Anda di halaman 1dari 21

FEMS Microbiology Reviews, fuv004, 39, 2015, 280300

doi: 10.1093/femsre/fuv004
Review Article

REVIEW ARTICLE

Bacterial transfer RNAs


Jennifer Shepherd and Michael Ibba
Department of Microbiology and the Center for RNA Biology, Ohio State University, Columbus, Ohio 43210, USA
Corresponding author: Department of Microbiology, Ohio State University 318 West 12th Avenue, Columbus, Ohio 43210, USA. Tel: +614-292-2120;

Fax: +614-292-8120; E-mail: ibba.1@osu.edu


One sentence summary: This review aims to bring together the results of intensive investigations into the synthesis, maturation, modification,
aminoacylation, editing and recycling of bacterial transfer RNAs.
Editor: Wolfgang Hess

ABSTRACT
Transfer RNA is an essential adapter molecule that is found across all three domains of life. The primary role of transfer
RNA resides in its critical involvement in the accurate translation of messenger RNA codons during protein synthesis and,
therefore, ultimately in the determination of cellular gene expression. This review aims to bring together the results of
intensive investigations into the synthesis, maturation, modification, aminoacylation, editing and recycling of bacterial
transfer RNAs. Codon recognition at the ribosome as well as the ever-increasing number of alternative roles for transfer
RNA outside of translation will be discussed in the specific context of bacterial cells.
Keywords: aminoacylation; codon; modification; protein synthesis; translation; tRNA

INTRODUCTION TO tRNA
Transfer ribonucleic acid (tRNA) is an essential component of
the cellular protein synthesis machinery that was found at the
and Abelson 1979; Soll
and
origin of life (Altman 1978; Soll
RajBhandary 1995). Primary research on tRNA can be dated back
to the early 1950s when Hoagland et al. first determined the presence of an enzyme in the pH 5 insoluble fraction of a rat liver extract that could activate amino acids in an ATP-dependent manner with the concurrent production of aminoacyl-adenylates
(Hoagland, Keller and Zamecnik 1956; Hoagland, Zamecnik and
Stephenson 1957; Hoagland et al. 1958). After this initial finding,
further work in the Zamecnik laboratory demonstrated that the
amino acid acceptor molecule in the extract was in fact tRNA (reviewed in RajBhandary and Kohrer 2006). The first sequence of
a tRNA molecule was made available in 1965 after extensive research in the yeast model system (Holley 1965; Holley et al. 1965).
Since then, enhancement in column chromatography and gel
electrophoresis methodologies has resulted in rapid progress in
both purification and sequence determination of individual cellular tRNAs (Cherayil and Bock 1965; Gillam et al. 1967; Holladay,
Pearson and Kelmers 1971; Pearson, Weiss and Kelmers 1971;

Ikemura and Dahlberg 1973). Functionally, each tRNA molecule


can be viewed as a specific adaptor that ultimately results in
the productive decoding of messenger RNA to protein. This is
achieved by transportation of appropriate amino acids to an
elongating polypeptide chain that is associated with the ribosome (Fig. 1) (Hopper and Phizicky 2003).
Before accurate recognition by the aminoacyl-tRNA synthetases (aaRSs) and other components of the protein synthesis machinery can occur, each tRNA must first undergo a specific maturation process. Maturation ensures that the structural
integrity and composition of the final tRNA molecule is conductive to its overall function. In all kingdoms of life, maturation of tRNA precursors is comprised of multiple steps. Prior
to aminoacylation of the 3 end by the aaRSs, each synthesized
tRNA precursor transcript must be efficiently processed at both
the 5 and the 3 ends followed, in most cases, by additional
modifications at specific residues within the structure (Grosjean
2009). Following these processes, mature aminoacylated tRNA
molecules will either proceed directly into protein synthesis or
a non-coding pathway such as those concerned with peptidoglycan biosynthesis, antibiotic biosynthesis and antibiotic resistance (Shepherd and Ibba 2013b). In a select number of cases, the

Received: 3 November 2014; Accepted: 21 January 2015



C FEMS 2015. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

280

Shepherd and Ibba

281

Figure 1. The typical life cycle of a bacterial transfer RNA. The numbering system corresponds to the specific sections of this review. Abbreviations include aminoacyltRNA synthetase (aaRS), inorganic pyrophosphate (PPi), amino acid (aa) and elongation factor-TU (EF-Tu).

aminoacylated tRNA molecule will be required to undergo additional maturation steps prior to participation in these pathways (Blanquet et al. 2003). This review will focus specifically on
bringing together the key findings that have enhanced our understanding of the processes involved in the production of mature functional bacterial tRNA molecules and their interaction
with the ribosome and other biosynthetic pathways.

Synthesis of tRNA
In bacteria, the genetic material encoding tRNA is most frequently found in clusters on the chromosome and the presence
of multiple gene copies for a single species is common. To date,
three different tRNA-encoding transcription units have been described in the literature encompassing those that just contain
tRNA genes, those that contain tRNA genes in the spacer regions of ribosomal RNA operons and tRNA operons that also
include specific protein-encoding genes such as elongation factor Tu (EF-Tu) (Toh et al. 2001). These genetic structures enable
RNA polymerase-mediated transcription of a tRNA from a single promoter into precursors that are typically longer than the
final mature molecule (Altman 1975). Whilst they contain the
information required for constitution of a functionally unique
cellular molecule, in structural terms the genes encoding for
transfer RNA are often small ranging from only 75 to 90 base
pairs in length. The overall nucleotide sequence varies significantly between each tRNA gene but there are always either
four or five inverted repeats that are responsible for formation
of the stem-loop elements found within each cloverleaf structure (Altman 1978; Fournier and Ozeki 1985). In bacteria, includ-

ing Escherichia coli, the tRNA gene often encodes the critical 3
CCA sequence. This removes the requirement seen in eukaryotic systems whereby this feature must be specifically added to
the molecule post-transcriptionally. Aside from all of these tertiary structure-encoding components, tRNA genes also contain
specific recognition elements that are necessary for recruitment
of enzymes involved in precursor maturation, protein synthesis
and, in some cases, other non-translational pathways.
In terms of the necessity of specific promoter elements, it has
been demonstrated that the upstream regions of both E. coli and
Mycobacterium capricolum tRNA transcription units contain the
consensus regions of typical bacterial promoters including the
35 and 10 elements. In addition, the spacing between these
elements is consistent with that found for other bacterial pro and RajBhandary
moters, ranging from 15 to 18 nucleotides (Soll
1995). A conserved domain consisting of a 7 base pair sequence
that is GC rich and associated with the stringent control response has been identified in the promoter region of all E. coli
tRNA genes (Ikemura and Dahlberg 1973a,b). When focusing on
termination of tRNA transcription, DNA sequence analysis has
determined that, in both E. coli and M. capricolum, there are rhoindependent terminator structures that consist of a dyad symmetrical sequence and T-residues downstream of most of the
and RajBhandary 1995). However, it is thought
tRNA genes (Soll
that transcription of some tRNA genes, such as tRNATyr in E. coli,
may require rho-dependent termination. Further research is required to determine the specific details of the transcriptional
initiation and termination processes that lead to the successful
generation of immature tRNA precursor molecules in different
bacteria.

282

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

Processing of tRNA precursors


Following their transcription, tRNA gene products often have
extensions at both the 5 and the 3 ends. In addition, many
bacterial transcripts are polycistronic in nature, containing
more than one tRNA species (Li and Deutscher 2002; Ow and
Kushner 2002). This results in the requirement for processing,
which proceeds by a series of endonucleolytic cleavage events
(Gegenheimer and Apirion 1981; Grosjean 2009). Substantial
amounts of intact tRNA precursor transcripts have not been observed in wild-type bacterial cells grown under normal conditions. This suggests that all of these endonucleolytic tRNA precursor processing events happen either during or immediately
after transcription (Altman and Smith 1971).
Across all domains of life, essential 5 tRNA transcript processing is carried out by a ribonucleoprotein called RNase P (reviewed in detail in Altman and Kirsebom 1999). To date, the
only known exception to the requirement for RNase P activity is
Nanoarchaeum equitans, which is presumed to be able to produce
leaderless tRNA transcripts (Randau and Soll 2008). In bacteria
RNase P was initially found to play a part in tRNA processing
when it was shown to specifically remove a 41 base pair fragment from the 5 end of E. coli tRNATyr leaving the 3 end intact
(Altman 1971; Altman and Smith 1971). Subsequent analysis of
tRNA precursors accumulated in a temperature-sensitive RNase
P mutant strain clarified the essential role of the enzyme in maturation of the 5 end of all E. coli tRNA species (Altman 1975).
With a few exceptions (Gupta 1984; Lee et al. 1987), maturation
of tRNA by RNase P processing results in the generation of a precursor molecule with a monophosphate at the 5 end and a terminal 2 -3 -cis glycol.
In structural terms, RNase P is defined as a ribozyme and
it has been demonstrated to have surprisingly versatile substrate specificity (reviewed in Kirsebom 2002; Harris and Christian 2003; Kazantsev and Pace 2006; Torres-Larios et al. 2006). Unlike many other naturally large ribozymes, RNase P can carry out
multiple turnovers in vivo, similar to the ribosome. Whilst RNase
P activity is conserved across all domains of life, the protein
composition of the enzyme is known to vary depending upon
whether it is bacterial, archaeal or eukaryotic in nature. In bacteria, RNase P is comprised of one small basic protein that is approximately 18 000 Daltons in size (Stark et al. 1978; Guthrie and
Atchison 1980). In archaea there are often 4 to 5 protein subunits
and in eukarya the number of protein subunits can be as many
as 10 (Hartmann and Hartmann 2003). The catalytic activity of
the 375377 nucleotide RNA component of RNase P was first described in 1983 and, in bacteria, this activity can be achieved
even in the absence of the protein component of the enzyme
(Stark et al. 1978; Guthrie and Atchison 1980; Guerrier-Takada
et al. 1983). Extensive biochemical characterization has indicated
that RNase P catalysis proceeds by an SN 2 nucleophilic substitution mechanism (Smith and Pace 1993; Beebe and Fierke 1994).
More specifically, magnesium ion-hydrate coordinate complexes
surround the target phosphodiester bond of the tRNA precursor
molecule resulting in the formation of a trigonal bipyramidal
transition state (Persson, Cuzic and Hartmann 2003; Cassano,
Anderson and Harris 2004). This subsequently enables the 2 hydroxyl group of the precursor tRNA to facilitate protonation
of the leaving 3 oxygen, thus finalizing removal of the leader sequence and signaling completion of the reaction (reviewed in
detail in Kazantsev and Pace 2006). Three specific universal features of the tRNA substrate have been implicated in accurate
recognition by the RNA component of bacterial RNase P. These
are the TC loop, the acceptor stem and the CCA end (Grosjean
2009).

Determination of the X-ray crystal structures of some examples of RNase P has resulted in the conclusion that the bacterial versions of the enzyme can be classified into two groups
(Krasilnikov et al. 2003, 2004; Kazantsev et al. 2005; Torres-Larios
et al. 2005; Kazantsev and Pace 2006; Baird et al. 2007). Type A, or
ancestral type RNase P, is widely dispersed throughout many of
the phylogenetic branches of the bacterial kingdom. In contrast,
type B or Bacillus type RNase P is restricted to Gram-positive bacteria (Pace and Brown 1995; Haas and Brown 1998; Baird et al.
2007). Whilst there are differences that focus around certain peripheral elements of the structures of type A and B RNase P proteins, both groups have a common catalytic core and tertiary fold
(reviewed in detail in Kazantsev and Pace 2006; Grosjean 2009).
Within the catalytic core, there are a series of highly conserved
residues. These residues occur within five separate regions of
sequence conservation encoding parts of the overall structure
that are involved in helix formation or the establishment of critical helical contacts. Structural studies on the protein components of type A and B RNase P have demonstrated that they
share both structural and functional similarity. This is further
supported by in vitro and in vivo studies that demonstrate efficient functional complementation of the protein component of
a type A RNase P with that of a type B RNase P and vice versa
(Waugh and Pace 1990; Wegscheid, Condon and Hartmann 2006).
The protein components of RNase P are known to complete the
typical ribonucleoprotein structure by folding into an - sandwich with a highly conserved electrostatic surface (Niranjanakumari et al. 1998; Stams et al. 1998; Spitzfaden et al. 2000; Kazantsev et al. 2003). A combination of mutational and biochemical
data has indicated that the unusual left-handed -- crossover
formed by the protein component produces a positively charged
face that binds to the RNA component of the enzyme (Stams
et al. 1998). Whilst the protein component of bacterial RNase P is
not required for catalytic activity in vitro, its overall importance
has been linked to acceleration of the tRNA processing reaction.
This suggests that it has a role in stabilization of the RNA component and possibly the reaction intermediates in addition to a
specific sequence in the precursor RNA in the C5 component of
RNase P (Walker and Engelke 2006; Smith, Hsieh and Fierke 2007;
Guenther et al. 2013).
When the tRNA precursor molecule contains one tRNA sequence, RNase P processing may be the only requirement for
efficient maturation of the 5 end. If the tRNA transcript contains more than one tRNA sequence other endoribonucleases,
such as RNase E and RNase III, may be required to generate
smaller precursor molecules. These smaller precursors can then
be much more efficiently processed by RNase P to generate a
tRNA molecule with a mature 5 end. Subsequent processing by
exoribonucleases, including RNases II, BN, D, PH, PNPase and T,
then account for generation of the mature 3 end (Sakano and
Shimura 1975, 1978; Schedl, Roberts and Primakoff 1976). Recently, RNase P-mediated separation of some E. coli transcripts
including valV valW, leuQ leuP leuV and secG leuU has been
demonstrated (Mohanty and Kushner 2007, 2008). One particular study reports RNase P-mediated separation of three precursor transcripts in E. coli that encode for the seven valine
tRNA molecules found within the cell (Agrawal, Mohanty and
Kushner 2014). Interestingly, specific focus on the ValU and LysT
transcripts has highlighted a previously uncharacterized role for
RNase P in 3 end processing. Unusually in this case, RNase P processing proceeds in the 3 to 5 direction generating one tRNA
precursor at a time with activity being initiated by specific endonucleolytic removal of the Rho-independent transcription terminators (Agrawal, Mohanty and Kushner 2014).

Shepherd and Ibba

Processing at the 3 end of the tRNA precursor is typically


initiated by an endonucleolytic cleavage event that takes place
downstream of the critical CCA terminus (Sekiya et al. 1979; Morl
and Marchfelder 2001). This cleavage event often happens before
RNase P processing of the 5 end and final exonucleolytic trimming of the 3 end. Endonucleolytic processing at the 3 end of
the precursor is carried out by a combination of two enzymes
termed RNase E and RNase III (Ray and Apirion 1981; Apirion
and Miczak 1993). Like RNase P, these enzymes can be found associated with the inner membrane of the cell (Miczak, Srivastava and Apirion 1991). In addition, fundamental aspects of primary and secondary RNA structure have been shown to determine the broad substrate specificities of both RNase E and RNase
III. More specifically, RNase E has been demonstrated to cleave
sequences that are both single stranded and AU-rich whilst
RNase III targets double-stranded RNA sequences (Robertson,
Webster and Zinder 1968; McDowall, Lin-Chao and Cohen 1994;
Nicholson 1999).
RNase E has a molecular weight of approximately 70 kDa
and was first identified as an important enzyme in the separation of precursor ribosomal RNA from larger transcripts (Ghora
and Apirion 1978). Subsequent work demonstrated that certain
tRNA precursors accumulate in E. coli cells that are deficient in
RNase E. One of these rRNA-independently transcribed precursors is known to encode tRNALeu and tRNAHis . Since this precursor cannot be processed in the absence of RNase E, this finding strongly supports a critical role for the enzyme in the initial
stages of tRNA processing (Ray and Apirion 1981). Extensive biochemical characterization has concluded that RNase E is primarily involved in the cleavage of multimeric and polycistronic tRNA
precursors in the spacer region and in the processing of 3 trailer
regions (Li and Deutscher 2002; Ow and Kushner 2002). However,
studies using tRNATyr Su3 precursors that lack a 3 RNase E cleavage site have indicated that this enzyme can also cleave the 5
leader region of certain tRNA precursor transcripts (Soderbom,
Svard and Kirsebom 2005). RNase E activity is critical to initial
tRNA precursor processing and results in the provision of accessible substrates for RNase P, which is inhibited by long trailing
regions at the 3 end of transcripts and, subsequently, the 3 exonucleases (Li and Deutscher 2002).
In contrast to what has been shown for RNase E, RNase III
has been reported to have a less significant role in polycistronic
tRNA transcript processing. In terms of overall structure, RNase
III is comprised of two identical polypeptide subunits that have
a molecular weight of 25 kDa (Dunn 1976). Since RNase III was
first described to be critical for cleavage of the large 30S polycistronic ribosomal RNA precursor, it was initially considered to
have a role in the processing of tRNA transcripts that are cotranscribed with rRNA (Dunn and Studier 1973; Nikolaev, Silengo
and Schlessinger 1973). This was later confirmed by the finding
that, in vitro, E. coli 30S rRNA transcripts that include tRNAGlu or
the tRNAIle -tRNAAla dimer in the spacer regions are cleaved by
RNase III to produce intermediate tRNA precursors that are then
suitable for RNase P processing (Lund and Dahlberg 1977). However, studies using mutants that are compromised in RNase III
activity demonstrated that this enzyme only has a minor role in
processing (Gegenheimer and Apirion 1981). In fact, early tRNA
maturation only becomes severely compromised if this mutation is introduced into a strain that is also deficient in RNase P
and RNase E activities, suggesting the latter two proteins have
the primary roles in this process (Plautz and Apirion 1981).
Whilst the activities of RNase E and RNase III generate precursors that allow RNase P to complete 5 end tRNA transcript
maturation, the endonucleolytic cleavage events that take place

283

at the 3 end of the transcript usually leave a stretch of extra residues that must be removed by exoribonucleases (Schedl,
Roberts and Primakoff 1976). Two types of intermediate tRNA
precursors have been identified in bacterial cells and they differ in terms of their requirements for exonucleolytic processing
at the 3 end. In the most common type of precursor, the CCA
end of the tRNA is already present and is followed by a series
of extra nucleotides. Final maturation of the 3 end of this precursor requires a nuclease that can trim right up to the critical
CCA end without disrupting it. This is supported by mutagenesis
studies that directly demonstrate that the CCA repair enzyme,
nucleotidyltransferase, is not involved in the maturation of this
type of precursor (Deutscher, Lin and Evans 1977). In the second
type of precursor, which is rare in bacterial cells but common in
eukaryotes, other residues substitute the CCA end. In this case,
3 processing requires a nuclease that will remove only the substituting residues leaving a product that can be repaired by nucleotidyltransferase such that it has a CCA terminus at the completion of maturation.
Originally, it was thought that RNase II, a well-known 3 exonuclease, was the sole enzyme involved in 3 exonucleolytic
processing of tRNA transcripts. Whilst this enzyme has been
shown in vitro to remove extra nucleotides at the 3 end of an E.
coli tRNATyr precursor, the fact that it is processive in nature and
cannot stop at the CCA end has led to searches for another more
specific exonuclease (Cudny and Deutscher 1980). A more likely
candidate was found with the discovery of a monomeric 38 kDa
protein termed RNase D. RNase D is specific for tRNA precursors
that have the 3 CCA end. This enzyme specifically removes all
extra residues that come before the CCA end in a non-processive
manner and, in doing so, generates a mature precursor tRNA
molecule that has amino acid accepting capability (Cudny and
Deutscher 1980). Perhaps more importantly, RNase D cannot hydrolyze the CCA end and the rate of its hydrolytic activity has
been shown to reduce by 30-fold when it reaches this sequence
(Ghosh and Deutscher 1978; Cudny and Deutscher 1980).
In the case of the second type of tRNA precursor, which has
an incomplete or absent CCA end, another 3 processing exonuclease was identified termed RNase BN. RNase BN has a molecular weight of approximately 35 kDa and, unlike RNase D, its
specificity for tRNA precursors is not determined by the presence of an intact 3 CCA end (Seidman et al. 1975; Schmidt and
McClain 1978; Asha et al. 1983; Asha and Deutscher 1983). Other
exonucleases that have been shown to play a role in 3 tRNA processing include RNases PH, T and PNPase (described in some de and RajBhandary 1995). Studies attempting to fully
tail in Soll
characterize all of the separate components of the processing
machinery have shown that the role and order of activity of the
different enzymes is entirely dependent on the specific tRNA
precursor in question (Li and Deutscher 1996). The ultimate conclusion of all of the processes discussed in this section is the
generation of a mature tRNA molecule, which can then successfully progress into the nucleotide modification and aminoacylation pathways.

Nucleotide modification of tRNA


Post-transcriptional modification has been shown to expand
the chemical diversity and fine-tune the functionality of tRNA
molecules across all three domains of life (see Table 1; reviewed
and RajBhandary 1995; Bjork
extensively in Bjork et al. 1987; Soll
et al. 1999; Gustilo, Vendeix and Agris 2008; Grosjean 2009; Motorin and Helm 2010; El Yacoubi, Bailly and de Crecy-Lagard
2012). To date, more than 100 different chemical modifications

284

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

Table 1. Summary of the potential modifications, their positions and


the enzymes required to make them in E. coli tRNA. The information
presented in this table was obtained from El Yacoubi, Bailly and de
Crecy-Lagard (2012).
Position

Modification

Enzyme

8
13
16
17
18
20
20a
32

S4 U

D
D
Gm
D
D
S2 C Cm, Um 

IscS, Thil
TruD
Dus A, B, C
Dus A, B, C
TrmH
Dus A, B, C
Dus A, B, C
IscS, TtcA Trml RluA

34

I k2 C ac4 C Um, Cm Q,
GluQ mnm5 U cmnm5 Um
cmnm5 U mnm5 s2 U
cmnm5 s2 U mnm5 se2 U
mcmo5 U cmo5 U

TadA TilS TmcA TrmL


FolE, QueDCEF, Tgt, QueA
and G+YadB MnmC1 and
C2+TrmL MnmE, GidA ? ?
? CmoA and B
AroBDEKLAC

37

m1 G m6 A m2 A i6 A ms2 i6 A
t 6 A m6 t 6 A

TrmD TrmN6? MiaA MiaB,


IscS, IscU TsaC, D, B and E
TsaA

38
39
40
46
47
54
55
65
67




m7 G
acp3 U
rT




TruA
TruA
TruA
TrmB
?
TrmA
TruB
TruC
TruC

have been identified, all of which generate various derivatives


of the four common nucleosides adenosine, guanosine, cytidine
and uridine. Therefore, it is no surprise that between 1 and
10% of the genes within a specific genome encode for proteins
that are involved in the tRNA modification process (Motorin and
Helm 2010; El Yacoubi, Bailly and de Crecy-Lagard 2012). Whilst
the vast majority of tRNA modifications were discovered in the
1970s, specific adaptation of both mass spectrometry and deepsequencing techniques are responsible for significant recent advances in our understanding of the critical functional roles these
modifications play within the cell (Chan et al. 2010; Findeiss et al.
2011; Globisch et al. 2011; Yi and Pan 2011; Wetzel and Limbach 2012). Modified nucleosides can be found at various locations in the tRNA cloverleaf and can increase the surface area of
the molecule by as much as 20% (Bjork et al. 1987) (Fig. 2). Notably, the highest frequency and diversity of modifications are
concentrated at positions 34 and 37 in the anticodon stem loop
(Grosjean 2009).
Modifications at specific locations within the cloverleaf
structure are associated with conferring different functions
upon the tRNA molecule (Bjork et al. 1987). Those found at or
around the 3 side of anticodon loop position 37 are associated with maintaining both the efficiency and fidelity of translation. This is achieved by altering the sensitivity of the tRNA
to the reading context of the ribosome-bound messenger RNA,
thereby ensuring that the correct reading frame is supported.
More specifically, ms2 and ms2 i6 tRNA modifications at position
37 are frequent when the base at position 36 is either adenine or
uracil. The nature of these modifications has been directly correlated with stabilization of interactions between the codon and

the anticodon particularly at the first base pair position (Bjork


et al. 1987; El Yacoubi, Bailly and de Crecy-Lagard 2012). This is
particularly critical for A:U and U:A. codon: anticodon interactions where the modification prevents the formation of intraloop
base pairs within the anticodon stem loop structure and reduces
the potential for frameshifting (Agris 2008; Grosjean 2009). In
Salmonella typhimurium miaA1 (or miaE) mutants, the tRNA pool
is missing the ms2 i6 o6 modification at adenine 37. Subsequently,
this strain has been found to have a significantly reduced growth
rate, altered regulation of various biosynthetic operons and a reduced rate of polypeptide chain elongation (Ericson and Bjork
1986). Complementary to these findings, tRNAs across all kingdoms of life that are involved in the recognition of triplet codons
that begin with adenine typically have a t6 A modification at position 37 (Bjork et al. 1987). Escherichia coli tRNAIle that does not
have this modification has been shown to decode less efficiently
in vitro. tRNAs that recognize codons beginning with cytosine or
guanine often have a more simple m1 G modification at position
37. A mutant of S. typhimurium that can produce tRNAs where
m1 G is present at 30 C but not at 41 C undergoes a higher rate
of frameshifting when it is presented with the challenge of decoding sequences containing runs of cytosines when it is grown
at the higher temperature (Bjork et al. 1987, 1999). These findings provide further evidence for the role of tRNA modifications
at and around this position in the maintenance of translational
efficiency (Miller, Hussain and Schweizer 1976).
Modifications at position 34 in the tRNA cloverleaf have been
shown to be involved in maintaining the fidelity of translation
by influencing codon choice and discrimination. This is particularly important when two amino acids are defined by codons
that differ by only one nucleotide in the triplet. For example,
methionine is encoded by AUG and isoleucine is encoded by
AUA, AUU and AUC. In bacteria, accurate recognition of the AUA
codon requires tRNAIle that has a k2 C modification at position 34.
This particular modification is critical for changing the amino
acid specificity and the anticodon identity of this tRNA molecule
from methionine to isoleucine (Soma et al. 2003). Conversely,
E. coli elongator tRNAMet has an ac4 C modification at the same
position, which compromises ribosome binding but provides a
huge advantage to the cell by preventing misreading of the AUA
isoleucine codon in vitro (Stern and Schulman 1978). Similarly,
modification of position 34 with Cm or Q has been reported to
enable tRNATrp or tRNATyr , respectively, to efficiently discriminate tryptophan (UGG) or tyrosine codons (UAU and UAC) from
stop codons during translation (Nakamura and Ito 2011; El Yacoubi, Bailly and de Crecy-Lagard 2012). Hydrolytic deamination
of the sixth carbon of adenosine 34 to inosine is a more unusual
form of modification that enables a single isoacceptor of E. coli
tRNAArg (ACG) to alternatively pair with U, C or A nucleotides in
the third position of the anticodon and thus successfully decode
all three arginine codons (CGU, CGC and CGA) (Crick 1966; Keller
et al. 1999). Modifications in the anticodon loop other than at
positions 34 and 37 have also been shown to influence both the
efficiency and fidelity of translation (Bjork et al. 1987).
Modified nucleotides outside of the anticodon region are
thought to be involved in stabilization of the overall structure of
the tRNA molecule (Bjork et al. 1987). In addition, the overall level
of tRNA modification is proposed to serve a regulatory function.
Recently, bacterial tRNA modification has been shown to have a
role in the host immune response. Fully modified native tRNATyr
isolated from E. coli was found to be non-immunostimulatory
towards human peripheral blood mononuclear cells. However,
unmodified tRNATyr induced interferon alpha production under
the same conditions suggesting that tRNA modification may also

Shepherd and Ibba

285

Figure 2. Known modifications and their positions in E. coli tRNA. The information presented in this figure was adapted from El Yacoubi, Bailly and de Crecy-Lagard,
(2012). Abbreviations for specific groups include m-, c-, n-, o-, t-, i-, K-, r- and s- for methyl, carbon, amino, oxy, threonine, isopentyl, lysine, ribose and thio groups,
respectively. Other abbreviations include acp: 3-amino-3-carboxypropyl, D: dihydrouridine, I: inosine, : pseudouridine, Q: queuosine. Other capital letters refer to
the appropriate bases uracil, cytosine, guanine and adenine. An index and an exponent indicate the number and the position of the substitution, for example 6dimethyladenosine is written as m6 2 A.

be a mechanism employed by pathogenic bacteria in host immune invasion (Gehrig et al. 2012). Further studies are required
to determine the nature and quantity of modifications involved
in this process.
Whilst tRNA modifications have a primary role in maintaining the efficiency and fidelity of translation, their importance is
generally underappreciated, and it is thought that they may also
take on a much more global regulatory role within the cell by
providing connections between protein synthesis, metabolism
and stress response pathways. This is reinforced by the finding
that many tRNA modifications are directly determined by cellular or environmental factors such as growth rate, oxygen levels
and nutrient levels. For example, the synthesis of some proteins
might be influenced by the proportion of tRNAs within the cell
pool that have a particular modification. This is because some
tRNA modifications give a bias for recognition of one codon over
another. In the case of the DNA damage response in yeast, tRNA
methyltransferase 9 has been found to enhance the translation
of DNA damage proteins, whose transcripts are overrepresented
by specific glutamic acid and arginine codons, by modifying the

uridine wobble bases of tRNAGlu (UUC) and tRNAArg (UCU)


(Begley et al. 2007). In addition, several thermophilic bacteria
show an increase in tRNA modification levels with growth temperature (Noon et al. 2003) and the proportion of thiolated tRNAs in E. coli increases with growth rate (Emilsson, Naslund and
Kurland 1992). These finding suggest a role for tRNA modifications in regulation of stress responses (El Yacoubi, Bailly and de
Crecy-Lagard 2012). Whatever their final function may be, fully
modified tRNA species produced at this stage are able to proceed into aminoacylation and subsequently enter translation
and other biological pathways in the cell.

Aminoacylation
The generation of aminoacylated tRNA is a critical function that
is essential for translation as well as a variety of other cellular
processes, which will be discussed in the section Other roles of
tRNA in the cell of this review. Across all three domains of life,
formation of an aminoacyl-tRNA is enzymatically a two-stage
process that is catalyzed by one of 23 aaRS. In the first stage of

286

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

Figure 3. Formation of an aminoacylated-tRNA molecule by an aaRS. An amino acid (blue) is activated by an aaRS to form an aminoacyl-adenylate. This process
requires ATP and results in the release of PPi. Following tRNA binding to the aaRS, the activated amino acid is transferred to the 3 end of the tRNA molecule forming
aminoacyl-tRNA with concurrent release of adenosine monophosphate (AMP). The aminoacyl-tRNA product is released from the aaRS and is either subject to binding
by EF-Tu for delivery to the ribosome or hijacking by other factors for diversion into biosynthetic pathways outside of translation. Figure reproduced from Reynolds,
Lazazzera and Ibba (2010).

the reaction, an amino acid is specifically activated by an aaRS to


form an aminoacyl-adenylate. Aminoacyl-adenylate formation
proceeds in an adenosine triphosphate (ATP)-dependent manner and results in the subsequent release of inorganic pyrophosphate (PPi). In the second stage of the reaction, the activated
amino acid moiety becomes transferred to either the 2 or the 3
hydroxyl group of the terminal adenine (A76) that is located at
the critical CCA terminus of the tRNA acceptor molecule. This results in the formation of an aminoacyl-ester bond and the reaction concludes with release of the aminoacylated-tRNA species
from the synthetase (summarized in Fig. 3) (Ibba and Soll 2000;
Reynolds, Lazazzera and Ibba 2010). Since the aaRS and their
roles in biology and medicine have recently been reviewed elsewhere (Pang, Poruri and Martinis 2014), the remainder of this
section will focus on providing a brief summary of the specific
identity elements within tRNA that are required for efficient
aminoacylation.
Accurate selection of the cognate tRNA from the cellular pool
by an aaRS is known to involve a dual discrimination mechanism
based upon velocity of the aminoacylation reaction and substrate binding affinity, both of which will only reach optimum
levels with the cognate molecule (Yarus and Berg 1967; Ebel et al.
1973; Roe, Sirover and Dudock 1973; Lam and Schimmel 1975). It
is also known that binding of the cognate tRNA enhances the
discriminatory power of some synthetases against non-cognate
amino acids (reviewed extensively in Hendrickson and Schimmel 2003). Early research characterizing various aspects of the
tRNA cloverleaf structure identified three regions that are important for interaction with and binding to the appropriate aaRS:
the acceptor stem, the D loop and the anticodon loop (Schimmel
1977; reviewed extensively in Giege, Sissler and Florentz 1998).
Involvement of the tRNA acceptor stem in recognition by and
interaction with the aaRS was first demonstrated using ribonuclease A protection assays and came as no surprise since the
activated amino acid is ultimately transferred to the 3 end of
this region (Dickson and Schimmel 1975). More specifically, correct alignment of the 3 end of the tRNA acceptor stem with the
catalytic residues of the enzyme is a requirement for productive
aminoacylation (Schimmel 1977). Examples of acceptor stem elements that specifically confer tRNA identity include the G3:U70
wobble in bacterial tRNAAla , which acts as a critical identity element for aminoacylation by AlaRS (Hou and Schimmel 1988),

and positions G1, G2, C81 and A82, which have been shown
to confer the amino acid accepting specificity of E. coli tRNATyr
(Hooper, Russell and Smith 1972; Shimura et al. 1972; Smith and
Celis 1973). The importance and relevance of specific tRNA acceptor stem elements can also vary between bacterial genera
and species. For example, it has recently been shown that Streptococcus pneumoniae AlaRS enzyme is less dependent upon the
G3:U70 acceptor stem wobble region for aminoacylation than
other bacterial AlaRS proteins (Shepherd and Ibba 2013b, 2014).
In addition, the same acceptor stem element can act as an antideterminant for aminoacylation by other aaRSs. For example, if
the G3:U70 element is put into yeast tRNAThr , then it becomes
an anti-determinant for aminoacylation by threonyl-tRNA synthetase (Nameki 1995; a full and comprehensive review of tRNA
identity elements is given in Giege, Sissler and Florentz 1998).
D-loop residues within tRNA can also act as aminoacylation
recognition elements for the aaRSs. This was first demonstrated
to be the case for yeast phenylalanyl-tRNA synthetase (PheRS),
which can aminoacylate heterologous tRNAPhe species that have
a specific sequence of bases in the D-loop that match that of its
own tRNA (Dudock et al. 1971; Roe, Sirover and Dudock 1973).
Strikingly, this region of tRNA structure has also been implicated
in editing of misaminoacylated-tRNA species by certain aaRSs
(see the Section Editing of aminoacyl-tRNA below for more details on the editing process). For example, D-loop residues 16,
20 and 21 in tRNAIle are implicated in the editing of mischarged
Val-tRNAIle by isoleucyl-tRNA synthetase (IleRS) (Hale et al. 1997;
Farrow, Nordin and Schimmel 1999). In addition, D-loop residue
16 in St. pneumoniae tRNAIle was recently shown to determine
the aminoacylation capacity of St. pneumoniae IleRS towards both
isoleucine and leucine (Shepherd and Ibba 2014).
Whilst the anticodon loop typically comes into close contact
with the aaRS in many systems (Schimmel et al. 1972; Schoemaker and Schimmel 1974; Schimmel 1977), its importance as a
specific identity element is dependent upon the particular tRNA
species. For example, early studies in yeast demonstrated that
the entire anticodon loop can be removed from tRNAPhe while
retaining the capacity for aminoacylation by PheRS (Thiebe,
Harbers and Zachau 1972). In contrast, mutation of the third
base in the anticodon of E. coli tRNAGly causes a significant decrease in the rate of aminoacylation of this species by glycyltRNA synthetase (Carbon and Squires 1971). In some instances,

Shepherd and Ibba

the anticodon loop also confers the amino acid identity of the
species. If the second base in E. coli tRNATrp is mutated, then it
is readily misaminoacylated with glutamine (Yaniv et al. 1974).
In addition, modification of either of the first two bases in the
anticodon loop of E. coli tRNAfmet inactivates the species entirely
(Schulman and Pelka 1977).
After completion of aminoacylation, most charged tRNA
species are sequestered by EF-Tu and subsequently proceed
into protein synthesis at the ribosome. Alternatively they can
enter other specific cellular processes, as discussed below in
the Section Other roles of tRNA in the cell. However, some
aminoacylated-tRNA species need to undergo further maturation events prior to participation in these terminal processes as
described below.

Maturation of aminoacyl-tRNA
After aminoacylation, most tRNA species are ready to directly participate in protein synthesis at the ribosome or in
other cellular pathways. In a relatively small number of cases,
aminoacylated-tRNA must first undergo a further maturation
step. The most common examples of additional maturation
events that take place in bacteria include transamidation,
which generates both asparaginylated and glutaminylated tRNA
species, production of selenocysteinyl-tRNA and formylation of
initiator methionyl-tRNA (summarized in Fig. 4) (Blanquet et al.
2003).
Transamidation is employed by some bacterial species as an
indirect mechanism for producing correctly aminoacylated GlntRNAGln from Glu-tRNAGln and Asn-tRNAAsn from Asp-tRNAAsn
in the absence of an encoded glutaminyl- or asparaginyl-tRNA
synthetase, respectively. Transamidation was first identified as a
major pathway for generating Gln-tRNAGln in Gram-positive bac-

287

teria, cyanobacteria and certain organelles (Wilcox and Nirenberg 1968; Wilcox 1969; Schon, Hottinger and Soll 1988a; Schon
et al. 1988b). Further studies have since indicated that this pathway, as well as that for Asn-tRNAAsn production, can also be
found in some Gram-negative bacteria and archaea (White and
Bayley 1972; Strauch, Zalkin and Aronson 1988; Curnow, Ibba and
Soll 1996; Gagnon et al. 1996; Becker and Kern 1998; Tumbula
et al. 2000) (reviewed in Blanquet et al. 2003). Regardless of bacterial origin, the transamidation reaction can be divided into two
steps. Initially, tRNAAsn and tRNAGln are aminoacylated with aspartate and glutamate, respectively. This activity requires either
an aspartyl- or a glutamyl-tRNA synthetase that has very poor
or no discriminatory power against the non-cognate tRNA but
maintains specificity for the cognate amino acid. The second
stage of the reaction is then accomplished by an amidotransferase that uses either L-asparagine, in the case of tRNAAsn , or Lglutamate, in the case of tRNAGln , as an amide nitrogen donor to
generate the final products (Fig. 4A). The aminoacylated tRNA intermediates produced during the first stage of the reaction typically bind EF-Tu very poorly and so are not efficiently recruited
to the ribosome before the next stage of the reaction, which requires the amidotransferase, can proceed. In the case of AsntRNAAsn production, it has also been shown that binding of EFTu to the mischarged intermediate is prevented by formation of
a protective shuttling complex, called the transamidosome, between the non-discriminatory AspRS and the amidotransferase
(Bailly et al. 2007).
Characterization of the glutamate amidotransferase (GluAdT) from Bacillus subtilis has demonstrated that the protein
is produced by expression from three genes, termed gatC, gatA
and gatB to produce a final product that is heterotrimeric
in nature (Strauch, Zalkin and Aronson 1988; Curnow et al.
1997). Since B. subtilis does not have a Gln-tRNA synthetase,

Figure 4. Indirect tRNA aminoacylation pathways. (A) The indirect pathways for generation of Gln-tRNAGln and Asn-tRNAAsn by transamidation. Abbreviations include
AdT for amidotransferase. (B) The selenocysteine tRNA modification pathway. (C) The formylation pathway for initiator Met-tRNAfMet . Abbreviations include FMT for
methionyl-tRNAf Met transformylase, FTHF for N-10 formyltetrahydrofolate and IF2 for initiation factor 2.

288

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

transamidation in the presence of a non-discriminatory GlutRNA synthetase is the only mechanism for producing correctly
acylated Gln-tRNAGln in this bacterium, thus indicating the importance of specific aminoacyl-tRNA maturation events for cell
survival (Lapointe, Duplain and Proulx 1986; Schon, Hottinger
and Soll 1988a; Chen et al. 1990). In many cases, including
Deinococcus radidurans, Thermus thermophilus and B. subtilis, GluAdt has been shown to have dual activity in vitro (Curnow et al.
1997, 1998; Becker et al. 2000b). Therefore, the in vivo specificity of
this enzyme against Asp-tRNAAsn or Glu-tRNAGln is entirely dependent on the cellular context, particularly with regard to the
presence or absence of the necessary tRNA-synthetase enzymes
(Becker et al. 1997, 2000a; Curnow et al. 1998). Maturation of GlutRNAGln and Asp-tRNAAsn to the correctly acylated species is essential for maintaining the fidelity of the genetic code within the
bacteria that possess these pathways.
Another form of aminoacyl-tRNA maturation is described
by the selenocysteinyl-tRNA pathway, which is found across all
three domains of life (reviewed in Commans and Bock 1999). The
requirement for this maturation pathway has been found to be
most notable in the synthesis of oxidoreductases, which often
substitute selenocysteine at cysteine codons because of its lower
redox potential (Blanquet et al. 2003). The first detection of a protein containing selenocysteine was made in the mid-1970s in
a glycine reductase isolated from the anaerobic Gram-positive
bacterium, Clostridium sticklandii (Cone et al. 1976). Since then the
pathway has been well characterized in E. coli, where it was first
shown that co-translational insertion of selenocysteine occurs
at UGA codons that are present within a specialized mRNA context (Chambers et al. 1986; Zinoni et al. 1986).
Detailed characterization of the E. coli selenocysteinyl-tRNA
pathway has indicated that this bacterium has a specific gene,
called selC, which encodes the genetic material required for production of a unique tRNA molecule that is specific for selenocysteine and displays an anticodon that is complimentary to UGA
(Leinfelder et al. 1988). This tRNASec molecule is a substrate for
serylation by seryl-tRNA synthetase. After the production of SertRNASec , a 500 kDa selenocysteine synthase (SelA) modifies the
serine into selenocysteine using phosphoselenate (produced by
SelD) as a donor (Fig. 4B).
The efficiency of the SerRS:tRNASec mischarging reaction has
been demonstrated to be as low as 1% compared to that of the
five cognate tRNASer isoacceptors in E. coli (Amberg et al. 1996;
Commans and Bock 1999). The low rate of serylation of tRNASec
by SerRS can be explained by the unusual and unique structure
of the tRNA molecule. Bacterial tRNASec is known to be larger
than other tRNAs and, in E. coli, reaches a total of 95 nucleotides
in length. The increased size is due to a combination of features including a long extra arm that is required for recognition by SerRS, and an extended 13 base pair acceptor helix. In
addition, tRNASec has been shown to have a purine: pyrimidine
pair between positions 11 and 24, which is rare except in initiator tRNAs, an extended 6 base pair D-stem and a reduced 4
nucleotide D-loop (Baron et al. 1993; Sturchler et al. 1993; Commans and Bock 1999). These specific and unique tRNA features
have been demonstrated to contribute to various stages of the
decoding process (Commans and Bock 1999).
Decoding and distinction of selenocysteine codons from
normal UGA stop codons also requires the presence of a
stem-loop structure, called the SECIS element, to be located
approximately 40 nucleotides away from the 3 end of the UGA
selenocysteine codon. In addition, a specific elongation factor,
called SelB, is required for recognition of the SECIS element and,
thus, efficient delivery of mature selenocysteinyl-tRNASec to the

ribosome (Forchhammer, Leinfelder and Bock 1989; Hilgenfeld,


Bock and Wilting 1996; Kromayer et al. 1996; Bock et al. 1997;
Haruna et al. 2014). Inappropriate sequestration of Ser- or SectRNASec by canonical EF-Tu is prevented by the presence of a specific anti-determinant box in the acceptor stem of tRNASec at the
8th, 9th and 10th base pair positions (Baron and Bock 1991).
A final, and critical, example of maturation prior to use
in protein synthesis is given by the highly specific formylation of initiator tRNA that has been pre-charged with methionine (Fig. 4C). This reaction is catalyzed by methionyl-tRNAfMet
transformylase (FMT) and requires the formyl-group donor, N10 formyltetrahydrofolate (FTHF). In E. coli, FMT activity is essential for cell viability at 42 C and its deletion also causes
marked growth defects at 37 C (Guillon et al. 1992a). Structural studies have indicated that FMT is an elongated protein
that is comprised of two separate domains. These domains are
joined together by an extended peptide (Schmitt, Blanquet and
Mechulam 1996a; Schmitt et al. 1996b). The N-terminal region
of the protein contains the catalytic center, is comprised of a
Rossmann fold and shares high homology with another FTHFbinding protein called glycinamide ribonucleotide transformylase. In contrast, the C-terminal domain adopts a beta-barrel
fold. Both domains of the protein and the extended linker region have been shown to make significant contacts with the
tRNA substrate. This accounts for the high specificity of FMT
for methionylated tRNAs over other aminoacylated species. The
activity of FMT has been shown to require specific features of
both the acceptor stem and the D-loop for accurate recognition
of the aminoacylated initiator tRNA. These features include a
mismatched base pair between positions 1 and 72 (Lee, Seong
and RajBhandary 1991; Guillon et al. 1992b), which along with
formylation itself, also prevents misappropriation of the species
by EF-Tu (Sundari et al. 1976; Petersen et al. 1979; Guillon et al.
1993, 1996). The maturation of initiator Met-tRNAmet by formylation is essential for initiation of translation in bacteria.

Editing of aminoacyl-tRNA
Under certain circumstances, non-cognate aminoacyl-tRNA
needs to be proofread and edited to ensure that the efficiency
and fidelity of translation is maintained. In bacteria, this is
achieved by a combination of enzymatic activities, which include the pre- and post-transfer editing mechanisms of aaRSs
as well as the functions of trans-editing factors, peptidyl-tRNA
hydrolase and D-aminoacyl-tRNA deacylase.
The aaRSs represent the first step in quality control of translation (reviewed extensively in Ling, Reynolds and Ibba 2009;
Reynolds, Lazazzera and Ibba 2010). Aminoacyl-tRNA synthesis
occurs in two stages (described in the Section Aminoacylation
and Fig. 3) and, if the accuracy of translation is to be adequately
maintained, the aaRSs must successfully sequester both cognate tRNA and amino acid from similar non-cognate molecules
present in the cellular pool. Generally, the aaRSs have high specificity for their cognate tRNA. This is due to a combination of
the large surface area that is available during recognition of this
substrate and also kinetic proof reading that occurs during the
aminoacylation reaction (Ibba and Soll 2000; Guth and Francklyn
2007). However, near-cognate amino acids can be much harder
for the aaRSs to discriminate against since they can differ from
the cognate substrate by as little as one methyl group. This particular problem has been investigated in detail for IleRS, which
must distinguish between two isosteric amino acids: isoleucine
and valine. In this case, IleRS is able to maintain the accuracy
of translation by employing both pre- and post-transfer editing

Shepherd and Ibba

289

Figure 5. Quality control steps during the formation of a non-cognate aminoacyl-tRNA. After activation of a non-cognate amino acid (red), the aminoacyl adenylate
may be hydrolyzed and released. A non-cognate aminoacyl-tRNA may be translocated into the editing site of the aaRS for hydrolysis. If the mischarged aminoacyl-tRNA
is released from the aaRS without being edited, it is subjected to editing by trans-editing factors or through resampling by the aaRS. Some non-cognate aminoacyltRNAs are discriminated against by EF-Tu, which either binds them too tightly or too weakly for efficient delivery and release in the ribosome. Figure reproduced from
Reynolds, Lazazzera and Ibba (2010).

mechanisms against the non-cognate amino acid (Baldwin and


Berg 1966; Eldred and Schimmel 1972).
Pre-transfer editing refers to specific hydrolysis of a noncognate aminoacyl-adenylate prior to transfer of the activated
amino acid moiety to the tRNA acceptor. In contrast, posttransfer editing occurs after the activated amino acid has been
attached to the designated tRNA isoacceptor. This type of editing
takes place either prior to (in cis) or after release (in trans) of the
aminoacylated tRNA from the synthetase (summarized in Fig. 5)
(Ling, Reynolds and Ibba 2009). Whilst many of the aaRSs possess either one or both of these editing mechanisms, inefficient
hydrolysis of some incorrectly acylated tRNAs can give rise to a
requirement for separate trans-editing factors if the accuracy of
translation is to be maintained (Ahel et al. 2003).
One of the most widespread trans-editing factors is AlaXp,
which can be found across all domains of life and shares homology with the editing domain of alanyl-tRNA synthetase (AlaRS)
(Fukunaga and Yokoyama 2007; reviewed in Schimmel 2011;
Guo and Schimmel 2012). The requirement for AlaXp is associated with the challenge presented by relatively efficient production and release of mischarged Gly-, and more frequently,
Ser-tRNAAla from AlaRS (Swairjo and Schimmel 2005; Chong,
Yang and Schimmel 2008). Whilst AlaRS does possess its own
editing domain against both of these species, serine mischarging is particularly problematic. Furthermore, examples across all
three domains of life have indicated that there is a high level
of serine sensitivity during protein synthesis, with elevated levels of serine substitution at alanine codons causing toxic effects
(Beebe, Ribas De Pouplana and Schimmel 2003; Lee et al. 2006;
Chong, Yang and Schimmel 2008). Other common examples of
bacterial trans-editing factors that are involved in proofreading
of misaminoacylated tRNA species included Ybak, which clears
ProRS-generated Cys-tRNAPro (An and Musier-Forsyth 2005) and
ProXp, which clears ProRS-generated Ala-tRNAPro (Ahel et al.
2003; Wong et al. 2003; Das et al. 2014).
Whilst most trans-editing factors proofread and edit
mischarged-tRNAs before they participate in protein synthesis,
there is also a requirement for another enzymatic activity that
can recycle correctly aminoacylated tRNA species that have
already associated with the ribosome and begun to participate

in peptide chain elongation. Peptidyl-tRNA hydrolase (PTH)


can be found across all three domains of life and the esterase
activity of the protein is known to be critical for bacterial cell
survival (reviewed in Blanquet et al. 2003; Das and Varshney
2006; McFeeters 2013). Requirement for PTH activity is based
upon the natural process by which peptidyl-tRNAs dissociate
prematurely from the ribosome. This occurs with a translational frequency of approximately 10% and results in ribosomal
stalling and sudden abortion of protein chain elongation
(Manley 1978; Kurland and Ehrenberg 1985; Jorgensen and
Kurland 1990; Heurgue-Hamard et al. 1998). If left to accumulate
in the cell cytoplasm, these disassociated peptidyl-tRNAs
can result in toxicity, imbalance in the aminoacyl-tRNA pool
and, ultimately, translational arrest (Atherly and Menninger
1972; Garcia-Villegas et al. 1991; Menninger and Coleman 1993;
Schmitt et al. 1997a, b). These undesirable outcomes are avoided
by PTH, which specifically hydrolyzes the peptidyl-tRNAs into
uncharged tRNA and N-acyl-L amino acids or peptides. This is
achieved by cleavage of the ester bond that is located between
the C-terminus of the peptide and the 2 or 3 hydroxyl group of
the terminal adenine (A76 ) that is situated at the 3 end of the
tRNA molecule (Cuzin et al. 1967; Kossel and RajBhandary 1968).
Structural studies focused on the E. coli enzyme have indicated that PTH is comprised of one alpha-beta globular domain that is folded around a mixed beta-sheet (Schmitt et al.
1997). PTH is specific for elongator N-acyl-aminoacyl tRNAs with
residues K105 and R133 of the protein responsible for recognition during docking of the 5 phosphate of the tRNA substrate
(Fromant et al. 1999). In addition, a cluster of four asparagine
residues at positions 10, 21, 68 and 114 are involved in interactions with the TC tRNA acceptor stem helix (Fromant et al.
1999). In bacteria, fMet-tRNAfmet has a unique wobble region in
its acceptor stem between positions 1 and 72 that protects it and
consequently the process of translation initiation, from PTH activity (Schulman and Pelka 1975; Dutka et al. 1993). This wobble
region is missing from bacterial elongator tRNAs thus enhancing the specificity of PTH for them. Residues N10, H20 and D39
have been identified as critical for the catalytic mechanism of
the enzyme (Schmitt et al. 1997; Goodall, Chen and Page 2004;
Das and Varshney 2006).

290

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

In bacteria, there is a widespread existence of D-amino acids


and they are most commonly found incorporated naturally into
some cell wall peptidoglycans or synthesized antibiotics (Shoji,
Hinoo and Sakazaki 1976; Peypoux et al. 1985; Sivonen et al. 1992;
Jack and Jung 1998; Friedman 1999). However, accumulation of Damino acids can be toxic to the cell since not all of the aaRS have
adequate discriminatory power against this alternative form of
their cognate amino-acid substrate (Friedman 1999; Soutourina,
Blanquet and Plateau 2000a). Consequently, there is a requirement for D-aminoacyl-tRNA deacylase activity, which was first
identified in bacterial cell extracts after the tyrosyl-tRNA synthetases of both E. coli and B. subtilis were shown to incorrectly
aminoacylate their cognate tRNA with D-Tyr in vitro. In E. coli,
this enzyme was subsequently named D-Tyr-tRNATyr deacylase
(DTD).
Since its identification, structural studies have indicated that
bacterial DTD is a homodimer comprised of two 16 kDa subunits. The enzyme has some structural similarity to the editing
domains of archaeal threonyl-tRNA synthetases and specifically
folds into a beta-barrel structure that is closed on one side by
a beta-sheet (Ferri-Fioni et al. 2001; Hussain et al. 2006). Catalytically, DTD has been shown to have broad substrate specificity
against the ester linkage found in D- but not L-aminoacyl tRNAs (Calendar and Berg 1967; Soutourina et al. 1999; Soutourina,
Blanquet and Plateau 2000a; Soutourina, Plateau and Blanquet
2000b). Although EF-Tu is able to exclude D-aminoacyl-tRNAs
from its active site (Yamane, Miller and Hopfield 1981), inactivation of the dtd gene has been shown to exacerbate the toxicity
of D-Gln, D-Tyr, D-Ser, D-Asp and D-Trp in E. coli. These findings
highlight the importance of DTD activity with regard to its specific ability to proofread and edit D-aminoacyl-tRNAs and thus
ensure that an adequately balanced tRNA pool is available for
L-aminoacylation by the synthetases and protein synthesis at
the ribosome (Soutourina et al. 1999; Soutourina, Blanquet and
Plateau 2000a).

Codon recognition at the ribosome


Ribosomes are found across all domains of life and represent
the primary cellular structure that participates in protein synthesis. Bacterial ribosomes are comprised of a 30S and a 50S
subunit. The 30S subunit is made up of 16S ribosomal RNA and
approximately 20 protein components. In contrast, the 50S subunit consists of both 23S and 5S ribosomal RNA in complex with
approximately 30 proteins. During the process of translation, aminoacylated tRNAs bind to the ribosome in a codondependent manner. This ultimately results in translation of the
genetic information encoded by a bound mRNA into protein.
Briefly, the process of protein synthesis can be divided into
three stages: initiation, elongation and termination. During the
first stage, the initiator tRNA, fMet-tRNAfmet , undergoes binding
to the 30S ribosomal subunit in the presence of an mRNA bearing the AUG codon. This is promoted by initiation factor two (IF2)
whereby appropriate interaction between the initiator tRNA and
the mRNA start codon signal rapid recruitment of the 50S ribosomal subunit (Grunberg-Manago et al. 1978). During elongation, aminoacyl-tRNA is brought to the ribosome in a complex
with GTP-activated EF-Tu (Miller and Weissbach 1977; Johnson,
Miller and Cantor 1978). Progression through this stage results
in translocation of peptidyl-tRNA from the aminoacyl-site (A) of
the ribosome to the peptidyl-site (P). This is then followed by release of deacylated tRNA to the exit site (E) of the ribosome with
each of these stages promoted by GTP-activated elongation factor G (Weissbach 1977). In the termination stage, release factors

one and two bind the ribosome in a process that is aided by GTPactivated release factor three. This occurs only in response to a
stop codon arriving in the aminoacylation site (Weissbach 1977)
(reviewed fully in Ogle, Carter and Ramakrishnan 2003; Khade
and Joseph 2010; Novoa and Ribas de Pouplana 2012).
During each stage of translation, tRNA undergoes a series of
specific interactions with the ribosome that participate directly
in the decoding process. In the earliest stages of translation, interactions between the anticodon loop of initiator fMet-tRNAfmet
and the P site of the 30S ribosomal subunit are directly responsible for the high accuracy of initiation factor three-mediated discrimination against elongator tRNA. More specifically, residues
G1338 and A1339 within the 16S rRNA component of the small
subunit interact with conserved GC base pairs found at initiator tRNA anticodon arm positions 2941 and 3040, respectively.
This stabilizes docking of initiator tRNA to the ribosome and results in full initiation of translation (Abdi and Fredrick 2005; Lancaster and Noller 2005; Berk et al. 2006; Korostelev et al. 2006;
Selmer et al. 2006; Qin, Abdi and Fredrick 2007; Voorhees et al.
2009).
After completion of early initiation, the acceptor stem of the
aminoacylated-tRNA is bound in a complex with EF-Tu and so
cannot interact with the A site of the 50S ribosomal subunit.
However, the anticodon loop of the tRNA is able to directly interact with the A site of the 30S ribosomal subunit (Moazed and
Noller 1989). Conserved residues G530, A1492 and A1493 within
the 16S rRNA of the small subunit interact with the resulting
codon-anticodon helix (Ogle et al. 2001; Cochella, Brunelle and
Green 2007). Together, these interactions result in the formation
of a decoding intermediate, which is most commonly referred
to as the A/T state. This state is not stable during interaction between the ribosome and aminoacyl-tRNA that does not match
the codon specified by the bound mRNA sequence (Pape, Wintermeyer and Rodnina 1999; Valle et al. 2003).
Interaction of the aminoacylated-tRNA with the A site of the
50S ribosomal subunit is only achieved upon successful codon
anticodon pairing, which triggers EF-Tu-mediated GTP hydrolysis. Accommodation of the aminoacylated-tRNA within the 50S
A site subsequently triggers peptide bond formation by the 23S
rRNA that comprises the peptidyl-transferase center (Ban et al.
2000; Nissen et al. 2000; Harms et al. 2001). This reaction is known
to be aided by the formation of a base pair between G2553 of 23S
rRNA and residue C75 of the aminoacyl-tRNA present in the ribosomal A site (Schmeing et al. 2005). After this reaction, deacylated tRNA becomes accommodated in the P site of the ribosome, eventually shifting to the E site where the acceptor stem
undergoes various interactions with the 50S ribosomal subunit.
Dipeptidyl-tRNA in the A site can then progress to the P site
making room for accommodation of another aminoacyl-tRNA,
thus allowing translation to proceed (fully reviewed in Khade
and Joseph 2010). Aminoacylated-tRNA species that are not recruited by the translation machinery typically proceed into other
cellular pathways as described below.

Other roles of tRNA in the cell


In addition to their primary role in translation, many bacteria
also require aminoacylated-tRNA species to enter the peptidoglycan, antibiotic resistance, antibiotic synthesis and membrane
phospholipid modification pathways (reviewed in Shepherd and
Ibba 2013b). Due to their critical role in cell survival, some
tRNA molecules are also confirmed toxin targets (Lu et al. 2005;
Jablonowski and Schaffrath 2007; Meineke et al. 2011; Nawrot,
Sochacka and Duchler 2011; Uthman et al. 2011; Lopes et al. 2014).

Shepherd and Ibba

In this section, a brief overview of the specific involvement of


mature bacterial tRNAs in processes outside of protein synthesis will be given.
Sequestration of aminoacylated-tRNA donors away from the
protein synthesis machinery and into the peptidoglycan crosslinking pathway has been demonstrated to occur throughout
many genera and species of bacteria. Peptidoglycan is a critical
component of the bacterial cell wall where it has an important
role in providing protection against osmotic stress and turgor
pressure. In Gram-positive bacteria, 3070% of the total cell wall
biomass is defined as peptidoglycan and the structure may be
heavily utilized as an anchoring platform for various virulence
factors (Schleifer and Kandler 1972; Dramsi et al. 2008). In contrast, peptidoglycan makes up 10% or less of the total cell wall
biomass in Gram-negative bacteria.
Despite these differences, the basic carbohydrate backbone
of peptidoglycan is always comprised of alternating -1, 4linked N-acetylglucosamine (GlcNAc) and N-acetylmuramic acid
(MurNAc) residues. Furthermore, the lactyl-ether appendage of
the MurNAc component is typically modified by the addition of
an L-Ala- -D-Glu-X-D-Ala-D-Ala-containing pentapeptide side
chain. Within this side chain, position X is usually represented
by meso-diaminopimelic acid in Gram-negative bacteria and Llysine in Gram-positive bacteria (Bugg 1999). An additional level
of structural rigidity is obtained by the formation of cross-links
between the pentapeptide side chains of adjacent peptidoglycan
strands. Such cross-links can either be direct, forming between
position X on one strand and L-Ala at the fourth position of another, or indirect, which requires the formation of an intersecting amino acid bridge. Overall, cross-linking tends to occur at a
higher frequency in Gram-positive bacteria (7090%) as opposed
to Gram-negative bacteria (2550%). In addition, indirect crosslinks are more common in bacteria that have L-lysine at position
X in the pentapeptide side chain and formation of the intersecting amino acid bridge usually requires appropriately charged
tRNA to serve as the amino acid donor (Garcia-Bustos, Chait and
Tomasz 1987; Bugg 1999).
The occurrence of indirect peptidoglycan cross-linking has
been reported in several Gram-positive pathogenic bacteria
where it is most frequently associated with increased antibiotic resistance levels (reviewed in Shepherd and Ibba 2013b).
Briefly, in St. pneumoniae, the peptidoglycan structure is comprised of a combination of both branched and linear muropeptides. Branched muropeptides have an L-Ala-L-Ala or an L-Ser-LAla dipeptide bridge attached to the third L-Lysine found at position X in the pentapeptide side chain (Fiser, Filipe and Tomasz
2003). Dipeptide bridge formation is associated with penicillin
resistance in this pathogen and requires the activities of the
aminoacyl-tRNA-dependent MurM and MurN proteins (Filipe
and Tomasz 2000; Weber et al. 2000). MurM adds either L-Ala or
L-Ser at the first position of the cross-link, which is then subsequently completed by the L-Ala adding activity of MurN (Filipe,
Pinho and Tomasz 2000). It has recently been demonstrated that
MurM is able to more efficiently use mischarged Ser-tRNAAla as
opposed to Ala-tRNAAla and Ser-tRNASer as a substrate for peptidoglycan cross-linking (Shepherd 2011). In addition, MurM can
also deacylate Ala- and Ser-tRNAs in the absence of the peptidoglycan substrate, Lipid II, suggesting that this protein has
dual functionality in cell-wall biosynthesis and quality control
of translation (Shepherd and Ibba 2013b, 2014).
In Staphylococcus aureus, indirect peptidoglycan cross-linking
involves the formation of a pentaglycine bridge, which is ultimately appended to the L-Lysine residue occupying position X
of the pentapeptide side chain. Formation of the pentaglycine

291

bridge requires the sequential activities of the Gly-tRNAGly dependent FemX, FemA and FemB proteins (Rohrer et al. 1999;
Tschierske et al. 1999; Schneider et al. 2004). Three out of the five
tRNAGly isoacceptors encoded in the genome of Sta. aureus have
sequence identity elements consistent with weak binding to
EF-Tu, thus ensuring their participation in peptidoglycan rather
than protein synthesis (Giannouli et al. 2009). Participation of
aminoacyl-tRNA in Sta. aureus cell-wall biogenesis is critical, and
selective inactivation of femA or femB is known to be lethal due
to a combination of phenotypic effects that include loss of methicillin resistance (Stranden et al. 1997; Ling and Berger-Bachi
1998; Rohrer and Berger-Bachi 2003). Two other Staphylococcal species, Sta. simulans and Sta. capitis, use the aminoacyltRNA-dependent Lif and Epr proteins, respectively, to substitute serine at the third and fifth positions of the standard pentaglycine bridge. By doing so, these proteins ultimately confer resistance against the glycylglycine endopeptidases that are produced by these bacteria, thus ensuring that peptidoglycan integrity is maintained during competition with other species for
occupation of the same niche (Sugai et al. 1997; Tschierske et al.
1997; Ehlert et al. 2000). Other well-characterized examples of the
importance of aminoacylated tRNAs in peptidoglycan biosynthesis include Weissella viridescens FemX (Ala addition), Enterococcus faecalis BppA1/BppA2 (Ala addition) and the vancomycindependent FemX/VanK system (Gly addition) in Streptomyces
coelicolor (reviewed in Shepherd and Ibba 2013a) (Bouhss et al.
2002; Hegde and Blanchard 2003; Hong et al. 2004, 2005; Maillard
et al. 2005; Fonvielle et al. 2009).
In addition to their involvement in peptidoglycan biosynthesis, aminoacylated-tRNAs also participate in antibiotic resistance and synthesis pathways. Many Gram-positive and Gramnegative bacteria have a specific type of virulence factor, called
MprF, encoded in their genome that can catalyze the aminoacylation of inner membrane lipids subsequently altering the
permeability of the cell wall to cationic antimicrobial agents
(reviewed in Dare and Ibba 2012; Shepherd and Ibba 2013a).
This process requires donor aminoacyl-tRNA molecules and is
known to adjust the overall negative charge of the cell membrane in a manner that is dependent upon the nature of the
amino acid that is being transferred to the lipid (Roy and Ibba
2008; Ernst and Peschel 2011). One of the most well characterized classes of MprF protein is comprised of the aminoacylphosphatidylglycerol synthases (aaPGSs). The aaPGSs are integral cytoplasmic membrane proteins that specifically transfer
amino acids from appropriately charged tRNA species to the
polar head group of phosphatidylglycerol, which is a common
bacterial membrane lipid (Roy and Ibba 2008; Ernst et al. 2009).
Lysylation of phosphatidylglyercol by the aaPGS enzymes of C.
perfringens, Sta. aureus and M. tuberculosis is associated with an
increase in the net positive charge of the cytoplasmic membranes of these bacteria. This change in membrane polarity has
been directly correlated with reduced permeability to defensins,
aminoglycosides, glycopeptides and -lactams (Oku et al. 2004;
Staubitz et al. 2004; Roy and Ibba 2008; Maloney et al. 2009). In
contrast, recent work focusing on Listeria monocytogenes aaPGS
has indicated that lysylation of phosphatidylglycerol in the
membrane of this pathogen has a much more global physiological effect on the cell that extends beyond antimicrobial resistance and into regulation of the motility operon (Dare et al.
2014). In addition, Pseudomonas aeruginosa has an aaPGS enzyme
that aminoacylates phosphatidylglycerol with alanine resulting
in neutralization of the overall charge of the membrane. This
subsequently causes enhancement in the resistance of the bacterium to a range of negatively charged -lactams and increased

292

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

survival upon exposure to acidic or osmotically stressful growth


conditions (Roy and Ibba 2008; Klein et al. 2009). Whilst the importance of aminoacylated-tRNAs in MprF-mediated antibiotic
resistance is clear, these findings suggest that the overall functional relevance of membrane lipid aminoacylation is dependent upon both the species of bacteria and the features of the
environment to which it is routinely exposed.
The involvement of aminoacyl-tRNA in antibiotic synthesis
has been well characterized in the cases of valanimycin biosynthesis by Str. viridifaciens, and pacidamycin biosynthesis by Str.
coeruleorubidus (reviewed with cyclodipeptide synthesis in Shepherd and Ibba 2013a). Essentially, valanimycin biosynthesis requires VlmA. VlmA is a dedicated SerRS synthetase that diverts
L-serine from a Ser-tRNASer donor molecule away from protein
synthesis and into this antibiotic synthesis pathway (Garg et al.
2002, 2008; Garg, Gonzalez and Parry 2006). In contrast, generation of pacidamycin requires PacB activity. PacB transfers Lalanine from donor aminoacyl-tRNA to a critical intermediate
in the pathway for synthesis of this antibiotic (Chen et al. 1989;
Zhang et al. 2011a,b). Aminoacyl-tRNA donors are also required
for the synthesis of cyclodipeptides by members of the Bacillus
and Streptomycete families of bacteria (Lautru et al. 2002; Leys
et al. 2003; Seward et al. 2006; Gondry et al. 2009; Bonnefond et al.
2011).
Given the clear importance of tRNAs to a variety of cellular
processes, it is no surprise that they are toxin targets in both eukaryotic and prokaryotic systems. In some cases, possession of
a toxinantitoxin system that targets tRNA can be an advantage
to the host bacterium. For example, Shigella flexneri and S. enterica are known to have the VapBC toxinantitoxin system. The
toxin component of this system, VapC, specifically targets the
anticodon loop of the initiator tRNAfmet cleaving it between nucleotides A38 and C39 (Winther and Gerdes 2011). In these bacteria, VapC activity has been shown to inhibit translation both
in vitro and in vivo in a manner that can be rescued either by
VapB, the antitoxin, or addition of extra charged initiator tRNA.
In addition, VapC production was specifically activated by an
abrupt reduction in growth rate. This suggests that it may be
involved in stress response to environmental conditions by affecting the global regulation of cellular translation and causing
reprogramming of translational initiation (Winther and Gerdes
2011). This toxin family has also recently been identified in the
Gram-negative pathogen Leptospira interrogans where it is believed to have the same regulatory role (Lopes et al. 2014).
Other toxins that target tRNAs, including colicin D, colicin E
and PrrC, are specifically transported out of the producing cell
with the aim of killing competitor cells during niche colonization. Colicins are defined as plasmid-encoded toxins that exert toxicity against other strains of E. coli that do not carry the
producing Col plasmid (Konisky 1982; Pugsley 1984a,b). Colicin
D is known to cleave the anticodon loops of four isoacceptors
of tRNAArg between positions 38 and 39 (Tomita et al. 2000). In
contrast, Colicin E5 cleaves the anticodon loop of four different tRNAs (His, Tyr, Asn and Asp) between positions 34 and 35
(Ogawa et al. 1999). Recently, tRNAs have also been shown to be
one of the targets for bacterial contact-dependent growth inhibition (Diner et al. 2012).
Targeted-tRNA cleavage is also a documented bacterial antiviral defense mechanism. For example, E. coli can produce an
antiviral defense toxin, called PrrC, which specifically cleaves
tRNALys at a single site in the anticodon loop just 5 of the modified wobble uridine base. PrrC activity is induced in response
to bacteriophage T4 infection (Amitsur, Morad and Kaufmann

1989; Penner et al. 1995; Kaufmann 2000). Subsequent depletion


of tRNALys severely impairs synthesis of the late stage viral proteins by the host cell and ultimately arrests rapid spread of the
virus throughout the entire bacterial population. Homologues
of the PrrC enzyme are found across a diverse range of bacteria, thus indicating the importance of tRNA in antiviral defense strategies. Streptococcal and E. coli PrrC proteins are also
known to have fungicidal activity against yeast suggesting that
they have the potential to confer a competitive advantage on
the host bacterium during niche colonization or pathogenic infection processes (Meineke et al. 2011).

OUTLOOK
Recent years have seen new fields opening up in tRNA biology, the success of which is due in no small part to the substantial body of knowledge that has been built up over the last
50 years. Advances in genomics and other technologies that can
be applied to the entire cellular population of tRNAs have started
to reveal new roles for tRNAs as regulators of numerous cellular processes (Raina and Ibba 2014). For example, the ability to
measure the aminoacylation status of all tRNAs and to monitor tRNA modification patterns globally have begun to uncover
new roles for tRNA in cellular stress responses (Pan 2013; Gu,
Begley and Dedon 2014). Another area of growing interest comes
from the increasing awareness that tRNAs directly and indirectly
function in numerous roles outside of translation of the genetic
code (Banerjee et al. 2010; Anderson and Ivanov 2014). Overall,
research in the tRNA field is entering a new and exciting era and
there are clearly many more surprises still to come from this
small but highly versatile RNA.

FUNDING
This work was supported by a grant from the National Institutes
of Health (GM065183).
Conflict of interest. None declared.

REFERENCES
Abdi NM, Fredrick K. Contribution of 16S rRNA nucleotides forming the 30S subunit A and P sites to translation in Escherichia
coli. RNA 2005;11:162432.
Agrawal A, Mohanty BK, Kushner SR. Processing of the seven valine tRNAs in Escherichia coli involves novel features of RNase
P. Nucleic Acids Res 2014;42:116679.
Agris PF. Bringing order to translation: the contributions of
transfer RNA anticodon-domain modifications. EMBO Rep
2008;9:62935.
Ahel I, Korencic D, Ibba M, et al. Trans-editing of mischarged tRNAs. P Natl Acad Sci USA 2003;100:154227.
Altman S. Isolation of tyrosine tRNA precursor molecules. Nat
New Biol 1971;229:1921.
Altman S. Biosynthesis of transfer RNA in Escherichia coli. Cell
1975;4:219.
Altman S Transfer RNA. Cambridge, MA: Massachusetts Institute
of Technology, 1978.
Altman S, Kirsebom L. 14 Ribonuclease P. Cold Spring Harbor M
1999;37:35180.
Altman S, Smith JD. Tyrosine tRNA precursor molecule polynucleotide sequence. Nat New Biol 1971;233:359.

Shepherd and Ibba

Amberg R, Mizutani T, Wu XQ, et al. Selenocysteine synthesis in


mammalia: an identity switch from tRNA(Ser) to tRNA(Sec).
J Mol Biol 1996;263:819.
Amitsur M, Morad I, Kaufmann G. In vitro reconstitution of anticodon nuclease from components encoded by phage T4 and
Escherichia coli CTr5X. EMBO J 1989;8:24115.
An S, Musier-Forsyth K. Cys-tRNA(Pro) editing by Haemophilus
influenzae YbaK via a novel synthetase.YbaK.tRNA ternary
complex. J Biol Chem 2005;280:3446572.
Anderson P, Ivanov P. tRNA fragments in human health and disease. FEBS Lett 2014;588:4297304.
Apirion D, Miczak A. RNA processing in prokaryotic cells. Bioessays 1993;15:11320.
Asha PK, Blouin RT, Zaniewski R, et al. Ribonuclease BN: identification and partial characterization of a new tRNA processing
enzyme. P Natl Acad Sci USA 1983;80:33014.
Asha PK, Deutscher MP. Escherichia coli CAN lacks a tRNAprocessing nuclease. J Bacteriol 1983;156:41920.
Atherly AG, Menninger JR. Mutant E. coli strain with temperature sensitive peptidyl-transfer RNA hydrolase. Nat New Biol
1972;240:2456.
Bailly M, Blaise M, Lorber B, et al. The transamidosome: a
dynamic ribonucleoprotein particle dedicated to prokaryotic tRNA-dependent asparagine biosynthesis. Mol Cell
2007;28:22839.
Baird NJ, Fang XW, Srividya N, et al. Folding of a universal
ribozyme: the ribonuclease P RNA. Q Rev Biophys 2007;40:
11361.
Baldwin AN, Berg P. Transfer ribonucleic acid-induced hydrolysis of valyladenylate bound to isoleucyl ribonucleic acid synthetase. J Biol Chem 1966;241:83945.
Ban N, Nissen P, Hansen J, et al. The complete atomic structure of the large ribosomal subunit at 2.4 A resolution. Science
2000;289:90520.
Banerjee R, Chen S, Dare K, et al. tRNAs: cellular barcodes for
amino acids. FEBS Lett 2010;584:38795.
Baron C, Bock A. The length of the aminoacyl-acceptor stem of
the selenocysteine-specific tRNA(Sec) of Escherichia coli is the
determinant for binding to elongation factors SELB or Tu. J
Biol Chem 1991;266:203759.
Baron C, Westhof E, Bock A, et al. Solution structure of
selenocysteine-inserting
tRNA(Sec)
from
Escherichia
coli. Comparison with canonical tRNA(Ser). J Mol Biol
1993;231:27492.
Becker HD, Kern D. Thermus thermophilus: a link in evolution of
the tRNA-dependent amino acid amidation pathways. P Natl
Acad Sci USA 1998;95:128327.
Becker HD, Min B, Jacobi C, et al. The heterotrimeric Thermus thermophilus Asp-tRNA(Asn) amidotransferase can also generate
Gln-tRNA(Gln). FEBS Lett 2000a;476:1404.
Becker HD, Reinbolt J, Kreutzer R, et al. Existence of two distinct
aspartyl-tRNA synthetases in Thermus thermophilus. Structural and biochemical properties of the two enzymes. Biochemistry 1997;36:878597.
Becker HD, Roy H, Moulinier L, et al. Thermus thermophilus contains an eubacterial and an archaebacterial aspartyl-tRNA
synthetase. Biochemistry 2000b;39:321630.
Beebe JA, Fierke CA. A kinetic mechanism for cleavage of
precursor tRNA(Asp) catalyzed by the RNA component
of Bacillus subtilis ribonuclease P. Biochemistry 1994;33:
10294304.
Beebe K, Ribas De Pouplana L, Schimmel P. Elucidation of tRNAdependent editing by a class II tRNA synthetase and significance for cell viability. EMBO J 2003;22:66875.

293

Begley U, Dyavaiah M, Patil A, et al. Trm9-catalyzed tRNA modifications link translation to the DNA damage response. Mol
Cell 2007;28:86070.
Berk V, Zhang W, Pai RD, et al. Structural basis for mRNA
and tRNA positioning on the ribosome. P Natl Acad Sci USA
2006;103:158304.
Bjork GR, Durand JM, Hagervall TG, et al. Transfer RNA
modification: influence on translational frameshifting and
metabolism. FEBS Lett 1999;452:4751.
Bjork GR, Ericson JU, Gustafsson CE, et al. Transfer RNA modification. Annu Rev Biochem 1987;56:26387.
Blanquet S, Mechulam Y, Schmitt E, Aminoacyl-transfer RNA
maturation. In Lapointe J, Brakier-Gingras L (eds) Translation
Mechanisms. New York: Kluwer Academic/Plenum Publishers
2003, 6579.
Bock A, Hilgenfeld R, Tormay P, et al. Domain structure of the
selenocysteine-specific translation factor SelB in prokaryotes. Biomed Environ Sci 1997;10:1258.
Bonnefond L, Arai T, Sakaguchi Y, et al. Structural basis for nonribosomal peptide synthesis by an aminoacyltRNA synthetase paralog. P Natl Acad Sci USA 2011;108:
39127.
Bouhss A, Josseaume N, Severin A, et al. Synthesis of the Lalanyl-L-alanine cross-bridge of Enterococcus faecalis peptidoglycan. J Biol Chem 2002;277:4593541.
Bugg TD. Bacterial peptidoglycan biosynthesis and its inhibition.
In: Pinto BM (ed.) Comprehensive Natural Products Chemistry,
Vol. 3. Elsevier: Amsterdam, 1999, 24194.
Calendar R, Berg P. D-Tyrosyl RNA: formation, hydrolysis and utilization for protein synthesis. J Mol Biol 1967;26:3954.
Carbon J, Squires C. Studies on genetically altered transfer RNA
species in Escherichia coli. Cancer Res 1971;31:6636.
Cassano AG, Anderson VE, Harris ME. Analysis of solvent nucleophile isotope effects: evidence for concerted mechanisms
and nucleophilic activation by metal coordination in nonenzymatic and ribozyme-catalyzed phosphodiester hydrolysis.
Biochemistry 2004;43:1054759.
Chambers I, Frampton J, Goldfarb P, et al. The structure of the
mouse glutathione peroxidase gene: the selenocysteine in
the active site is encoded by the termination codon, TGA.
EMBO J 1986;5:12217.
Chan CT, Dyavaiah M, DeMott MS, et al. A quantitative systems approach reveals dynamic control of tRNA modifications during cellular stress. PLoS Genet 2010;6:e1001247.
Chen MW, Jahn D, Schon A, et al. Purification and characterization of Chlamydomonas reinhardtii chloroplast glutamyltRNA synthetase, a natural misacylating enzyme. J Biol Chem
1990;265:40547.
Chen RH, Buko AM, Whittern DN, et al. Pacidamycins, a novel
series of antibiotics with anti-Pseudomonas aeruginosa activity. II. Isolation and structural elucidation. J Antibiot 1989;42:
51220.
Cherayil JD, Bock RM. A column chromatographic procedure for
the fractionation of s-RNA. Biochemistry 1965;4:117483.
Chong YE, Yang XL, Schimmel P. Natural homolog of tRNA synthetase editing domain rescues conditional lethality caused
by mistranslation. J Biol Chem 2008;283:300738.
Cochella L, Brunelle JL, Green R. Mutational analysis reveals two
independent molecular requirements during transfer RNA
selection on the ribosome. Nat Struct Mol Biol 2007;14:306.
Commans S, Bock A. Selenocysteine inserting tRNAs: an
overview. FEMS Microbiol Rev 1999;23:33551.
Cone JE, Del Rio RM, Davis JN, et al. Chemical characterization of the selenoprotein component of clostridial glycine

294

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

reductase: identification of selenocysteine as the organoselenium moiety. P Natl Acad Sci USA 1976;73:265963.
Crick FH. Codonanticodon pairing: the wobble hypothesis. J Mol
Biol 1966;19:54855.
Cudny H, Deutscher MP. Apparent involvement of ribonuclease
D in the 3 processing of tRNA precursors. P Natl Acad Sci USA
1980;77:37841.
Curnow AW, Hong K, Yuan R, et al. Glu-tRNAGln amidotransferase: a novel heterotrimeric enzyme required for correct
decoding of glutamine codons during translation. P Natl Acad
Sci USA 1997;94:1181926.
Curnow AW, Ibba M, Soll D. tRNA-dependent asparagine formation. Nature 1996;382:58990.
Curnow AW, Tumbula DL, Pelaschier JT, et al. GlutamyltRNA(Gln) amidotransferase in Deinococcus radiodurans may
be confined to asparagine biosynthesis. P Natl Acad Sci USA
1998;95:1283843.
Cuzin F, Kretchmer N, Greenberg RE, et al. Enzymatic hydrolysis of N-substituted aminoacyl-tRNA. P Natl Acad Sci USA
1967;58:207986.
Dare K, Ibba M. Roles of tRNA in cell wall biosynthesis. Wiley Interdiscip Rev RNA 2012;3:24764.
Dare K, Shepherd J, Roy H, et al. LysPGS formation in Listeria monocytogenes has broad roles in maintaining membrane
integrity beyond antimicrobial peptide resistance. Virulence
2014;5:53446.
Das G, Varshney U. Peptidyl-tRNA hydrolase and its critical role
in protein biosynthesis. Microbiology 2006;152:21915.
Das M, Vargas-Rodriguez O, Goto Y, et al. Distinct tRNA recognition strategies used by a homologous family of editing
domains prevent mistranslation. Nucleic Acids Res 2014;42:
394353.
Deutscher MP, Lin JJ, Evans JA. Transfer RNA metabolism in Escherichia coli cells deficient in tRNA nucleotidyltransferase. J
Mol Biol 1977;117:108194.
Dickson LA, Schimmel PR. Structure of transfer RNAAMINOACYL TRANSFER RNA synthetase complexes investigated by nuclease digestion. Arch Biochem Biophys
1975;167:63845.
Diner EJ, Beck CM, Webb JS, et al. Identification of a target cell
permissive factor required for contact-dependent growth inhibition (CDI). Gene Dev 2012;26:51525.
Dramsi S, Magnet S, Davison S, et al. Covalent attachment of proteins to peptidoglycan. FEMS Microbiol Rev 2008;32:30720.
Dudock B, DiPeri C, Scileppi K, et al. The yeast phenylalanyltransfer RNA synthetase recognition site: the region adjacent to the dihydrouridine loop. P Natl Acad Sci USA 1971;68:
6814.
Dunn JJ. RNase III cleavage of single-stranded RNA. Effect of
ionic strength on the fideltiy of cleavage. J Biol Chem 1976;251:
380714.
Dunn JJ, Studier FW. T7 early RNAs and Escherichia coli ribosomal
RNAs are cut from large precursor RNAs in vivo by ribonuclease 3. P Natl Acad Sci USA 1973;70:3296300.
Dutka S, Meinnel T, Lazennec C, et al. Role of the 1-72 base pair
in tRNAs for the activity of Escherichia coli peptidyl-tRNA hydrolase. Nucleic Acids Res 1993;21:402530.
Ebel JP, Giege R, Bonnet J, et al. Factors determining the specificity of the tRNA aminoacylation reaction. Non-absolute
specificity of tRNA-aminoacyl-tRNA synthetase recognition
and particular importance of the maximal velocity. Biochimie
1973;55:54757.
Ehlert K, Tschierske M, Mori C, et al. Site-specific serine incorporation by Lif and Epr into positions 3 and 5 of the

Staphylococcal peptidoglycan interpeptide bridge. J Bacteriol


2000;182:26358.
El Yacoubi B, Bailly M, de Crecy-Lagard V. Biosynthesis and function of posttranscriptional modifications of transfer RNAs.
Annu Rev Genet 2012;46:6995.
Eldred EW, Schimmel PR. Rapid deacylation by isoleucyl transfer ribonucleic acid synthetase of isoleucine-specific transfer ribonucleic acid aminoacylated with valine. J Biol Chem
1972;247:29614.
Emilsson V, Naslund AK, Kurland CG. Thiolation of transfer RNA
in Escherichia coli varies with growth rate. Nucleic Acids Res
1992;20:4499505.
Ericson JU, Bjork GR. Pleiotropic effects induced by modification
deficiency next to the anticodon of tRNA from Salmonella typhimurium LT2. J Bacteriol 1986;166:101321.
Ernst CM, Peschel A. Broad-spectrum antimicrobial peptide resistance by MprF-mediated aminoacylation and flipping of
phospholipids. Mol Microbiol 2011;80:2909.
Ernst CM, Staubitz P, Mishra NN, et al. The bacterial defensin resistance protein MprF consists of separable domains for lipid
lysinylation and antimicrobial peptide repulsion. PLoS Pathog
2009;5:e1000660.
Farrow MA, Nordin BE, Schimmel P. Nucleotide determinants for
tRNA-dependent amino acid discrimination by a class I tRNA
synthetase. Biochemistry 1999;38:16898903.
Ferri-Fioni ML, Schmitt E, Soutourina J, et al. Structure of
crystalline D-Tyr-tRNA(Tyr) deacylase. A representative of
a new class of tRNA-dependent hydrolases. J Biol Chem
2001;276:4728590.
Filipe SR, Pinho MG, Tomasz A. Characterization of the murMN
operon involved in the synthesis of branched peptidoglycan peptides in Streptococcus pneumoniae. J Biol Chem
2000;275:2776874.
Filipe SR, Tomasz A. Inhibition of the expression of penicillin
resistance in Streptococcus pneumoniae by inactivation of cell
wall muropeptide branching genes. P Natl Acad Sci USA
2000;97:48916.
Findeiss S, Langenberger D, Stadler PF, et al. Traces of posttranscriptional RNA modifications in deep sequencing data.
Biol Chem 2011;392:30513.
Fiser A, Filipe SR, Tomasz A. Cell wall branches, penicillin resistance and the secrets of the MurM protein. Trends Microbiol
2003;11:54753.
Fonvielle M, Chemama M, Villet R, et al. Aminoacyl-tRNA recognition by the FemXWv transferase for bacterial cell wall synthesis. Nucleic Acids Res 2009;37:1589601.
Forchhammer K, Leinfelder W, Bock A. Identification of a novel
translation factor necessary for the incorporation of selenocysteine into protein. Nature 1989;342:4536.
Fournier MJ, Ozeki H. Structure and organization of the transfer ribonucleic acid genes of Escherichia coli K-12. Microbiol Rev
1985;49:37997.
Friedman M. Chemistry, nutrition, and microbiology of D-amino
acids. J Agr Food Chem 1999;47:345779.
Fromant M, Plateau P, Schmitt E, et al. Receptor site for the 5 phosphate of elongator tRNAs governs substrate selection by
peptidyl-tRNA hydrolase. Biochemistry 1999;38:49827.
Fukunaga R, Yokoyama S. Structure of the AlaX-M transediting enzyme from Pyrococcus horikoshii. Acta Crystallogr
D 2007;63:390400.
Gagnon Y, Lacoste L, Champagne N, et al. Widespread use of
the glu-tRNAGln transamidation pathway among bacteria. A
member of the alpha purple bacteria lacks glutaminyl-trna
synthetase. J Biol Chem 1996;271:1485663.

Shepherd and Ibba

Garcia-Bustos JF, Chait BT, Tomasz A. Structure of the peptide network of pneumococcal peptidoglycan. J Biol Chem
1987;262:154005.
Garcia-Villegas MR, De La Vega FM, Galindo JM, et al. PeptidyltRNA hydrolase is involved in lambda inhibition of host protein synthesis. EMBO J 1991;10:354955.
Garg RP, Gonzalez JM, Parry RJ. Biochemical characterization of
VlmL, a Seryl-tRNA synthetase encoded by the valanimycin
biosynthetic gene cluster. J Biol Chem 2006;281:2678591.
Garg RP, Ma Y, Hoyt JC, et al. Molecular characterization and
analysis of the biosynthetic gene cluster for the azoxy antibiotic valanimycin. Mol Microbiol 2002;46:50517.
Garg RP, Qian XL, Alemany LB, et al. Investigations of valanimycin biosynthesis: elucidation of the role of seryl-tRNA. P
Natl Acad Sci USA 2008;105:65437.
Gegenheimer P, Apirion D. Processing of procaryotic ribonucleic
acid. Microbiol Rev 1981;45:50241.
Gehrig S, Eberle ME, Botschen F, et al. Identification of modifications in microbial, native tRNA that suppress immunostimulatory activity. J Exp Med 2012;209:22533.
Ghora BK, Apirion D. Structural analysis and in vitro processing
to p5 rRNA of a 9S RNA molecule isolated from an rne mutant
of E. coli. Cell 1978;15:105566.
Ghosh RK, Deutscher MP. Purification of potential 3 processing
nucleases using synthetic tRNA precursors. Nucleic Acids Res
1978;5:383142.
Giannouli S, Kyritsis A, Malissovas N, et al. On the role of an unusual tRNAGly isoacceptor in Staphylococcus aureus. Biochimie
2009;91:34451.
Giege R, Sissler M, Florentz C. Universal rules and idiosyncratic
features in tRNA identity. Nucleic Acids Res 1998;26:501735.
Gillam I, Millward S, Blew D, et al. The separation of soluble ribonucleic acids on benzoylated diethylaminoethylcellulose.
Biochemistry 1967;6:304356.
Globisch D, Pearson D, Hienzsch A, et al. Systems-based analysis
of modified tRNA bases. Angew Chem Int Edit 2011;50:973942.
Gondry M, Sauguet L, Belin P, et al. Cyclodipeptide synthases are
a family of tRNA-dependent peptide bond-forming enzymes.
Nat Chem Biol 2009;5:41420.
Goodall JJ, Chen GJ, Page MG. Essential role of histidine 20 in the
catalytic mechanism of Escherichia coli peptidyl-tRNA hydrolase. Biochemistry 2004;43:458391.
Grosjean H. DNA and RNA Modification Enzymes: Structure, Mechanism, Function and Evolution. Austin, TX: Landes Bioscience,
2009.
Grunberg-Manago M, Buckingham R, Cooperman B, et al. Structure and function of the translation Machinery. Symp Soc Gen
Microbi 1978;28:27110.
Gu C, Begley TJ, Dedon PC. tRNA modifications regulate translation during cellular stress. FEBS Lett 2014;588:428796.
Guenther UP, Yandek LE, Niland CN, et al. Hidden specificity
in an apparently nonspecific RNA-binding protein. Nature
2013;502:3858.
Guerrier-Takada C, Gardiner K, Marsh T, et al. The RNA moiety
of ribonuclease P is the catalytic subunit of the enzyme. Cell
1983;35:84957.
Guillon JM, Heiss S, Soutourina J, et al. Interplay of methionine tRNAs with translation elongation factor Tu and translation initiation factor 2 in Escherichia coli. J Biol Chem
1996;271:223215.
Guillon JM, Mechulam Y, Blanquet S, et al. Importance of formylability and anticodon stem sequence to give a tRNA(Met)
an initiator identity in Escherichia coli. J Bacteriol 1993;175:
450714.

295

Guillon JM, Mechulam Y, Schmitter JM, et al. Disruption of the


gene for Met-tRNA(fMet) formyltransferase severely impairs
growth of Escherichia coli. J Bacteriol 1992a;174:4294301.
Guillon JM, Meinnel T, Mechulam Y, et al. Nucleotides of
tRNA governing the specificity of Escherichia coli methionyltRNA(fMet) formyltransferase. J Mol Biol 1992b;224:35967.
Guo M, Schimmel P. Structural analyses clarify the complex control of mistranslation by tRNA synthetases. Curr Opin Struct
Biol 2012;22:11926.
Gupta R. Halobacterium volcanii tRNAs. Identification of 41 tRNAs covering all amino acids, and the sequences of 33 class
I tRNAs. J Biol Chem 1984;259:946171.
Gustilo EM, Vendeix FA, Agris PF. tRNAs modifications bring order to gene expression. Curr Opin Microbiol 2008;11:13440.
Guth EC, Francklyn CS. Kinetic discrimination of tRNA identity
by the conserved motif 2 loop of a class II aminoacyl-tRNA
synthetase. Mol Cell 2007;25:53142.
Guthrie C, Atchison R. Biochemical characterization of RNase
P: a tRNA processing activity with protein and RNA components. Cold Spring Harbor M 1980;9:8397.
Haas ES, Brown JW. Evolutionary variation in bacterial RNase P
RNAs. Nucleic Acids Res 1998;26:40939.
Hale SP, Auld DS, Schmidt E, et al. Discrete determinants
in transfer RNA for editing and aminoacylation. Science
1997;276:12502.
Harms J, Schluenzen F, Zarivach R, et al. High resolution structure of the large ribosomal subunit from a mesophilic eubacterium. Cell 2001;107:67988.
Harris ME, Christian EL. Recent insights into the structure and
function of the ribonucleoprotein enzyme ribonuclease P.
Curr Opin Struct Biol 2003;13:32533.
Hartmann E, Hartmann RK. The enigma of ribonuclease P evolution. Trends Genet 2003;19:5619.
Haruna K, Alkazemi MH, Liu Y, et al. Engineering the elongation
factor Tu for efficient selenoprotein synthesis. Nucleic Acids
Res 2014;42:997683.
Hegde SS, Blanchard JS. Kinetic and mechanistic characterization of recombinant Lactobacillus viridescens FemX (UDP-Nacetylmuramoyl pentapeptide-lysine N6-alanyltransferase).
J Biol Chem 2003;278:228617.
Hendrickson TL, Schimmel P. Transfer RNA-dependent amino
acid discrimination by aminoacyl-tRNA synthetases. In
Lapointe J, Brakier-Gingras L (eds) Translation Mechanisms.
New York: Kluwer Academic/Plenum Publishers 2003;
3464.
Heurgue-Hamard V, Karimi R, Mora L, et al. Ribosome release factor RF4 and termination factor RF3 are involved in
dissociation of peptidyl-tRNA from the ribosome. EMBO J
1998;17:80816.
Hilgenfeld R, Bock A, Wilting R. Structural model for the
selenocysteine-specific elongation factor SelB. Biochimie
1996;78:9718.
Hoagland MB, Keller EB, Zamecnik PC. Enzymatic carboxyl activation of amino acids. J Biol Chem 1956;218:34558.
Hoagland MB, Stephenson ML, Scott JF, et al. A soluble ribonucleic acid intermediate in protein synthesis. J Biol Chem
1958;231:24157.
Hoagland MB, Zamecnik PC, Stephenson ML. Intermediate
reactions in protein biosynthesis. Biochim Biophys Acta
1957;24:2156.
Holladay DW, Pearson RL, Kelmers AD. A general method for
the separation of isoaccepting transfer ribonucleic acids: purification of five leucine transfer ribonucleic acids from Escherichia coli. Biochim Biophys Acta 1971;240:54153.

296

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

Holley RW. Structure of an alanine transfer ribonucleic acid.


JAMA 1965;194:86871.
Holley RW, Apgar J, Everett GA, et al. Structure of a ribonucleic
acid. Science 1965;147:14625.
Hong HJ, Hutchings MI, Hill LM, et al. The role of the novel Fem
protein VanK in vancomycin resistance in Streptomyces coelicolor. J Biol Chem 2005;280:1305561.
Hong HJ, Hutchings MI, Neu JM, et al. Characterization of an inducible vancomycin resistance system in Streptomyces coelicolor reveals a novel gene (vanK) required for drug resistance.
Mol Microbiol 2004;52:110721.
Hopper AK, Phizicky EM. tRNA transfers to the limelight. Gene
Dev 2003;17:16280.
Hooper ML, Russell RL, Smith JD. Mischarging in mutant tyrosine
transfer RNAs. FEBS Lett 1972;22:14955.
Hou YM, Schimmel P. A simple structural feature is a major determinant of the identity of a transfer RNA. Nature
1988;333:1405.
Hussain T, Kruparani SP, Pal B, et al. Post-transfer editing mechanism of a D-aminoacyl-tRNA deacylase-like domain in threonyl-tRNA synthetase from archaea. EMBO J
2006;25:415262.
Ibba M, Soll D. Aminoacyl-tRNA synthesis. Annu Rev Biochem
2000;69:61750.
Ikemura T, Dahlberg JE. Small ribonucleic acids of Escherichia
coli. I. Characterization by polyacrylamide gel electrophoresis and fingerprint analysis. J Biol Chem 1973a;248:
502432.
Ikemura T, Dahlberg JE. Small ribonucleic acids of Escherichia coli.
II. Noncoordinate accumulation during stringent control. J
Biol Chem 1973b;248:503341.
Jablonowski D, Schaffrath R. Zymocin, a composite chitinase and
tRNase killer toxin from yeast. Biochem Soc T 2007;35:15337.
Jack RW, Jung G. Natural peptides with antimicrobial activity.
CHIMIA 1998;52:4855.
Johnson AE, Miller DL, Cantor CR. Functional covalent complex
between elongation factor Tu and an analog of lysyl-tRNA. P
Natl Acad Sci USA 1978;75:30759.
Jorgensen F, Kurland CG. Processivity errors of gene expression
in Escherichia coli. J Mol Biol 1990;215:51121.
Kaufmann G. Anticodon nucleases. Trends Biochem Sci
2000;25:704.
Kazantsev AV, Krivenko AA, Harrington DJ, et al. High-resolution
structure of RNase P protein from Thermotoga maritima. P
Natl Acad Sci USA 2003;100:7497502.
Kazantsev AV, Krivenko AA, Harrington DJ, et al. Crystal structure of a bacterial ribonuclease P RNA. P Natl Acad Sci USA
2005;102:133927.
Kazantsev AV, Pace NR. Bacterial RNase P: a new view of an ancient enzyme. Nat Rev Microbiol 2006;4:72940.
Keller W, Wolf J, Gerber A. Editing of messenger RNA precursors
and of tRNAs by adenosine to inosine conversion. FEBS Lett
1999;452:716.
Khade P, Joseph S. Functional interactions by transfer RNAs in
the ribosome. FEBS Lett 2010;584:4206.
Kirsebom LA. RNase P RNA-mediated catalysis. Biochem Soc T
2002;30:11538.
Klein S, Lorenzo C, Hoffmann S, et al. Adaptation of Pseudomonas aeruginosa to various conditions includes tRNAdependent formation of alanyl-phosphatidylglycerol. Mol Microbiol 2009;71:55165.
Konisky J. Colicins and other bacteriocins with established modes of action. Annu Rev Microbiol 1982;36:125
44.

Korostelev A, Trakhanov S, Laurberg M, et al. Crystal structure


of a 70S ribosome-tRNA complex reveals functional interactions and rearrangements. Cell 2006;126:106577.
Kossel H, RajBhandary UL. Studies on polynucleotides. LXXXVI.
Enzymic hydrolysis of N-acylaminoacyl-transfer RNA. J Mol
Biol 1968;35:53960.
Krasilnikov AS, Xiao Y, Pan T, et al. Basis for structural diversity
in homologous RNAs. Science 2004;306:1047.
Krasilnikov AS, Yang X, Pan T, et al. Crystal structure of the specificity domain of ribonuclease P. Nature 2003;421:7604.
Kromayer M, Wilting R, Tormay P, et al. Domain structure of the
prokaryotic selenocysteine-specific elongation factor SelB. J
Mol Biol 1996;262:41320.
Kurland CG, Ehrenberg M. Constraints on the accuracy of messenger RNA movement. Q Rev Biophys 1985;18:42350.
Lam SS, Schimmel PR. Equilibrium measurements of cognate and noncognate interactions between aminoacyl
transfer RNA synthetases and transfer RNA. Biochemistry
1975;14:277580.
Lancaster L, Noller HF. Involvement of 16S rRNA nucleotides
G1338 and A1339 in discrimination of initiator tRNA. Mol Cell
2005;20:62332.
Lapointe J, Duplain L, Proulx M. A single glutamyl-tRNA synthetase aminoacylates tRNAGlu and tRNAGln in Bacillus subtilis and efficiently misacylates Escherichia coli tRNAGln1 in
vitro. J Bacteriol 1986;165:8893.
Lautru S, Gondry M, Genet R, et al. The albonoursin gene Cluster of S noursei biosynthesis of diketopiperazine metabolites
independent of nonribosomal peptide synthetases. Chem Biol
2002;9:135564.
Lee BJ, de laPena P, Tobian JA, et al. Unique pathway of expression
of an opal suppressor phosphoserine tRNA. P Natl Acad Sci
USA 1987;84:63848.
Lee CP, Seong BL, RajBhandary UL. Structural and sequence elements important for recognition of Escherichia coli formylmethionine tRNA by methionyl-tRNA transformylase are clustered in the acceptor stem. J Biol Chem 1991;266:180127.
Lee JW, Beebe K, Nangle LA, et al. Editing-defective tRNA synthetase causes protein misfolding and neurodegeneration.
Nature 2006;443:505.
Leinfelder W, Zehelein E, Mandrand-Berthelot MA, et al. Gene
for a novel tRNA species that accepts L-serine and cotranslationally inserts selenocysteine. Nature 1988;331:7235.
Leys D, Mowat CG, McLean KJ, et al. Atomic structure of Mycobacterium tuberculosis CYP121 to 1.06 A reveals novel features of
cytochrome P450. J Biol Chem 2003;278:51417.
Li Z, Deutscher MP. Maturation pathways for E. coli tRNA
precursors: a random multienzyme process in vivo. Cell
1996;86:50312.
Li Z, Deutscher MP. RNase E plays an essential role in the maturation of Escherichia coli tRNA precursors. RNA 2002;8:97109.
Ling B, Berger-Bachi B. Increased overall antibiotic susceptibility in Staphylococcus aureus femAB null mutants. Antimicrob
Agents Ch 1998;42:9368.
Ling J, Reynolds N, Ibba M. Aminoacyl-tRNA synthesis and translational quality control. Annu Rev Microbiol 2009;63:6178.
Lopes AP, Lopes LM, Fraga TR, et al. VapC from the leptospiral
VapBC toxin-antitoxin module displays ribonuclease activity
on the initiator tRNA. PLoS One 2014;9:e101678.
Lu J, Huang B, Esberg A, et al. The Kluyveromyces lactis gammatoxin targets tRNA anticodons. RNA 2005;11:164854.
Lund E, Dahlberg JE. Spacer transfer RNAs in ribosomal RNA
transcripts of E. coli: processing of 30S ribosomal RNA in vitro.
Cell 1977;11:24762.

Shepherd and Ibba

McDowall KJ, Lin-Chao S, Cohen SN. A+U content rather


than a particular nucleotide order determines the
specificity of RNase E cleavage. J Biol Chem 1994;269:
107906.
McFeeters RL. Recent antimicrobial developments targeting peptidyl-tRNA hydrolases. JSM Biotech Biomed Eng
2013;1:1006.
Maillard AP, Biarrotte-Sorin S, Villet R, et al. Structure-based sitedirected mutagenesis of the UDP-MurNAc-pentapeptidebinding cavity of the FemX alanyl transferase from Weissella
viridescens. J Bacteriol 2005;187:38338.
Maloney E, Stankowska D, Zhang J, et al. The two-domain
LysX protein of Mycobacterium tuberculosis is required for
production of lysinylated phosphatidylglycerol and resistance to cationic antimicrobial peptides. PLoS Pathog 2009;5:
e1000534.
Manley JL. Synthesis and degradation of termination and
premature-termination fragments of beta-galactosidase in
vitro and in vivo. J Mol Biol 1978;125:40732.
Meineke B, Schwer B, Schaffrath R, et al. Determinants of eukaryal cell killing by the bacterial ribotoxin PrrC. Nucleic Acids
Res 2011;39:687700.
Menninger JR, Coleman RA. Lincosamide antibiotics stimulate
dissociation of peptidyl-tRNA from ribosomes. Antimicrob
Agents Ch 1993;37:20279.
Miczak A, Srivastava RA, Apirion D. Location of the RNAprocessing enzymes RNase III, RNase E and RNase P in the
Escherichia coli cell. Mol Microbiol 1991;5:180110.
Miller D, Weissbach H. Factors involved in the transfer of
aminoacyl-tRNA to the ribosome. In: Weissbach H, Moore S
(eds). Molecular Mechanisms of Protein Biosynthesis. New York:
Academic Press, 1977, 32373.
Miller JP, Hussain Z, Schweizer MP. The involvement of
the anticodon adjacent modified nucleoside N-(9-(BETA-Dribofuranosyl) purine-6-ylcarbamoyl)-threonine in the biological function of E. coli tRNAile. Nucleic Acids Res
1976;3:1185201.
Moazed D, Noller HF. Intermediate states in the movement of
transfer RNA in the ribosome. Nature 1989;342:1428.
Mohanty BK, Kushner SR. Ribonuclease P processes polycistronic tRNA transcripts in Escherichia coli independent of
ribonuclease E. Nucleic Acids Res 2007;35:761425.
Mohanty BK, Kushner SR. Rho-independent transcription terminators inhibit RNase P processing of the secG leuU and metT
tRNA polycistronic transcripts in Escherichia coli. Nucleic Acids
Res 2008;36:36475.
Morl M, Marchfelder A. The final cut. The importance of tRNA
3 -processing. EMBO Rep 2001;2:1720.
Motorin Y, Helm M. tRNA stabilization by modified nucleotides.
Biochemistry 2010;49:493444.
Nakamura Y, Ito K. tRNA mimicry in translation termination and
beyond. Wiley Interdiscip Rev RNA 2011;2:64768.
Nameki N. Identity elements of tRNA(Thr) towards Saccharomyces cerevisiae threonyl-tRNA synthetase. Nucleic Acids
Res 1995;23:28316.
Nawrot B, Sochacka E, Duchler M. tRNA structural and functional changes induced by oxidative stress. Cell Mol Life Sci
2011;68:402332.
Nicholson AW. Function, mechanism and regulation of bacterial
ribonucleases. FEMS Microbiol Rev 1999;23:37190.
Nikolaev N, Silengo L, Schlessinger D. A role for ribonuclease
3 in processing of ribosomal ribonucleic acid and messenger ribonucleic acid precursors in Escherichia coli. J Biol Chem
1973;248:79679.

297

Niranjanakumari S, Stams T, Crary SM, et al. Protein component


of the ribozyme ribonuclease P alters substrate recognition
by directly contacting precursor tRNA. P Natl Acad Sci USA
1998;95:152127.
Nissen P, Hansen J, Ban N, et al. The structural basis of ribosome activity in peptide bond synthesis. Science 2000;289:
92030.
Noon KR, Guymon R, Crain PF, et al. Influence of temperature
on tRNA modification in archaea: Methanococcoides burtonii (optimum growth temperature [Topt], 23 degrees C)
and Stetteria hydrogenophila (Topt, 95 degrees C). J Bacteriol
2003;185:548390.
Novoa EM, Ribas de Pouplana L. Speeding with control: codon
usage, tRNAs, and ribosomes. Trends Genet 2012;28:57481.
Ogawa T, Tomita K, Ueda T, et al. A cytotoxic ribonuclease targeting specific transfer RNA anticodons. Science 1999;283:
2097100.
Ogle JM, Brodersen DE, Clemons WM, et al. Recognition of cognate transfer RNA by the 30S ribosomal subunit. Science
2001;292:897902.
Ogle JM, Carter AP, Ramakrishnan V. Insights into the decoding
mechanism from recent ribosome structures. Trends Biochem
Sci 2003;28:25966.
Oku Y, Kurokawa K, Ichihashi N, et al. Characterization of the
Staphylococcus aureus mprF gene, involved in lysinylation of
phosphatidylglycerol. Microbiology 2004;150:4551.
Ow MC, Kushner SR. Initiation of tRNA maturation by RNase
E is essential for cell viability in E. coli. Gene Dev 2002;16:
110215.
Pace NR, Brown JW. Evolutionary perspective on the structure and function of ribonuclease P, a ribozyme. J Bacteriol
1995;177:191928.
Pan T. Adaptive translation as a mechanism of stress response
and adaptation. Annu Rev Genet 2013;47:12137.
Pang YL, Poruri K, Martinis SA. tRNA synthetase: tRNA aminoacylation and beyond. Wiley Interdiscip Rev RNA 2014;5:46180.
Pape T, Wintermeyer W, Rodnina M. Induced fit in initial selection and proofreading of aminoacyl-tRNA on the ribosome.
EMBO J 1999;18:38007.
Pearson RL, Weiss JF, Kelmers AD. Improved separation
of transfer RNAs on polychlorotrifuoroethylene-supported
reversed-phase chromatography columns. Biochim Biophys
Acta 1971;228:7704.
Penner M, Morad I, Snyder L, et al. Phage T4-coded Stp: doubleedged effector of coupled DNA and tRNA-restriction systems. J Mol Biol 1995;249:85768.
Persson T, Cuzic S, Hartmann RK. Catalysis by RNase P RNA:
unique features and unprecedented active site plasticity. J
Biol Chem 2003;278:43394401.
Petersen HU, Roll T, Grunberg-Manago M, et al. Specific interaction of initiation factor IF2 of E. coli with formylmethionyltRNA f Met. Biochem Bioph Res Co 1979;91:106874.
Peypoux F, Marion D, Maget-Dana R, et al. Structure of bacillomycin F, a new peptidolipid antibiotic of the iturin group.
Eur J Biochem 1985;153:33540.
Plautz G, Apirion D. Processing of RNA in Escherichia coli is limited
in the absence of ribonuclease III, ribonuclease E and ribonuclease P. J Mol Biol 1981;149:8139.
Pugsley AP. The ins and outs of colicins. Part I: production,
and translocation across membranes. Microbiol Sci 1984a;1:
16875.
Pugsley AP. The ins and outs of colicins. Part II. Lethal action,
immunity and ecological implications. Microbiol Sci 1984b;1:
2035.

298

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

Qin D, Abdi NM, Fredrick K. Characterization of 16S rRNA mutations that decrease the fidelity of translation initiation. RNA
2007;13:234855.
Raina M, Ibba M. tRNAs as regulators of biological processes.
Front Genet 2014;5:171.
RajBhandary UL, Kohrer C. Early days of tRNA research: discovery, function, purification and sequence analysis. J Biosci
2006;31:43951.
Randau L, Soll D. Transfer RNA genes in pieces. EMBO Rep
2008;9:6238.
Ray BK, Apirion D. Transfer RNA precursors are accumulated
in Escherichia coli in the absence of RNase E. Eur J Biochem
1981;114:51724.
Reynolds NM, Lazazzera BA, Ibba M. Cellular mechanisms that
control mistranslation. Nat Rev Microbiol 2010;8:84956.
Robertson HD, Webster RE, Zinder ND. Purification and properties of ribonuclease III from Escherichia coli. J Biol Chem
1968;243:8291.
Roe B, Sirover M, Dudock B. Kinetics of homologous and heterologous aminoacylation with yeast phenylalanyl transfer
ribonucleic acid synthetase. Biochemistry 1973;12:414654.
Rohrer S, Berger-Bachi B. FemABX peptidyl transferases: a link
between branched-chain cell wall peptide formation and
beta-lactam resistance in gram-positive cocci. Antimicrob
Agents Ch 2003;47:83746.
Rohrer S, Ehlert K, Tschierske M, et al. The essential Staphylococcus aureus gene fmhB is involved in the first step of peptidoglycan pentaglycine interpeptide formation. P Natl Acad Sci
USA 1999;96:93516.
Roy H, Ibba M. RNA-dependent lipid remodeling by bacterial
multiple peptide resistance factors. P Natl Acad Sci USA
2008;105:466772.
Sakano H, Shimura Y. Sequential processing of precursor tRNA
molecules in Escherichia coli. P Natl Acad Sci USA 1975;72:
336973.
Sakano H, Shimura Y. Characterization and in vitro processing
of transfer RNA precursors accumulated in a temperaturesensitive mutant of Escherichia coli. J Mol Biol 1978;123:
287326.
Schedl P, Roberts J, Primakoff P. In vitro processing of E. coli tRNA
precursors. Cell 1976;8:58194.
Schimmel P. Mistranslation and its control by tRNA synthetases.
Philos Tr R Soc Lon B 2011;366:296571.
Schimmel PR. Approaches to understanding the mechanism of
specific protein-transfer RNA interactions. Accounts Chem Res
1977;10:4118.
Schimmel PR, Uhlenbeck OC, Lewis JB, et al. Binding of complementary oligonucleotides to free and aminoacyl transfer ribonucleic acid synthetase bound transfer ribonucleic acid.
Biochemistry 1972;11:6426.
Schleifer KH, Kandler O. Peptidoglycan types of bacterial
cell walls and their taxonomic implications. Bacteriol Rev
1972;36:40777.
Schmeing TM, Huang KS, Strobel SA, et al. An induced-fit mechanism to promote peptide bond formation and exclude hydrolysis of peptidyl-tRNA. Nature 2005;438:5204.
Schmidt FJ, McClain WH. An Escherichia coli ribonuclease which
removes an extra nucleotide from a biosynthetic intermediate of bacteriophage T4 proline transfer RNA. Nucleic Acids
Res 1978;5:412939.
Schmitt E, Blanquet S, Mechulam Y. Structure of crystalline Escherichia coli methionyl-tRNA(f)Met formyltransferase: comparison with glycinamide ribonucleotide formyltransferase.
EMBO J 1996a;15:474958.

Schmitt E, Fromant M, Plateau P, et al. Crystallization and preliminary X-ray analysis of Escherichia coli peptidyl-tRNA hydrolase. Proteins 1997a;28:1356.
Schmitt E, Mechulam Y, Fromant M, et al. Crystal structure at
1.2 A resolution and active site mapping of Escherichia coli
peptidyl-tRNA hydrolase. EMBO J 1997b;16:47609.
Schmitt E, Mechulam Y, Ruff M, et al. Crystallization and preliminary X-ray analysis of Escherichia coli methionyl-tRNA(fMet)
formyltransferase. Proteins 1996b;25:13941.
Schneider T, Senn MM, Berger-Bachi B, et al. In vitro assembly
of a complete, pentaglycine interpeptide bridge containing
cell wall precursor (lipid II-Gly5) of Staphylococcus aureus. Mol
Microbiol 2004;53:67585.
Schoemaker HJ, Schimmel PR. Photo-induced joining of a transfer RNA with its cognate aminoacyl-transfer RNA synthetase.
J Mol Biol 1974;84:50313.
Schon A, Hottinger H, Soll D. Misaminoacylation and transamidation are required for protein biosynthesis in Lactobacillus
bulgaricus. Biochimie 1988a;70:3914.
Schon A, Kannangara CG, Gough S, et al. Protein biosynthesis
in organelles requires misaminoacylation of tRNA. Nature
1988b;331:18790.
Schulman LH, Pelka H. The structural basis for the resistance of
Escherichia coli formylmethionyl transfer ribonucleic acid to
cleavage by Escherichia coli peptidyl transfer ribonucleic acid
hydrolase. J Biol Chem 1975;250:5427.
Schulman LH, Pelka H. Structural requirements for aminoacylation of Escherichia coli formylmethionine transfer RNA. Biochemistry 1977;16:425665.
Seidman JG, Schmidt FJ, Foss K, et al. A mutant of escherichia coli defective in removing 3 terminal nucleotides
from some transfer RNA precursor molecules. Cell 1975;5:
389400.
Sekiya T, Contreras R, Takeya T, et al. Total synthesis of a tyrosine
suppressor transfer RNA gene. XVII. Transcription, in vitro, of
the synthetic gene and processing of the primary transcript
to transfer RNA. J Biol Chem 1979;254:580216.
Selmer M, Dunham CM, Murphy FVt, et al. Structure of the
70S ribosome complexed with mRNA and tRNA. Science
2006;313:193542.
Seward HE, Roujeinikova A, McLean KJ, et al. Crystal structure of
the Mycobacterium tuberculosis P450 CYP121-fluconazole complex reveals new azole drug-P450 binding mode. J Biol Chem
2006;281:3943743.
Shepherd J. Characterisation of Pneumococcal Peptidoglycan CrossLinking Enzymology. Thesis, University of Warwick, 2011.
Shepherd J, Ibba M. Direction of aminoacylated transfer RNAs
into antibiotic synthesis and peptidoglycan-mediated antibiotic resistance. FEBS Lett 2013a;587:2895904.
Shepherd J, Ibba M. Lipid II-independent trans editing of mischarged tRNAs by the penicillin resistance factor MurM. J Biol
Chem 2013b;288:2591523.
Shepherd J, Ibba M. Relaxed substrate specificity leads to extensive tRNA Mischarging by Streptococcus pneumoniae class
I and Class II aminoacyl-tRNA synthetases. MBio 2014;5:
e0165614.
Shimura Y, Aono H, Ozeki H, et al. Mutant tyrosine tRNA of altered amino acid specificity. FEBS Lett 1972;22:1448.
Shoji J, Hinoo H, Sakazaki R. The constituent amino acids and
fatty acid of antibiotic 333-25. (Studies on antibiotics from
the genus Bacillus. XII. J Antibiot (Tokyo) 1976;29:5215.
Sivonen K, Namikoshi M, Evans WR, et al. Three new microcystins, cyclic heptapeptide hepatotoxins, from Nostoc sp.
strain 152. Chem Res Toxicol 1992;5:4649.

Shepherd and Ibba

Smith D, Pace NR. Multiple magnesium ions in the ribonuclease


P reaction mechanism. Biochemistry 1993;32:527381.
Smith JD, Celis JE. Mutant tyrosine transfer RNA that can be
charged with glutamine. Nat New Biol 1973;243:6671.
Smith JK, Hsieh J, Fierke CA. Importance of RNA-protein interactions in bacterial ribonuclease P structure and catalysis.
Biopolymers 2007;87:32938.
Soderbom F, Svard SG, Kirsebom LA. RNase E cleavage in the 5
leader of a tRNA precursor. J Mol Biol 2005;352:227.
D, Abelson J Transfer RNA: Structure, Properties, and Recognition.
Soll
New York: Cold Spring Harbor Laboratory 1979.
D, RajBhandary UL. tRNA: Structure, Biosynthesis, and Function.
Soll
Washington, DC: ASM Press 1995.
Soma A, Ikeuchi Y, Kanemasa S, et al. An RNA-modifying enzyme that governs both the codon and amino acid specificities of isoleucine tRNA. Mol Cell 2003;12:68998.
Soutourina J, Blanquet S, Plateau P. D-tyrosyl-tRNA(Tyr)
metabolism in Saccharomyces cerevisiae. J Biol Chem
2000a;275:1162630.
Soutourina J, Plateau P, Blanquet S. Metabolism of D-aminoacyltRNAs in Escherichia coli and Saccharomyces cerevisiae cells. J
Biol Chem 2000b;275:3253542.
Soutourina J, Plateau P, Delort F, et al. Functional characterization of the D-Tyr-tRNATyr deacylase from Escherichia coli. J Biol
Chem 1999;274:1910914.
Spitzfaden C, Nicholson N, Jones JJ, et al. The structure of ribonuclease P protein from Staphylococcus aureus reveals a unique
binding site for single-stranded RNA. J Mol Biol 2000;295:
10515.
Stams T, Niranjanakumari S, Fierke CA, et al. Ribonuclease P protein structure: evolutionary origins in the translational apparatus. Science 1998;280:7525.
Stark BC, Kole R, Bowman EJ, et al. Ribonuclease P: an enzyme with an essential RNA component. P Natl Acad Sci USA
1978;75:371721.
Staubitz P, Neumann H, Schneider T, et al. MprF-mediated
biosynthesis of lysylphosphatidylglycerol, an important determinant in staphylococcal defensin resistance. FEMS Microbiol Lett 2004;231:6771.
Stern L, Schulman LH. The role of the minor base N4acetylcytidine in the function of the Escherichia coli noninitiator methionine transfer RNA. J Biol Chem 1978;253:61329.
Stranden AM, Ehlert K, Labischinski H, et al. Cell wall monoglycine cross-bridges and methicillin hypersusceptibility in
a femAB null mutant of methicillin-resistant Staphylococcus
aureus. J Bacteriol 1997;179:916.
Strauch MA, Zalkin H, Aronson AI. Characterization of the
glutamyl-tRNA(Gln)-to-glutaminyl-tRNA(Gln) amidotransferase reaction of Bacillus subtilis. J Bacteriol 1988;170:
91620.
Sturchler C, Westhof E, Carbon P, et al. Unique secondary and
tertiary structural features of the eucaryotic selenocysteine
tRNA(Sec). Nucleic Acids Res 1993;21:10739.
Sugai M, Fujiwara T, Ohta K, et al. epr, which encodes glycylglycine endopeptidase resistance, is homologous to femAB
and affects serine content of peptidoglycan cross bridges
in Staphylococcus capitis and Staphylococcus aureus. J Bacteriol
1997;179:43118.
Sundari RM, Stringer EA, Schulman LH, et al. Interaction of
bacterial initiation factor 2 with initiator tRNA. J Biol Chem
1976;251:333845.
Swairjo MA, Schimmel PR. Breaking sieve for steric exclusion
of a noncognate amino acid from active site of a tRNA synthetase. P Natl Acad Sci USA 2005;102:98893.

299

Thiebe R, Harbers K, Zachau HG. Aminoacylation of fragment combinations from yeast tRNA phe. Eur J Biochem
1972;26:14452.
Toh Y, Hori H, Tomita K, et al. Transfer RNA synthesis and regulation. In: eLS. John Wiley & Sons Ltd, Chichester. 2001. http://
www.els.net [doi:10.1002/9780470015902.a0000529.pub3].
Tomita K, Ogawa T, Uozumi T, et al. A cytotoxic ribonuclease which specifically cleaves four isoaccepting arginine tRNAs at their anticodon loops. P Natl Acad Sci USA 2000;97:
827883.
Torres-Larios A, Swinger KK, Krasilnikov AS, et al. Crystal structure of the RNA component of bacterial ribonuclease P. Nature 2005;437:5847.
Torres-Larios A, Swinger KK, Pan T, et al. Structure of ribonuclease Pa universal ribozyme. Curr Opin Struct Biol 2006;16:
32735.
Tschierske M, Ehlert K, Stranden AM, et al. Lif, the lysostaphin
immunity factor, complements FemB in staphylococcal peptidoglycan interpeptide bridge formation. FEMS Microbiol Lett
1997;153:2614.
Tschierske M, Mori C, Rohrer S, et al. Identification of three additional femAB-like open reading frames in Staphylococcus aureus. FEMS Microbiol Lett 1999;171:97102.
Tumbula DL, Becker HD, Chang WZ, et al. Domain-specific recruitment of amide amino acids for protein synthesis. Nature
2000;407:10610.
C, Growth inhibition strategies based
Uthman S, Kheir E, Bar
on antimicrobial microbes/toxins. In: Mendez-Vilas A (ed.).
Science against Microbial Pathogens: Communicating Current Research and Technological Advances: Spain: Formatex Research
Center, 2011, 1321-9.
Valle M, Zavialov A, Li W, et al. Incorporation of aminoacyl-tRNA
into the ribosome as seen by cryo-electron microscopy. Nat
Struct Biol 2003;10:899906.
Voorhees RM, Weixlbaumer A, Loakes D, et al. Insights into substrate stabilization from snapshots of the peptidyl transferase center of the intact 70S ribosome. Nat Struct Mol Biol
2009;16:52833.
Walker SC, Engelke DR. Ribonuclease P: the evolution of an
ancient RNA enzyme. Crit Rev Biochem Mol 2006;41:77
102.
Waugh DS, Pace NR. Complementation of an RNase P RNA
(rnpB) gene deletion in Escherichia coli by homologous
genes from distantly related eubacteria. J Bacteriol 1990;172:
631622.
Weber B, Ehlert K, Diehl A, et al. The fib locus in Streptococcus pneumoniae is required for peptidoglycan crosslinking
and PBP-mediated beta-lactam resistance. FEMS Microbiol Lett
2000;188:815.
Wegscheid B, Condon C, Hartmann RK. Type A and B RNase P
RNAs are interchangeable in vivo despite substantial biophysical differences. EMBO Rep 2006;7:4117.
Weissbach H. Molecular Mechanisms of Protein Biosynthesis. New
York: Academic Press 1977.
Wetzel C, Limbach PA. Global identification of transfer RNAs
by liquid chromatography-mass spectrometry (LC-MS). J Proteomics 2012;75:345064.
White BN, Bayley ST. Further codon assignments in an extremely halophilic bacterium using a cell-free proteinsynthesizing system and a ribosomal binding assay. Can J
Biochem 1972;50:6009.
Wilcox M. Gamma-glutamyl phosphate attached to glutaminespecific tRNA. A precursor of glutaminyl-tRNA in Bacillus subtilis. Eur J Biochem 1969;11:40512.

300

FEMS Microbiology Reviews, 2015, Vol. 39, No. 3

Wilcox M, Nirenberg M. Transfer RNA as a cofactor coupling


amino acid synthesis with that of protein. P Natl Acad Sci USA
1968;61:22936.
Winther KS, Gerdes K. Enteric virulence associated protein VapC
inhibits translation by cleavage of initiator tRNA. P Natl Acad
Sci USA 2011;108:74037.
Wong FC, Beuning PJ, Silvers C, et al. An isolated class
II aminoacyl-tRNA synthetase insertion domain is functional in amino acid editing. J Biol Chem 2003;278:52857
64.
Yamane T, Miller DL, Hopfield JJ. Discrimination between D- and
L-tyrosyl transfer ribonucleic acids in peptide chain elongation. Biochemistry 1981;20:705964.
Yaniv M, Folk WR, Berg P, et al. A single mutational modification
of a tryptophan-specific transfer RNA permits aminoacylation by glutamine and translation of the codon UAG. J Mol
Biol 1974;86:24560.

Yarus M, Berg P. Recognition of tRNA by aminoacyl tRNA synthetases. J Mol Biol 1967;28:47990.
Yi C, Pan T. Cellular dynamics of RNA modification. Accounts
Chem Res 2011;44:13808.
Zhang W, Ntai I, Bolla ML, et al. Nine enzymes are required for assembly of the pacidamycin group of peptidyl nucleoside antibiotics. J Am Chem Soc 2011a;133:
52403.
Zhang W, Ntai I, Kelleher NL, et al. tRNA-dependent peptide
bond formation by the transferase PacB in biosynthesis of the
pacidamycin group of pentapeptidyl nucleoside antibiotics.
P Natl Acad Sci USA 2011b;108:1224953.
Zinoni F, Birkmann A, Stadtman TC, et al. Nucleotide sequence
and expression of the selenocysteine-containing polypeptide of formate dehydrogenase (formate-hydrogen-lyaselinked) from Escherichia coli. P Natl Acad Sci USA 1986;83:
46504.

Anda mungkin juga menyukai