Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

3166±3173 Nucleic Acids Research, 2003, Vol. 31, No.

12
DOI: 10.1093/nar/gkg427

Characterization of the 2¢±5¢-oligoadenylate


synthetase ubiquitin-like family
Signe Eskildsen1, Just Justesen1, Mikkel Heide Schierup2 and Rune Hartmann1,3,*
1
Department of Molecular Biology, University of Aarhus, CF Moellers Alle 130, DK-8000 Aarhus C, Denmark,
2
Bioinformatics Research Center, University of Aarhus, Ny Munkegade 116, DK-8000 Aarhus C, Denmark and
3
Department of Molecular Cardiology/NB20, Lerner Research Institute, The Cleveland Clinic Foundation, OH, USA

Received February 14, 2003; Revised April 5, 2003; Accepted April 22, 2003

ABSTRACT activation of the OAS enzymes results in the synthesis of


2¢±5¢-linked oligoadenylates (2±5As) from ATP (6). In turn,
The interferon-induced 2¢±5¢-oligoadenylate synthet- these 2±5As can bind to the latent RNase L which subse-
ases (OAS) are important for the antiviral activity quently dimerises into the active form. The activated RNase L
of interferons. The human and murine OAS gene then degrades viral and cellular RNAs, suppressing protein
families each contain four genes: OAS1, OAS2, synthesis and viral growth (7). Most viruses produce dsRNA at
OAS3 and OASL, all having one or more conserved some stage in their life cycle, and it is generally believed that
OAS units composed of ®ve translated exons. The the activating dsRNA in infected cells is of viral origin (8).
OASL gene has both an OAS unit and a C-terminus Furthermore, the OAS are involved in the induction of
of two ubiquitin-like repeats. In this study, we apoptosis and control of cell growth (9,10), and studies show
demonstrate that murine Oasl1 protein is inactive that RNase L and the 2±5A system play a role in tumorigenesis
while murine Oasl2 is active as an OAS. Further- (11).
more, murine Oasl2 requires double-stranded RNA In humans, the OAS gene family is composed of four genes
located on chromosome 12 (6,12,13) The hOAS1, hOAS2 and
as co-factor. The af®nity of murine Oasl2 for the
hOAS3 genes are encoded by a tightly coupled locus on
double-stranded RNA activator is higher than that of 12q24.1. The additional member of the OAS family in humans
human OAS1 (p42 isoform). We propose a model for is the hOASL gene which is located on 12q24.2. Each OAS
the evolutionary origin of the murine Oasl1 and gene consists of a conserved OAS unit composed of ®ve
Oasl2 genes. The identi®cation of a human ortho- translated exons (exons A±E). OAS1 has one unit, whereas
logue (hOASL2) to the murine Oasl2 gene estab- OAS2 and OAS3 have two and three units, respectively, and all
lishes that the OASL gene was duplicated prior to three genes encode active 2±5A synthetases. The hOASL gene
the radiation of the rodent and primate groups. encodes a two-domain protein composed of an OAS unit fused
We suggest that murine Oasl2, which has both to a 164 amino acid C-terminal domain, homologous to a
enzymatic activity and a ubiquitin-like domain, is a tandem repeat of ubiquitin. In contrast to the different
functional intermediate between the active OAS enzymes encoded by the hOAS1, hOAS2 and hOAS3 genes,
species and the inactive human OASL1/murine no enzymatic activity has been detected for the hOASL
protein (14,15).
Oasl1 proteins. In addition, we propose that murine
Another member of the OASL class proteins was identi®ed
Oasl1 appears to have gained a hitherto unchar- in chicken (16). The ChOASL protein is active upon induction
acterized function independent of 2¢±5¢-linked by poly(I)´poly(C), and the identi®cation of this protein
oligoadenylate synthesis. divided the OASL proteins into two classes: the active and the
inactive. We have recently described the structure of the
murine Oas gene family which included two mOasl genes
INTRODUCTION (mOasl1 and mOasl2) that exist head to tail on chromosome
Interferons (IFNs) constitute an important part of the mam- 5F (17,18).
malian innate immune system (1). They confer resistance to Exons A and B of the OAS genes encode a ®ve-stranded
viral infections by regulating the transcription of a large antiparallel b-sheet domain (19, R.Hartmann, unpublished).
number of genes (2). The 2¢±5¢-oligoadenylate synthetases This domain is structurally homologous to the `palm' domain
(OAS) were among the ®rst characterised IFN-induced found in other polymerases and contains a number of amino
antiviral proteins (3±5). The OAS gene family is induced by acids crucial for nucleotide transferase activity (20). The exon
both type I and type II IFNs. The OAS proteins are expressed B of the OAS family contains a P-loop, which is a short
as latent enzymes, which require double-stranded RNA ¯exible stretch of amino acids forming a helical turn; this
(dsRNA) for activation; however, they do not harbour any motif is present in many nucleoside triphosphate-binding
of the characteristic dsRNA-binding motifs (6). This proteins such as kanamycin nucleotidyltransferase and

*To whom correspondence should be addressed. Tel: +1 216 445 6909; Fax: +1 216 445 1466; Email: rh@mb.au.dk
Correspondence may also be addressed to Just Justesen. Tel: +45 89422682; Fax: +45 89422637; Email: jj@mb.au.dk

Nucleic Acids Research, Vol. 31 No. 12 ã Oxford University Press 2003; all rights reserved
Nucleic Acids Research, 2003, Vol. 31, No. 12 3167

different polymerases (20). The P-loop is involved in the were lysed in 25 ml lysis buffer [50 mM Na2HPO4 pH 8.0,
binding of the triphosphate of the nucleotide. Mutational 500 mM NaCl, 10% glycerol, 20 mM imidazole, 0.1% (v/v)
studies have demonstrated the importance of the P-loop in NP-40, 5 mM b-mercaptoethanol and protease inhibitor
hOAS1 protein for catalytic activity (21). The second motif cocktail without EDTA (CompleteÔ, Roche Molecular
present in exon B is composed of three conserved aspartic Biochemicals, Indianapolis, IN)] using a French press. The
acids in b-strands 2 and 5; these three aspartic acids coordinate lysate was clari®ed by centrifugation, and the supernatant was
the catalytically active magnesium ions (19,21). When mixed with Ni2+-NTA±agarose beads (Qiagen) and rotated
considering the amino acid sequence of exon B encoded by for 1 h. The beads were washed (pH 8.0, 50 mM Na2HPO4,
mOasl1 and mOasl2 genes, notable differences exist. In 500 mM NaCl, 10% glycerol and 50 mM imidazole), applied
mOasl1 protein, two of the three aspartic acids are replaced, to a column (Bio-Rad, Hercules, CA) and eluted with 15 ml of
and in hOASL1 all three are substituted. Furthermore, in both elution buffer (pH 8.0, 50 mM Na2HPO4, 500 mM NaCl, 10%
mOasl1 and hOASL1, several mutations are introduced into glycerol, 250 mM imidazole) in 2 ml fractions. The fractions
the P-loop motif, explaining why the hOASL1 protein is were analysed by 10% SDS±PAGE. The eluted His-tagged
inactive as expected for mOasl1. In contrast, mOasl2 and protein was loaded on a Heparin HiTrap (5 ml) column
ChOASL proteins have both motifs conserved, suggesting that (Amersham Biosciences) (buffer A: 20 mM Tris±HCl pH 7.0,
the mOasl2 protein belongs to the class of active OASL 5% glycerol). Bound protein was eluted in a linear gradient of
proteins. an increasing concentration of NaCl (20±100%) (buffer B:
In this study, we have investigated the biochemical 20 mM Tris±HCl pH 7.0, 5% glycerol, 1 M NaCl). The
properties of mOasl1 and mOasl2 proteins. We puri®ed volume of the gradient was 10.054 ml. The fractions were
recombinant protein for examination of the 2±5A synthetase collected in 0.5 ml and analysed by SDS±PAGE (10%). The
activity and demonstrated that while mOasl1 has no activity, eluted protein was dialysed for 2 h (600 mM NaCl, 25 mM
mOasl2 is an active 2±5A synthetase that requires dsRNA. HEPES pH 6.8, 5% glycerol), diluted 1:2 with cold 100%
Furthermore, we have determined the af®nity of mOasl2 for glycerol and stored at ±20°C.
poly(I)´poly(C) in comparison with hOAS1 (p42 isoform). We
propose a model for the evolutionary origin of the mOasl1 and 2¢±5¢-Oligoadenylate synthetase activity assay by PEI
mOasl2 genes. Our data suggest that mOasl2 protein is a thin-layer chromatography
functional intermediate between the active hOAS and the The activity of the recombinant proteins was determined as
inactive hOASL1/mOasl1 proteins. In addition, mOasl1 previously described (22) with the following overall reaction
protein has gained several substitutions in exon B, resulting mixture: 4 ml of 53 OAS buffer, 4 ml of water, 4 ml of
in loss of 2±5A synthetase activity, suggesting that mOasl1 recombinant protein, 4 ml of ATP pH 7.5 including 0.025 mCi
may have obtained an novel function independent of 2±5A of [a-32P]ATP/ml, 4 ml of poly(I)´poly(C) (Amersham
synthesis. Biosciences) or water. The 53 OAS buffer contains 20 mM
Tris±HCl pH 7.5, 75 mM Mg(OAc)2, 1 mM dithiothreitol,
0.2 mM EDTA, 0.5 mg/ml bovine serum albumin, 10% (v/v)
MATERIALS AND METHODS glycerol, 30 mM creatine phosphate (Boehringer Mannheim
GmbH) and 0.5 mg/ml creatine kinase (Boehringer Mannheim
RT±PCR for determination of IFN induction GmbH). The reactions were stopped with 10 ml of 50 mM
Total RNA was puri®ed from L929 (DSMZ: accession 2) EDTA after either 1 or 2 h. Separation of the ATP and the
cells using the Midi RNEasy puri®cation kit (Qiagen, 2¢±5¢-oligomers was performed by thin-layer chromatography
Valencia, CA) according to the manufacturer's instructions. (TLC) as described previously (22). The TLC plate was
The cells were either untreated, or treated with murine IFN-a visualised using a PhosphoImagerÔ (Molecular Dynamics,
(500 U/ml) or IFN-g (100 U/ml) for 24 h. A 5 mg aliquot of Sunnyvale, CA).
total RNA was reverse-transcribed using the First strand
Activity assay using Mono Q to detect 2±5As
cDNA synthesis kit (Amersham Biosciences, Piscataway, NJ).
The PCR was carried out with 30 cycles of 96°C for 30 s, 60°C The incubations to make 2±5As were prepared in a similar
for 30 s, and 72°C for 2 min, resulting in a 1535 bp PCR way as the activity assay for TLC; however, the total volume
product for mOasl1 and a 1526 bp product for mOasl2. The was 100 ml. After incubation for 4 h, the OAS enzymes
murine b-actin gene (GenBank accession no. M12481) was were heat-inactivated, treated with alkaline phosphatase
included as control. The mOasl and control reactions were mixed (Boehringer Mannheim GmbH) and applied to a Mono Q
in equal amounts before gel electrophoresis (1% agarose). HR5/5 column (Amersham Biosciences). The different 2¢±5¢-
oligomers were eluted by a linear gradient of NaCl (0±70%)
Cloning, expression and puri®cation of recombinant (buffer B: Tris±HCl pH 7.5, 700 mM NaCl) of 19.64 ml. The
His-tagged protein 2±5As were detected at 254 nm.
The IMAGE clone 1546490 (GenBank accession no.
BE13926) was used as a template for mOasl1, and cDNA Determination of kinetic parameters
from IFN-a-induced L929 cells was used as template for In order to estimate the kinetic parameters of both mOasl2
mOasl2. The expression vectors (pET30b vector, Novagen, and hOAS1 (p42 isoform) enzymes, we used the
Madison, WI) were introduced into BL21 pRI cells by ANEMONA.XLT which is an Excel template (23). This
electroporation using standard conditions. Protein synthesis template implements among others the Michaelis±Menten
was induced by addition of 0.5 mM isopropyl-b-D- model, and the KM and Vmax parameters are calculated by non-
thiogalactoside (IPTG) overnight at room temperature. Cells linear regression performed by the least-squares method. The
3168 Nucleic Acids Research, 2003, Vol. 31, No. 12

Figure 1. (A) Schematic drawing of the hOASL and mOasl genes on human chromosome 12 and mouse chromosome 5. Genes with reading frames in the
same direction as the annotated nucleotide sequence are indicated by squares above the lines; genes below are in the reverse direction. The genes are not
drawn to scale. CEN, centromere; TEL, telomere. (B) Amino acid alignments of hOAS1 p42 (D00068), ChOASL (AB037592), hOASL1 (AJ22089), hOASL2
(NT_028327), mOasl1 (AY089728) and mOasl2 (AK010034) (numbers in parentheses indicate the GenBank accession no. for each protein). Black boxes
indicate conserved residues, grey boxes related sequences. Gaps are indicated by ±. The middle part of exon B has been cut out as indicated by black dots.
The three catalytic aspartate residues are indicated by asterisks. (C) Transcription of mOasl2 (lanes 1±3) and mOasl1 (lanes 4±6) genes. Products of PCRs
(30 cycles) with cDNA from untreated (lanes 1 and 4), IFN-a-induced (lanes 2 and 5) and IFN-g-induced (lanes 3 and 6) mouse L929 cells, analysed by gel
electrophoresis (1% agarose). The mOasl reaction and the control reaction (murine b-actin) were mixed in equal amounts before gel electrophoresis.

kcat constant de®nes the number of substrate molecules (ATP) version of the relative rate test was used employing either the
oligomerised per molecule of enzyme per second. hOAS1 sequence or ChOAS1 sequence as outgroup. All
analyses were done using MEGA 2.1 (25).
Western blotting
The His-tagged and heparin-puri®ed protein was subjected to
10% SDS±PAGE and blotted onto polyvinylidene di¯uoride RESULTS
(Immobilon-P, Millipore) membranes. The puri®ed,
recombinant proteins were detected using the monoclonal A pseudogene corresponding to mOasl2 in humans
mouse anti-His6 antibody (Boehringer Mannheim GmbH); To date, only a single OASL gene has been identi®ed in
monoclonal goat anti-mouse horseradish peroxidase (HRP)- humans, in contrast to the two genes (mOasl1 and mOasl2)
conjugated secondary antibody (Dako) was used. The blots characterised in mice. Therefore, we searched the human
were visualised using the enhanced chemiluminescence genome for a counterpart of the mOasl2 gene. This was done
method (ECL; Amersham Biosciences) according to the by performing a BLAST search of the translated human
manufacturer's instructions. genome with the amino acid sequence of mOasl2. Using this
approach, we identi®ed two exons (exons A and B) located
Alignment and phylogenetic analysis
downstream of the hOASL gene on chromosome 12 position
The alignment of sequences was done at the amino acid level 127.482 kb (exon A) and 127.476 kb (exon B) (Fig. 1A).
using default settings of the ClustalX 1.82 program, followed These exons exhibit 54 and 53% identity to exons A and B in
by manual adjustments. The following sequences were mOasl2, respectively, and their positions correspond to the
used: hOAS1 (D00068), hOASL1 (AJ225089), mOasl2 homologous position of the mOasl2 gene. The N- and
(AK010034), mOasl1 (AY089728), ChOASL (AB037592), C-termini of exon B from human, mouse and chicken OASL
Geodia cydonium OAS1 (Y18497) and a pseudogene proteins are aligned in Figure 1B, showing the important
sequence (hOASL2) from the human genome (NT_028327). amino acids in the active site. As expected for a pseudogene,
Phylogenetic trees were reconstructed with and without the several stop codons are present in hOASL2 exon A, but no
partial hOASL2 sequence, using the Geodia sequence as an traces of the remaining sequences of the hOASL2 gene were
outgroup, and the minimum evolution criterion for choosing found. We believe that the identi®ed exons represent an
the tree topology. Pairwise distances between sequences were hOASL2 pseudogene, which has not previously been
calculated assuming rate heterogeneity in evolutionary rate described.
modelled by a gamma distribution with shape parameter
Interferon induction of mOasl1 and mOasl2
a = 1.5. Only alignment columns without gaps (complete
deletion) were used. Standard bootstrap was done using 1000 In order to investigate whether the mOasl genes are induced by
replicates. For testing for differences in evolutionary rates IFNs, we extracted total RNA from IFN-a- and IFN-g-induced
between human and mouse OASL genes (24), a one-parameter and untreated mouse L929 cells. By RT±PCR studies, we
Nucleic Acids Research, 2003, Vol. 31, No. 12 3169

loaded on the heparin column and then the eluted fractions


containing the highest amount of protein were pooled (lane 2).
The identity of the protein was veri®ed by western blotting
using an anti-His tag antibody (Fig. 2C). mOasl1 protein did
not bind to the heparin; therefore, the puri®cation was limited
to nickel af®nity, followed by dialysis removing the imidazole
(data not shown).
mOasl2 possesses true 2¢±5¢-oligoadenylate synthetase
activity
The putative OAS activities of mOasl1 and mOasl2 were
tested by assays using synthetic dsRNA [poly(I)´poly(C)] as
activator. In the following experiments, the total amount of
2±5A production was measured by TLC. As shown in
Figure 3A, mOasl2 is highly active and the protein is
enzymatically active after 22 h at 37°C (Fig. 3A and data
not shown). As expected for an enzyme, a linear correlation
exists between the activity and concentration of mOasl2
enzyme (Fig. 3C). In contrast, mOasl1 was unable to
synthesize 2±5A (Fig. 3B) even after longer periods of
incubation (22 h at 37°C), hence this protein possesses no
2±5A synthetase activity.
In order to verify that the products synthesised by mOasl2
indeed are 2±5A oligomers, we employed a second method.
The reaction products were treated with alkaline phosphatase
and the oligonucleotides were separated by an anion exchange
Figure 2. Puri®cation of recombinant mOasl2 protein. (A) The elution pro- column (Mono Q). The migration of the mOasl2 products was
®le of mOasl2 using the heparin±Sepharose column for puri®cation. The
graphs show the absorption at 280 nm and the NaCl gradient from 0 to 1 M. compared with standard 2±5As produced by hOAS1. The
(B) A 10% SDS±polyacrylamide gel with 5 mM of pooled His tag fractions superimposed elution pro®les of products synthesised by
(lane 1) and 5 mM of pooled fractions from the heparin column (lane 2) was mOasl2 and hOAS1 were identical (Fig. 3D). mOasl2 is able
stained with Coomassie brilliant blue. The size of the molecular markers is to synthesise dimeric (pppApA), trimeric (pppApApA) and
indicated. (C) Western blot analysis of the samples loaded in (B). They
were visualised using an anti-His tag antibody and a secondary goat
longer oligomers after an extended incubation time (22 h)
anti-mouse HRP-conjugated antibody. (data not shown).
The activity of mOasl2 is dependent on dsRNA
In the previously described experiments, we have used the
show that mOasl1 and mOasl2 are induced by IFN-a in synthetic dsRNA poly(I)´poly(C) as an activator. Next, we
murine L929 cells; additionally, mOasl2 is induced by IFN-g investigated whether mOasl2, like all the human 2±5A
although to a lower extent (Fig. 1C). No transcript was synthetases, needs dsRNA as co-factor. Dose±response curves
detected in untreated L929 cells with the PCR conditions used using poly(I)´poly(C) as activator of mOasl2 and hOAS1 are
here. We have no evidence of the p54/mOasl2 gene identi®ed presented in Figure 4A and B, respectively. These curves
by Tiefenthaler et al. (26) (GenBank accession no. AF068835) clearly indicate that the 2±5A synthetase activity of
in IFN-induced cells. This p54/mOasl2 variant has a shorter mOasl2 is activated by increasing amounts of dsRNA and
ubiquitin-like domain (1.5 ubiquitin-like repeat). A poly- that without dsRNA, no 2±5A was produced. hOAS1 (p42
morphism giving allelic variation in the different mouse cells isoform) was included for comparison. Hence mOasl2 needs
may explain the two forms of mOasl2. The following analysis dsRNA/poly(I)´poly(C) as a cofactor for 2±5A synthetase
is based solely on the mOasl2 gene with two ubiquitin-like activity.
repeats (GenBank accession no. AK010034). From the dose±response curves, we have estimated the
maximal velocity (Vmax) and the apparent dissociation
Puri®cation of active murine Oasl2 protein constant (Kapp) by non-linear curve ®t. The Kapp de®nes the
concentration of dsRNA activator, which induces 1/2 Vmax
To explore the biochemical properties of mOasl2, we cloned activity. The estimated parameters using either poly(I)´
the cDNA from IFN-a-induced L929 cells. N-terminal His- poly(C), poly(A)´poly(U) or poly(GU) are presented in
tagged protein was expressed in Escherichia coli at room Table 1. mOasl2 has a lower Kapp for poly(I)´poly(C) than
temperature to avoid storage of protein in inclusion bodies. hOAS1 (p42 isoform); consequently, our data suggest that
The ®rst puri®cation step was His tag puri®cation using Ni2+- mOasl2 has higher af®nity for poly(I)´poly(C) than hOAS1.
NTA beads. mOasl2 was puri®ed further by af®nity We continued the investigation of the dsRNA binding of
chromatography using a heparin column to remove several mOasl2 by using poly(A)´poly(U) which has been shown to
putative degradation products. The elution pro®le of the activate hOAS1, although to a lower extent than poly(I)´
heparin column is shown in Figure 2A. A sample of the pooled poly(C) (27). Our experiments show that mOasl2, on the
His tag fractions is shown in Figure 2B (lane 1). This was contrary, is not activated by poly(A)´poly(U) to any extent.
3170 Nucleic Acids Research, 2003, Vol. 31, No. 12

Figure 4. mOasl2 is induced by poly(I)´poly(C). (A) Dose±response curve


for poly(I)´poly(C) induction of mOasl2 (2.5 ng/ml). (B) Dose±response
curve for poly(I)´poly(C) induction of hOAS1 (2.5 ng/ml).

Table 1. Kapp for induction by different dsRNA


Kappa mOasl2 p42

Poly(I)´poly(C) 0.13 mg/ml 1.40 mg/ml


Poly(A)´poly(U) No activation 25.00 mg/ml
Poly(GU) No activation No activation
Figure 3. mOasl2 is active whereas mOasl1 is not. (A) ATP was separated aThe
from the produced 2±5A products (pppApA and pppApApA) synthesised by apparent dissociation constant (Kapp) was estimated by a non-linear
mOasl2 using TLC. Increasing amounts of mOasl2 protein were added (0, curve ®t.
0.0007, 0.0014, 0.0028, 0.0056, 0.0112 and 0.0224 mg/ml) with a ®nal con-
centration of 100 mg/ml poly(I)´poly(C). Cont., contaminations present in all
reactions. (B) 2±5A synthetase activity assay using mOasl1 protein, and Table 2. Kinetic parameters
nucleotides separated by TLC. The same experimental conditions as in (A). mOasl2 p42
(C) The amount of 2±5A produced by mOasl2 in (A) was quanti®ed and
normalised to the ATP spot. A linear correlation exists between the activity KM(ATP) 0.66 mM 0.31 mM
(nmol ATP/min) and the concentration of mOasl2 (mg). The experiment was kcat 0.39 s±1 7.20 s±1
repeated twice. (D) The heat-inactivated 2±5A synthetase reaction [®nal
concentration of 100 mg/ml poly(I)´poly(C)] was treated with alkaline phos-
phatase and separated using a Mono Q column. The elution pro®les of the
products produced by mOasl2 (2.5 ng/ml) (blue) and p42 (2.5 ng/ml) (red)
are superimposed. 1, adenylate; 2, pppApA; 3, pppA(pA)2; 4, pppA(pA)3; 5,
pppA(pA)4; 6, pppA(pA)5; and 7, pppA(pA)6. enzyme. By ®tting our data to this equation, we were able to
estimate the KM values as shown in Table 2. The KM for
mOasl2 with ATP as substrate is 0.66 mM and is 0.31 mM for
hOAS1.
We also investigated whether poly(GU) can activate mOasl2
even though it does not activate hOAS1. Our studies show no Phylogenetic analysis
activation of mOasl2 with poly(GU). The identi®cation of the hOASL2 pseudogene establishes that
the duplication of the OASL gene has occurred prior to the
Determination of kinetic parameters [KM(ATP) and kcat] radiation of the rodent and primate groups. However, subse-
We continued our biochemical analyses of mOasl2 by quently during evolution, the hOASL2 gene has evolved into a
determination of the kinetic parameters. We plotted the pseudogene and, during this process, exons C±E and T have
amount of synthesized 2±5A as a function of the ATP been lost and several stop codons have arisen in exon A. We
substrate concentration. The Michaelis±Menten equation performed a phylogenetic analysis by aligning mOasl1,
proposes a model for estimation of substrate af®nity of an mOasl2, human p42 (OAS1), hOASL1, hOASL2 (exon B
Nucleic Acids Research, 2003, Vol. 31, No. 12 3171

DISCUSSION
This study was performed to understand the biochemical
properties of the two mOasl proteins. We have cloned and
puri®ed protein encoded by the mOasl1 and mOasl2 genes and
characterised their 2±5A synthetase activity. Our results
demonstrate that the mOasl2 protein is an active 2±5A
synthetase requiring dsRNA as cofactor for activity. As
expected from sequence comparisons, mOasl1 is inactive like
its human orthologue, the hOASL1 protein (15). Thus it is
tempting to speculate that mOasl1 is the functional
counterpart of hOASL1.
The OASL proteins consist of an OAS domain and a
C-terminal domain of two ubiquitin-like repeats. They have
been identi®ed in human, mouse and chicken, and thus we
expect this family to exist throughout the mammalian and
avian classes. The function of the ubiquitin-like domain of the
OASL proteins is unknown. Mice have two mOasl genes,
mOasl1 and mOasl2, with striking differences observed in the
sequences of exon B of these genes. As described in the
Introduction, the P-loop motif and a triad of aspartic acids
coordinating the catalytic active magnesium ions are present
Figure 5. Phylogenetic analyses. (A) Phylogenetic tree of exons A±E. in exon B. Both these motifs have been altered in the mOasl1
Bootstrap values are based on 1000 bootstrap replications. Sequences from protein, whereas they are retained in the mOasl2 protein (see
mOasl2 (AK010034), mOasl1 (AY089725), hOAS1 p42 (D00068), hOASL also Fig. 1B).
(AJ22089), hOASL2 (NT_028327), ChOASL (AB037592) and Geodia
OAS1 (Y18497) are aligned (numbers in parentheses indicate the GenBank
In comparison with hOAS1, the speci®c activity of mOasl2
accession no. for each protein). (B) Phylogenetic tree of exon B. is ~20-fold lower; this could at least partly be due to evolution
of the mOasl2 protein towards a 2±5A-independent function.
The KM we have determined for mOasl2 is marginally higher
than for human p42; however, it is still signi®cantly lower than
only), ChOASL and the OAS gene from the marine sponge the KM for both human p69 OAS2 and p100 OAS3 proteins
G.cydonium (28). In the subsequent phylogenetic analysis, the (30).
Geodia OAS sequence was used as an outgroup to root the Previous workers have shown that mOasl2 protein only
trees. synthesises dimeric 2±5As (pppApA) (18) whereas we report
Phylogenetic trees were constructed for the complete gene that mOasl2 is able to synthesise up to pentamers, although the
(Fig. 5A) and for exon B (Fig. 5B) only, using the minimum major product is dimeric 2±5As. We use creatine kinase and
evolution criterion on a gamma distance matrix (a = 1.5). creatine phosphate in our OAS assays to regenerate ATP from
Analysis on exon B alone allowed us to include the hOASL2 ADP and AMP. Since ADP and AMP may be inhibitory to
psudogene. Figure 5B shows that mOasl1/hOASL1 and mOasl2, this can explain the lower activity observed in the
mOasl2/hOASL2 group together with high bootstrap support, experiments of Kakuta et al. (18). The activity of mOasl2 is
suggesting that they indeed represent orthologous pairs of more sensitive towards ADP and AMP inhibition than human
genes and that the duplication occurred prior to the diversi- p42 under the same experimental conditions (data not shown).
®cation of human and mouse but before the diversi®cation of Thus it is crucial to avoid degradation of ATP in the reaction
mammals and birds. Bootstrap support of the shown topology when investigating the biochemistry of mOasl2. The assay
was robust to other choices of a (between 1 and 5) and to other system used here has the advantage that an eventual degrad-
principles of phylogenetic reconstruction (including likeli- ation of ATP is visualised directly on the chromatogram.
hood methods). The non-parametric relative rates test (24) We have measured a higher af®nity of mOasl2 for
revealed that mOasl1 has evolved signi®cantly faster than poly(I)´poly(C) compared with hOAS1. This supports pub-
mOasl2 since their divergence (45 versus 28 changes, P < lished results from other groups showing that the hOAS1
0.05). This agrees well with the fact that mOasl2 has retained protein in general requires the highest concentrations of
activity and therefore has more common constraints with the dsRNA for activation among the OAS family members (30).
ChOASL and hOAS1 sequences. Previous analysis of the OAS The hOAS2 protein requires intermediate concentrations of
family made by Kumar et al. (29) showed that the evolution- dsRNA for activation, and the OAS3 proteins are the most
ary distance between hOASL1 and mOasl2 is longer than sensitive to dsRNA (31). It should be noted that large batch to
expected for two orthologous genes, which led them to suggest batch variations of commercially available poly(I)´poly(C)
that the two genes were paralogues. This was later proven to exist, making comparison of the absolute values for
be true by the identi®cation of the mOasl1 gene. For exon B, poly(I)´poly(C) dif®cult. In addition, mOasl2 was not activ-
the relative rate test suggests not surprisingly that the hOASL2 ated by poly(A)´poly(U), in contrast to hOAS1 which was
pseudogene has evolved faster than the mOASL2 sequence activated by this dsRNA although higher concentrations were
since their divergence, though not signi®cantly so (15 versus 7 required. This suggests that the dsRNA recognition mechan-
changes, P = 0.08). ism might differ between the mOasl2 and hOAS1 proteins.
3172 Nucleic Acids Research, 2003, Vol. 31, No. 12

non-essential. Thus we suggest that mOasl2 having both


activity and an ubiquitin-like domain is a functional inter-
mediate between the active hOAS proteins and the inactive
hOASL1/mOasl1 proteins.
In contrast, accumulations of amino acid substitutions in the
mOasl1 gene led to loss of 2±5A synthetase activity. Since
both hOASL1 and mOasl1 proteins are inactive, the loss of
activity occurred in the common ancestor of man and mouse.
Sequence comparisons and functional data demonstrate that
the hOASL1 gene is an orthologue to the mOasl1 gene. The
most common evolutionary fate of gene duplicates is silencing
(32); since this does not seem to be the case for the mOasl1
gene, it appears that mOasl1 has acquired a putative novel
function which is independent of 2±5A synthetase activity.
Preliminary data suggest that this function requires the
ubiquitin-like domain and that hOASL1 has an antiviral
activity conferred by the ubiquitin-like domain (33).
Figure 6. Model for the evolutionary origin of the OASL1 and OASL2
genes. ACKNOWLEDGEMENTS
The authors thank Lisbet Kjeldberg for excellent assistance
with enzymatic measurements, Niels Ole Kjeldgaard for
Previously published results by Tiefenthaler et al. claimed stimulating and strong interest, and Robert H. Silverman for
that the activity of mOasl2 was independent of dsRNA (26). critical reading of this manuscript. This work was supported
We were unable to detect any activity of the mOasl2 protein in by the Danish Natural Science Research Council, The Novo-
the absence of dsRNA. Furthermore, Tiefenthaler et al. did not Nordisk Foundation and the Danish Cancer Society.
observe any increase in the 2±5A synthetase activity upon
stimulation with poly(I)´poly(C), whereas we observed a
strong and dose-dependent activation of the mOasl2 enzyme REFERENCES
by poly(I)´poly(C). The speci®c activity of the enzyme used 1. Stark,G.R., Kerr,I.M., Williams,B.R., Silverman,R.H. and Schreiber,R.D.
by Tiefenthaler et al., although not stated in the paper, appears (1998) How cells respond to interferons. Annu. Rev. Biochem., 67,
low, since they use 0.5 mg of protein in contrast to the 0.05 mg 227±264.
2. Der,S.D., Zhou,A., Williams,B.R. and Silverman,R.H. (1998)
used here. The method we utilised for puri®cation of Identi®cation of genes differentially regulated by interferon alpha, beta,
recombinant mOasl2 protein is optimised for expressing the or gamma using oligonucleotide arrays. Proc. Natl Acad. Sci. USA, 95,
protein at low temperatures, which is essential for recovering 15623±15628.
active protein. However, at present, we are unable to explain 3. Roberts,W.K., Hovanessian,A., Brown,R.E., Clemens,M.J. and Kerr,I.M
the dsRNA-independent activity observed by Tiefenthaler (1976) Interferon-mediated protein kinase and low-molecular-weight
inhibitor of protein synthesis. Nature, 264, 477±480.
et al. (26) 4. Hovanessian,A., Brown,R.E. and Kerr,I.M. (1977) Synthesis of low
Based on our phylogenetic analyses, we propose a model molecular weight inhibitor of protein synthesis with enzyme from
for the evolutionary origin of the mOasl1 and mOasl2 genes. A interferon-treated cells. Nature, 268, 537±540.
duplication of the ancestral OAS gene followed by fusion to 5. Chebath,J., Benech,P., Revel,M. and Vigneron,M. (1987) Constitutive
expression of (2¢±5¢) oligo A synthetase confers resistance to
the ubiquitin-like domain by exon shuf¯ing resulted in the picornavirus infection. Nature, 330, 587±588.
formation of the original OASL gene (Fig. 6). This gene 6. Justesen,J., Hartmann,R. and Kjeldgaard,N.O. (2000) Gene structure and
possesses both the ubiquitin-like domain and a catalytically function of the 2¢±5¢-oligoadenylate synthetase family. Cell. Mol. Life
active OAS domain, as is the case for both mOasl2 and Sci., 57, 1593±1612.
7. Dong,B. and Silverman,R.H. (1995) 2±5A-dependent RNase molecules
ChOASL genes. Duplication of the ancestral OASL gene led to dimerize during activation by 2±5A. J. Biol. Chem., 270, 4133±4137.
the precursors of mOasl1 and mOasl2. This duplication 8. Coccia,E.M., Romeo,G., Nissim,A., Marziali,G., Albertini,R.,
occurred either prior to the establishment of the mammalian Affabris,E., Battistini,A., Fiorucci,G., Orsatti,R. and Rossi,G.B. (1990) A
class or early during mammalian evolution prior to the full-length murine 2±5A synthetase cDNA transfected in NIH-3T3 cells
radiation of the rodent and primate groups, as demonstrated by impairs EMCV but not VSV replication. Virology, 179, 228±233.
9. Ghosh,A., Sarkar,S.N., Rowe,T.M. and Sen,G.C. (2001) A speci®c
the identi®cation of an hOASL2 pseudogene. After the isozyme of 2¢±5¢ oligoadenylate synthetase is a dual function
duplication of the ancestral OASL gene, mOasl2 has main- proapoptotic protein of the Bcl-2 family. J. Biol. Chem., 276,
tained the 2±5A synthetase activity, which is the putative 25447±25455.
original function of the OASL protein. This is manifested in 10. Castelli,J.C., Hassel,B.A., Maran,A., Paranjape,J., Hewitt,J.A., Li,X.L.,
Hsu,Y.T., Silverman,R.H. and Youle,R.J. (1998) The role of 2¢±5¢
the conservation of the catalytically important amino acids in oligoadenylate-activated ribonuclease L in apoptosis. Cell Death Differ.,
the sequence of exon B. Some substitutions of amino acids 5, 313±320.
have occurred outside exon B, explaining why mOasl2 is more 11. Carpten,J., Nupponen,N., Isaacs,S., Sood,R., Robbins,C., Xu,J.,
closely related to the mOasl1/hOASL1 proteins when con- Faruque,M., Moses,T., Ewing,C., Gillanders,E. et al. (2002) Germline
mutations in the ribonuclease L gene in families showing linkage with
sidering the entire OAS unit. The hOASL2 gene is not HPC1. Nature Genet., 30, 181±184.
functional in humans, and only exons A and B remain as a 12. Hovnanian,A., Rebouillat,D., Mattei,M.G., Levy,E.R., Marie,I.,
pseudogene. Hence, at least in humans, the hOASL2 protein is Monaco,A.P. and Hovanessian.A.G. (1998) The human 2¢,5¢-
Nucleic Acids Research, 2003, Vol. 31, No. 12 3173

oligoadenylate synthetase locus is composed of three distinct genes products, 2¢5¢ oligoadenylates and 2¢5¢ co-oligonucleotides. Nucleic
clustered on chromosome 12q24.2 encoding the 100-, 69- and 40-kDa Acids Res., 8, 3073±3085.
forms. Genomics, 52, 267±277. 23. HernaÂndez,A. and Ruiz,M.T. (1998) An EXCEL template for calculation
13. Hovnanian,A,, Rebouillat,D,, Levy,E,R,, Mattei,M,G. and of enzyme kinetic parameters by non-linear regression. Bioinformatics,
Hovanessian,A.G. (1999) The human 2¢,5¢-oligoadenylate synthetase-like 14, 227±228.
gene (OASL) encoding the interferon-induced 56-kDa protein maps to 24. Tajima,F. (1993) Simple methods for testing the molecular evolutionary
chromosome 12q24.2 in the proximity of the 2¢,5¢-OAS locus. Genomics, clock hypothesis. Genetics, 135, 599±607.
56, 362±363. 25. Kumar,S., Tamura,K., Jakobsen,I.B. and Nei,M. (2001) MEGA2:
14. Hartmann,R., Olsen,H.S., Widder,S., Jorgensen,R. and Justesen,J. (1998) molecular evolutionary genetics analysis software. Bioinformatics, 17,
p59OASL, a 2¢±5¢ oligoadenylate synthetase like protein: a novel human 1244±1245.
gene related to the 2¢±5¢ oligoadenylate synthetase family. Nucleic Acids 26. Tiefenthaler,M., Marksteiner,R., Neyer,S., Koch,F., Hofer,S., Schuler,G.,
Res., 26, 4121±4128. Nussenzweig,M., Schneider,R. and Heu¯er,C. (1999) M1204, a novel
15. Rebouillat,D., Marie,I. and Hovanessian,A.G. (1998) Molecular cloning 2¢,5¢ oligoadenylate synthetase with a ubiquitin-like extension, is induced
and characterization of two related and interferon-induced 56-kDa and during maturation of murine dendritic cells. J. Immunol., 163, 760±765.
30-kDa proteins highly similar to 2¢±5¢ oligoadenylate. Eur. J. Biochem., 27. Baglioni,C., Minks,M.A. and De,Clercq (1981) Structural requirements
257, 319±330. of polynucleotides for the activation of (2¢±5¢)An polymerase and protein
16. Yamamoto,A., Iwata,A., Koh,Y., Kawai,S., Murayama,S., Hamada,K., kinase. Nucleic Acids Res., 9, 4939±4950.
28. Wiens,M., Kuusksalu,A., Kelve,M. and Muller,W.E. (1999) Origin of the
Maekawa,S., Ueda,S. and Sokawa,Y. (1998) Two types of chicken 2¢,5¢-
interferon-inducible (2¢±5¢)oligoadenylate synthetases: cloning of the
oligoadenylate synthetase mRNA derived from alleles at a single locus.
(2¢±5¢)oligoadenylate synthetase from the marine sponge Geodia
Biochim. Biophys Acta, 1395, 181±191.
cydonium. FEBS Lett., 462, 12±18.
17. Eskildsen,S., Hartmann,R., Kjeldgaard,N.O. and Justesen,J. (2002) Gene 29. Kumar,S., Mitnik,C., Valente,G. and Smith,F. (2000) Expansion and
structure of the murine 2¢±5¢-oligoadenylate synthetase family. Cell. Mol. molecular evolution of the interferon-induced 2¢±5¢ oligoadenylate
Life Sci., 59, 1212±1222. synthetase gene family. Mol. Biol. Evol., 17, 738±750.
18. Kakuta,S., Shibata,S. and Iwakura,Y. (2002) Genomic structure of the 30. Sarkar,S.N. and Sen,G.C. (1998) Production, puri®cation and
mouse 2¢,5¢-oligoadenylate synthetase gene family. J. Interferon characterization of recombinant 2¢, 5¢-oligoadenylate synthetases.
Cytokine Res., 22, 981±993. Methods, 15, 233±242.
19. Sarkar,S.N., Ghosh,A., Wang,H.W., Sung,S.S. and Sen,G.C. (1999) The 31. Marie,I., Blanco,J., Rebouillat,D. and Hovanessian,A.G. (1997) 69-kDa
nature of the catalytic domain of 2¢±5¢-oligoadenylate synthetases. J. Biol. and 100-kDa isoforms of interferon-induced (2¢±5¢)oligoadenylate
Chem., 274, 25535±25542. synthetase exhibit differential catalytic parameters. Eur. J. Biochem.,
20. Holm,L. and Sander,C. (1995) DNA polymerase beta belongs to an 248, 558±566.
ancient nucleotidyltransferase superfamily. Trends Biochem. Sci., 20, 32. Lynch,M. and Conery,J.S. (2000) The evolutionary fate and
345±347. consequences of duplicate genes. Science, 290, 1151±1155.
21. Yamamoto,Y., Sono,D. and Sokawa,Y. (2000) Effects of speci®c 33. Hartmann,R., Rebouillat,D., Justesen,J., Sen,S. and Williams,B. (2001)
mutations in active site motifs of 2¢,5¢-oligoadenylate synthetase on The p59 oligoadenylate synthetase like protein (p59OASL) does not
enzymatic activity. J. Interferon Cytokine Res., 20, 337±340. display oligoadenylate synthetase activity but possesses antiviral
22. Justesen,J., Ferbus,D. and Thang,M.N. (1980) 2¢5¢ oligoadenylate properties conferred by an ubiquitin-like domain. J. Interferon Cytokine
synthetase, an interferon induced enzyme: direct assay methods for the Res., 9 (suppl.), 6±8.

You might also like