Heme trafficking

  • 文章类型: Journal Article
    铁原卟啉IX(血红素)是一种氧化还原活性辅因子,在哺乳动物细胞中通过GAPDH结合,并通过受NO生理水平影响的过程分配。这会影响许多血红素蛋白的活性,包括吲哚胺双加氧酶-1(IDO1),一种参与免疫反应和肿瘤生长的氧化还原酶。为了获得进一步的理解,我们创建了四Cys人GAPDH报告构建体(TC-hGAPDH),其在标记后可以通过荧光猝灭指示其血红素结合。当在人细胞系中纯化或表达时,TC-hGAPDH具有天然GAPDH和血红素结合使其荧光猝灭45-65%的特性,允许它实时报告活细胞中线粒体生成血红素的GAPDH结合。在线粒体血红素合成活跃的细胞中,低水平的NO暴露会增加血红素对IDO1的分配,同时由于线粒体的补充而使TC-hGAPDH血红素水平保持恒定。当线粒体血红素合成被阻断时,在需要IDO1能够结合血红素并存在活性hsp90的过程中,低NO导致TC-hGAPDH中现有血红素几乎完全转移到IDO1。较高的NO暴露具有相反的作用,并导致IDO1血红素转移回TC-hGAPDH。这证明:(i)线粒体血红素通过GAPDH的流动与靶标递送紧密耦合,(ii)NO通过促进与GAPDH的保守血红素交换来上调或下调IDO1活性,该交换根据NO暴露水平朝任一方向发展。驱动浓度依赖性的能力,可逆蛋白血红素交换是前所未有的,揭示了NO在生物学中的新作用。
    Iron protoporphyrin IX (heme) is a redox-active cofactor that is bound in mammalian cells by GAPDH and allocated by a process influenced by physiologic levels of NO. This impacts the activity of many heme proteins including indoleamine dioxygenase-1 (IDO1), a redox enzyme involved in immune response and tumor growth. To gain further understanding we created a tetra-Cys human GAPDH reporter construct (TC-hGAPDH) which after labeling could indicate its heme binding by fluorescence quenching. When purified or expressed in a human cell line, TC-hGAPDH had properties like native GAPDH and heme binding quenched its fluorescence by 45-65%, allowing it to report on GAPDH binding of mitochondrially-generated heme in live cells in real time. In cells with active mitochondrial heme synthesis, low-level NO exposure increased heme allocation to IDO1 while keeping the TC-hGAPDH heme level constant due to replenishment by mitochondria. When mitochondrial heme synthesis was blocked, low NO caused a near complete transfer of the existing heme in TC-hGAPDH to IDO1 in a process that required IDO1 be able to bind the heme and have an active hsp90 present. Higher NO exposure had the opposite effect and caused IDO1 heme to transfer back to TC-hGAPDH. This demonstrated: (i) flow of mitochondrial heme through GAPDH is tightly coupled to target delivery, (ii) NO up- or down-regulates IDO1 activity by promoting a conserved heme exchange with GAPDH that goes in either direction according to the NO exposure level. The ability to drive a concentration-dependent, reversible protein heme exchange is unprecedented and reveals a new role for NO in biology.
    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
    细胞色素P4503A4和2D6(EC1.14.13.97和1.14.14;CYP3A4和2D6)是含血红素的酶,其催化大量异生物质和药物底物的氧化,因此广泛影响人类生物学和药物疗法。尽管它们的活动与血红素含量成正比,人们对使血红素成熟到功能形式的细胞血红素递送和插入过程知之甚少。基于我们先前的将这些蛋白质与细胞内血红素分配联系起来的研究,我们调查了GAPDH和伴侣Hsp90的潜在参与。我们研究了血红素在HEK293T和GlyACHO细胞中瞬时表达或在HEPG2细胞中自然表达以响应利福平后,血红素的递送和插入CYP3A4和2D6中。并研究了它们与细胞中GAPDH和Hsp90的关联。结果表明,GAPDH及其血红素结合功能参与线粒体生成的血红素向apo-CYP3A4和2D6的递送,并且细胞伴侣Hsp90还参与驱动其血红素插入。揭示细胞如何将血红素分配给CYP3A4和2D6,为它们的成熟过程以及这如何有助于调节它们在健康和疾病中的功能提供了新的见解。
    Cytochrome P450 3A4 and 2D6 (EC 1.14.13.97 and 1.14.14.1; CYP3A4 and 2D6) are heme-containing enzymes that catalyze the oxidation of a wide number of xenobiotic and drug substrates and thus broadly impact human biology and pharmacologic therapies. Although their activities are directly proportional to their heme contents, little is known about the cellular heme delivery and insertion processes that enable their maturation to functional form. We investigated the potential involvement of GAPDH and chaperone Hsp90, based on our previous studies linking these proteins to intracellular heme allocation. We studied heme delivery and insertion into CYP3A4 and 2D6 after they were transiently expressed in HEK293T and GlyA CHO cells or when naturally expressed in HEPG2 cells in response to rifampicin, and also investigated their associations with GAPDH and Hsp90 in cells. The results indicate that GAPDH and its heme binding function is involved in delivery of mitochondria-generated heme to apo-CYP3A4 and 2D6, and that cell chaperone Hsp90 is additionally involved in driving their heme insertions. Uncovering how cells allocate heme to CYP3A4 and 2D6 provides new insight on their maturation processes and how this may help to regulate their functions in health and disease.
    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
    Heme,几乎所有生物体的必需分子,主要作为大量蛋白质的辅因子。然而,血红素是如何从合成位点转移到血红蛋白组装的位置的,在细胞中仍然是未知的,尤其是细菌。在这项研究中,以Shewanellaoneidensis为模型,我们将HtpA(SO0126)鉴定为真核生物中发现的血红素运输蛋白和TANGO2蛋白的同源物。我们表明,HtpA同源物广泛分布在生物体的所有域中,并经历了平行进化。在缺席的情况下,细胞色素(cyt)c含量和过氧化氢酶活性显著下降。我们进一步表明HtpA和代表性TANGO2蛋白都以1:1的化学计量和相对低的解离常数结合血红素。蛋白质相互作用分析证实HtpA直接与细胞色素c成熟系统相互作用。我们的发现揭示了血红素在细菌中的跨膜运输,并扩展了对TANGO2蛋白的理解。重要性血红素的细胞内运输,血液蛋白的重要辅助因子,即使在真核生物中,更别说细菌了.在这里,我们开发了一种高通量方法,真核细胞TANGO2蛋白的同源物,被鉴定为血红素结合蛋白,可增强细胞色素c的生物合成和过氧化氢酶活性。HtpA直接与细胞色素c生物合成系统相互作用,支持这种蛋白质,像TANGO2一样,在细胞内血红素运输中起作用。HtpA同源物分布广泛,但其中绝大多数被发现是不可交换的,可能是平行进化的结果。通过证实HtpA及其真核同源物的血红素运输性质,我们的发现提供了对血红素运输过程的一般见解,并强调了所有生物体在进化过程中的功能保护。
    Heme, an essential molecule for virtually all living organisms, acts primarily as a cofactor in a large number of proteins. However, how heme is mobilized from the site of synthesis to the locations where hemoproteins are assembled remains largely unknown in cells, especially bacterial ones. In this study, with Shewanella oneidensis as the model, we identified HtpA (SO0126) as a heme-trafficking protein and homolog of TANGO2 proteins found in eukaryotes. We showed that HtpA homologs are widely distributed in all domains of living organisms and have undergone parallel evolution. In its absence, the cytochrome (cyt) c content and catalase activity decreased significantly. We further showed that both HtpA and representative TANGO2 proteins bind heme with 1:1 stoichiometry and a relatively low dissociation constant. Protein interaction analyses substantiated that HtpA directly interacts with the cytochrome c maturation system. Our findings shed light on cross-membrane transport of heme in bacteria and extend the understanding of TANGO2 proteins. IMPORTANCE The intracellular trafficking of heme, an essential cofactor for hemoproteins, remains underexplored even in eukaryotes, let alone bacteria. Here we developed a high-throughput method by which HtpA, a homolog of eukaryotic TANGO2 proteins, was identified to be a heme-binding protein that enhances cytochrome c biosynthesis and catalase activity in Shewanella oneidensis. HtpA interacts with the cytochrome c biosynthesis system directly, supporting that this protein, like TANGO2, functions in intracellular heme trafficking. HtpA homologs are widely distributed, but a large majority of them were found to be non-exchangeable, likely a result of parallel evolution. By substantiating the heme-trafficking nature of HtpA and its eukaryotic homologs, our findings provide general insight into the heme-trafficking process and highlight the functional conservation along evolution in all living organisms.
    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
    吲哚胺-2,3-双加氧酶(IDO1)和色氨酸-2,3-双加氧酶(TDO)催化L-色氨酸转化为N-甲酰-犬尿氨酸,因此在代谢中起主要作用,炎症,和肿瘤免疫监视。因为它们的活动取决于血红素含量,它们在生物学环境中有所不同,并以动态的方式上升或下降,我们研究了哺乳动物细胞中血红素水平如何受到一氧化氮(NO)的影响。我们利用天然或通过转染表达TDO或IDO1的细胞,并确定其活性,血红素含量,和表达水平作为NO暴露的函数。我们发现NO具有双峰效应:狭窄范围的低NO暴露促进细胞将血红素分配到无血红素的TDO和IDO1群体中,从而提高其血红素含量和活性4至6倍,尽管超出此范围,但NO暴露会对其血红素含量和活性产生负面影响。NO没有改变双加氧酶蛋白表达水平,当NO由化学供体释放或由免疫刺激的巨噬细胞自然产生时,观察到其双峰影响。NO驱动的血红素分配给IDO1和TDO需要GAPDH-血红素复合物的参与,而IDO1则需要伴侣Hsp90活性。因此,细胞可以通过NO对血红素分配的双峰控制来上调或下调其IDO1和TDO活性。这种机制具有重要的生物医学意义,并有助于解释为什么动物中的IDO1和TDO活性随着免疫刺激而上升和下降。
    Indoleamine-2, 3-dioxygenase (IDO1) and Tryptophan-2, 3-dioxygenase (TDO) catalyze the conversion of L-tryptophan to N-formyl-kynurenine and thus play primary roles in metabolism, inflammation, and tumor immune surveillance. Because their activities depend on their heme contents, which vary in biological settings and go up or down in a dynamic manner, we studied how their heme levels may be impacted by nitric oxide (NO) in mammalian cells. We utilized cells expressing TDO or IDO1 either naturally or via transfection and determined their activities, heme contents, and expression levels as a function of NO exposure. We found NO has a bimodal effect: a narrow range of low NO exposure promoted cells to allocate heme into the heme-free TDO and IDO1 populations and consequently boosted their heme contents and activities 4- to 6-fold, while beyond this range the NO exposure transitioned to have a negative impact on their heme contents and activities. NO did not alter dioxygenase protein expression levels, and its bimodal impact was observed when NO was released by a chemical donor or was generated naturally by immune-stimulated macrophage cells. NO-driven heme allocations to IDO1 and TDO required participation of a GAPDH-heme complex and for IDO1 required chaperone Hsp90 activity. Thus, cells can up- or downregulate their IDO1 and TDO activities through a bimodal control of heme allocation by NO. This mechanism has important biomedical implications and helps explain why the IDO1 and TDO activities in animals go up and down in response to immune stimulation.
    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
    血红素b(铁原卟啉IX)作为金属因子和信号分子在生物学中起着重要作用。然而,血红素信号的目标和介导血红素从合成或摄取位点到血红素依赖性或受调节蛋白的交换的蛋白质网络知之甚少。在这里,我们描述了一种基于质谱的定量化学蛋白质组学策略,用于鉴定人胚肾HEK293细胞中可能与血红素信号传导和运输相关的交换不稳定血液蛋白.该策略涉及用血红素生物合成抑制剂琥珀酰丙酮(SA)消耗内源性血红素,使推定的血红素结合蛋白处于apo状态,然后使用血红素琼脂糖树脂捕获这些蛋白质,最后洗脱并通过质谱鉴定。通过仅鉴定相对于对照珠状琼脂糖以SA依赖性方式与血红素琼脂糖高特异性相互作用的蛋白质,我们已经扩大了可能参与结合和缓冲不稳定血红素的蛋白质和本体的数量,或者是血红素信号的靶标。值得注意的是,这些包括参与染色质重塑的蛋白质,DNA损伤反应,RNA剪接,细胞骨架组织和囊泡运输,通过最近发表的免费研究,其中许多与血红素有关。一起来看,这些结果为血红素在从基因组完整性到蛋白质运输等一系列扩展的细胞过程中的新兴作用提供了支持。
    Heme b (iron protoporphyrin IX) plays important roles in biology as a metallocofactor and signaling molecule. However, the targets of heme signaling and the network of proteins that mediate the exchange of heme from sites of synthesis or uptake to heme dependent or regulated proteins are poorly understood. Herein, we describe a quantitative mass spectrometry (MS)-based chemoproteomics strategy to identify exchange labile hemoproteins in human embryonic kidney HEK293 cells that may be relevant to heme signaling and trafficking. The strategy involves depleting endogenous heme with the heme biosynthetic inhibitor succinylacetone (SA), leaving putative heme-binding proteins in their apo-state, followed by the capture of those proteins using hemin-agarose resin, and finally elution and identification by MS. By identifying only those proteins that interact with high specificity to hemin-agarose relative to control beaded agarose in an SA-dependent manner, we have expanded the number of proteins and ontologies that may be involved in binding and buffering labile heme or are targets of heme signaling. Notably, these include proteins involved in chromatin remodeling, DNA damage response, RNA splicing, cytoskeletal organization, and vesicular trafficking, many of which have been associated with heme through complementary studies published recently. Taken together, these results provide support for the emerging role of heme in an expanded set of cellular processes from genome integrity to protein trafficking and beyond.
    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
    血红素调节基序(HRMs)存在于多种具有不同生物学功能的蛋白质中。在血红素加氧酶-2(HO2)中,血红素与HRM结合,并容易转移到蛋白质核心的催化位点。为了进一步定义这种血红素转移机制,我们评估了GAPDH的能力,一个已知的血红素伴侣,将血红素转移到HO2的HRM和/或催化核心。我们的结果表明GAPDH和HO2在体外形成复合物。我们已经通过在具有HO2报告构建体的HEK293细胞中的荧光猝灭来跟踪两个位点处的血红素插入。在我们的报告构建体中每个位点血红素结合所必需的残基突变后,我们发现HO2结合血红素的核心和活细胞中的HRM,血红素向HO2的递送取决于GAPDH的存在,GAPDH能够结合血红素。总之,GAPDH参与血红素向HO2的输送,但是,令人惊讶的是,不在HO2的特定地点。因此,我们的结果强调了血红素与核心和HRM结合以及HO2通过GAPDH与血红素池相互作用以维持细胞血红素稳态的重要性。
    Heme regulatory motifs (HRMs) are found in a variety of proteins with diverse biological functions. In heme oxygenase-2 (HO2), heme binds to the HRMs and is readily transferred to the catalytic site in the core of the protein. To further define this heme transfer mechanism, we evaluated the ability of GAPDH, a known heme chaperone, to transfer heme to the HRMs and/or the catalytic core of HO2. Our results indicate GAPDH and HO2 form a complex in vitro. We have followed heme insertion at both sites by fluorescence quenching in HEK293 cells with HO2 reporter constructs. Upon mutation of residues essential for heme binding at each site in our reporter construct, we found that HO2 binds heme at the core and the HRMs in live cells and that heme delivery to HO2 is dependent on the presence of GAPDH that is competent for heme binding. In sum, GAPDH is involved in heme delivery to HO2 but, surprisingly, not to a specific site on HO2. Our results thus emphasize the importance of heme binding to both the core and the HRMs and the interplay of HO2 with the heme pool via GAPDH to maintain cellular heme homeostasis.
    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
    生物学中的一氧化氮(NO)信号传导依赖于其通过NO受体可溶性鸟苷酸环化酶(sGC)激活环磷酸鸟苷(cGMP)的产生。sGC必须获得血红素并形成异二聚体才能起作用,但自相矛盾的是,在细胞和组织中通常以不成熟的无血红素形式存在。根据我们之前的发现,NO可以驱动sGC成熟,我们通过利用荧光sGC构建体研究了其基础,该构建体的血红素水平可以在活细胞中监测。我们发现,在生理水平产生的NO会迅速触发细胞将血红素动员到未成熟的sGC。当NO在细胞内或相邻细胞产生时,就会发生这种情况。在没有暴露的几秒钟内开始,并引导细胞构建sGC异二聚体,从而将其活性sGC水平提高几倍。NO触发的血红素部署涉及细胞甘油醛-3-磷酸脱氢酶(GAPDH)-血红素复合物,需要伴侣hsp90,并且新形成的sGC异二聚体在NO产生停止后很长时间仍具有功能。我们得出的结论是,生理水平的NO通过引起细胞血红素的快速部署来触发其自身受体的组装。响应于NO重定向细胞血红素是细胞和组织调节其cGMP信号传导和更一般地调节其血红素蛋白活性的一种方式,无论NO生物合成发生在哪里。
    Nitric oxide (NO) signaling in biology relies on its activating cyclic guanosine monophosphate (cGMP) production by the NO receptor soluble guanylyl cyclase (sGC). sGC must obtain heme and form a heterodimer to become functional, but paradoxically often exists as an immature heme-free form in cells and tissues. Based on our previous finding that NO can drive sGC maturation, we investigated its basis by utilizing a fluorescent sGC construct whose heme level can be monitored in living cells. We found that NO generated at physiologic levels quickly triggered cells to mobilize heme to immature sGC. This occurred when NO was generated within cells or by neighboring cells, began within seconds of NO exposure, and led cells to construct sGC heterodimers and thus increase their active sGC level by several-fold. The NO-triggered heme deployment involved cellular glyceraldehyde-3-phosphate dehydrogenase (GAPDH)-heme complexes and required the chaperone hsp90, and the newly formed sGC heterodimers remained functional long after NO generation had ceased. We conclude that NO at physiologic levels triggers assembly of its own receptor by causing a rapid deployment of cellular heme. Redirecting cellular heme in response to NO is a way for cells and tissues to modulate their cGMP signaling and to more generally tune their hemeprotein activities wherever NO biosynthesis takes place.
    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
    血红素加氧酶(HO)通过将血红素氧化降解为一氧化碳来解毒,铁,和胆绿素,被还原成胆红素排出体外.人类表达HO的两种同工型:诱导型HO-1,其响应于过量血红素和其他应激源而上调,和组成型HO-2。对HO-1的调节和生理功能了解很多,而对HO-2在调节血红素稳态中的作用知之甚少。表达组成型HO-2的生化必要性取决于血红素在HO-1不被诱导的条件下是否足够丰富和可作为底物。通过测量不稳定的血红素,总血红素,和胆红素在人胚肾HEK293细胞沉默或过表达的HO-2,以及各种HO-2突变等位基因,我们发现内源性血红素太限制了底物,无法观察到HO-2依赖性血红素降解。相反,我们发现了HO-2在血红素的结合和缓冲中的新作用。一起来看,在没有过量血红素的情况下,我们建议HO-2通过充当控制血红素生物利用度的血红素缓冲因子来调节血红素稳态。当血红素过量时,HO-1被诱导,HO-2和HO-1均可通过酶促降解保护血红素毒性。我们的结果解释了为什么HO-2的无催化活性突变体对氧化应激具有细胞保护作用。此外,HO-2过度表达导致的生物可利用血红素的变化,选择性结合三价铁血红素,与不稳定血红素被氧化一致,从而为血红素贩运和信号传递提供了新的见解。
    Heme oxygenases (HOs) detoxify heme by oxidatively degrading it into carbon monoxide, iron, and biliverdin, which is reduced to bilirubin and excreted. Humans express two isoforms of HO: the inducible HO-1, which is upregulated in response to excess heme and other stressors, and the constitutive HO-2. Much is known about the regulation and physiological function of HO-1, whereas comparatively little is known about the role of HO-2 in regulating heme homeostasis. The biochemical necessity for expressing constitutive HO-2 is dependent on whether heme is sufficiently abundant and accessible as a substrate under conditions in which HO-1 is not induced. By measuring labile heme, total heme, and bilirubin in human embryonic kidney HEK293 cells with silenced or overexpressed HO-2, as well as various HO-2 mutant alleles, we found that endogenous heme is too limiting a substrate to observe HO-2-dependent heme degradation. Rather, we discovered a novel role for HO-2 in the binding and buffering of heme. Taken together, in the absence of excess heme, we propose that HO-2 regulates heme homeostasis by acting as a heme buffering factor that controls heme bioavailability. When heme is in excess, HO-1 is induced, and both HO-2 and HO-1 can provide protection from heme toxicity via enzymatic degradation. Our results explain why catalytically inactive mutants of HO-2 are cytoprotective against oxidative stress. Moreover, the change in bioavailable heme due to HO-2 overexpression, which selectively binds ferric over ferrous heme, is consistent with labile heme being oxidized, thereby providing new insights into heme trafficking and signaling.
    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
    Aerobic respiration is a key energy-producing pathway in many prokaryotes and virtually all eukaryotes. The final step of aerobic respiration is most commonly catalyzed by heme-copper oxidases embedded in the cytoplasmic or mitochondrial membrane. The majority of these terminal oxidases contain a prenylated heme (typically heme a or occasionally heme o) in the active site. In addition, many heme-copper oxidases, including mitochondrial cytochrome c oxidases, possess a second heme a cofactor. Despite the critical role of heme a in the electron transport chain, the details of the mechanism by which heme b, the prototypical cellular heme, is converted to heme o and then to heme a remain poorly understood. Recent structural investigations, however, have helped clarify some elements of heme a biosynthesis. In this review, we discuss the insight gained from these advances. In particular, we present a new structural model of heme o synthase (HOS) based on distance restraints from inferred coevolutionary relationships and refined by molecular dynamics simulations that are in good agreement with the experimentally determined structures of HOS homologs. We also analyze the two structures of heme a synthase (HAS) that have recently been solved by other groups. For both HOS and HAS, we discuss the proposed catalytic mechanisms and highlight how new insights into the heme-binding site locations shed light on previously obtained biochemical data. Finally, we explore the implications of the new structural data in the broader context of heme trafficking in the heme a biosynthetic pathway and heme-copper oxidase assembly.
    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
    可溶性鸟苷酸环化酶(sGC)是哺乳动物NO-cGMP信号传导的关键组分。尽管血红素必须结合在sGCβ1亚基(sGCβ)中才能使sGC起作用,血红素如何传递给sGCβ仍然未知。鉴于GAPDH显示血红素伴侣的诱导型NO合酶的特性,在这里,我们调查血红素传递给apo-sGCβ是否涉及GAPDH。我们利用了sGCβ报告基因构建体,四半胱氨酸sGCβ,其血红素插入可以在活细胞中进行荧光猝灭,评估降低细胞GAPDH表达如何影响血红素传递,并检查表达WTGAPDH或血红素结合缺陷的GAPDH变体是否恢复血红素递送。我们还使用纯化的蛋白质研究了细胞中GAPDH和sGCβ之间的相互作用及其复合物形成和潜在的血红素转移。我们发现血红素传递给apo-sGCβ与细胞GAPDH表达水平相关,并取决于GAPDH结合细胞内血红素的能力,apo-sGCβ与细胞中的GAPDH结合,当血红素结合sGCβ时解离,并且纯化的GAPDH-血红素复合物与apo-sGCβ结合并将其血红素转移到sGCβ。根据这些结果,我们提出了一个模型,其中GAPDH获得线粒体血红素,然后与apo-sGCβ形成复合物,以完成血红素向sGCβ的递送。我们的发现阐明了sGC成熟的关键步骤,并揭示了调节其在健康和疾病中的活性的另一种机制。
    Soluble guanylyl cyclase (sGC) is a key component of NO-cGMP signaling in mammals. Although heme must bind in the sGC β1 subunit (sGCβ) for sGC to function, how heme is delivered to sGCβ remains unknown. Given that GAPDH displays properties of a heme chaperone for inducible NO synthase, here we investigated whether heme delivery to apo-sGCβ involves GAPDH. We utilized an sGCβ reporter construct, tetra-Cys sGCβ, whose heme insertion can be followed by fluorescence quenching in live cells, assessed how lowering cell GAPDH expression impacts heme delivery, and examined whether expressing WT GAPDH or a GAPDH variant defective in heme binding recovers heme delivery. We also studied interaction between GAPDH and sGCβ in cells and their complex formation and potential heme transfer using purified proteins. We found that heme delivery to apo-sGCβ correlates with cellular GAPDH expression levels and depends on the ability of GAPDH to bind intracellular heme, that apo-sGCβ associates with GAPDH in cells and dissociates when heme binds sGCβ, and that the purified GAPDH-heme complex binds to apo-sGCβ and transfers its heme to sGCβ. On the basis of these results, we propose a model where GAPDH obtains mitochondrial heme and then forms a complex with apo-sGCβ to accomplish heme delivery to sGCβ. Our findings illuminate a critical step in sGC maturation and uncover an additional mechanism that regulates its activity in health and disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号