Complement Factor I

补体因子 I
  • 文章类型: Journal Article
    背景:结肠癌(CC)的发生和进展受肿瘤微环境的影响很大。然而,确切的潜在监管机制尚不清楚.
    目的:研究免疫浸润相关差异表达基因(DEGs)在CC中的作用,探讨补体因子I(CFI)的作用及其可能的分子机制。
    方法:使用生物信息学筛选与免疫浸润相关的DEGs的CC。定量逆转录聚合酶链反应用于检查CC细胞系中hubDEGs的表达。构建了稳定的CFI敲低HT29和HCT116细胞系,使用CCK-8,5-乙炔基-2'-脱氧尿苷评估CFI在体外的不同作用,伤口愈合,和transwell分析。采用苏木精和伊红染色和免疫组织化学染色来评估CFI对使用BALB/c雄性裸鼠构建的CC异种移植模型的肿瘤发生的影响。使用蛋白质印迹法测量与糖酵解和Wnt途径相关的关键蛋白。
    结果:筛选了六个关键的免疫浸润相关的DEGs,其中CFI的表达,补体因子B,淋巴增强子结合因子1和SRY相关的高迁移率组框4上调,而脂肪酸结合蛋白1和骨形态发生蛋白2的表达下调。此外,CFI可以用作CC的诊断生物标志物。功能上,CFI沉默抑制CC细胞增殖,迁移,入侵,和肿瘤生长。机械上,CFI敲除下调关键糖酵解相关蛋白(葡萄糖转运蛋白1型,己糖激酶2,乳酸脱氢酶A,和丙酮酸激酶M2)和Wnt途径相关蛋白(β-catenin和c-Myc)。进一步的研究表明,CFI敲低通过阻断Wnt/β-catenin/c-Myc途径抑制CC中的糖酵解。
    结论:本研究的发现表明,CFI通过影响糖酵解和Wnt/β-catenin/c-Myc途径在CC发育中起关键作用,表明它可以作为CC治疗干预的有希望的目标。
    BACKGROUND: Colon cancer (CC) occurrence and progression are considerably influenced by the tumor microenvironment. However, the exact underlying regulatory mechanisms remain unclear.
    OBJECTIVE: To investigate immune infiltration-related differentially expressed genes (DEGs) in CC and specifically explored the role and potential molecular mechanisms of complement factor I (CFI).
    METHODS: Immune infiltration-associated DEGs were screened for CC using bioinformatics. Quantitative reverse transcription polymerase chain reaction was used to examine hub DEGs expression in the CC cell lines. Stable CFI-knockdown HT29 and HCT116 cell lines were constructed, and the diverse roles of CFI in vitro were assessed using CCK-8, 5-ethynyl-2\'-deoxyuridine, wound healing, and transwell assays. Hematoxylin and eosin staining and immunohistochemistry staining were employed to evaluate the influence of CFI on the tumorigenesis of CC xenograft models constructed using BALB/c male nude mice. Key proteins associated with glycolysis and the Wnt pathway were measured using western blotting.
    RESULTS: Six key immune infiltration-related DEGs were screened, among which the expression of CFI, complement factor B, lymphoid enhancer binding factor 1, and SRY-related high-mobility-group box 4 was upregulated, whereas that of fatty acid-binding protein 1, and bone morphogenic protein-2 was downregulated. Furthermore, CFI could be used as a diagnostic biomarker for CC. Functionally, CFI silencing inhibited CC cell proliferation, migration, invasion, and tumor growth. Mechanistically, CFI knockdown downregulated the expression of key glycolysis-related proteins (glucose transporter type 1, hexokinase 2, lactate dehydrogenase A, and pyruvate kinase M2) and the Wnt pathway-related proteins (β-catenin and c-Myc). Further investigation indicated that CFI knockdown inhibited glycolysis in CC by blocking the Wnt/β-catenin/c-Myc pathway.
    CONCLUSIONS: The findings of the present study demonstrate that CFI plays a crucial role in CC development by influencing glycolysis and the Wnt/β-catenin/c-Myc pathway, indicating that it could serve as a promising target for therapeutic intervention in CC.
    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
    编码调节酶补体因子I(CFI;FI)的基因中的罕见变体(RV),其降低蛋白质功能或水平增加年龄相关性黄斑变性(AMD)风险。共有3357名受试者在SCOPE自然史研究中进行了AMD继发的地理萎缩(GA)筛查,包括CFI测序,然后进行血清FI测量。在C3b和C4b液相裂解测定和表面结合C3b裂解的新型基于珠子的功能测定(BBFA)的纯FI蛋白的背景下,表征了将其归类为I型(低血清水平)或II型(正常血清水平但酶功能降低)的11种具有挑战性的CFIRV基因型。预测或先前表征为良性的另外4种变体,使用BBFA进行分析,以增加其分类的置信度。总之,3个变体[W51S,C67R,I370T]导致低表达。另外4个变体[P64L,R339Q,G527V和P528T]被鉴定为对于C3b分解>1log相对于WT蛋白增加的IC50是高度有害的,而2个变体[K476E和R474Q]的功能降低了约1个对数。同时,6个变体[P50A,T203I,K441R,E548Q,P553S,S570T]具有与野生型(WT)相似的IC50。Odds比值(ORs)与BBFAIC50呈正相关(r=0.76,P<0.01),而ORs与联合注释依赖性耗竭(CADD)评分无差异(r=0.43,P=0.16)。总的来说,15个CFIRV在功能上进行了表征,这可以帮助将来患者进行补体靶向治疗的分层方法。纯蛋白质体外分析仍然是确定CFIRV功能结果的金标准。
    Rare variants (RVs) in the gene encoding the regulatory enzyme complement factor I (CFI; FI) that reduce protein function or levels increase age-related macular degeneration risk. A total of 3357 subjects underwent screening in the SCOPE natural history study for geographic atrophy secondary to age-related macular degeneration, including CFI sequencing and serum FI measurement. Eleven CFI RV genotypes that were challenging to categorize as type I (low serum level) or type II (normal serum level, reduced enzymatic function) were characterized in the context of pure FI protein in C3b and C4b fluid phase cleavage assays and a novel bead-based functional assay (BBFA) of C3b cleavage. Four variants predicted or previously characterized as benign were analyzed by BBFA for comparison. In all, three variants (W51S, C67R, and I370T) resulted in low expression. Furthermore, four variants (P64L, R339Q, G527V, and P528T) were identified as being highly deleterious with IC50s for C3b breakdown >1 log increased versus the WT protein, while two variants (K476E and R474Q) were ∼1 log reduced in function. Meanwhile, six variants (P50A, T203I, K441R, E548Q, P553S, and S570T) had IC50s similar to WT. Odds ratios and BBFA IC50s were positively correlated (r = 0.76, p < 0.01), while odds ratios versus combined annotation dependent depletion (CADD) scores were not (r = 0.43, p = 0.16). Overall, 15 CFI RVs were functionally characterized which may aid future patient stratification for complement-targeted therapies. Pure protein in vitro analysis remains the gold standard for determining the functional consequence of CFI RVs.
    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
    非典型溶血性尿毒综合征(aHUS)是一种罕见的肾脏疾病,由于补体替代途径(AP)的失调。补体因子I(CFI)负调节AP和CFI基因罕见变体与aHUS相关,具有较低的疾病外显率。我们报告了10例与罕见CFI变异p.Ile357Met(c.1071T>G)相关的HUS无关病例。在2007年1月至2022年1月在法国HUS注册中纳入的患者中,回顾性鉴定了所有具有孤立的p.Ile357MetCFI错义变异的患者。我们确定了10名无关患者(70%为女性;HUS诊断时的中位年龄,36.5岁),在CFI基因中携带相同的罕见变异p.Ile357Met。在5例患者中,有7例患者(病例1-7)在与恶性高血压相关的天然肾脏中出现aHUS。没有人接受C5抑制剂。其中2例发生在围产期,肾功能完全恢复,而这些患者中有5例达到肾衰竭,需要替代疗法(KFRT)。四名KFRT患者随后接受了肾脏移植。Threelater在肾脏移植物中出现了C3肾小球病,但没有人出现aHUS复发。其他三名患者(病例8-10)在肾移植后经历了从头TMA,由各种触发器沉淀。罕见的CFI变体p.Ile357Met似乎是HUS和某些形式的继发性HUS的促进遗传因素。
    Atypical hemolytic uremic syndrome (aHUS) is a rare kidney disease due to a dysregulation of the complement alternative pathway. Complement factor I (CFI) negatively regulates the alternative pathway and CFI gene rare variants have been associated to aHUS with a low disease penetrance. We report 10 unrelated cases of HUS associated to a rare CFI variant, p.Ile357Met (c.1071T>G). All patients with isolated p.Ile357Met CFI missense variant were retrospectively identified among patients included between January 2007 and January 2022 in the French HUS Registry. We identified 10 unrelated patients (70% women; median age at HUS diagnosis, 36.5 years) who carry the same rare variant p.Ile357Met in the CFI gene. Seven patients (cases 1-7) presented with aHUS in the native kidney associated with malignant hypertension in 5 patients. None received a C5 inhibitor. Two of these cases occurred in the peripartum period with complete recovery of kidney function, while 5 of these patients reached kidney failure requiring replacement therapy (KFRT). Four patients with KFRT subsequently underwent kidney transplantation. Three later developed C3 glomerulopathy in their kidney graft, but none had aHUS recurrence. Three other patients (cases 8-10) experienced de novo thrombotic microangiopathy after kidney transplantation, precipitated by various triggers. The rare CFI variant p.Ile357Met appears to be a facilitating genetic factor for HUS and for some forms of secondary HUS.
    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
    虽然许多膜结合补体抑制剂保护身体细胞免受先天免疫的自身攻击,可溶性抑制剂如补体因子I(FI)很少在肝脏外产生。以前,我们报道了非小细胞肺癌(NSCLC)细胞系中FI的表达。现在,我们评估了肺癌患者癌活检组织中的FI含量,并将结果与临床病理特征和临床结局相关联.免疫组织化学染色强度与年龄无关,吸烟状况,肿瘤大小,舞台,分化等级,和T细胞浸润,但与无进展生存期(PFS)相关,总生存期(OS)和疾病特异性生存期(DSS)。高与高的多变量Cox分析低FI含量显示PFS的HR0.55,95%CI0.32-0.95,p=0.031,HR0.51,95%CI0.25-1.02,OSp=0.055,DSS的HR0.32,95%CI0.12-0.84,p=0.021。不良预后可能源于FI的非规范作用,因为染色模式与活性补体成分C4b的FI支持降解的产物C4d不相关。为了阐明这一点,我们用CRISPR/Cas9技术改造了三种天然表达FI的人NSCLC细胞系,并比较了每个细胞系中FI缺陷和FI充足克隆的转录组。RNA测序揭示了参与细胞内信号通路控制增殖的差异表达基因,凋亡,和对生长因子的反应能力。此外,体外集落形成实验表明,与FI充足的NSCLC细胞相比,FI缺乏的细胞形成的病灶较小,但是当将纯化的FI蛋白添加到培养基中时,它们的大小增加。我们假设FI的非规范活性会影响细胞生理学,并导致肺癌患者的不良预后。
    While numerous membrane-bound complement inhibitors protect the body\'s cells from innate immunity\'s autoaggression, soluble inhibitors like complement factor I (FI) are rarely produced outside the liver. Previously, we reported the expression of FI in non-small cell lung cancer (NSCLC) cell lines. Now, we assessed the content of FI in cancer biopsies from lung cancer patients and associated the results with clinicopathological characteristics and clinical outcomes. Immunohistochemical staining intensity did not correlate with age, smoking status, tumor size, stage, differentiation grade, and T cell infiltrates, but was associated with progression-free survival (PFS), overall survival (OS) and disease-specific survival (DSS). Multivariate Cox analysis of low vs. high FI content revealed HR 0.55, 95 % CI 0.32-0.95, p=0.031 for PFS, HR 0.51, 95 % CI 0.25-1.02, p=0.055 for OS, and HR 0.32, 95 % CI 0.12-0.84, p=0.021 for DSS. Unfavorable prognosis might stem from the non-canonical role of FI, as the staining pattern did not correlate with C4d - the product of FI-supported degradation of active complement component C4b. To elucidate that, we engineered three human NSCLC cell lines naturally expressing FI with CRISPR/Cas9 technology, and compared the transcriptome of FI-deficient and FI-sufficient clones in each cell line. RNA sequencing revealed differentially expressed genes engaged in intracellular signaling pathways controlling proliferation, apoptosis, and responsiveness to growth factors. Moreover, in vitro colony-formation assays showed that FI-deficient cells formed smaller foci than FI-sufficient NSCLC cells, but their size increased when purified FI protein was added to the medium. We postulate that a non-canonical activity of FI influences cellular physiology and contributes to the poor prognosis of lung cancer 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
    因子I(FI)是补体系统的重要调节剂。加上共同因素,FI降低C3b,抑制补体的进一步激活。FI中的基因突变与病理状况相关,如年龄相关性黄斑变性和非典型溶血性尿毒症综合征。这里,我们评估了在患者中发现的8种重组FI遗传变异.我们在两种辅因子存在下评估了FI的辅因子活性;因子H和可溶性CR1。采用不同的分析测定;SDS-PAGE评估C3b的降解,使用新颖的基于Luminex珠的测定来测量流体相iC3b的产生和表面结合的C3b的降解的ELISA。我们证明,FIMAC和FI的SP域中的突变导致蛋白酶活性显着降低,而LDLRA2结构域中的两个分析突变并未导致FI功能的任何深刻变化。采用的不同测定法显示出强正相关,但是遗传变异Ile55Phe和Gly261Asp的活性差异只能通过结合不同的方法和辅因子来评估FI活性来观察。总之,我们的结果为评估FI突变影响的可用诊断工具提供了新的视角.
    Factor I (FI) is an essential regulator of the complement system. Together with co-factors, FI degrades C3b, which inhibits further complement activation. Genetic mutations in FI are associated with pathological conditions like age-related macular degeneration and atypical hemolytic uremic syndome. Here, we evaluated eight recombinant FI genetic variants found in patients. We assessed FI\'s co-factor activity in the presence of two co-factors; Factor H and soluble CR1. Different analytical assays were employed; SDS-PAGE to evaluate the degradation of C3b, ELISA to measure the generation of fluid phase iC3b and the degradation of surface-bound C3b using a novel Luminex bead-based assay. We demonstrate that mutations in the FIMAC and SP domains of FI led to significantly reduced protease activity, whereas the two analyzed mutations in the LDLRA2 domain did not result in any profound changes in FI\'s function. The different assays employed displayed a strong positive correlation, but differences in the activity of the genetic variants Ile55Phe and Gly261Asp could only be observed by combining different methods and co-factors for evaluating FI activity. In conclusion, our results provide a new perspective regarding available diagnostic tools for assessing the impact of mutations in FI.
    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
    一名34岁的日本男子视力模糊,头痛,恶心,贫血,血小板减少症,和严重的肾功能不全.最初怀疑血栓性微血管病是由恶性高血压引起的。降压药不能改善他的血小板减少症或肾功能不全,和其他引起血栓性微血管病的疾病被排除。因此,患者被诊断为非典型溶血性尿毒综合征。肾活检显示血栓性微血管病和C3肾小球病重叠。基因检测显示c.848A>G(p。Asp283Gly),编码补体因子I的基因中的错义杂合变体。用补体因子I突变重叠非典型溶血性尿毒综合征和C3肾小球病非常罕见,尤其是在日本。
    A 34-year-old Japanese man presented with blurred vision, headache, nausea, anemia, thrombocytopenia, and severe renal dysfunction. Thrombotic microangiopathy was initially suspected to have been caused by malignant hypertension. Antihypertensive medications did not improve his thrombocytopenia or renal dysfunction, and other diseases causing thrombotic microangiopathy were ruled out. Therefore, the patient was diagnosed with atypical hemolytic uremic syndrome. A renal biopsy revealed an overlap of thrombotic microangiopathy and C3 glomerulopathy. Genetic testing revealed c.848A>G (p.Asp283Gly), a missense heterozygous variant in the gene encoding complement factor I. Overlapping atypical hemolytic uremic syndrome and C3 glomerulopathy with complement factor I mutation is very rare, especially in Japan.
    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
    非典型溶血性尿毒综合征(aHUS)是一种以急性肾损伤(AKI)为特征的疾病,血小板减少症,和继发于补体途径失调的微血管病性溶血性贫血(MAHA)。已经确定了几种触发因素在遗传易感患者中引起aHUS;然而,嗜酸性粒细胞增多综合征(HES)引发的aHUS尚未报道。在这篇文章中,我们介绍了一例aHUS,表现为全身性荨麻疹皮疹和血管性水肿。初步调查显示嗜酸性粒细胞增多(最大绝对嗜酸性粒细胞计数为6840细胞/µL),骨髓分析正常;因此,诊断为特发性HES。住院期间,病人出现抽搐,studopore,和全面的血栓性微血管病(TMA),AKI需要临时血液透析。肾活检证实存在肾TMA。对aHUS相关基因的编码区进行下一代测序,揭示潜在的补体因子I(CFI)缺乏症,CFI基因的杂合变体p.P64L。患者成功接受了高剂量类固醇和延长血浆置换持续时间的治疗。
    Atypical hemolytic uremic syndrome (aHUS) is a condition characterized by acute kidney injury (AKI), thrombocytopenia, and microangiopathic hemolytic anemia secondary to complement pathway dysregulation. Several triggers have been identified as causing aHUS in genetically susceptible patients; however, hypereosinophilia syndrome (HES)-triggered aHUS has not been reported. In this article, we present a case of aHUS presented with generalized urticarial rashes and angioedema. The initial investigations revealed hypereosinophilia (maximal absolute eosinophil count of 6,840 cells/µL) with normal bone-marrow analyses; hence, idiopathic HES was diagnosed. During hospitalization, the patient developed convulsion, stuporous, and full-blown thrombotic microangiopathy (TMA), with AKI requiring temporary hemodialysis. A kidney biopsy confirmed the existence of renal TMA. Next-generation sequencing of the coding regions of aHUS-related genes was performed, revealing an underlying complement factor I (CFI) deficiency, a heterozygous variant p.P64L of CFI gene. The patient was successfully treated with high-dose steroids and extended duration of plasmapheresis.
    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
    补充因子I(FI)是经典,凝集素和替代途径补体调节。FI是一种88kDa血浆蛋白,以非活性构型循环,直到与其辅因子和底物形成三分子复合物,因此结构重组允许催化三联体裂解其底物,C3b和C4b。为了保持其作为主要补体调节酶的作用,缺乏与免疫病理学有关。在完全FI缺乏的情况下,消耗性C3缺乏导致包囊微生物的反复感染。无菌脑炎症和血管表现也较不常见。已证明因子I基因(CFI)的杂合突变在非典型溶血性尿毒症综合征中富集,尽管外显率很低.CFI的单倍功能不全也与视网膜厚度减少有关,并且是年龄相关性黄斑变性发展的强危险因素。使用血浆纯化或重组蛋白补充FI早已被假定,然而,技术困难阻碍了进入临床试验的进展。仅使用基因疗法,在地理萎缩的I/II期临床试验中,CFI补充已与GT005一起进入临床。
    Complement factor I (FI) is the nexus for classical, lectin and alternative pathway complement regulation. FI is an 88 kDa plasma protein that circulates in an inactive configuration until it forms a trimolecular complex with its cofactor and substrate whereupon a structural reorganization allows the catalytic triad to cleave its substrates, C3b and C4b. In keeping with its role as the master complement regulatory enzyme, deficiency has been linked to immunopathology. In the setting of complete FI deficiency, a consumptive C3 deficiency results in recurrent infections with encapsulated microorganisms. Aseptic cerebral inflammation and vasculitic presentations are also less commonly observed. Heterozygous mutations in the factor I gene (CFI) have been demonstrated to be enriched in atypical haemolytic uraemic syndrome, albeit with a very low penetrance. Haploinsufficiency of CFI has also been associated with decreased retinal thickness and is a strong risk factor for the development of age-related macular degeneration. Supplementation of FI using plasma purified or recombinant protein has long been postulated, however, technical difficulties prevented progression into clinical trials. It is only using gene therapy that CFI supplementation has reached the clinic with GT005 in phase I/II clinical trials for geographic atrophy.
    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
    非典型溶血性尿毒综合征(aHUS)是一种罕见的血栓性微血管病变。在约60%的患者中发现补体蛋白的遗传变异。其中,约15%携带补体因子I中的突变。因子I(FI)是一种多结构域丝氨酸蛋白酶,其在辅因子蛋白存在下切割并由此失活C3b和C4b。晶体结构表明FI具有两个钙结合域,LDLRA1和LDLRA2。然而,钙在FI中的作用尚不清楚。我们确定在aHUS中鉴定出9种遗传变异(N151S,G162D,G188A,V230E,A240G,G243R,C247G,A258T,和Q260D)聚集在LDLRA1的钙结合位点周围。使用定点诱变,我们确定除A258T外的所有合成都受到损害,暗示有缺陷的蛋白质折叠,也许是由于钙结合的损失。为了进一步探索这种可能性,我们产生了12个与LDLRA1和LDLRA2中的钙协调的丙氨酸突变体(K239A,D242A,I244A,D246A,D252A,E253A,Y276A,N279A,E281A,D283A,D289A,和D290A),预计会干扰钙结合。除K239A和Y276A外,没有一个突变体被分泌。这些观察结果表明,钙离子在FI中起着关键的结构和功能作用。
    Atypical hemolytic uremic syndrome (aHUS) is a rare thrombotic microangiopathy. Genetic variants in complement proteins are found in ≈60% of patients. Of these patients, ≈15% carry mutations in complement factor I (CFI). Factor I (FI) is a multidomain serine protease that cleaves and thereby inactivates C3b and C4b in the presence of cofactor proteins. Crystal structures have shown that FI possesses 2 calcium-binding domains, low-density lipoprotein receptor class A (LDLRA) 1 and LDLRA2. Yet, the role of calcium in FI is unknown. We determined that 9 genetic variants identified in aHUS (N151S, G162D, G188A, V230E, A240G, G243R, C247G, A258T, and Q260D) cluster around the calcium-binding site of LDLRA1. Using site-directed mutagenesis, we established that the synthesis of all, except A258T, was impaired, implying defective protein folding, perhaps due to loss of calcium binding. To further explore this possibility, we generated 12 alanine mutants that coordinate with the calcium in LDLRA1 and LDLRA2 (K239A, D242A, I244A, D246A, D252A, E253A, Y276A, N279A, E281A, D283A, D289A, and D290A) and are expected to perturb calcium binding. Except for K239A and Y276A, none of the mutants was secreted. These observations suggest that calcium ions play key structural and functional roles in FI.
    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
    补体因子I(CFI)基因中的突变先前已被鉴定为复发性CNS炎症的原因。我们介绍了一个26岁的男性患有18次复发性脑膜炎的病例,谁在CFI中有一个变体(c.859G>A,P.Gly287Arg)以前与神经系统表现无关。他用canakinumab缓解了,针对白细胞介素-1β的人单克隆抗体。
    Mutations in the complement factor I (CFI) gene have previously been identified as causes of recurrent CNS inflammation. We present a case of a 26-year-old man with 18 episodes of recurrent meningitis, who had a variant in CFI(c.859G>A,p.Gly287Arg) not previously associated with neurologic manifestations. He achieved remission with canakinumab, a human monoclonal antibody targeted at interleukin-1 beta.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号