Browse > Article
http://dx.doi.org/10.5483/BMBRep.2020.53.9.141

How Z-DNA/RNA binding proteins shape homeostasis, inflammation, and immunity  

Kim, Chun (Department of Molecular and Life Science, Hanyang University [ERICA Campus])
Publication Information
BMB Reports / v.53, no.9, 2020 , pp. 453-457 More about this Journal
Abstract
The right-handed double-helical structure of DNA (B-DNA), which follows the Watson-Crick model, is the canonical form of DNA existing in normal physiological settings. Even though an alternative left-handed structure of DNA (Z-DNA) was discovered in the late 1970s, Z-form nucleic acid has not received much attention from biologists, because it is extremely unstable under physiological conditions, has an ill-defined mechanism of its formation, and has obscure biological functions. The debate about the physiological relevance of Z-DNA was settled only after a class of proteins was found to potentially recognize the Z-form architecture of DNA. Interestingly, these Z-DNA binding proteins can bind not only the left-handed form of DNA but also the equivalent structure of RNA (Z-RNA). The Z-DNA/RNA binding proteins present from viruses to humans function as important regulators of biological processes. In particular, the proteins ADAR1 and ZBP1 are currently being extensively re-evaluated in the field to understand potential roles of the noncanonical Z-conformation of nucleic acids in host immune responses and human disease. Despite a growing body of evidence supporting the biological importance of Z-DNA/RNA, there remain many unanswered principal questions, such as when Z-form nucleic acids arise and how they signal to downstream pathways. Understanding Z-DNA/RNA and the sensors in different pathophysiological conditions will widen our view on the regulation of immune responses and open a new door of opportunity to develop novel types of immunomodulatory therapeutic possibilities.
Keywords
ADAR1; Inflammation; Necroptosis; ZBP1; Z-DNA; Z-RNA;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Ishii KJ, Kawagoe T, Koyama S et al (2008) TANK-binding kinase-1 delineates innate and adaptive immune responses to DNA vaccines. Nature 451, 725-729   DOI
2 Lippmann J, Rothenburg S, Deigendesch N et al (2008) IFNbeta responses induced by intracellular bacteria or cytosolic DNA in different human cells do not require ZBP1 (DLM-1/DAI). Cell Microbiol 10, 2579-2588   DOI
3 Kuriakose T and Kanneganti TD (2018) ZBP1: Innate Sensor Regulating Cell Death and Inflammation. Trends Immunol 39, 123-134   DOI
4 Pasparakis M and Vandenabeele P (2015) Necroptosis and its role in inflammation. Nature 517, 311-320   DOI
5 Kaiser WJ, Upton JW and Mocarski ES (2008) Receptor-interacting protein homotypic interaction motif-dependent control of NF-kappa B activation via the DNA-dependent activator of IFN regulatory factors. J Immunol 181, 6427-6434   DOI
6 Rebsamen M, Heinz LX, Meylan E et al (2009) DAI/ZBP1 recruits RIP1 and RIP3 through RIP homotypic interaction motifs to activate NF-kappaB. EMBO Rep 10, 916-922   DOI
7 Upton JW, Kaiser WJ and Mocarski ES (2012) DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe 11, 290-297   DOI
8 Pham TH, Kwon KM, Kim YE, Kim KK and Ahn JH (2013) DNA sensing-independent inhibition of herpes simplex virus 1 replication by DAI/ZBP1. J Virol 87, 3076-3086   DOI
9 Kuriakose T, Man SM, Malireddi RK et al (2016) ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways. Sci Immunol 1, aag2045   DOI
10 Thapa RJ, Ingram JP, Ragan KB et al (2016) DAI Senses Influenza A Virus Genomic RNA and Activates RIPK3-Dependent Cell Death. Cell Host Microbe 20, 674-681   DOI
11 Wang AH, Quigley GJ, Kolpak FJ et al (1979) Molecular structure of a left-handed double helical DNA fragment at atomic resolution. Nature 282, 680-686   DOI
12 Drew H, Takano T, Tanaka S, Itakura K and Dickerson RE (1980) High-salt d(CpGpCpG), a left-handed Z' DNA double helix. Nature 286, 567-573   DOI
13 Herbert A, Lowenhaupt K, Spitzner J and Rich A (1995) Chicken double-stranded RNA adenosine deaminase has apparent specificity for Z-DNA. Proc Natl Acad Sci U S A 92, 7550-7554   DOI
14 Berger I, Winston W, Manoharan R et al (1998) Spectroscopic characterization of a DNA-binding domain, Z alpha, from the editing enzyme, dsRNA adenosine deaminase: evidence for left-handed Z-DNA in the Z alpha-DNA complex. Biochemistry 37, 13313-13321   DOI
15 Schwartz T, Behlke J, Lowenhaupt K, Heinemann U and Rich A (2001) Structure of the DLM-1-Z-DNA complex reveals a conserved family of Z-DNA-binding proteins. Nat Struct Biol 8, 761-765   DOI
16 Schwartz T, Rould MA, Lowenhaupt K, Herbert A and Rich A (1999) Crystal structure of the Zalpha domain of the human editing enzyme ADAR1 bound to left-handed Z-DNA. Science 284, 1841-1845   DOI
17 Placido D, Brown BA 2nd, Lowenhaupt K, Rich A and Athanasiadis A (2007) A left-handed RNA double helix bound by the Z alpha domain of the RNA-editing enzyme ADAR1. Structure 15, 395-404   DOI
18 Brown BA 2nd, Lowenhaupt K, Wilbert CM, Hanlon EB and Rich A (2000) The zalpha domain of the editing enzyme dsRNA adenosine deaminase binds left-handed Z-RNA as well as Z-DNA. Proc Natl Acad Sci U S A 97, 13532-13536   DOI
19 Rothenburg S, Deigendesch N, Dittmar K et al (2005) A PKR-like eukaryotic initiation factor 2alpha kinase from zebrafish contains Z-DNA binding domains instead of dsRNA binding domains. Proc Natl Acad Sci U S A 102, 1602-1607   DOI
20 Kim YG, Muralinath M, Brandt T et al (2003) A role for Z-DNA binding in vaccinia virus pathogenesis. Proc Natl Acad Sci U S A 100, 6974-6979   DOI
21 Strehblow A, Hallegger M and Jantsch MF (2002) Nucleocytoplasmic distribution of human RNA-editing enzyme ADAR1 is modulated by double-stranded RNA-binding domains, a leucine-rich export signal, and a putative dimerization domain. Mol Biol Cell 13, 3822-3835   DOI
22 Koehler H, Cotsmire S, Langland J et al (2017) Inhibition of DAI-dependent necroptosis by the Z-DNA binding domain of the vaccinia virus innate immune evasion protein, E3. Proc Natl Acad Sci U S A 114, 11506-11511   DOI
23 Kim U, Garner TL, Sanford T, Speicher D, Murray JM and Nishikura K (1994) Purification and characterization of double-stranded RNA adenosine deaminase from bovine nuclear extracts. J Biol Chem 269, 13480-13489   DOI
24 O'Connell MA, Krause S, Higuchi M et al (1995) Cloning of cDNAs encoding mammalian double-stranded RNA-specific adenosine deaminase. Mol Cell Biol 15, 1389-1397   DOI
25 Patterson JB and Samuel CE (1995) Expression and regulation by interferon of a double-stranded-RNA-specific adenosine deaminase from human cells: evidence for two forms of the deaminase. Mol Cell Biol 15, 5376-5388   DOI
26 Poulsen H, Nilsson J, Damgaard CK, Egebjerg J and Kjems J (2001) CRM1 mediates the export of ADAR1 through a nuclear export signal within the Z-DNA binding domain. Mol Cell Biol 21, 7862-7871   DOI
27 Gallo A, Vukic D, Michalik D, O'Connell MA and Keegan LP (2017) ADAR RNA editing in human disease; more to it than meets the I. Hum Genet 136, 1265-1278   DOI
28 Athanasiadis A, Placido D, Maas S, Brown BA 2nd, Lowenhaupt K and Rich A (2005) The crystal structure of the Zbeta domain of the RNA-editing enzyme ADAR1 reveals distinct conserved surfaces among Z-domains. J Mol Biol 351, 496-507   DOI
29 Lin J, Kumari S, Kim C et al (2016) RIPK1 counteracts ZBP1-mediated necroptosis to inhibit inflammation. Nature 540, 124-128   DOI
30 Maelfait J, Liverpool L, Bridgeman A, Ragan KB, Upton JW and Rehwinkel J (2017) Sensing of viral and endogenous RNA by ZBP1/DAI induces necroptosis. EMBO J 36, 2529-2543   DOI
31 Newton K, Wickliffe KE, Maltzman A et al (2016) RIPK1 inhibits ZBP1-driven necroptosis during development. Nature 540, 129-133   DOI
32 Schoggins JW, MacDuff DA, Imanaka N et al (2014) Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity. Nature 505, 691-695   DOI
33 Daniels BP, Kofman SB, Smith JR et al (2019) The Nucleotide Sensor ZBP1 and Kinase RIPK3 Induce the Enzyme IRG1 to Promote an Antiviral Metabolic State in Neurons. Immunity 50, 64-76 e64   DOI
34 Lladser A, Mougiakakos D, Tufvesson H et al (2011) DAI (DLM-1/ZBP1) as a genetic adjuvant for DNA vaccines that promotes effective antitumor CTL immunity. Mol Ther 19, 594-601   DOI
35 Hirvinen M, Capasso C, Guse K et al (2016) Expression of DAI by an oncolytic vaccinia virus boosts the immunogenicity of the virus and enhances antitumor immunity. Mol Ther Oncolytics 3, 16002   DOI
36 Hartner JC, Schmittwolf C, Kispert A, Muller AM, Higuchi M and Seeburg PH (2004) Liver disintegration in the mouse embryo caused by deficiency in the RNA-editing enzyme ADAR1. J Biol Chem 279, 4894-4902   DOI
37 Chung H, Calis JJA, Wu X et al (2018) Human ADAR1 Prevents Endogenous RNA from Triggering Translational Shutdown. Cell 172, 811-824.e14   DOI
38 Rice GI, Kasher PR, Forte GM et al (2012) Mutations in ADAR1 cause Aicardi-Goutieres syndrome associated with a type I interferon signature. Nat Genet 44, 1243-1248   DOI
39 Wang Q, Khillan J, Gadue P and Nishikura K (2000) Requirement of the RNA editing deaminase ADAR1 gene for embryonic erythropoiesis. Science 290, 1765-1768   DOI
40 Jiao H, Wachsmuth L, Kumari S et al (2020) Z-nucleic-acid sensing triggers ZBP1-dependent necroptosis and inflammation. Nature 580, 391-395   DOI
41 Wang R, Li H, Wu J et al (2020) Gut stem cell necroptosis by genome instability triggers bowel inflammation. Nature 580, 386-390   DOI
42 Marshall PR, Zhao Q, Li X et al (2020) Dynamic regulation of Z-DNA in the mouse prefrontal cortex by the RNAediting enzyme Adar1 is required for fear extinction. Nat Neurosci 23, 718-729   DOI
43 Suzuki N, Suzuki T, Inagaki K et al (2005) Mutation analysis of the ADAR1 gene in dyschromatosis symmetrica hereditaria and genetic differentiation from both dyschromatosis universalis hereditaria and acropigmentatio reticularis. J Invest Dermatol 124, 1186-1192   DOI
44 Wang Q, Miyakoda M, Yang W et al (2004) Stress-induced apoptosis associated with null mutation of ADAR1 RNA editing deaminase gene. J Biol Chem 279, 4952-4961   DOI
45 Liddicoat BJ, Piskol R, Chalk AM et al (2015) RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself. Science 349, 1115-1120   DOI
46 Mannion NM, Greenwood SM, Young R et al (2014) The RNA-editing enzyme ADAR1 controls innate immune responses to RNA. Cell Rep 9, 1482-1494   DOI
47 Koeris M, Funke L, Shrestha J, Rich A and Maas S (2005) Modulation of ADAR1 editing activity by Z-RNA in vitro. Nucleic Acids Res 33, 5362-5370   DOI
48 Herbert A (2019) Z-DNA and Z-RNA in human disease. Commun Biol 2, 7   DOI
49 Ng SK, Weissbach R, Ronson GE and Scadden AD (2013) Proteins that contain a functional Z-DNA-binding domain localize to cytoplasmic stress granules. Nucleic Acids Res 41, 9786-9799   DOI
50 Protter DS and Parker R (2016) Principles and Properties of Stress Granules. Trends Cell Biol 26, 668-679   DOI
51 Fu Y, Comella N, Tognazzi K, Brown LF, Dvorak HF and Kocher O (1999) Cloning of DLM-1, a novel gene that is up-regulated in activated macrophages, using RNA differential display. Gene 240, 157-163   DOI
52 Takaoka A, Wang Z, Choi MK et al (2007) DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature 448, 501-505   DOI