intracellular parasitism

细胞内寄生
  • 文章类型: Journal Article
    新生隐球菌是兼性细胞内真菌病原体。与年轻的子细胞相比,十代大(10GEN)的新型念珠菌细胞对巨噬细胞的吞噬作用和杀伤作用更强。然而,介导这种抗性和细胞内寄生的机制知之甚少。这里,我们确定了10GEN新型梭菌细胞内存活的重要因素,如脲酶活性,胶囊合成,和DNA含量使用流式细胞术和荧光显微镜技术。应用延时成像的实时可视化来确定吞噬体酸度,膜渗透性,和吞噬不同世代新生梭菌的J774巨噬细胞的呕吐(非裂解性胞吐)率。我们的结果表明,老年新生C。表现出更高的脲酶活性和增强的高尔基体活性。此外,老C.新生者更有可能在G2阶段被捕,导致偶尔形成异常三聚体样细胞。最后,酵母细胞的晚期年龄略微减少宿主细胞内的呕吐症事件,这可能与吞噬溶酶体pH和膜通透性增加有关。总之,我们的研究结果表明,在酸性吞噬溶酶体中,古老的新型C.这些策略可能被老的新生梭菌用于抵抗吞噬体杀死并驱动隐球菌病的发病机理。对这些必需的宿主-病原体相互作用的理解可以进一步阐明为新型抗真菌治疗设计带来新见解的机制。
    Cryptococcus neoformans is a facultative intracellular fungal pathogen. Ten-generation-old (10GEN) C. neoformans cells are more resistant to phagocytosis and killing by macrophages than younger daughter cells. However, mechanisms that mediate this resistance and intracellular parasitism are poorly understood. Here, we identified important factors for the intracellular survival of 10GEN C. neoformans, such as urease activity, capsule synthesis, and DNA content using flow cytometry and fluorescent microscopy techniques. The real-time visualization of time-lapse imaging was applied to determine the phagosomal acidity, membrane permeability, and vomocytosis (non-lytic exocytosis) rate in J774 macrophages that phagocytosed C. neoformans of different generational ages. Our results showed that old C. neoformans exhibited higher urease activity and enhanced Golgi activity. In addition, old C. neoformans were more likely to be arrested in the G2 phase, resulting in the occasional formation of aberrant trimera-like cells. To finish, the advanced generational age of the yeast cells slightly reduced vomocytosis events within host cells, which might be associated with increased phagolysosome pH and membrane permeability. Altogether, our results suggest that old C. neoformans prevail within acidic phagolysosomes and can manipulate the phagosome pH. These strategies may be used by old C. neoformans to resist phagosomal killing and drive cryptococcosis pathogenesis. The comprehension of these essential host-pathogen interactions could further shed light on mechanisms that bring new insights for novel antifungal therapeutic design.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    单核吞噬细胞促进专性细胞内寄生虫弓形虫的传播。这里,我们报道了一组来自致密颗粒细胞器(GRA)的分泌的寄生虫效应蛋白如何协调树突状细胞样趋化和促炎激活被寄生的巨噬细胞.这些效应使得能够有效传播II型弓形虫谱系,人类中非常普遍的基因型。我们确定了效应子GRA15和GRA24通过作用于NF-κB和p38MAPK信号通路促进CCR7介导的巨噬细胞趋化的新功能,分别,GRA16/18的贡献和效应TEEGR的反调节。Further,GRA28增强了原代巨噬细胞中Ccr7基因位点的染色质可及性和GRA15/24/NF-κB依赖性转录。在体内,过继转移的感染野生型弓形虫的巨噬细胞在迁移到小鼠的次级器官中胜过感染GRA15/24双突变体的巨噬细胞。数据显示弓形虫,而不是被动地穿梭,通过协同多态GRA效应物在寄生的人和鼠吞噬细胞中诱导精细调节的前迁移状态,积极促进其传播。
    细胞内病原体可以利用被感染宿主细胞的细胞功能,例如,细胞内生存和传播。然而,微生物如何协调复杂细胞过程的劫持,如宿主细胞迁移,仍然知之甚少。因此,常见的寄生虫弓形虫积极侵入人类和其他脊椎动物的免疫细胞并改变其迁移特性。这里,我们表明,来自寄生虫的许多分泌效应蛋白的协同作用,主要是GRA15和GRA24,作用于宿主细胞信号通路以激活趋化性。Further,蛋白质效应GRA28选择性地作用于宿主细胞核中的染色质可接近性,以选择性地增强宿主基因表达。效应子的联合活动最终导致受感染的吞噬细胞内的促迁移信号传导。我们提供了一个分子框架,描述弓形虫如何协调复杂的生物学表型,例如吞噬细胞的迁移激活以促进传播。
    Mononuclear phagocytes facilitate the dissemination of the obligate intracellular parasite Toxoplasma gondii. Here, we report how a set of secreted parasite effector proteins from dense granule organelles (GRA) orchestrates dendritic cell-like chemotactic and pro-inflammatory activation of parasitized macrophages. These effects enabled efficient dissemination of the type II T. gondii lineage, a highly prevalent genotype in humans. We identify novel functions for effectors GRA15 and GRA24 in promoting CCR7-mediated macrophage chemotaxis by acting on NF-κB and p38 MAPK signaling pathways, respectively, with contributions of GRA16/18 and counter-regulation by effector TEEGR. Further, GRA28 boosted chromatin accessibility and GRA15/24/NF-κB-dependent transcription at the Ccr7 gene locus in primary macrophages. In vivo, adoptively transferred macrophages infected with wild-type T. gondii outcompeted macrophages infected with a GRA15/24 double mutant in migrating to secondary organs in mice. The data show that T. gondii, rather than being passively shuttled, actively promotes its dissemination by inducing a finely regulated pro-migratory state in parasitized human and murine phagocytes via co-operating polymorphic GRA effectors.
    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
    李斯特菌溶血素O(LLO)介导的快速吞噬体逃逸是单核细胞增生李斯特菌细胞内复制和发病的先决条件。在从对早期内体Rab5GTP酶呈阴性且对晚期内体Rab7呈阳性的空泡内化后几分钟内发生逃逸。使用突变分析,我们发现单核细胞增生李斯特菌的最佳细胞内增殖需要李斯特菌侵入素InlB。从这个观察开始,我们在HeLa细胞中确定InlB促进含李斯特菌液泡(LCV)的早期吞噬体逃逸和有效的Rab7获取。向LCV募集III类磷酸肌醇3激酶(PI3K)Vps34并积累其脂质产物,磷脂酰肌醇3-磷酸酯(PI3P),两个关键的内体成熟介质,也依赖于InlB。小干扰RNA(siRNA)敲低实验表明,Rab7募集和早期(LLO介导的)逃逸需要Vps34,并支持InlB依赖性细胞内增殖。一起,我们的数据表明,InlB通过破坏III类PI3K/Vps34信号传导加速LCV转化为逃逸有利的Rab7晚期吞噬体。我们的发现揭示了InlBinvasin在李斯特菌发病机制中作为细胞内促进增殖的毒力因子的新功能。重要性通过操纵内体区室避免溶酶体杀死是一种被认为主要限于针内细胞内病原体的毒力机制。我们的发现很重要,因为它们表明细胞溶质病原体,如单核细胞增生李斯特菌,内化后迅速逃离吞噬体,作为生存策略的一部分,也可以广泛颠覆内吞贩运。他们还澄清说,而不是延迟吞噬体成熟(为LLO依赖性破坏留出时间,正如目前所认为的),通过InlBL.单核细胞增多性细胞似乎有助于吞噬液泡快速转化为有利于逃逸的晚期吞噬体。我们的数据强调了细菌毒力因子的多功能性。在细胞表面,InlBinvasin通过I类PI3K激活诱导受体介导的吞噬作用,而在内化后,它利用III类PI3K(Vsp34)来促进细胞内存活。系统地阐明李斯特菌在整个胞吞途径中干扰PI3K信号传导的机制可能会导致新的抗感染疗法。
    Rapid phagosomal escape mediated by listeriolysin O (LLO) is a prerequisite for Listeria monocytogenes intracellular replication and pathogenesis. Escape takes place within minutes after internalization from vacuoles that are negative to the early endosomal Rab5 GTPase and positive to the late endosomal Rab7. Using mutant analysis, we found that the listerial invasin InlB was required for optimal intracellular proliferation of L. monocytogenes. Starting from this observation, we determined in HeLa cells that InlB promotes early phagosomal escape and efficient Rab7 acquisition by the Listeria-containing vacuole (LCV). Recruitment of the class III phosphoinositide 3-kinase (PI3K) Vps34 to the LCV and accumulation of its lipid product, phosphatidylinositol 3-phosphate (PI3P), two key endosomal maturation mediators, were also dependent on InlB. Small interfering RNA (siRNA) knockdown experiments showed that Vps34 was required for Rab7 recruitment and early (LLO-mediated) escape and supported InlB-dependent intracellular proliferation. Together, our data indicate that InlB accelerates LCV conversion into an escape-favorable Rab7 late phagosome via subversion of class III PI3K/Vps34 signaling. Our findings uncover a new function for the InlB invasin in Listeria pathogenesis as an intracellular proliferation-promoting virulence factor. IMPORTANCE Avoidance of lysosomal killing by manipulation of the endosomal compartment is a virulence mechanism assumed to be largely restricted to intravacuolar intracellular pathogens. Our findings are important because they show that cytosolic pathogens like L. monocytogenes, which rapidly escape the phagosome after internalization, can also extensively subvert endocytic trafficking as part of their survival strategy. They also clarify that, instead of delaying phagosome maturation (to allow time for LLO-dependent disruption, as currently thought), via InlB L. monocytogenes appears to facilitate the rapid conversion of the phagocytic vacuole into an escape-conducive late phagosome. Our data highlight the multifunctionality of bacterial virulence factors. At the cell surface, the InlB invasin induces receptor-mediated phagocytosis via class I PI3K activation, whereas after internalization it exploits class III PI3K (Vsp34) to promote intracellular survival. Systematically elucidating the mechanisms by which Listeria interferes with PI3K signaling all along the endocytic pathway may lead to novel anti-infective therapies.
    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
    病原体检测后,巨噬细胞通常在组织中保持固着状态,而树突状细胞(DC)迁移到次级淋巴组织。专性细胞内原生动物弓形虫通过不清楚的机制利用单核吞噬细胞的运输进行传播。我们报告说,弓形虫感染后,巨噬细胞启动通常归因于DC的转录因子的表达,通过趋化反应上调CCR7表达,并在过继转移到小鼠体内时进行系统迁移。我们表明,通过MYR1分泌途径释放的寄生虫效应物GRA28,与宿主细胞核中的宿主染色质重塑剂合作,以驱动被寄生的巨噬细胞的趋化迁移。在体内挑战研究中,感染野生型弓形虫的骨髓源性巨噬细胞在迁移和到达次级器官方面优于MYR1或GRA28缺陷菌株攻击的巨噬细胞.这项工作揭示了细胞内寄生虫如何劫持吞噬细胞中的趋化性,并强调了单核吞噬细胞系统分化细胞中显着的迁移可塑性。
    Upon pathogen detection, macrophages normally stay sessile in tissues while dendritic cells (DCs) migrate to secondary lymphoid tissues. The obligate intracellular protozoan Toxoplasma gondii exploits the trafficking of mononuclear phagocytes for dissemination via unclear mechanisms. We report that, upon T. gondii infection, macrophages initiate the expression of transcription factors normally attributed to DCs, upregulate CCR7 expression with a chemotactic response, and perform systemic migration when adoptively transferred into mice. We show that parasite effector GRA28, released by the MYR1 secretory pathway, cooperates with host chromatin remodelers in the host cell nucleus to drive the chemotactic migration of parasitized macrophages. During in vivo challenge studies, bone marrow-derived macrophages infected with wild-type T. gondii outcompeted those challenged with MYR1- or GRA28-deficient strains in migrating and reaching secondary organs. This work reveals how an intracellular parasite hijacks chemotaxis in phagocytes and highlights a remarkable migratory plasticity in differentiated cells of the mononuclear phagocyte system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    组织胞浆菌病是由热双态真菌荚膜组织胞浆菌病引起的全身性真菌病。虽然健康个体可以发展组织胞浆菌病,这种疾病在免疫功能低下的患者中特别危及生命,根据感染菌株的接种物和毒力,具有广泛的临床表现。在这次审查中,我们讨论了已建立的毒力因子和致病特征,使荚膜H.包括哺乳动物。理解和整合这些机制是制定新的预防和治疗干预措施的关键一步。
    Histoplasmosis is a systemic mycosis caused by the thermally dimorphic fungus Histoplasma capsulatum. Although healthy individuals can develop histoplasmosis, the disease is particularly life-threatening in immunocompromised patients, with a wide range of clinical manifestations depending on the inoculum and virulence of the infecting strain. In this review, we discuss the established virulence factors and pathogenesis traits that make H. capsulatum highly adapted to a wide variety of hosts, including mammals. Understanding and integrating these mechanisms is a key step toward devising new preventative and therapeutic interventions.
    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
    溶酶体中病原体的吞噬和破坏构成先天免疫防御的核心要素。这里,我们展示了布鲁氏菌,布鲁氏菌病的病原体,全球最普遍的细菌人畜共患病,通过激活编码BLOS1的Bloc1s1mRNA的受调节的IRE1α依赖性衰变(RIDD)来破坏这种免疫防御途径,BLOS1是一种促进内体-溶酶体融合的蛋白质。RIDD缺陷型细胞和携带IRE1αRIDD无能力变体的小鼠对感染具有抗性。Bloc1s1基因的失活损害了组装BLOC-1相关复合物(BORC)的能力,导致BORC相关溶酶体贩运成分的不同招募,含有布鲁氏菌的液泡(BCV)的核周运输,和增强对感染的易感性。抗RIDD的Bloc1s1变体保持了BORC的完整性和促进离心溶酶体运输的BORC相关成分的更高水平的关联,导致BCV外周运输和溶酶体破坏增强,和抵抗感染。这些发现表明,BLOS1上的宿主RIDD活性通过破坏BORC指导的溶酶体运输来调节布鲁氏菌细胞内寄生。值得注意的是,冠状病毒鼠肝炎病毒也破坏了RIDD-BLOS1轴以促进细胞内复制。我们的工作将BLOS1确立为一种新型的免疫防御因子,其活性被多种病原体劫持。
    The phagocytosis and destruction of pathogens in lysosomes constitute central elements of innate immune defense. Here, we show that Brucella, the causative agent of brucellosis, the most prevalent bacterial zoonosis globally, subverts this immune defense pathway by activating regulated IRE1α-dependent decay (RIDD) of Bloc1s1 mRNA encoding BLOS1, a protein that promotes endosome-lysosome fusion. RIDD-deficient cells and mice harboring a RIDD-incompetent variant of IRE1α were resistant to infection. Inactivation of the Bloc1s1 gene impaired the ability to assemble BLOC-1-related complex (BORC), resulting in differential recruitment of BORC-related lysosome trafficking components, perinuclear trafficking of Brucella-containing vacuoles (BCVs), and enhanced susceptibility to infection. The RIDD-resistant Bloc1s1 variant maintains the integrity of BORC and a higher-level association of BORC-related components that promote centrifugal lysosome trafficking, resulting in enhanced BCV peripheral trafficking and lysosomal destruction, and resistance to infection. These findings demonstrate that host RIDD activity on BLOS1 regulates Brucella intracellular parasitism by disrupting BORC-directed lysosomal trafficking. Notably, coronavirus murine hepatitis virus also subverted the RIDD-BLOS1 axis to promote intracellular replication. Our work establishes BLOS1 as a novel immune defense factor whose activity is hijacked by diverse pathogens.
    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
    Arginine homeostasis in lysosomes is critical for the growth and metabolism of mammalian cells. Phagolysosomes of macrophages are the niche where the parasitic protozoan Leishmania resides and causes human leishmaniasis. During infection, parasites encounter arginine deprivation, which is monitored by a sensor on the parasite cell surface. The sensor promptly activates a mitogen-activated protein kinase 2 (MAPK2)-mediated arginine deprivation response (ADR) pathway, resulting in upregulating the abundance and activity of the Leishmania arginine transporter (AAP3). Significantly, the ADR is also activated during macrophage infection, implying that arginine levels within the host phagolysosome are limiting for growth. We hypothesize that ADR-mediated upregulation of AAP3 activity is necessary to withstand arginine starvation, suggesting that the ADR is essential for parasite intracellular development. CRISPR/Cas9-mediated disruption of the AAP3 locus yielded mutants that retain a basal level of arginine transport but lack the ability to respond to arginine starvation. While these mutants grow normally in culture, they were impaired in their ability to develop inside THP-1 macrophages and were ∼70 to 80% less infective in BALB/c mice. Hence, inside the host macrophage, Leishmania must overcome the arginine \"hunger games\" by upregulating the transport of arginine via the ADR. We show that the ability to monitor and respond to changes in host metabolite levels is essential for pathogenesis.IMPORTANCE In this study, we report that the ability of the human pathogen Leishmania to sense and monitor the lack of arginine in the phagolysosome of the host macrophage is essential for disease development. Phagolysosomes of macrophages are the niche where Leishmania resides and causes human leishmaniasis. During infection, the arginine concentration in the phagolysosome decreases as part of the host innate immune response. An arginine sensor on the Leishmania cell surface activates an arginine deprivation response pathway that upregulates the expression of a parasite arginine transporter (AAP3). Here, we use CRISPR/Cas9-mediated disruption of the AAP3 locus to show that this response enables Leishmania parasites to successfully compete with the host macrophage in the \"hunger games\" for arginine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    热休克蛋白70(Hsp70),一个高度保守的蛋白质家族,在生物体中广泛分布,在生物和非生物胁迫反应中起着重要作用。然而,关于Hsp70基因的报道在微孢子虫中很少,一个非常大的专性细胞内寄生虫,可以感染几乎所有的动物,包括人类。在这项研究中,我们从8个小孢子虫基因组中鉴定出37种Hsp70蛋白,并将它们分为4个亚家族(A-D).这些微孢子虫中Hsp70基因的数量明显少于Rozellaallomycis和酵母。所有微孢子虫物种都包含来自每个亚家族和相似亚细胞位置的基因(线粒体,内质网,胞质溶胶,和胞质溶胶和/或细胞核),表明每个Hsp70成员可以履行不同的职能。同一亚家族中Hsp70蛋白的保守结构和基序高度相似。表达分析表明,亚家族C胞质溶胶(cyto)相关的Hsp70在微孢子虫发育过程中具有功能。免疫荧光测定显示,Cyto-NbHsp70在细胞质中位于家蚕Nosema的增殖阶段。Cyto-NbHsp70抗血清还标记了受感染细胞内的头孢菌素,表明该抗血清是标记不同微孢子虫物种增殖阶段的潜在分子标记。Cyto-NbHsp70的RNAi后,家蚕原虫的繁殖受到显着抑制,表明Cyto-NbHsp70对于病原体增殖很重要。我们的系统发育数据表明,Hsp70蛋白在微孢子虫适应细胞内寄生过程中进化,它们在病原体的发育中起着重要作用。
    Heat shock protein 70 (Hsp70), a highly conserved protein family, is widely distributed in organisms and plays fundamental roles in biotic and abiotic stress responses. However, reports on Hsp70 genes are scarce in microsporidia, a very large group of obligate intracellular parasites that can infect nearly all animals, including humans. In this study, we identified 37 Hsp70 proteins from eight microsporidian genomes and classified them into four subfamilies (A-D). The number of Hsp70 genes in these microsporidia was significantly fewer than in Rozella allomycis and yeast. All microsporidian species contained genes from each subfamily and similar subcellular locations (mitochondria, endoplasmic reticulum, cytosol, and cytosol and/or nucleus), indicating that each Hsp70 member may fulfil distinct functions. The conserved structures and motifs of the Hsp70 proteins in the same subfamily were highly similar. Expression analysis indicated that the subfamily C cytosol (cyto)-associated Hsp70s is functional during microsporidia development. Immunofluorescence assays revealed that Cyto-NbHsp70 was cytoplasmically located in the proliferation-stage of Nosema bombycis. Cyto-NbHsp70 antiserum also labeled Encephalitozoon hellem within infected cells, suggesting that this antiserum is a potential molecular marker for labeling the proliferative phases of different microsporidian species. The propagation of N. bombycis was significantly inhibited following RNAi of Cyto-NbHsp70, indicating that Cyto-NbHsp70 is important for pathogen proliferation. Our phylogenetic data suggest that Hsp70 proteins evolved during microsporidia adaption to intracellular parasitism, and they play important roles in pathogen development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    一种新的微孢子虫物种,白斑孢子虫。11月。,sp.11月。,根据形态学描述了草履虫,精细结构,和SSUrRNA基因序列。这是迄今为止报道的第一例草履虫微孢子虫病。所有观察到的生命周期阶段都是单核的。寄生虫在细胞质中发育,至少部分人口在内质网及其衍生物。Meronts除以二元裂变。疟原虫孢子虫通过莲座状出芽分裂。早期的孢子母细胞表现出发育良好的外孢子形成泡状结构。具有独特球形的孢子的大小为二态(3.7±0.2和1.9±0.2μm)。这两种类型的孢子的特征是薄的内生孢子,一个短的等线极管制造一个不完整的线圈,两分极化体,和一个大的后空泡。对于10个测试的草履虫物种复合体菌株中的5个,实验感染是成功的。所有易感菌株都属于密切相关的P.primaurelia和P.pentaurelia物种。系统发育分析将新物种置于微孢子虫的进化枝4中,并揭示了其与Euplotesporabinucleata(来自纤毛虫Euploteswoodruffi的微孢子虫)的密切关系,对赫尔米西亚和大环哺乳动物来说,水生无脊椎动物的微孢子虫。
    A new microsporidian species, Globosporidium paramecii gen. nov., sp. nov., from Paramecium primaurelia is described on the basis of morphology, fine structure, and SSU rRNA gene sequence. This is the first case of microsporidiosis in Paramecium reported so far. All observed stages of the life cycle are monokaryotic. The parasites develop in the cytoplasm, at least some part of the population in endoplasmic reticulum and its derivates. Meronts divide by binary fission. Sporogonial plasmodium divides by rosette-like budding. Early sporoblasts demonstrate a well-developed exospore forming blister-like structures. Spores with distinctive spherical shape are dimorphic in size (3.7 ± 0.2 and 1.9 ± 0.2 μm). Both types of spores are characterized by a thin endospore, a short isofilar polar tube making one incomplete coil, a bipartite polaroplast, and a large posterior vacuole. Experimental infection was successful for 5 of 10 tested strains of the Paramecium aurelia species complex. All susceptible strains belong to closely related P. primaurelia and P. pentaurelia species. Phylogenetic analysis placed the new species in the Clade 4 of Microsporidia and revealed its close relationship to Euplotespora binucleata (a microsporidium from the ciliate Euplotes woodruffi), to Helmichia lacustris and Mrazekia macrocyclopis, microsporidia from aquatic invertebrates.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Apicomplexan寄生虫携带嵌合蛋白,体现P4型ATPase和鸟苷酸环化酶结构域。这些蛋白质-在这组重要病原体中充当cGMP信号的执行器-就其庞大的大小而言,确实是不寻常的。作案手法,和进化的再利用。就像神话中的狮身人面像,一种人狮嵌合生物,构成具有挑战性的谜语,P4型ATPase-鸟苷酸环化酶嵌合体存在结构和功能难题。在这里我们回顾一下函数,拓扑,机制,以及顶孔丛寄生虫中肺泡特异性嵌合体的分子内协调。了解这些分子狮身人面像的艰巨技术挑战肯定会让许多跨学科研究人员在未来几十年忙碌。
    Apicomplexan parasites harbor chimeric proteins embodying P4-type ATPase and guanylate cyclase domains. Such proteins - serving as the actuator of cGMP signaling in this group of important pathogens - are indeed unusual in terms of their sheer size, modus operandi, and evolutionary repurposing. Much like the mythological Sphinx, a human-lion chimeric creature that posed challenging riddles, the P4-type ATPase-guanylate cyclase chimeras present both structural and functional conundrums. Here we review the function, topology, mechanism, and intramolecular coordination of the alveolate-specific chimeras in apicomplexan parasites. The steep technological challenge to understand these molecular Sphinxes will surely keep many interdisciplinary researchers busy in the next decades.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号