期刊论文详细信息
Particle and Fibre Toxicology
Engineered single nucleotide polymorphisms in the mosquito MEK docking site alter Plasmodium berghei development in Anopheles gambiae
Shirley Luckhart1  Aaron C Brault2  Michael Anishchenko2  Kong Cheung1  Lattha Souvannaseng1  Ashley A Brenton1 
[1] Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, 95616 Davis, CA, USA;Division of Vector-Borne Diseases, National Center for Emerging Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 3156 Rampart Rd, 80521 Fort Collins, CO, USA
关键词: Immunity;    Single nucleotide polymorphism;    Malaria;    Plasmodium;    MAPK;    Mosquito;    Anopheles;   
Others  :  803920
DOI  :  10.1186/1756-3305-7-287
 received in 2014-02-11, accepted in 2014-06-13,  发布年份 2014
PDF
【 摘 要 】

Background

Susceptibility to Plasmodium infection in Anopheles gambiae has been proposed to result from naturally occurring polymorphisms that alter the strength of endogenous innate defenses. Despite the fact that some of these mutations are known to introduce non-synonymous substitutions in coding sequences, these mutations have largely been used to rationalize knockdown of associated target proteins to query the effects on parasite development in the mosquito host. Here, we assay the effects of engineered mutations on an immune signaling protein target that is known to control parasite sporogonic development. By this proof-of-principle work, we have established that naturally occurring mutations can be queried for their effects on mosquito protein function and on parasite development and that this important signaling pathway can be genetically manipulated to enhance mosquito resistance.

Methods

We introduced SNPs into the A. gambiae MAPK kinase MEK to alter key residues in the N-terminal docking site (D-site), thus interfering with its ability to interact with the downstream kinase target ERK. ERK phosphorylation levels in vitro and in vivo were evaluated to confirm the effects of MEK D-site mutations. In addition, overexpression of various MEK D-site alleles was used to assess P. berghei infection in A. gambiae.

Results

The MEK D-site contains conserved lysine residues predicted to mediate protein-protein interaction with ERK. As anticipated, each of the D-site mutations (K3M, K6M) suppressed ERK phosphorylation and this inhibition was significant when both mutations were present. Tissue-targeted overexpression of alleles encoding MEK D-site polymorphisms resulted in reduced ERK phosphorylation in the midgut of A. gambiae. Furthermore, as expected, inhibition of MEK-ERK signaling due to D-site mutations resulted in reduction in P. berghei development relative to infection in the presence of overexpressed catalytically active MEK.

Conclusion

MEK-ERK signaling in A. gambiae, as in model organisms and humans, depends on the integrity of conserved key residues within the MEK D-site. Disruption of signal transmission via engineered SNPs provides a purposeful proof-of-principle model for the study of naturally occurring mutations that may be associated with mosquito resistance to parasite infection as well as an alternative genetic basis for manipulation of this important immune signaling pathway.

【 授权许可】

   
2014 Brenton et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140708051214760.pdf 1305KB PDF download
Figure 6. 70KB Image download
Figure 5. 68KB Image download
Figure 4. 54KB Image download
Figure 3. 86KB Image download
Figure 2. 70KB Image download
【 图 表 】

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

【 参考文献 】
  • [1]World Health Organization Global Malaria Program: World malaria report: 2013. Geneva: World Health Organization; 2013.
  • [2]Li M, Liu J, Zhang C: Evolutionary history of the vertebrate mitogen activated protein kinases family. PLoS ONE 2011, 6:e26999.
  • [3]Horton AA, Wang B, Camp L, Price MS, Arshi A, Nagy M, Nadler SA, Faeder JR, Luckhart S: The mitogen-activated protein kinome from Anopheles gambiae: identification, phylogeny and functional characterization of the ERK, JNK and p38 MAP kinases. BMC Genomics 2011, 12:574.
  • [4]Zahoor Z, Davies AJ, Kirk RS, Rollinson D, Walker AJ: Nitric oxide production by Biomphalaria glabrata haemocytes: effects of Schistosoma mansoni ESPs and regulation through the extracellular signal-regulated kinase pathway. Parasit Vectors 2009, 2:18.
  • [5]Ibraim IC, De Assis RR, Pessoa NL, Campos MA, Melo MN, Turco SJ, Soares RP: Two biochemically distinct lipophosphoglycans from Leishmania braziliensis and Leishmania infantum trigger different innate immune responses in murine macrophages. Parasit Vectors 2013, 6:54.
  • [6]Chang L, Karin M: Mammalian MAP kinase signalling cascades. Nature 2001, 410:37-40.
  • [7]Krishna M, Narang H: The complexity of mitogen-activated protein kinases (MAPKs) made simple. Cell Mol Life Sci 2008, 65:3525-3544.
  • [8]Rubinfeld H, Seger R: The ERK cascade: a prototype of MAPK signaling. Mol Biotechnol 2005, 31:151-174.
  • [9]Fukuda M, Gotoh Y, Nishida E: Interaction of MAP kinase with MAP kinase kinase: its possible role in the control of nucleocytoplasmic transport of MAP kinase. EMBO J 1997, 16:1901-1908.
  • [10]Adachi M, Fukuda M, Nishida E: Two co-existing mechanisms for nuclear import of MAP kinase: passive diffusion of a monomer and active transport of a dimer. EMBO J 1999, 18:5347-5358.
  • [11]Wolf I, Rubinfeld H, Yoon S, Marmor G, Hanoch T, Seger R: Involvement of the activation loop of ERK in the detachment from cytosolic anchoring. J Biol Chem 2001, 276:24490-24497.
  • [12]Xu B, Wilsbacher JL, Collisson T, Cobb MH: The N-terminal ERK-binding site of MEK1 is required for efficient feedback phosphorylation by ERK2 in vitro and ERK activation in vivo. J Biol Chem 1999, 274:34029-34035.
  • [13]Bromberg-White JL, Andersen NJ, Duesbery NS: MEK genomics in development and disease. Brief Funct Genomics 2012, 11:300-310.
  • [14]Mayor F Jr, Jurado-Pueyo M, Campos PM, Murga C: Interfering with MAP kinase docking interactions: implications and perspective for the p38 route. Cell Cycle 2007, 6:528-533.
  • [15]Zhou T, Sun L, Humphreys J, Goldsmith EJ: Docking interactions induce exposure of activation loop in the MAP kinase ERK2. Structure 2006, 14:1011-1019.
  • [16]Tanoue T, Adachi M, Moriguchi T, Nishida E: A conserved docking motif in MAP kinases common to substrates, activators and regulators. Nat Cell Biol 2000, 2:110-116.
  • [17]Bardwell AJ, Flatauer LJ, Matsukuma K, Thorner J, Bardwell L: A conserved docking site in MEKs mediates high-affinity binding to MAP kinases and cooperates with a scaffold protein to enhance signal transmission. J Biol Chem 2001, 276:10374-10386.
  • [18]Xu B, Stippec S, Robinson FL, Cobb MH: Hydrophobic as well as charged residues in both MEK1 and ERK2 are important for their proper docking. J Biol Chem 2001, 276:26509-26515.
  • [19]Lim J, Gowda DC, Krishnegowda G, Luckhart S: Induction of nitric oxide synthase in Anopheles stephensi by Plasmodium falciparum: mechanism of signaling and the role of parasite glycosylphosphatidylinositols. Infect Immun 2005, 73:2778-2789.
  • [20]Surachetpong W, Pakpour N, Cheung KW, Luckhart S: Reactive oxygen species-dependent cell signaling regulates the mosquito immune response to Plasmodium falciparum. Antioxid Redox Signal 2011, 14:943-955.
  • [21]Surachetpong W, Singh N, Cheung KW, Luckhart S: MAPK ERK signaling regulates the TGF-beta1-dependent mosquito response to Plasmodium falciparum. PLoS Pathog 2009, 5:e1000366.
  • [22]Price I, Ermentrout B, Zamora R, Wang B, Azhar N, Mi Q, Constantine G, Faeder JR, Luckhart S, Vodovotz Y: In vivo, in vitro, and in silico studies suggest a conserved immune module that regulates malaria parasite transmission from mammals to mosquitoes. J Theor Biol 2013, 334:173-186.
  • [23]Peterson TM, Gow AJ, Luckhart S: Nitric oxide metabolites induced in Anopheles stephensi control malaria parasite infection. Free Radic Biol Med 2007, 42:132-142.
  • [24]Ramiro RS, Alpedrinha J, Carter L, Gardner A, Reece SE: Sex and death: the effects of innate immune factors on the sexual reproduction of malaria parasites. PLoS Pathog 2011, 7:e1001309.
  • [25]Muller HM, Dimopoulos G, Blass C, Kafatos FC: A hemocyte-like cell line established from the malaria vector Anopheles gambiae expresses six prophenoloxidase genes. J Biol Chem 1999, 274:11727-11735.
  • [26]Geneious version 5.5.8 created by Biomatters [http://www.geneious.com/ webcite]
  • [27]Peng R, Maklokova VI, Chandrashekhar JH, Lan Q: In vivo functional genomic studies of sterol carrier protein-2 gene in the yellow fever mosquito. PLoS ONE 2011, 6:e18030.
  • [28]Moreira LA, Ghosh AK, Abraham EG, Jacobs-Lorena M: Genetic transformation of mosquitoes: a quest for malaria control. Int J Parasitol 2002, 32:1599-1605.
  • [29]Corby-Harris V, Drexler A, Watkins De Jong L, Antonova Y, Pakpour N, Ziegler R, Ramberg F, Lewis EE, Brown JM, Luckhart S, Riehle MA: Activation of Akt signaling reduces the prevalence and intensity of malaria parasite infection and lifespan in Anopheles stephensi mosquitoes. PLoS Pathog 2010, 6:e1001003.
  • [30]Mansour SJ, Matten WT, Hermann AS, Candia JM, Rong S, Fukasawa K, Vande Woude GF, Ahn NG: Transformation of mammalian cells by constitutively active MAP kinase kinase. Science 1994, 265:966-970.
  • [31]Huang W, Erikson RL: Constitutive activation of Mek1 by mutation of serine phosphorylation sites. Proc Natl Acad Sci U S A 1994, 91:8960-8963.
  • [32]Attardo GM, Hansen IA, Raikhel AS: Nutritional regulation of vitellogenesis in mosquitoes: implications for anautogeny. Insect Biochem Mol Biol 2005, 35:661-675.
  • [33]Anderson WA, Spielman A: Permeability of the ovarian follicle of Aedes aegypti mosquitoes. J Cell Biol 1971, 50:201-221.
  • [34]Hauck ES, Antonova-Koch Y, Drexler A, Pietri J, Pakpour N, Liu D, Blacutt J, Riehle MA, Luckhart S: Overexpression of phosphatase and tensin homolog improves fitness and decreases Plasmodium falciparum development in Anopheles stephensi. Microbes Infect 2013, 15:775-787.
  • [35]McCubrey JA, Steelman LS, Abrams SL, Chappell WH, Russo S, Ove R, Milella M, Tafuri A, Lunghi P, Bonati A, Stivala F, Nicoletti F, Libra M, Martelli AM, Montalto G, Cervello M: Emerging MEK inhibitors. Expert Opin Emerg Drugs 2010, 15:203-223.
  • [36]Bardwell AJ, Frankson E, Bardwell L: Selectivity of docking sites in MAPK kinases. J Biol Chem 2009, 284:13165-13173.
  • [37]Turk BE: Manipulation of host signalling pathways by anthrax toxins. Biochem J 2007, 402:405-417.
  • [38]Vitale G, Pellizzari R, Recchi C, Napolitani G, Mock M, Montecucco C: Anthrax lethal factor cleaves the N-terminus of MAPKKs and induces tyrosine/threonine phosphorylation of MAPKs in cultured macrophages. Biochem Biophys Res Commun 1998, 248:706-711.
  • [39]Duesbery NS, Webb CP, Leppla SH, Gordon VM, Klimpel KR, Copeland TD, Ahn NG, Oskarsson MK, Fukasawa K, Paull KD, Vande Woude GF: Proteolytic inactivation of MAP-kinase-kinase by anthrax lethal factor. Science 1998, 280:734-737.
  • [40]Koo HM, VanBrocklin M, McWilliams MJ, Leppla SH, Duesbery NS, Vande Woude GF: Apoptosis and melanogenesis in human melanoma cells induced by anthrax lethal factor inactivation of mitogen-activated protein kinase kinase. Proc Natl Acad Sci U S A 2002, 99:3052-3057.
  • [41]Huang D, Ding Y, Luo WM, Bender S, Qian CN, Kort E, Zhang ZF, VandenBeldt K, Duesbery NS, Resau JH, Teh BT: Inhibition of MAPK kinase signaling pathways suppressed renal cell carcinoma growth and angiogenesis in vivo. Cancer Res 2008, 68:81-88.
  • [42]Abi-Habib RJ, Urieto JO, Liu S, Leppla SH, Duesbery NS, Frankel AE: BRAF status and mitogen-activated protein/extracellular signal-regulated kinase kinase 1/2 activity indicate sensitivity of melanoma cells to anthrax lethal toxin. Mol Cancer Ther 2005, 4:1303-1310.
  • [43]Nimnual A, Bar-Sagi D: The two hats of SOS. Sci STKE 2002, 2002:pe36.
  • [44]Langlois WJ, Sasaoka T, Saltiel AR, Olefsky JM: Negative feedback regulation and desensitization of insulin- and epidermal growth factor-stimulated p21ras activation. J Biol Chem 1995, 270:25320-25323.
  • [45]Aksamitiene E, Kholodenko BN, Kolch W, Hoek JB, Kiyatkin A: PI3K/Akt-sensitive MEK-independent compensatory circuit of ERK activation in ER-positive PI3K-mutant T47D breast cancer cells. Cell Signal 2010, 22:1369-1378.
  • [46]Wu Z, Jiao P, Huang X, Feng B, Feng Y, Yang S, Hwang P, Du J, Nie Y, Xiao G, Xu H: MAPK phosphatase-3 promotes hepatic gluconeogenesis through dephosphorylation of forkhead box O1 in mice. J Clin Invest 2010, 120:3901-3911.
  • [47]Kim M, Cha GH, Kim S, Lee JH, Park J, Koh H, Choi KY, Chung J: MKP-3 has essential roles as a negative regulator of the Ras/mitogen-activated protein kinase pathway during Drosophila development. Mol Cell Biol 2004, 24:573-583.
  • [48]Doddareddy MR, Rawling T, Ammit AJ: Targeting mitogen-activated protein kinase phosphatase-1 (MKP-1): structure-based design of MKP-1 inhibitors and upregulators. Curr Med Chem 2012, 19:163-173.
  • [49]Horton AA, Lee Y, Coulibaly CA, Rashbrook VK, Cornel AJ, Lanzaro GC, Luckhart S: Identification of three single nucleotide polymorphisms in Anopheles gambiae immune signaling genes that are associated with natural Plasmodium falciparum infection. Malar J 2010, 9:160.
  • [50]Harris C, Lambrechts L, Rousset F, Abate L, Nsango SE, Fontenille D, Morlais I, Cohuet A: Polymorphisms in Anopheles gambiae immune genes associated with natural resistance to Plasmodium falciparum. PLoS Pathog 2010, 6:e1001112.
  • [51]Riehle MM, Markianos K, Niare O, Xu J, Li J, Toure AM, Podiougou B, Oduol F, Diawara S, Diallo M, Coulibaly B, Ouatara A, Kruglyak L, Traore SF, Vernick KD: Natural malaria infection in Anopheles gambiae is regulated by a single genomic control region. Science 2006, 312:577-579.
  • [52]Li J, Wang X, Zhang G, Githure JI, Yan G, James AA: Genome-block expression-assisted association studies discover malaria resistance genes in Anopheles gambiae. Proc Natl Acad Sci U S A 2013, 110:20675-20680.
  文献评价指标  
  下载次数:46次 浏览次数:17次