DMF) promoted FSP1 expression, thereby improving cellular antioxidant capacity and alleviating SA-induced ferroptosis. These results suggest that SA-triggers renal injury through oxidative stress and ferroptosis, driven by the suppression of the Nrf2/FSP1/CoQ10 axis.
    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
    卵巢老化是女性不孕的主要因素。多种抗氧化剂已应用于不同的临床场景,但它们对卵巢老化女性生育能力的影响尚不清楚。为了解决这个问题,本研究进行了一项荟萃分析,以评估抗氧化剂对卵巢老化女性生育的有效性和安全性.共纳入20项随机临床试验(RCT),有2617名参与者。结果表明,使用抗氧化剂不仅显着增加了回收的卵母细胞数量和高质量的胚胎率,而且减少了促性腺激素的剂量,有助于提高临床妊娠率。根据亚组分析,关于不同的剂量设置,较低剂量的效果更明显;就抗氧化剂类型而言,辅酶Q10(CoQ10)往往比褪黑激素更有效,肌醇和维生素,因为与安慰剂或不治疗相比,辅酶Q10表现出更多的优势,而在其他药物中观察到小的改善。此外,根据CoQ10的亚组分析,CoQ10改善妊娠率的最佳治疗方案是在控制性促排卵周期前三个月30mg/d,卵巢储备减少的女性明显受益于辅酶Q10治疗,尤其是35岁以下的人。我们的研究表明,抗氧化剂是卵巢衰老女性的有效和安全的补充疗法。应根据患者年龄和卵巢储备,从低剂量提供适当的抗氧化治疗。本研究在PROSPERO注册(CRD42022359529)。
    Ovarian aging is a major factor for female subfertility. Multiple antioxidants have been applied in different clinical scenarios, but their effects on fertility in women with ovarian aging are still unclear. To address this, a meta-analysis was performed to evaluate the effectiveness and safety of antioxidants on fertility in women with ovarian aging. A total of 20 randomized clinical trials with 2617 participants were included. The results showed that use of antioxidants not only significantly increased the number of retrieved oocytes and high-quality embryo rates but also reduced the dose of gonadotropin, contributing to higher clinical pregnancy rates. According to the subgroup analysis of different dose settings, better effects were more pronounced with lower doses; in terms of antioxidant types, coenzyme Q10 (CoQ10) tended to be more effective than melatonin, myo-inositol, and vitamins. When compared with placebo or no treatment, CoQ10 showed more advantages, whereas small improvements were observed with other drugs. In addition, based on subgroup analysis of CoQ10, the optimal treatment regimen of CoQ10 for improving pregnancy rate was 30 mg/d for 3 mo before the controlled ovarian stimulation cycle, and women with diminished ovarian reserve clearly benefited from CoQ10 treatment, especially those aged <35 y. Our study suggests that antioxidant consumption is an effective and safe complementary therapy for women with ovarian aging. Appropriate antioxidant treatment should be offered at a low dose according to the patient\'s age and ovarian reserve. This study was registered at PROSPERO as CRD42022359529.
    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
    COQ8A在辅酶Q10(CoQ10)的生物合成中起重要作用,COQ8A基因的变异与原发性CoQ10缺乏症-4(COQ10D4)有关,也称为COQ8A-共济失调。当前对特定变体类型之间关联的理解,辅酶Q10缺乏的严重程度,原发性辅酶Q10缺乏个体的氧化应激程度仍不确定。在这里,我们提供了一个18岁的COQ8A-共济失调患者的临床和遗传特征的综合分析,谁在COQ8A基因中表现出新的复合杂合变体(c.1904_1906del和c.637C>T)。这些变体降低了患者肌肉和皮肤成纤维细胞样品中COQ8A和线粒体蛋白的表达水平,导致线粒体呼吸不足,增加ROS产生和改变线粒体膜电位。值得注意的是,COQ8A-共济失调的最佳治疗方法仍不确定。目前,治疗包括补充辅酶Q10,然而,我们的患者症状并未得到显著改善。此外,我们详细回顾了以往文献中补充辅酶Q10的反应和患者的演变。我们发现,只有一半的患者可以在共济失调方面得到显着改善。本研究旨在扩大COQ10D4的基因型-表型谱,解决以前关于CoQ10在这些疾病中的有效性的评论中的差异,并有助于建立COQ8A-共济失调的标准化治疗方案。
    COQ8A plays an important role in the biosynthesis of coenzyme Q10 (CoQ10), and variations in COQ8A gene are associated with primary CoQ10 deficiency-4 (COQ10D4), also known as COQ8A-ataxia. The current understanding of the association between the specific variant type, the severity of CoQ10 deficiency, and the degree of oxidative stress in individuals with primary CoQ10 deficiencies remains uncertain. Here we provide a comprehensive analysis of the clinical and genetic characteristics of an 18-year-old patient with COQ8A-ataxia, who exhibited novel compound heterozygous variants (c.1904_1906del and c.637C > T) in the COQ8A gene. These variants reduced the expression levels of COQ8A and mitochondrial proteins in the patient\'s muscle and skin fibroblast samples, contributed to mitochondrial respiration deficiency, increased ROS production and altered mitochondrial membrane potential. It is worth noting that the optimal treatment for COQ8A-ataxia remains uncertain. Presently, therapy consists of CoQ10 supplementation, however, it did not yield significant improvement in our patient\'s symptoms. Additionally, we reviewed the response of CoQ10 supplementation and evolution of patients in previous literatures in detail. We found that only half of patients could got notable improvement in ataxia. This research aims to expand the genotype-phenotype spectrum of COQ10D4, address discrepancies in previous reviews regarding the effectiveness of CoQ10 in these disorders, and help to establish a standardized treatment protocol for COQ8A-ataxia.
    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
    辅酶Q10(CoQ10)是广泛存在于线粒体内膜中的自然成分。CoQ10作为三磷酸腺苷(ATP)生产的关键辅因子,并在体内表现出抗氧化特性。线粒体,作为电池的能量供应中心,在生殖细胞成熟和胚胎发育中起着至关重要的作用,细胞分裂和细胞分化的复杂过程,从单细胞(受精卵)转变为多细胞生物(胎儿)。这里,我们讨论了CoQ10对卵母细胞成熟的影响以及CoQ10在胎儿发育不同阶段各器官生长中的重要作用。这些使我们对胚胎发育的病理生理学以及CoQ10在改善生育质量中的潜在作用有了更深入的了解。为进一步开展其在临床治疗中的应用提供了参考。
    Coenzyme Q10 (CoQ10) is a natural component widely present in the inner membrane of mitochondria. CoQ10 functions as a key cofactor for adenosine triphosphate (ATP) production and exhibits antioxidant properties in vivo. Mitochondria, as the energy supply center of cells, play a crucial role in germ cell maturation and embryonic development, a complicated process of cell division and cellular differentiation that transforms from a single cell (zygote) to a multicellular organism (fetus). Here, we discuss the effects of CoQ10 on oocyte maturation and the important role of CoQ10 in the growth of various organs during different stages of fetal development. These allowed us to gain a deeper understanding of the pathophysiology of embryonic development and the potential role of CoQ10 in improving fertility quality. They also provide a reference for further developing its application in clinical treatments.
    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:心脏移植(HT)已被批准为终末期心力衰竭患者的最佳治疗方案。然而,冷缺血再灌注(I/R)损伤仍然是一个不可避免和突出的挑战,这是早期移植物功能障碍的主要因素,也是HT长期存活的障碍。冷I/R损伤通过促进线粒体功能障碍和增加自由基产生和炎症反应来诱导心脏移植物损伤。因此,我们设计了一种负载有辅酶Q10(CoQ10)(CoQ10@TNP)的线粒体靶向纳米载体,用于在鼠异位心脏移植模型中治疗心脏移植后的冷I/R损伤。
    方法:使用共沉淀法合成了由CaCO3/CaP/生物素化-羧甲基壳聚糖(CaCO3/CaP/BCMC)组成的杂化纳米颗粒,并通过生物素-抗生物素蛋白相互作用将靶向线粒体的四肽SS31掺入到杂化纳米颗粒的表面上。透射电子显微镜(TEM)和动态光散射(DLS)分析用于表征。体外,H9c2细胞的缺氧-复氧模型用于复制体内冷I/R损伤,并用CoQ10@TNPs治疗。CoQ10@TNPs对H9c2细胞损伤的影响通过氧化损伤和凋亡的分析来评估。在体内,用含有CoQ10@TNP的保存液灌注供体心脏(DHs),并在4°C下体外储存12小时。将DHs异位移植并分析移植物功能,氧化损伤,凋亡,和移植后1天的炎症标志物。
    结果:成功合成了CoQ10@TNP,并将CoQ10递送至冷缺血心肌的线粒体。体外实验表明,CoQ10@TNP在4℃时被H9c2细胞吸收并位于线粒体内,从而改善冷I/R损伤中的氧化应激损伤和线粒体损伤。体内实验表明,CoQ10@TNP在4℃时在DH组织中积累,在冷藏过程中位于线粒体内,并通过减轻线粒体氧化损伤和炎症来改善心脏移植物功能。
    结论:CoQ10@TNPs可以精确地将CoQ10递送至冷I/R损伤的心肌细胞的线粒体,从而有效地消除线粒体活性氧(mtROS),从而减少冷I/R损伤的移植组织的氧化损伤和炎症反应,最终改善心脏移植功能。因此,CoQ10@TNP提供了一种有效的方法来保护心脏移植物免受长时间的冷缺血,强调缓解HT期间冷I/R损伤的治疗潜力。这些发现为增强HT后的现有结果和扩大移植的可行移植物的范围提供了机会。
    Heart transplantation (HT) has been approved as an optimal therapeutic regimen for patients with terminal-stage cardiac failure. However, cold ischaemia‒reperfusion (I/R) injury remains an unavoidable and outstanding challenge, which is a major factor in early graft dysfunction and an obstacle to long-term survival in HT. Cold I/R injury induces cardiac graft injury by promoting mitochondrial dysfunction and augmenting free radical production and inflammatory responses. We therefore designed a mitochondrion-targeted nanocarrier loaded with Coenzyme Q10 (CoQ10) (CoQ10@TNPs) for treatment of cold I/R injury after cardiac graft in a murine heterotopic cardiac transplantation model.
    Hybrid nanoparticles composed of CaCO3/CaP/biotinylated-carboxymethylchitosan (CaCO3/CaP/BCMC) were synthesized using the coprecipitation method, and the mitochondria-targeting tetrapeptide SS31 was incorporated onto the surface of the hybrid nanoparticles through biotin-avidin interactions. Transmission electron microscopy (TEM) and dynamic light scattering (DLS) analysis were used for characterisation. In vitro, the hypoxia-reoxygenation model of H9c2 cells was employed to replicate in vivo cold I/R injury and treated with CoQ10@TNPs. The impact of CoQ10@TNPs on H9c2 cell injury was assessed by analysis of oxidative damage and apoptosis. In vivo, donor hearts (DHs) were perfused with preservation solution containing CoQ10@TNPs and stored in vitro at 4 °C for 12 h. The DHs were heterotopically transplanted and analysed for graft function, oxidative damage, apoptosis, and inflammatory markers 1 day post-transplantation.
    CoQ10@TNPs were successfully synthesized and delivered CoQ10 to the mitochondria of the cold ischaemic myocardium. In vitro experiments demonstrated that CoQ10@TNPs was taken up by H9c2 cells at 4 °C and localized within the mitochondria, thus ameliorating oxidative stress damage and mitochondrial injury in cold I/R injury. In vivo experiments showed that CoQ10@TNPs accumulated in DH tissue at 4 °C, localized within the mitochondria during cold storage and improved cardiac graft function by attenuating mitochondrial oxidative injury and inflammation.
    CoQ10@TNPs can precisely deliver CoQ10 to the mitochondria of cold I/R-injured cardiomyocytes to effectively eliminate mitochondrial reactive oxygen species (mtROS), thus reducing oxidative injury and inflammatory reactions in cold I/R-injured graft tissues and finally improving heart graft function. Thus, CoQ10@TNPs offer an effective approach for safeguarding cardiac grafts against extended periods of cold ischaemia, emphasizing the therapeutic potential in mitigating cold I/R injury during HT. These findings present an opportunity to enhance existing results following HT and broaden the range of viable grafts for transplantation.
    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
    在高脂血症引起的骨质疏松症中,骨髓间充质干细胞(BMSCs)分化为更多的脂肪细胞比成骨细胞,导致骨形成减少。阐明高脂血症对骨代谢的影响并寻求调节脂肪细胞-成骨细胞谱系分配的新药物至关重要。辅酶Q10,线粒体呼吸链的限速辅酶,据报道,通过充当线粒体抗氧化剂来减少氧化应激和脂质过氧化。然而,其对高脂血症所致骨质疏松症的影响尚不清楚.这里,我们通过使用高脂饮食(HFD)处理的ApoE-/-小鼠或氧化低密度脂蛋白(ox-LDL)处理的BMSCs,分析了CoQ10对高脂血症诱导的骨质疏松症的治疗机制.在HFD处理的ApoE-/-小鼠和ox-LDL处理的BMSCs中,血脂水平升高,骨形成相关标志物降低,这可以被CoQ10逆转。此外,在HFD处理的ApoE-/-小鼠和ox-LDL处理的BMSCs中PGC-1α蛋白表达降低,伴有线粒体功能障碍,降低ATP含量和活性氧(ROS)的过度产生,这也可以被CoQ10拮抗。此外,PGC-1α敲低体外促进ROS生成,BMSC凋亡,和脂肪分化,同时减弱BMSCs的成骨分化。机械上,提示miR-130b-3p抑制剂治疗高脂血症骨质疏松可增加PGC1-α蛋白的表达,从而改善线粒体功能。总的来说,CoQ10缓解ApoE-/-小鼠高脂血症诱导的骨质疏松症,并调节脂肪细胞-成骨细胞谱系分配。其潜在机制可能涉及通过调节miR-130b-3p/PGC-1α通路改善线粒体功能。
    In hyperlipidemia-induced osteoporosis, bone marrow mesenchymal stem cells (BMSCs) differentiate into more adipocytes than osteoblasts, leading to decreased bone formation. It is vital to elucidate the effects of hyperlipidemia on bone metabolism and seek new agents that regulate adipocyte-osteoblast lineage allocation. CoQ10, a rate-limiting coenzyme of the mitochondrial respiratory chain, has been reported to decrease oxidative stress and lipid peroxidation by functioning as a mitochondrial antioxidant. However, its effect on hyperlipidemia-induced osteoporosis remains unknown. Here, we analyzed the therapeutic mechanisms of CoQ10 on hyperlipidemia-induced osteoporosis by using high-fat diet (HFD)-treated ApoE-/- mice or oxidized low-density lipoprotein (ox-LDL)-treated BMSCs. The serum lipid levels were elevated and bone formation-related markers were decreased in HFD-treated ApoE-/- mice and ox-LDL-treated BMSCs, which could be reversed by CoQ10. Additionally, PGC-1α protein expression was decreased in HFD-treated ApoE-/- mice and ox-LDL-treated BMSCs, accompanied by mitochondrial dysfunction, decreased ATP content and overgeneration of reactive oxygen species (ROS), which could also be antagonized by CoQ10. Furthermore, PGC-1α knockdown in vitro promoted ROS generation, BMSC apoptosis, and adipogenic differentiation while attenuating osteogenic differentiation in BMSCs. Mechanistically, it suggested that the expression of PGC1-α protein was increased with miR-130b-3p inhibitor treatment in osteoporosis under hyperlipidemia conditions to improve mitochondrial function. Collectively, CoQ10 alleviates hyperlipidemia-induced osteoporosis in ApoE-/- mice and regulates adipocyte-osteoblast lineage allocation. The possible underlying mechanism may involve the improvement of mitochondrial function by modulating the miR-130b-3p/PGC-1α pathway.
    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
    辅酶Q10(CoQ10)对人类至关重要,尤其是在生物学和医学领域。本实验的目的是研究提高CoQ10产量的条件。目前,微生物发酵是辅酶Q10的主要生产方法,而微生物辅酶Q10代谢控制发酵的生产工艺非常关键。代谢通量是代谢工程中细胞生理学最重要的决定因素之一。代谢通量分析(MFA)用于估计代谢网络中的细胞内通量。在这个实验中,以球形红杆菌为研究对象,分析了氨水(NH3·H2O)和碳酸钙(CaCO3)对CoQ10代谢通量的影响。当使用CaCO3调节pH时,辅酶Q10的产量为274.43mg·L-1(8.71mg·g-1DCW),高于NH3·H2O调整。结果表明,当CaCO3用于调节pH时,更多的葡萄糖-6-磷酸(G6P)进入戊糖磷酸(HMP)途径,产生更多的NADPH,这增强了CoQ10的合成。在抗震酸节点处,更多的代谢通量参与对羟基苯甲酸(pHBA;CoQ10的合成前体)的合成,增强CoQ10的合成代谢流。此外,CaCO3与有机酸反应产生的Ca2+促进CoQ10的合成。总之,CaCO3调整比NH3·H2O调整更有利于R.sphaeroides合成CoQ10。分析了由培养条件的扰动引起的代谢通量的迁移,以比较合成CoQ10的细胞内代谢通量分布的变化。因此,代谢网络的主要节点被确定为G6P和分支酸。这为CoQ10合成途径相关基因的修饰提供了理论依据。
    Coenzyme Q10 (CoQ10) is crucial for human beings, especially in the fields of biology and medicine. The aim of this experiment was to investigate the conditions for increasing CoQ10 production. At present, microbial fermentation is the main production method of CoQ10, and the production process of microbial CoQ10 metabolism control fermentation is very critical. Metabolic flux is one of the most important determinants of cell physiology in metabolic engineering. Metabolic flux analysis (MFA) is used to estimate the intracellular flux in metabolic networks. In this experiment, Rhodobacter sphaeroides was used as the research object to analyze the effects of aqueous ammonia (NH3·H2O) and calcium carbonate (CaCO3) on the metabolic flux of CoQ10. When CaCO3 was used to adjust the pH, the yield of CoQ10 was 274.43 mg·L-1 (8.71 mg·g-1 DCW), which was higher than that of NH3·H2O adjustment. The results indicated that when CaCO3 was used to adjust pH, more glucose-6-phosphate (G6P) entered the pentose phosphate (HMP) pathway and produced more NADPH, which enhanced the synthesis of CoQ10. At the chorismic acid node, more metabolic fluxes were involved in the synthesis of p-hydroxybenzoic acid (pHBA; the synthetic precursor of CoQ10), enhancing the anabolic flow of CoQ10. In addition, Ca2+ produced by the reaction of CaCO3 with organic acids promotes the synthesis of CoQ10. In summary, the use of CaCO3 adjustment is more favorable for the synthesis of CoQ10 by R. sphaeroides than NH3·H2O adjustment. The migration of metabolic flux caused by the perturbation of culture conditions was analyzed to compare the changes in the distribution of intracellular metabolic fluxes for the synthesis of CoQ10. Thus, the main nodes of the metabolic network were identified as G6P and chorismic acid. This provides a theoretical basis for the modification of genes related to the CoQ10 synthesis pathway.
    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
    Hepatic ischemia-reperfusion injury (IRI) has been concerned as a main complication of liver surgery and transplantation. Previous studies show that reactive oxygen species (ROS) associated inflammation response and contribute to the liver damage during IRI. Coenzyme Q10 (CoQ10) has shown many beneficial effects on abrogating ROS production and ameliorating liver injury. This study found lower CoQ10 level in the process of liver IRI in a mouse model of hepatic IRI. Meanwhile, our results showed that CoQ10 administration significantly attenuate hepatic IRI proved by HE staining, serum ALT/AST. The NOD-like receptor protein 3 (NLRP3) inflammasome is activated by ROS which triggers the activation of inflammatory caspases. In this study, NLRP3 was significantly suppressed by CoQ10 while Foxp3 exhibited increased expression in liver. Furthermore, Kupffer cells (KCs) pretreated with CoQ10 under the condition of hypoxia and reoxygenation contributed to improved CD4+CD25+Foxp3+ regulatory T cells (Tregs) ratio in co-culture system. Furthermore, NLRP3 inflammasome activator treatment in vivo resulted in higher expression of caspase-1 and NLRP3 and reduction of Tregs in liver, which reversed the protection of CoQ10 in the liver injury. Taken together, our study discovered that CoQ10 can suppress NLRP3 activity in KCs and improves Foxp3+ Tregs differentiation depending on M2 macrophage polarization of KCs to ameliorate hepatic IRI.
    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
    血小板高反应性和氧化应激是COVID-19患者血栓性疾病的重要原因。氧化应激,由活性氧(ROS)的过度产生引起,可以增加血小板功能和血栓形成的风险。辅酶Q10(CoQ10),具有较强的抗氧化活性和抗血小板作用。然而,CoQ10抑制刺突蛋白诱导的血小板聚集的作用及机制尚未研究。本研究旨在研究SARS-CoV-2刺突蛋白是否通过ROS信号增强人血小板功能以及CoQ10的体外保护作用。使用一系列血小板功能测定,我们发现刺突蛋白增强了血小板聚集和氧化应激,如ROS级别,线粒体膜电位去极化,和体外脂质损伤水平(MDA和8-iso-PGF2α)。此外,辅酶Q10减弱刺突蛋白诱导的血小板聚集。作为一种抗血小板机制,我们发现CoQ10显著降低了刺突蛋白诱导的ROS的过量产生。我们的发现表明,CoQ10对刺突蛋白增强的血小板聚集的保护作用可能与其强大的抗氧化能力有关。
    Platelet hyperreactivity and oxidative stress are the important causes of thrombotic disorders in patients with COVID-19. Oxidative stress, induced by the excessive generation of reactive oxygen species (ROS), could increase platelet function and the risk of thrombus formation. Coenzyme Q10 (CoQ10), exhibits strong antioxidative activity and anti-platelet effect. However, the effects and mechanisms of CoQ10 on attenuating platelet aggregation induced by spike protein have never been studied. This study aims to investigate whether the SARS-CoV-2 spike protein potentiates human platelet function via ROS signaling and the protective effect of CoQ10 in vitro. Using a series of platelet function assays, we found that spike protein potentiated platelet aggregation and oxidative stress, such as ROS level, mitochondrial membrane potential depolarization, and lipid damage level (MDA and 8-iso-PGF2α) in vitro. Furthermore, CoQ10 attenuated platelet aggregation induced by spike protein. As an anti-platelet mechanism, we showed that CoQ10 significantly decreased the excess production of ROS induced by spike protein. Our findings show that the protective effect of CoQ10 on spike protein-potentiated platelet aggregation is probably associated with its strong antioxidative ability.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号