Symporters

Symborters
  • 文章类型: Journal Article
    成纤维细胞的胶原蛋白合成减少是皮肤老化的关键方面。聚L-乳酸(PLLA)是一种能连续释放乳酸的生物可吸收材料,刺激皮肤内源性胶原蛋白合成。在这里,这项研究旨在研究释放PLLA的乳酸对成纤维细胞中胶原蛋白生成的影响。在体外将人成纤维细胞暴露于不同浓度的PLLA,同时将PLLA体内注射到老年小鼠的背部皮肤中。通过钙黄绿素-AM/PI染色评价PLLA对胶原蛋白合成和嫩肤的安全性和有效性。EdU增殖试验,并使用蛋白质印迹和免疫荧光分析成纤维细胞中胶原蛋白I和胶原蛋白III的表达。为了阐明潜在的机制,PLLA处理的成纤维细胞的无细胞上清液和细胞裂解物中的乳酸含量,以及测量总赖氨酸的乳酸化(PanKla)水平。此外,我们发现,成纤维细胞可以通过单羧酸转运蛋白1(MCT1)摄取从PLLA释放的细胞外乳酸,以促进潜伏转化生长因子β结合蛋白1(LTBP1)通过KAT8依赖性机制在赖氨酸752(K752)处的乳酸化,然后增加成纤维细胞中胶原蛋白I和胶原蛋白III的蛋白质水平。总的来说,这项研究强调了对非组蛋白蛋白的乳酸化修饰用于嫩肤的有价值的见解。
    Decreased collagen synthesis by fibroblasts is a key aspect of skin aging. Poly-L-Lactic Acid (PLLA) is a bioabsorbable material that can release lactate continuously, stimulating endogenous collagen synthesis in the skin. Herein, this study aimed to investigate the impact of PLLA-released lactate on collagen production in fibroblasts for skin rejuvenation. Human fibroblasts were exposed to varying concentrations of PLLA in vitro, while PLLA was injected into the back skin of aged mice in vivo. Safety and efficacy of PLLA on collagen synthesis and skin rejuvenation were evaluated through Calcein-AM/PI staining, EdU proliferation assay, and analysis of collagen I and collagen III expression in fibroblasts using western blotting and immunofluorescence. To elucidate the underlying mechanisms, lactate contents in cell-free supernatant and cell lysates from PLLA-treated fibroblasts, as well as total lysine lactylation (Pan Kla) levels were measured. Additionally, we found that fibroblasts can uptake extracellular lactate released from PLLA through monocarboxylate transporter-1 (MCT1) to facilitate latent-transforming growth factor beta-binding protein 1 (LTBP1) lactylation at lysine 752 (K752) via a KAT8-dependent mechanism, then increases the protein levels of collagen I and collagen III in fibroblasts. Overall, this study highlights a valuable insight into lactylation modification of non-histone protein for skin rejuvenation.
    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
    乙型肝炎病毒(HBV)表达共末端大(L),中间(M),和含有preS1/preS2/S的小(S)包膜蛋白,preS2/S,和S域单独,分别。S和preS1结构域介导与硫酸乙酰肝素蛋白聚糖和牛磺胆酸钠共转运多肽(NTCP)的序列病毒体附着,分别,可被抗S和抗preS1抗体阻断。抗preS2抗体如何中和HBV感染性仍然是神秘的。慢性HBV感染的晚期通常选择突变的preS2翻译起始密码子,以防止M蛋白表达,或框内preS2缺失以缩短L和M蛋白。当引入基因型C或D的感染性克隆时,M-负突变和大多数5'preS2缺失都维持了病毒体的产生。这种突变子代病毒颗粒在NTCP重建的HepG2细胞中具有感染性。对基因型D克隆进行中和实验。尽管仍然易感抗preS1和抗S中和抗体,M-负突变体仅被测试的两种抗preS2抗体部分中和,而preS2缺失突变体具有抗性。通过感染实验,使用具有丢失与增加的M蛋白表达的病毒颗粒,或仅存在于L或M蛋白上的中和逃逸preS2缺失,我们发现全长L和M蛋白均有助于两种抗preS2抗体中和病毒.因此,免疫逃逸可能是选择M-负突变的驱动力,尤其是preS2删除。L和M蛋白均可介导抗preS2抗体的中和,这一事实可能揭示了潜在的分子机制。重要的大(L),中间(M),和乙型肝炎病毒(HBV)的小(S)包膜蛋白含有preS1/preS2/S,preS2/S,和S域单独,分别。硫酸乙酰肝素蛋白聚糖和牛磺胆酸钠协同转运多肽(NTCP)作为低和高亲和力HBV受体的发现可以解释抗S和抗preS1抗体的中和潜力,分别,但是抗preS2中和抗体是如何工作的仍然是神秘的。在这项研究中,我们在基因型D的背景下发现了两个M-负突变体,在NTCP重建的HepG2细胞中部分逃脱了两个抗preS2中和抗体,而几个天然存在的preS2缺失突变体逃脱了这两种抗体。通过点突变来消除或增强M蛋白的表达,通过选择性地将preS2缺失引入L或M蛋白,我们发现抗preS2抗体与L和M蛋白的结合有助于中和野生型HBV感染性。我们的发现可能揭示了抗preS2抗体中和HBV感染性的可能机制。
    Hepatitis B virus (HBV) expresses co-terminal large (L), middle (M), and small (S) envelope proteins containing preS1/preS2/S, preS2/S, and S domain alone, respectively. S and preS1 domains mediate sequential virion attachment to heparan sulfate proteoglycans and sodium taurocholate cotransporting polypeptide (NTCP), respectively, which can be blocked by anti-S and anti-preS1 antibodies. How anti-preS2 antibodies neutralize HBV infectivity remains enigmatic. The late stage of chronic HBV infection often selects for mutated preS2 translation initiation codon to prevent M protein expression, or in-frame preS2 deletions to shorten both L and M proteins. When introduced to infectious clone of genotype C or D, both M-minus mutations and most 5\' preS2 deletions sustained virion production. Such mutant progeny viral particles were infectious in NTCP-reconstituted HepG2 cells. Neutralization experiments were performed on the genotype D clone. Although remaining susceptible to anti-preS1 and anti-S neutralizing antibodies, M-minus mutants were only partially neutralized by two anti-preS2 antibodies tested while preS2 deletion mutants were resistant. By infection experiments using viral particles with lost versus increased M protein expression, or a neutralization escaping preS2 deletion only present on L or M protein, we found that both full-length L and M proteins contributed to virus neutralization by the two anti-preS2 antibodies. Thus, immune escape could be a driving force for the selection of M-minus mutations, and especially preS2 deletions. The fact that both L and M proteins could mediate neutralization by anti-preS2 antibodies may shed light on the underlying molecular mechanism.IMPORTANCEThe large (L), middle (M), and small (S) envelope proteins of hepatitis B virus (HBV) contain preS1/preS2/S, preS2/S, and S domain alone, respectively. The discovery of heparan sulfate proteoglycans and sodium taurocholate cotransporting polypeptide (NTCP) as the low- and high-affinity HBV receptors could explain neutralizing potential of anti-S and anti-preS1 antibodies, respectively, but how anti-preS2 neutralizing antibodies work remains enigmatic. In this study, we found two M-minus mutants in the context of genotype D partially escaped two anti-preS2 neutralizing antibodies in NTCP-reconstituted HepG2 cells, while several naturally occurring preS2 deletion mutants escaped both antibodies. By point mutations to eliminate or enhance M protein expression, and by introducing preS2 deletion selectively to L or M protein, we found binding of anti-preS2 antibodies to both L and M proteins contributed to neutralization of wild-type HBV infectivity. Our finding may shed light on the possible mechanism(s) whereby anti-preS2 antibodies neutralize HBV infectivity.
    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
    长期服用抗结核药物会引起药物性胆汁淤积性肝损伤。利福平导致的肝损伤可能与胆汁酸核受体法尼醇X受体(FXR)有关。为了调查这一点,通过连续7天的灌胃给药利福平(200mg/kg),在野生型(C57BL/6N)小鼠和FXR基因敲除(FXR-null)小鼠中均可诱导胆汁淤积。与C57BL/6N小鼠比拟,FXR-null小鼠在利福平给药后表现出更严重的肝损伤,以肝脏尺寸增大为特征,转氨酶升高,增加炎症。此外,在利福平治疗下,FXR敲除会损害脂质分泌并加剧肝脏脂肪变性。重要的是,代谢分子BSEP的表达增加,C57BL/6N小鼠服用利福平后NTCP和CYP7A1下降,而这些变化在FXR敲除小鼠中不存在。此外,利福平治疗C57BL/6N和FXR-null小鼠与c-JunN末端激酶磷酸化(p-JNK)水平升高有关,在FXR无效小鼠中具有更明显的升高。我们的研究表明,利福平引起的肝损伤,脂肪变性,胆汁淤积与FXR功能障碍和胆汁酸代谢改变有关,并且JNK信号通路部分参与了这种损伤。基于这些结果,我们认为FXR可能是解决药物性肝损伤的新治疗靶点。
    Antituberculosis drugs induce pharmacologic cholestatic liver injury with long-term administration. Liver injury resulting from rifampicin is potentially related to the bile acid nuclear receptor Farnesoid X Receptor (FXR). To investigate this, cholestasis was induced in both wild-type (C57BL/6N) mice and FXR knockout (FXR-null) mice through administration of rifampicin (200 mg/kg) via gavage for 7 consecutive days. Compared with C57BL/6N mice, FXR-null mice exhibited more severe liver injury after rifampicin administration, characterized by enlarged liver size, elevated transaminases, and increased inflammation. Moreover, under rifampicin treatment, FXR knockout impairs lipid secretion and exacerbates hepatic steatosis. Significantly, the expression of metabolism molecules BSEP increased, while NTCP and CYP7A1 decreased following rifampicin administration in C57BL/6N mice, whereas these changes were absent in FXR knockout mice. Furthermore, rifampicin treatment in both C57BL/6N and FXR-null mice was associated with elevated c-Jun N-terminal kinase phosphorylation (p-JNK) levels, with a more pronounced elevation in FXR-null mice. Our study suggests that rifampicin-induced liver injury, steatosis, and cholestasis are associated with FXR dysfunction and altered bile acid metabolism, and that the JNK signaling pathway is partially implicated in this injury. Based on these results, we propose that FXR might be a novel therapeutic target for addressing drug-induced liver injury.
    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
    患儿 男,1月11日龄,因发现皮肤黄染1个月余就诊,肝功能示转氨酶升高、胆汁淤积。因患儿经治疗后粪便色浅,肝功能无改善,肝脏穿刺病理示细胆管增生,遂行胆道探查术,但胆道通畅。术后继续给予护肝利胆治疗。患儿3月龄时发现其在黄疸消退后仍存在显著增高的胆汁酸水平,最终行基因检测确诊钠牛磺胆酸共转运多肽缺陷病。.
    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
    背景:Allan-Herndon-Dudley综合征(AHDS)是由SLC16A2基因的半合子亚基的致病性变异引起的,它编码单羧酸转运蛋白8,并遵循X连锁隐性模式。AHDS表现为神经精神运动发育迟缓,智力残疾,运动障碍,甲状腺激素异常.常误诊为脑瘫或甲状腺功能减退症。
    方法:一名9个月大的男婴头部控制不佳,乏力,电机延迟,四肢高渗,甲状腺异常.尽管补充了左旋甲状腺素和康复治疗,没有观察到改善。全外显子组测序在SLC16A2中发现了一个新的无义突变(c.124G>T,p.E42X),明确地确定了诊断。
    方法:确认了AHDS。
    方法:左甲状腺素治疗在婴儿期早期开始,接着是3个月的康复治疗,从5个月大开始。左甲状腺素和甲咪唑的联合给药在1岁和10个月大时开始,分别。
    结果:虽然甲状腺激素水平有所改善,神经发育迟缓持续存在.
    结论:AHDS应适用于表现为不典型神经系统特征和甲状腺激素异常的患者,如三碘甲状腺原氨酸升高和甲状腺素水平降低。外显子组测序的早期利用有助于及时诊断。鉴定的SLC16A2无义突变与严重的神经学表型相关,并增加与AHDS相关的遗传变异谱。
    BACKGROUND: Allan-Herndon-Dudley syndrome (AHDS) results from a pathogenic variant in the hemizygous subunit of the SLC16A2 gene, which encodes monocarboxylate transporter 8 and follows an X-linked recessive pattern. AHDS manifests as neuropsychomotor developmental delay, intellectual disability, movement disorders, and thyroid hormone abnormalities. It is frequently misdiagnosed as cerebral palsy or hypothyroidism.
    METHODS: A 9-month-old male infant exhibited poor head control, hypodynamia, motor retardation, hypertonic limbs, and thyroid abnormalities. Despite levothyroxine supplementation and rehabilitation therapy, no improvements were observed. Whole-exome sequencing identified a novel nonsense mutation in SLC16A2 (c.124G > T, p.E42X), which unequivocally established the diagnosis.
    METHODS: AHDS was confirmed.
    METHODS: Levothyroxine treatment commenced early in infancy, followed by 3 months of rehabilitation therapy, starting at 5 months of age. The combined administration of levothyroxine and methimazole was initiated at 1 year and 10 months of age, respectively.
    RESULTS: While improvements were noted in thyroid hormone levels, neurological developmental delays persisted.
    CONCLUSIONS: AHDS should be considered in patients presenting with atypical neurological features and thyroid hormone abnormalities such as elevated triiodothyronine and decreased thyroxine levels. The early utilization of exome sequencing aids in prompt diagnosis. The identified SLC16A2 nonsense mutation correlates with severe neurological phenotypes and adds to the spectrum of genetic variations associated with AHDS.
    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
    甲状腺乳头状癌(PTC)手术后复发的可能性很高,术后应用促甲状腺激素(TSH)抑制和放射性碘(131I)治疗。本研究探讨了淋巴结转移(LNM)的潜在机制,旨在开发PTC的分化治疗方法。
    本研究通过分析2073例PTC患者的临床资料,探讨PTC淋巴结转移的危险因素。分析了癌症基因组图谱甲状腺癌(TCGA-THCA)和基因表达的基因表达综合(GEO)数据库,以鉴定基因表达与表型之间的相互关系。
    分析临床数据,我们发现男性性别,年龄较小,肿瘤较大,甲状腺外延伸(ETE)是淋巴结转移的危险因素(P<0.05)。相反,甲状腺功能参数,如TSH,FT3,FT4,TSH/FT3和TSH/FT4与LNM无关(P>0.05)。女性TSH水平较高(P<0.05)。基因表达分析显示男性SLC5A5表达下调,年轻的个人,那些有淋巴结转移的人,较低的SLC5A5水平与较差的无病生存率相关(P<0.05)。此外,我们对单细胞RNA测序(scRNA-seq)数据的检查表明,SLC5A5在肿瘤和淋巴结转移样本中的表达降低,与TSHR的表达呈正相关。
    TSH对PTC行为的影响尚不清楚,而依赖于SLC5A5的吸收131I的能力显示出不同性别和年龄的差异。我们得出结论,PTC的术后治疗应考虑性别和年龄引起的差异。
    UNASSIGNED: Thyroid papillary carcinoma (PTC) had a high possibility of recurrence after surgery, and thyroid stimulating hormone (TSH) suppression and radioactive iodine (131I) were used for postoperative therapy. This study explored the potential mechanism of lymph node metastasis (LNM) and aimed to develop differentiated treatments for PTC.
    UNASSIGNED: This study explored the risk factors of lymph node metastasis in PTC by analyzing the clinical information of 2073 cases. The Cancer Genome Atlas Thyroid Cancer (TCGA-THCA) and the Gene Expression Omnibus (GEO) databases of gene expression were analyzed to identify the interrelationships between gene expression to phenotype.
    UNASSIGNED: Analyzing clinical data, we found that male gender, younger age, larger tumor size, and extra-thyroidal extension (ETE) were risk significant risk factors for lymph node metastasis(P<0.05). Conversely, thyroid function parameters such as TSH, FT3, FT4, TSH/FT3, and TSH/FT4 didn\'t correlate with LNM(P>0.05), and TSH levels were observed to be higher in females(P<0.05). Gene expression analysis revealed that SLC5A5 was down-regulated in males, younger individuals, and those with lymph node metastasis, and a lower level of SLC5A5 was associated with a worse disease-free survival(P<0.05). Additionally, our examination of single-cell RNA sequencing (scRNA-seq) data indicated that SLC5A5 expression was reduced in tumors and lymph node metastasis samples, correlating positively with the expression of TSHR.
    UNASSIGNED: The impact of TSH on PTC behavior remained unclear, while the capacity for absorbing 131I in dependence on SLC5A5 showed variations across different genders and ages. We conclude that postoperative treatment of PTC should take into account the differences caused by gender and age.
    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
    阿尔茨海默病(AD)的标志是淀粉样β(Aβ)斑块在大脑中沉积,导致认知功能的缺陷。淀粉样β寡聚体(AβOs),产生Aβ斑块的可溶性前体肽,还会产生神经毒性和小胶质细胞增生以及糖酵解重编程。最近,单羧酸转运蛋白1(MCT1),一个关键的糖酵解调节剂,和它的辅助蛋白质,发现CD147在外泌体的分泌中起重要作用,30-200nm大小的囊泡,被认为是AD中的毒性分子载体。然而,低浓度AβOs(1nM)对小胶质细胞MCT1和CD147表达的影响以及1nMAβOs处理的小胶质细胞来源的外泌体对神经元毒性的影响在很大程度上仍然难以捉摸。在这项研究中,1nMAβOs诱导显著的轴突病变和小胶质细胞增生。此外,1nMAβOs处理的神经元或小胶质细胞来源的外泌体通过神经元自体或异源摄取产生轴突病变,支持外泌体作为AD中神经毒性介质的作用。有趣的是,通过用1nMAβOs或来自1nMAβOs治疗的小胶质细胞或神经元的外泌体治疗,MCT1和CD147在小胶质细胞中得到增强,提示AβOs诱导的增强的MCT1和CD147在小胶质细胞与AD神经发病机制中的意义,这与来自AD和AD患者的小鼠模型中的小胶质细胞的单细胞RNA测序数据的计算机模拟分析一致。
    A hallmark of Alzheimer\'s disease (AD) is amyloid-β (Aβ) plaque deposition in the brain, causing deficits in cognitive function. Amyloid-beta oligomers (AβOs), the soluble precursor peptides producing Aβ plaques, also produce neurotoxicity and microgliosis together with glycolytic reprogramming. Recently, monocarboxylate transporter 1 (MCT1), a key glycolysis regulator, and its ancillary protein, CD147, are found to play an important role in the secretion of exosomes, 30-200 nm vesicles in size, which are considered as toxic molecule carriers in AD. However, the effect of low-concentration AβOs (1 nM) on microglia MCT1 and CD147 expression as well as 1 nM AβOs-treated microglia-derived exosomes on neuronal toxicity remain largely elusive. In this study, 1 nM AβOs induce significant axonopathy and microgliosis. Furthermore, 1 nM AβOs-treated neurons- or microglia-derived exosomes produce axonopathy through their autologous or heterologous uptake by neurons, supporting the role of exosomes as neurotoxicity mediators in AD. Interestingly, MCT1 and CD147 are enhanced in microglia by treatment with 1 nM AβOs or exosomes from 1 nM AβOs-treated- microglia or neurons, suggesting the implication of AβOs-induced enhanced MCT1 and CD147 in microglia with AD neuropathogenesis, which is consistent with the in-silico analysis of the single cell RNA sequencing data from microglia in mouse models of AD and AD patients.
    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
    曾经被认为是代谢废物,“乳酸现在被认为是三羧酸(TCA)循环的主要燃料。我们的代谢通量分析表明,骨骼肌主要利用乳酸来促进TCA循环。乳酸盐通过单羧酸转运蛋白(MCTs)转运通过细胞膜,其中MCTl在肌肉中高度表达。我们使用骨骼肌中MCT1特异性缺失的小鼠分析了MCT1如何影响肌肉功能。MCT1删除增强了运行性能,增加氧化纤维,同时减少糖酵解纤维,并增强葡萄糖向TCA循环的通量。MCT1缺乏增加线粒体蛋白的表达,增加细胞呼吸速率,并提高肌肉中的线粒体活性。机械上,PGC-1α的蛋白质水平,线粒体生物发生的主要调节因子,在MCT1缺失时通过细胞NAD+水平和SIRT1活性的增加而升高。总的来说,这些结果表明,MCT1介导的乳酸穿梭通过调节线粒体生物发生和TCA通量在调节肌肉功能中起关键作用。
    Once considered as a \"metabolic waste,\" lactate is now recognized as a major fuel for tricarboxylic acid (TCA) cycle. Our metabolic flux analysis reveals that skeletal muscle mainly uses lactate to fuel TCA cycle. Lactate is transported through the cell membrane via monocarboxylate transporters (MCTs) in which MCT1 is highly expressed in the muscle. We analyzed how MCT1 affects muscle functions using mice with specific deletion of MCT1 in skeletal muscle. MCT1 deletion enhances running performance, increases oxidative fibers while decreasing glycolytic fibers, and enhances flux of glucose to TCA cycle. MCT1 deficiency increases the expression of mitochondrial proteins, augments cell respiration rate, and elevates mitochondrial activity in the muscle. Mechanistically, the protein level of PGC-1α, a master regulator of mitochondrial biogenesis, is elevated upon loss of MCT1 via increases in cellular NAD+ level and SIRT1 activity. Collectively, these results demonstrate that MCT1-mediated lactate shuttle plays a key role in regulating muscle functions by modulating mitochondrial biogenesis and TCA flux.
    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
    抑制性神经传递的强度取决于细胞内神经元氯化物浓度,主要受阳离子-氯化物共转运蛋白NKCC1(钠-氯化钾共转运蛋白1)和KCC2(氯化钾共转运蛋白2)的活性调节。脑源性神经营养因子(BDNF)影响这些协同转运蛋白的功能。BDNF是从前体蛋白(proBDNF)合成的,其经历蛋白水解切割以产生成熟BDNF(mBDNF)。虽然先前的研究表明BDNF信号参与KCC2的活性,但其具体机制尚不清楚。我们研究了大鼠海马神经元和大鼠幼鼠子宫内电穿孔皮质中两种形式的BDNF和氯化物稳态之间的相互作用,跨越行为,细胞,和分子水平。我们发现pro-和mBDNF通过抑制神经元挤出氯化物的能力在未成熟神经元中起着相当的作用。此外,proBDNF增加KCC2的内吞作用,同时维持成熟神经元中EGABA的去极化。行为上,体感皮层中proBDNF电穿孔的大鼠幼崽表现出感觉缺陷,延迟拥挤,避免悬崖。这些发现强调了BDNF信号传导在通过调节KCC2调节氯化物转运中的作用。总之,这项研究为BDNF之间复杂的相互作用提供了有价值的见解,氯化物稳态,和抑制性突触传递,揭示所涉及的潜在细胞机制。
    The strength of inhibitory neurotransmission depends on intracellular neuronal chloride concentration, primarily regulated by the activity of cation-chloride cotransporters NKCC1 (Sodium-Potassium-Chloride Cotransporter 1) and KCC2 (Potassium-Chloride Cotransporter 2). Brain-derived neurotrophic factor (BDNF) influences the functioning of these co-transporters. BDNF is synthesized from precursor proteins (proBDNF), which undergo proteolytic cleavage to yield mature BDNF (mBDNF). While previous studies have indicated the involvement of BDNF signaling in the activity of KCC2, its specific mechanisms are unclear. We investigated the interplay between both forms of BDNF and chloride homeostasis in rat hippocampal neurons and in utero electroporated cortices of rat pups, spanning the behavioral, cellular, and molecular levels. We found that both pro- and mBDNF play a comparable role in immature neurons by inhibiting the capacity of neurons to extrude chloride. Additionally, proBDNF increases the endocytosis of KCC2 while maintaining a depolarizing shift of EGABA in maturing neurons. Behaviorally, proBDNF-electroporated rat pups in the somatosensory cortex exhibit sensory deficits, delayed huddling, and cliff avoidance. These findings emphasize the role of BDNF signaling in regulating chloride transport through the modulation of KCC2. In summary, this study provides valuable insights into the intricate interplay between BDNF, chloride homeostasis, and inhibitory synaptic transmission, shedding light on the underlying cellular mechanisms involved.
    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:SLC5A7基因突变导致先天性肌无力,一种罕见的遗传病.SLC5A7基因中的突变点在个体之间不同,并且包含各种遗传变异;然而,相关病例中尚未报道外显子缺失变异.本研究旨在探讨SLC5A7基因变异导致的先天性肌无力综合征患者及其家庭成员的临床表型和遗传特征。
    方法:我们描述了一例患有先天性肌无力综合征的中国男性,表现为四肢波动无力。遗传检测揭示了SLC5A7基因中跨越外显子1-9的杂合缺失突变。QPCR证实患者母亲和兄弟的SLC5A7基因外显子9缺失。用吡啶斯的明治疗后,肌无力的临床症状得到改善。
    结论:SLC5A7基因的外显子1、5和9编码胆碱转运蛋白的跨膜区。这些外显子中的突变可以影响胆碱转运蛋白的稳定性和质膜水平。因此,SLC5A7基因外显子1-9的杂合缺失可能是该患者的致病原因。在表现出波动无力的患者中,RNS阳性,和重症肌无力抗体的血清阴性,应该考虑详细的家族史,建议加强基因检测以确定病因。
    BACKGROUND: Mutations in the SLC5A7 gene cause congenital myasthenia, a rare genetic disorder. Mutation points in the SLC5A7 gene differ among individuals and encompass various genetic variations; however, exon deletion variants have yet to be reported in related cases. This study aims to explore the clinical phenotype and genetic traits of a patient with congenital myasthenic syndrome due to SLC5A7 gene variation and those of their family members.
    METHODS: We describe a case of a Chinese male with congenital myasthenic syndrome presenting fluctuating limb weakness. Genetic testing revealed a heterozygous deletion mutation spanning exons 1-9 in the SLC5A7 gene. QPCR confirmed a deletion in exon 9 of the SLC5A7 gene in the patient\'s mother and brother. Clinical symptoms of myasthenia improved following treatment with pyridostigmine.
    CONCLUSIONS: Exons 1, 5, and 9 of the SLC5A7 gene encode the choline transporter\'s transmembrane region. Mutations in these exons can impact the stability and plasma membrane levels of the choline transporter. Thus, a heterozygous deletion in exons 1-9 of the SLC5A7 gene could be the pathogenic cause for this patient. In patients exhibiting fluctuating weakness, positive RNS, and seronegativity for myasthenia gravis antibodies, a detailed family history should be considered, and enhanced genetic testing is recommended to determine the cause.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号