Jump to content

ENU: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
No edit summary
Kmug176 (talk | contribs)
No edit summary
Line 17: Line 17:
*5. ENU targets spermatogonial stem cells.<ref name=nolan/>
*5. ENU targets spermatogonial stem cells.<ref name=nolan/>


[[Image:Overviewfig1.jpg|right|thumb|250px|Figure 1:Overview of ENU mutagenesis screen. In a non-complementation screen, an ENU-induced male is crossed with a female carrying a mutant allele (a) of the gene of interest (A). If the mutation is dominant, then it will be present in every generation. However, if the mutation is recessive or if the G1 progeny are non-viable, then a different strategy is used to identify the mutation. An ENU-treated male is crossed with a wild type female. From the pool of G1 individuals, a heterozygous male is crossed to a female carrying the mutant allele (a). If the G2 progeny are infertile or non-viable, they can be recovered again from the G1 male.
[[Image:Overviewfig1.jpg|right|thumb|250px|Figure 1:Overview of ENU mutagenesis screen.]]
]]


=='''Overview of ENU mutagenesis screen'''==
=='''Overview of ENU mutagenesis screen'''==

Revision as of 17:00, 16 March 2008

File:ENU.png
ENU

ENU, also known as N-ethyl-N-nitrosourea (chemical formula C3H7N3O2), is a highly potent mutagen. For a given gene in mice, ENU can induce 1 new mutation in every 700 gametes. It is also toxic at high doses.

The chemical is an alkylating agent, and acts by transferring the ethyl group of ENU to nucleobases (usually thymine) in nucleic acids. Its main targets are the spermatogonial stem cells, from which mature sperm are derived.

ENU mutagenesis

N-ethyl-N-nitrosourea (ENU) is an alkylating agent which causes point mutations across the genome at an interval of approximately 1-2 Mb.[1]. Traditionally spontaneous mouse mutants were used by mouse geneticists for research purposes, which limited their research to a small repertoire of genetic variants. The introduction of mutagens like X-irradiation was able to expedite the process to some extent. However, X-irradiation caused unwanted deletions and other mutations. ENU was introduced as a mutagen in 1979 by Bill Russell et al and has revolutionized the field of mouse genetics ever since.[2].

Properties and advantages of ENU mutagenesis

  • 1. ENU is an alkylating agent and has preference for A->T base transversions and also for AT->GC transitions.[3] However it is also shown to cause GC->AT transitions.[4].
  • 2.It is known to induce point mutations, which implies that by mapping for the desired phenotype, the researcher can identify a single candidate gene responsible for the phenotype.[2]
  • 3.The point mutations are approximately are at 1-2 Mb interval and occur at an approximate rate ranging from 1 in 300 genomes to 1 in 5000 genomes.[3]
  • 4. Point mutations induced by ENU can either be gain-of-function mutations or loss-of function mutations in a gene as against deletions, which can induce only loss-of-function mutations.
  • 5. ENU targets spermatogonial stem cells.[3]
Figure 1:Overview of ENU mutagenesis screen. In a non-complementation screen, an ENU-induced male is crossed with a female carrying a mutant allele (a) of the gene of interest (A). If the mutation is dominant, then it will be present in every generation. However, if the mutation is recessive or if the G1 progeny are non-viable, then a different strategy is used to identify the mutation. An ENU-treated male is crossed with a wild type female. From the pool of G1 individuals, a heterozygous male is crossed to a female carrying the mutant allele (a). If the G2 progeny are infertile or non-viable, they can be recovered again from the G1 male.

Overview of ENU mutagenesis screen

The ENU mutagenesis screen is a type of forward (phenotype based) genetic screen which is used to identify and study a phenotype of interest. As illustrated in Figure 1, the screening process begins with mutagenising a male mouse with ENU. This is followed by systematic phenotypic analysis of the progeny. The progeny are assessed for behavioral, physiological or dysmorphological changes. The abnormal phenotype is identified. Identification of the candidate gene is then achieved by positional cloning of the mutant mice with the phenotype of interest.

Figure 2:Types of screens.

Types of screens

Depending on the region being assessed, forward genetic screens can be classified as illustrated in Figure 2 as:[2]

  • 1. Region Specific screens: Studies are designed specifically to obtain a gradient of phenotypes by generating an allelic series which are helpful in studying the region of interest.
  • 2 Genome-wide screens: These comprise of simple dominant or recessive screens and are often useful in understanding specific genetic and biochemical pathways.

REGION SPECIFIC SCREENS Region specific can be classified as follows:

Figure 3:Non-complementation screens.
  • a. Non-complementation screens

Complementation is the phenomenon which enables generation of the wild type phenotype when organisms carrying recessive mutations in different genes are crossed.[2] Thus if an organism has one functional copy of the gene, then this functional copy is capable of complementing the mutated or the lost copy of the gene. In contrast, if both the copies of the gene are mutated or lost, then this will lead to allelic non-complementation (Figure 3) and thus manifestation of the phenotype. The phenomenon of redundancy explains that many a times multiple genes are able to compensate for the loss of a particular gene. However, if two or more genes involved in the same biological processes or pathways are lost, then this leads to non-allelic non-complementation. In a non-complementation screen, an ENU-induced male is crossed with a female carrying a mutant allele (a) of the gene of interest (A). If the mutation is dominant, then it will be present in every generation. However, if the mutation is recessive or if the G1 progeny are non-viable, then a different strategy is used to identify the mutation. An ENU-treated male is crossed with a wild type female. From the pool of G1 individuals, a heterozygous male is crossed to a female carrying the mutant allele (a). If the G2 progeny are infertile or non-viable, they can be recovered again from the G1 male.

Figure 4:Deletion Screens.
  • b. Deletion screens

Deletions on chromosomes can be spontaneous or induced. In this screen, ENU-treated males are crossed to females homozygous for a deletion of the region of interest. The G1 progeny are compound heterozygotes for the ENU-induced mutation (Figure 4). Also, they are haploid with respect to the genes in the deleted region and thus loss-of-function or gain-of-function due to the ENU-induced mutation is expressed dominantly. Thus deletion screens have an advantage over other recessive screens due to the identification of the mutation in the G1 progeny itself. Rinchik et al performed a deletion screen and complementation analysis and were able to isolate 11 independent recessive loci, which were grouped into seven complementation groups on chromosome 7, a region surrounding the albino (Tyr) gene and the pink-eyed dilution (p) gene.[2]

Figure 5:Balancer Screens.
  • c. Balancer screens

A chromosome carrying a balancer region is termed as a balancer chromosome. A balancer is a region which prevents recombination between homologous chromosomes during meiosis. This is possible due to the presence of an inverted region or a series of inversions. Balancer chromosome was primalrily used for studies in Drosophila melanogaster genetics. Monica Justice et al efficiently carried out a balancer screen using a balancer chromosome constructed by Alan Bradley et al on mouse chromosome 11. In this screen, a ENU-induced male is crossed with a female heterozygous for the balancer chromosome.[2]The mice carrying the balancer chromosome have yellow ears and tail. The G1 heterozygotes are (Figure 5) are crossed to females carrying the rex mutation (Rex in figure 5), which confers a curly coat. In G2, homozygotes for the balancer are non-viable and are not recovered. Mice carrying the rex mutation trans to the balancer or ENU-induced mutation have a curly coat and are discarded. Mice that are compound heterozygotes for the balancer and the ENU-induced mutation are brother-sister mated to obtain homozygotes for the ENU-induced mutation in G3.

GENOME-WIDE SCREENS Genome-wide screens are most often useful for studying genetic diseases in which multiple genetic and biochemical pathways may be involved. Thus with this approach, candidate genes or regions across the genome, that are associated with the phenotype can be identified.

Figure 6:Conventional screens
  • a. Conventional screens

These screens can be designed to identify simple dominant and recessive phenotypes. (Figure 6). Thus an ENU-induced G0 male is crossed with a wild type female. The G1 progeny can be screened to identify dominant mutations. However, if the mutation is recessive, then G3 individuals homozygous for the mutation can be recovered from the G1 males in two ways:

    • A] The G1 male is crossed with a wild type female to generate a pool of G2 progeny. The G3 individuals can be obtained by crossing the G1 male to the G2 daughters. This will yield a proportion of the G3 individuals which resemble the G1 male to a large extent.
    • B] G1 male is crossed to a wild type female to obtain a pool of G2 animals., which are then brother-sister mated to obtain the G3 progenies. This approach yields a variety of mutants in the G3 progeny.

A number of organizations around the world are performing genome-wide mutagenesis screens using ENU. Some of them include German ENU mouse mutagenesis screen project performed by the GSF, Munich; Jackson laboratory, USA; Australian phenomics facility; Molecular neurobiology at Northwestern university, USA; Oak Ridge National Laboratory, USA; National centre for mouse genetics, MRC Harwell, UK and many more.[3]

Figure 7:Modifier screens.
  • b. Modifier screens

A modifier such as an enhancer or suppressor can alter the function of a gene. In a modifier screen, an organism with a pre-existing phenotype is selected. Thus, any mutations caused by the mutagen (ENU) can be assessed for their enhancive or suppressive activity.[2] Screening for dominant and recessive mutations is performed in a way similar to the conventional genome-wide screen (Figure 7). A number of modifier screens have been performed on Drosophila. Recently, Aliga et al performed a dominant modifier screen using ENU-induced mice to identify modifiers of the Notch signaling pathway.[5] Delta 1 is a ligand for the Notch receptor. A homozygous loss-of-function of Delta 1 (Dll1lacZ/lacZ) is embryonically lethal. ENU-treated mice were crossed to Dll1lacZ heterozygotes. 35 mutant lines were generated in G1 of which 7 revealed modifiers of the Notch signaling pathway.

  • c. Sensitized screens

In the case of genetic diseases involving multiple genes, mutations in multiple genes contributes to the progression of a disease. Mutation in just one of these genes however, might not contribute significantly to any phenotype. Such "predisposing genes" can be identified using sensitized screens.[6] In this type of a screen, the genetic or environmental background is modified so as to sensitize the mouse to these changes. The idea is that the predisposing genes can be unraveled on a modified genetic or environmental background. Rinchik et al performed a sensitized screen of mouse mutants predisposed to Diabetic nephropathy. Mice were treated with ENU on a sensitized background of type-1 diabetes. These diabetic mice had a dominant Akita mutation in the insulin-2 gene (Ins2Akita). These mice developed albuminuria, a phenotype that was not observed in the non-diabetic offsprings.[7]

Notes

  1. ^ Concepcion,D, Seburn, KL, Wen,G, Frankel, WN & Hamilton,BA 2004, p.953-9
  2. ^ a b c d e f g Kile,BT & Hilton, DJ 2005, p.557-67
  3. ^ a b c d Nolan,P, Hugill, A & Cox,RD,2002,p.278-89
  4. ^ Coghill,EL et al,2002,p.255-6
  5. ^ Rubio-Aliaga, I. et al. A genetic screen for modifiers of the delta1-dependent notch signaling function in the mouse. Genetics 175, 1451-1463 (2007)
  6. ^ Cordes, S.P. N-ethyl-N-nitrosourea mutagenesis: boarding the mouse mutant express. Microbiol Mol Biol Rev 69, 426-439 (2005).
  7. ^ Tchekneva, E.E. et al. A sensitized screen of N-ethyl-N-nitrosourea-mutagenized mice identifies dominant mutants predisposed to diabetic nephropathy. J Am Soc Nephrol 18, 103-112 (2007).

References

  • 1. Concepcion,D, Seburn, KL, Wen,G, Frankel, WN & Hamilton,BA: Mutation rate and predicted phenotypic target sizes in ethylnitrosourea-treated mice. Genetics, 168, 953-9 (2004)
  • 2. Kile,BT & Hilton, DJ: The art and design of genetic screens: mouse.Nat Rev Genet,6,557-67 (2005)
  • 3. Nolan,P, Hugill, A & Cox,RD: ENU mutagenesis in the mouse: application to human genetic diseases. Brief Funct Genomic Proteomic,1,278-89 (2002)
  • 4.Coghill,EL et al: A gene-driven approach to the identification of ENU mutants in mouse. Nat Genet, 30,255-6 (2002)
  • 5.Cordes, S.P. N-ethyl-N-nitrosourea mutagenesis: boarding the mouse mutant express. Microbiol Mol Biol Rev 69, 426-439 (2005).
  • 6.Rinchik, E.M., Carpenter, D.A. & Johnson, D.K. Functional annotation of mammalian genomic DNA sequence by chemical mutagenesis: a fine-structure genetic mutation map of a 1- to 2-cM segment of mouse chromosome 7 corresponding to human chromosome 11p14-p15. Proc Natl Acad Sci U S A 99, 844-849 (2002).
  • 7.Tchekneva, E.E. et al. A sensitized screen of N-ethyl-N-nitrosourea-mutagenized mice identifies dominant mutants predisposed to diabetic nephropathy. J Am Soc Nephrol 18, 103-112 (2007).
  • 8.Rubio-Aliaga, I. et al. A genetic screen for modifiers of the delta1-dependent notch signaling function in the mouse. Genetics 175, 1451-1463 (2007)

External links