Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

International Journal of Biological Macromolecules 257 (2024) 128646

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules


journal homepage: www.elsevier.com/locate/ijbiomac

Review

Biological importance of arginine: A comprehensive review of the roles in


structure, disorder, and functionality of peptides and proteins
Munishwar Nath Gupta a, 1, Vladimir N. Uversky b, *
a
Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
b
Department of Molecular Medicine, USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Arginine shows Jekyll and Hyde behavior in several respects. It participates in protein folding via ionic and H-
Molecular tweezers bonds and cation-pi interactions; the charge and hydrophobicity of its side chain make it a disorder-promoting
Cation-pi interactions amino acid. Its methylation in histones; RNA binding proteins; chaperones regulates several cellular processes.
Intrinsic disorder
The arginine-centric modifications are important in oncogenesis and as biomarkers in several cardiovascular
Protein-protein interactions
diseases. The cross-links involving arginine in collagen and cornea are involved in pathogenesis of tissues but
Hydrophobic interaction chromatography
Monoclonal antibody production have also been useful in tissue engineering and wound-dressing materials. Arginine is a part of active site of
Protein aggregation several enzymes such as GTPases, peroxidases, and sulfotransferases. Its metabolic importance is obvious as it is
Protein refolding involved in production of urea, NO, ornithine and citrulline. It can form unusual functional structures such as
Arginine methyltransferases molecular tweezers in vitro and sprockets which engage DNA chains as part of histones in vivo. It has been used
Post-translational modification in design of cell-penetrating peptides as drugs. Arginine has been used as an excipient in both solid and injectable
drug formulations; its role in suppressing opalescence due to liquid-liquid phase separation is particularly very
promising. It has been known as a suppressor of protein aggregation during protein refolding. It has proved its
usefulness in protein bioseparation processes like ion-exchange, hydrophobic and affinity chromatographies.
Arginine is an amino acid, whose importance in biological sciences and biotechnology continues to grow in
diverse ways.

1. Introduction debatable. These applications are diverse and include its function in
protein refolding, protein stabilization, as additive in freeze-drying of
Each of the amino acids present in proteins contributes in a different biologically active proteins such as enzymes, hormones, and protein
way to the structure and function of the proteins. This contribution due pharmaceuticals. This review is aimed at covering at a single place what
to their distinct side chains, of course, is modulated by the place of that we know about arginine functioning in different contexts. Apart from
amino acid in the primary sequence and the milieu around the protein. updating information about the intriguing behavior of this most basic
Arginine, with a guanidino group in its side chain and the highest pKa amino acid, this should stimulate cross-fertilization of ideas from
value (13.8) of all amino acids [1], is perhaps “more unequal among different sectors, which happen to be an eclectic mix. One aspect which
unequals”. In line with these considerations, Arakawa et al. (2007) has become important in recent years is related to roles of arginine in
wrote “there is insufficient information on arginine to elucidate its intrinsic disorder of proteins, and the present review devotes adequate
unique properties” [2]. While the subsequent period has seen some more attention to that as well.
insights into the behavior of this intriguing amino acid being gained; its
footprint in terms of its role in protein structure (and intrinsic disorder], 2. Arginine in protein structure and intrinsic disorder
biological processes, and many biotechnological applications seem to be
expanding as an unfinished story. It has been reported that “arginine is an abundant (5.1%) amino
Aside from its role in shaping structure or disorder in proteins (see acid… second most enriched amino acid in protein-protein interactions
below), the way arginine acts in several applications continues to be (after tryptophan)” [3]. In fact, as per Composition Profiler portal

* Corresponding author.
E-mail address: vuversky@usf.edu (V.N. Uversky).
1
Current address: 508/ block 3, Kirti Apartments, Mayur Vihar Phase 1 Extension,Delhi 110,091, India.

https://doi.org/10.1016/j.ijbiomac.2023.128646
Received 7 November 2023; Received in revised form 2 December 2023; Accepted 4 December 2023
Available online 6 December 2023
0141-8130/© 2023 Elsevier B.V. All rights reserved.
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

(http://www.cprofiler.org/help.html) [4], where amino acid composi­ correlation between thermodynamic and kinetic effects of salt bridges is
tions of the standard datasets have been pre-computed, as means and not a general phenomenon, as although the rates of α-helix formation at
standard deviations over 100,000 bootstrap iterations, arginine ac­ neutral pH follow the order (i + 4) Glu− /Arg+ > (i + 4) Asp− /Arg+ ≫ (i
counts for 5.40 ± 0.04 %, 4.93 ± 0.06 %, and 6.56 ± 0.13 % of residues + 4) Glu− /Lys+, the conformational stability forms a different order, (i
in SwissProt 51 [5], PDB Select 25 [6], and surface residues determined + 4) Glu− /Arg+ > (i + 4) Asp− /Arg+ ≈ (i + 4) Glu− /Lys+, suggesting
by the Molecular Surface Package over a sample of PDB structures of that the salt bridges, which do not contribute positively to thermody­
monomeric proteins, suitable for analyzing phenomena on protein sur­ namic stability may still play a kinetic role in formation or unfolding of
faces, such as binding [4], respectively. Among all amino acids, arginine secondary structures in proteins [15].
was shown to have the highest mutability in the case of missense mu­ One of the important paradigm shifts in our understanding of pro­
tations linked to the genetic disorders [7]. This is in spite of the fact that teins has been the discovery of intrinsic disorder in proteins. It is now
this amino acid can be encoded by 6 codons. clear that disorder (or unstructure, as some people prefer to call it), just
Structurally, lysine is closest to arginine and has the highest fre­ like structure, plays a number of crucial roles in functions of proteins
quency among amino acids in replacing arginine in the primary se­ [19–49]. Since disordered proteins do not fold at physiological condi­
quences of the same protein from different organisms. However, that tions, there are noticeable differences between the ordered and disor­
number is merely 48, as compared to frequency of 83 for glutamic acid dered proteins at the level of their amino acid sequences in terms of their
being replaced by aspartic acid [8]. At all physiologically relevant pH, amino acid compositions, charge, flexibility, hydrophobicity, sequence
arginine remains protonated. Unlike lysine, irrespective of pH, arginine complexity, and type and rate of amino acid substitutions over evolu­
serves exclusively as an H-donor [9]. In line with these considerations, tionary time scale. In fact, intrinsically disordered proteins (IDPs) and
Harms et al. (2011) conducted a systematic study of the ionizable groups intrinsically disordered regions (IDRs) are significantly depleted in
buried in the hydrophobic interior of proteins using staphylococcal order-promoting residues Asn, Cys, His, Ile, Leu, Met, Phe, Trp, Tyr, and
nuclease as a model [10]. This analysis revealed that lysine, aspartic Val and are substantially enriched in disorder-promoting residues Ala,
acid, and glutamic acid residues at 25 internal positions in this protein Arg, Gln, Glu, Gly, Lys, Pro, Ser, and Thr [4,23,29,50]. Therefore, Arg is
can have highly anomalous pKa values, with some being shifted by as among the disorder-promoting amino acid. The charge combined with
many as 5.7 pH units relative to normal pKa values in water [10]. On the hydrophobicity and side chain flexibility are deciding factors for this
contrary, arginine residues at the same internal positions exhibit no propensity [51].
detectable shifts in pKa, all being charged at pH ≤ 10 [10]. It was also It was pointed out that the arginine-rich motifs (ARMs) are among
emphasized that the remarkable potential of arginine residues to remain the common RNA binding domains in proteins and can function as in­
protonated in environments otherwise incompatible with charges is dependent recognition units [10,52]. Although ARMs in different pro­
determined by the capability of the guanidinium moiety of the arginine teins are all characterized by a preponderance of arginine residues, they
side chain to be he effectively neutralized via multiple hydrogen bonds have low overall sequence similarity and appear to have arisen inde­
to protein polar atoms and to site-bound water molecules [10]. The pendently throughout phylogeny [10]. It was pointed out that since
authors argued that “this unique capacity of Arg side chains to retain ARMs are intrinsically disordered, “individual arginine residues govern
their charge in dehydrated environments likely contributes toward the binding to an RNA ligand, and the inherent flexibility of the peptide
important functional roles of internal Arg residues in situations where a backbone may make it possible for ‘semi-specific’ recognition of a
charge is needed in the interior of a protein, in a lipid bilayer, or in discrete set of RNAs by a discrete set of ARM peptides and proteins”
similarly hydrophobic environments” [10]. [10].
Arginine, along with histidine and methionine, facilitates compact­ Protamines constitute an important example of proteins with high
ness of the protein tertiary structure by bringing together various sec­ arginine content [53]. These are small proteins which are actually most
ondary structure elements. These amino acids, thus also contribute to abundant proteins in sperm nucleus and were isolated by Freidrich
the enhanced stability of proteins from thermophiles [11]. The relative Miescher as early as about 150 years ago [54]. In fact, 48 % residues in
hydrophobicity of arginine is lower than that of lysine, being slightly the amino acid sequences of human protamines are arginines [53], and
higher than that of the glutamic acid and close to the hydrophobicity of the true protamines can contain up to 70 % arginine [55]. In line with
alanine [12]. While arginine shows no preference in its positioning in these observations, Fig. 1A shows that as a protein family, protamines
α-helices, β-sheets or β-turns, among α-helices, it is more often found are characterized by the exceptionally high arginine content. Based on
(along with glutamic acid, glutamine, and lysine) in α-helices near the their highly biased amino acid composition (see Fig. 1A), it is not sur­
surface of the globular proteins [13]. In terms of the propensity to form prising to find that almost all members of the protamine family are
α-helix, arginine is better than lysine and glutamic acid; whereas both intrinsically disordered (see Fig. 1B and 1C). This is agreement with
alanine and leucine are better than it [14]: rather limited experimental data showing that the free protamine is
Ala > Leu > Arg > Met > Lys > Gln > Glu unstructured in solution [56].
Based on the analysis of the stability and the folding and unfolding Being synthesized in the late-stage spermatids of many animals and
rates of 12 alanine-based α-helical peptides with a nearly identical plants, protamines are responsible for condensation of the spermatid
composition and containing three pairs of positively and negatively genome into a genetically inactive state. In addition, at least seven major
charged residues (either Glu− /Arg+, Asp− /Arg+, or Glu− /Lys+), Meu­ functions were ascribed to protamines, such as tight packaging and
zelaar et al. (2016) reported that the Glu− /Arg+ salt bridge promoted condensation of a paternal genome into a compact and hydrodynamic
α-helix content and stability at neutral pH, with the relative helicity and nucleus required for the fast movements of spermatozoa; protecting the
thermodynamic stability of the Glu− /Arg+ peptides following the trend paternal genetic message by making it inaccessible to nucleases or
(i + 4) Glu− /Arg+ > (i + 3) Glu− /Arg+ ≈ (i + 4) Arg+/Glu− > (i + 3) mutagens; removal of transcription factors and other proteins leading to
Arg+/Glu− [15]. a blank paternal genetic message that is devoid of epigenetic informa­
These observations were in line with previous studies [16–18] and tion; the imprinting of the paternal genome during spermatogenesis;
confirmed that a Glu− /Arg+-oriented salt bridge with Glu− and Arg+ acting as an epigenetic mark on some regions of the sperm genome;
spaced four peptide units apart is most favorable for the folded α-helical acting as a checkpoint during spermiogenesis, and potentially playing
conformation [15]. Furthermore, Meuzelaar et al. (2016) showed that some specific roles in the fertilized eggs [53,57]. All vertebrate prot­
the optimized Glu− /Arg+-oriented salt bridge noticeably accelerated amines contain a set of small “anchoring” domains containing multiple
α-helix formation, and slowed down the unfolding of the α-helix, acting arginine or lysine amino acids and are utilized in DNA binding, and also
in these respects much better than the optimized Asp− /Arg+ and Glu− / have multiple serine and threonine residues, which are often targeted
Lys+ salt bridges [15]. However, these authors also found that the for phosphorylation [55].

2
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

Fig. 1. Characteristic sequence features of a protamine family. A. Amino acid composition profile of 1880 protamines generated by Composition Profiler (http://
Cx − Corder
www.cprofiler.org/) [4]. The fractional difference is calculated as Corder , where Cx is the content of a given amino acid in the query set (1880 protamines or
known intrinsically disordered proteins), and Corder is the content of a given amino acid in the background set (Protein Data Bank Select 25). The amino acid residues
are ranked from most order-promoting residue to most disorder-promoting residue. Positive values indicate enrichment, and negative values indicate depletion of a
particular amino acid. The composition profile generated for experimentally validated disordered proteins from the DisProt database (black bars) is shown for
comparison. In both cases, error bars correspond to the standard deviations over 10,000 bootstrap iterations. The composition profile analysis showed that 8 out of 10
order-promoting residues (W, I, Y, F, L, V, N, and M) were statistically depleted in the protamines (p-value ≤0.05), and 6 out of 10 disorder-promoting residue (R, A,
Q, S, E, and P) were significantly enriched in these proteins. Note an extremely high relative content of arginine residues in protamines. B. Charge-hydropathy and
cumulative distribution function (CH-CDF) plot for 1880 protamines. The Y-coordinate is calculated as the distance of the corresponding protein from the boundary
in the CH plot [35,58]. The X-coordinate is calculated as the average distance of the corresponding protein’s CDF curve from the CDF boundary [58]. The resulting
CH-CDF plot allows distinguishing hybrid proteins containing moderate order and disorder from entirely ordered or entirely disordered proteins based on their
position within the CH-CDF phase space, with the quadrant where the proteins are located determines their classification [59,60]. Q1, 14 proteins (0.74 %) predicted
to be ordered by CDF and compact/ordered by CH-plot. Q2, 153 proteins (8.14 %) predicted to be ordered/compact by CH-plot and disordered by CDF (native molten
globules or hybrid proteins containing ordered domains and high levels of intrinsic disorder). Q3, 1705 proteins (90.69 %) predicted to be disordered by CH-plot and
CDF (native coils and pre-molten globules). Q4, 8 proteins (0.44 %) predicted to be disordered by CH-plot and ordered by CDF. Herein, we utilized web platform
Rapid Intrinsic Disorder Analysis Online (RIDAO) [61] to generate data for the CH-CDF plot. C. PONDR® VSL2 output for 1880 protamines. PONDR® VSL2 score is
the mean disorder score (MDS) for a protein, which is calculated as a sequence-length normalized sum of its per-residue scores. PONDR® VSL2 (%) is a percent of
predicted intrinsically disordered residues (PPIDR); i.e., percent of residues with disorder scores above 0.5. Color blocks indicate regions in which proteins are mostly
ordered (blue and light blue), moderately disordered (pink and light pink), or mostly disordered (red). If the two parameters agree, the corresponding part of the
background is dark (blue or pink), whereas light blue and light pink reflect areas in which only one of these criteria applies. The boundaries of the colored regions
represent arbitrary and accepted cutoffs for MDS (y-axis) and the percentage of predicted disordered residues (PPDR; x-axis). Generally, a PPIDR value of <10 % is
taken to correspond to a highly ordered protein, PPIDR between 10 % and 30 % is ascribed to moderately disordered protein, and PPIDR >30 % corresponds to a
highly disordered protein [62,63]. Mean disorder score (MDS) was calculated for each query protein as a protein length-normalized sum of all the per-residue
disorder scores. The per-residue disorder score ranges from 0 to 1, where a score of 0 indicates fully ordered residues and a score of 1 indicates fully disordered
residues. Residues with scores above the threshold of 0.5 were considered disordered residues. Residues with disorder scores between 0.25 and 0.5 were categorized
as highly flexible, while those with scores between 0.1 and 0.25 were classified as moderately flexible. Of the 1880 protamines, none are predicted to be highly
ordered, only 51 (2.7 %) demonstrate moderate disorder or conformational flexibility, whereas the vast majority of this set (1823 protein or 97.3 %) are predicted to
be highly disordered.

Protamine binding to DNA generates a remarkably stable complex, 3. Cation-π interactions often involve arginine
where a protamine wraps around the DNA helix in the major groove
[64], and tight binding of one protamine molecule per turn of DNA helix Apart from forming ionic bonds and H-bonds, the cation-π in­
[65] is achieved via “the combination of hydrogen bonds and electro­ teractions of arginine with aromatic ring containing amino acids and
static bonds that form between the guanidinium groups of each arginine ligands (of proteins containing arginines) are rather important in
residue in the anchoring domains of the protamine and the phosphate structural biology. The cation-π interactions originate because “electron-
groups in both DNA strands” [55]. rich π system above and below the benzene ring is partially negative and
this negatively charged region of the quadrupole interacts with posi­
tively charged amino acids” [66,67]. Such cation-π interactions have
long ago emerged as an important type of non-covalent bonds, which

3
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

play a number of crucial roles in functions of both structured and backbone and the major-groove edge of guanine, there are also simul­
intrinsically disordered proteins. This includes their involvement in taneous cation-π contacts between the guanidinium group of arginine
molecular recognition of ligands by proteins as evidenced by cation-π and flanking nucleobases [77]. This is illustrated by Fig. 2 representing
interaction between the ammonium ion of choline with the indole group different modes of the two-pronged arginine interaction with RNA [77].
of tryptophan in cholinesterase [68]. Similarly, the recognition of Rev. Here, Fig. 2A shows arginine-fork model proposed for the explanation of
peptide by the HIV Rev-responsive element was found to have a sig­ the extraordinary specificity of arginine for interaction between the
nificant contribution of cation-π interactions involving arginine residues HIV-1 Tat protein and TAR RNA [76]. Fig. 2B underscores the possibility
[69]. In proteins, phenylalanine, tyrosine and tryptophan can act as the of the guanidinium interactions with both phosphate and nucleobases as
π-systems, and lysine and arginine can be the cations, whereas histidine evidenced by the results of computational modeling of arginine bound to
represents a unique system, as it can be a cation as well as a π-system. TAR [78]. Finally, Fig. 2C shows the related details of TAR in complex
Hohlweg et al. (2018) discussed how the cation-π interaction facilitates with TBP6.7, where “R47 salt-bridges to the Uri23 phosphate with
placement of arginine in the transmembrane helical environment in an simultaneous cation-π interactions between the guanidinium and
ATPase [70]. More important, arginine has a special role in the proton nucleobases Ade22 and Uri23” [79].
translocation by the ATPase [70]. Involvement of cation-π interactions between aromatic amino acids,
Early studies focused on just geometrical criterion in identifying the such as tyrosine, and basic residues, such as arginine, in liquid-liquid
occurrence of the cation-π interaction in protein structures. Justin P. phase separation (LLPS) of several intrinsically disordered proteins has
Gallivan and Dennis A. Dougherty [71] discussed why that is problem­ been reported [80–82]. Recently, it was shown that the multiple cation-π
atic and factored in energetic considerations. Their estimates showed interactions are responsible for LLPS of the complex between synapto­
that >25 % of tryptophan residues in PDB can form cation-π in­ physin and synapsin [67]. Here, the C-terminal region of synaptophysin
teractions, and arginine is more probable than lysine in these in­ (residues 219–308) contains 10 repeat regions, 9 of which start with
teractions [71]. Kumar et al. (2018) explained why cation-π interactions tyrosine (Y-G-P/Q-Q-G) [83] and therefore obviously acts as a donor of π
maybe more common with arginine as compared to lysine [72]. In the systems. On the other hand, synapsin contains 85 positively charged
case of lysine, the interaction has a much higher contribution from amino acids and has a polybasic C-terminal intrinsically disordered re­
electrostatic forces, whereas in the case of arginine, there is about an gion that contains 31 positively charged residues, most of which are
equal contribution from electrostatic and dispersion forces. This means arginine (21/31) [84,85]. The crucial role of the multiple cation-π in­
that the arginine-π interactions are not that much affected by the polar teractions in the synaptophysin-synapsin coacervation was supported by
environments [72]. A recent review provides an update on importance the observation that the synaptophysin mutant form, where all nine
of cation-π interactions in diverse areas including healthcare [73]. The tyrosine residues (Y245, Y250, Y257, Y263, Y269, Y273, Y284, Y290,
authors pointed out that stabilization due to the cation-π interaction is and Y295) were replaced with serine failed to coacervate synapsin
higher at higher temperature. This is seen to be exploited in enzymes despite the fact this mutant form retains the negative charge of syn­
from thermophiles, which have more cation-π interactions than their aptophysin (− 8.3) [67]. Based on these findings, the authors concluded
mesophilic counterparts [73]. “these results are consistent with the possibility that multivalent elec­
Also, cation-π interactions play an important role in protein-nucleic trostatic π–cation interactions rather than simple negative–positive
acid interactions. For example, it is recognized that 72–87 % of known charge interactions mainly govern the coacervation between synapto­
protein-RNA interfaces contain arginine [74,75]. This enrichment of the physin and synapsin in living cells” [67].
protein-RNA interfaces in arginine is not random, as substitution of In the same vein, the phase separation of fused in sarcoma (FUS)
arginine with lysine often decreases the specificity of protein-RNA protein was shown to be regulated by cation-π interaction between
complexes [76]. Furthermore, although for a long time, it was known arginine and tyrosine residues [82]. The methylation of arginine
that interaction of arginine-containing proteins and RNA can be affected this cation-π interaction and can prevent the phase separation
described within the framework of an arginine-fork model [76], where a [73] (please see the section on post-translational modifications below,
single arginine forms four complementary contacts with nearby phos­ wherein arginine methyltransferases have been discussed). Further­
phates, yielding a two-pronged backbone readout, recently, it was more, a novel arginine-based interactions, arginine π-stacking, was
established that in addition to arginine interactions with the phosphate recently described [86]. This interaction mode involves arginine’s own

Fig. 2. Peculiarities of the arginine-based two-pronged arginine interactions with RNA. A. Original arginine-fork model wherein the guanidinium group salt-bridges
to two TAR phosphate groups [76]. B. Computational modeling supports nucleobase and backbone interactions [78]. C. TAR binding of R47 from TBP6.7 (PDB entry
6cmn [79]).
Reprinted (adapted) with permission from S.S. Chavali, C.E. Cavender, D.H. Mathews, J.E. Wedekind, Arginine Forks Are a Widespread Motif to Recognize Phosphate
Backbones and Guanine Nucleobases in the RNA Major Groove, J Am Chem Soc 142(47) (2020) 19835-19839. Copyright © 2020 American Chemical Society.

4
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

π-cloud in the guanidino group. The tau protein aggregation leading to


the formation of amyloid-like fibrils represents the basis of the patho­
genesis of various taupathies including Alzhiemer’s disease [86]. It was
proposed that π-stacking by arginine residues can promote aberrant
fibril interactions and also can drive the binding of other proteins to tau
fibrils, thereby contributing to the formation of toxic aggregates [86].
Although both cationic arginine and lysine residues are commonly
found in proteins capable of phase separation, arginine-rich proteins are
observed to undergo LLPS more readily than lysine-rich proteins
[82,87–92]. This observation supports the accepted notion that argi­
nine, which is abundant in the RNA-binding proteins (RBPs), is
considered as an important LLPS driver [82,88–92]. Although the dif­
ference between the lysine and arginine potentials to drive LLPS was
ascribed to the fact that in addition to the ability to form cation− π in­
teractions (with arginine forming stronger cation-π interactions with
aromatic groups), arginine is capable of formingπ− π interactions [92]
[88,93–97], recently it was established that the reentrant phase
behavior and tunable viscoelastic properties of the dense LLPS phase are
determined by the arginine hydrophobicity [87].
An important case of the arginine-centric pathogenesis is given by a
set of dipeptide repeat proteins generated as a result of a hexanucleotide
repeat expansion (HRE) GGGGCC (G4C2) mutation in the 5′ non-coding
region of the gene C9 open reading frame 72 (C9orf72) gene, which is
the most common genetic cause of amyotrophic lateral sclerosis (ALS)
and frontotemporal lobar degeneration (FTLD) [98–101]. Despite its Fig. 3. Schematic representation of an interplay between the poly(Gly-Arg),
intronic localization and lack of an ATG start codon, this expanded RNA, RNA-binding proteins (RBPs), G3BP1, YTHDF, and m6A-mRNA leading
microsatellite region was shown to encode a series of dipeptide repeat to the development of the cytoplasmic poly(Gly-Arg) biomolecular condensates.
(DPR) proteins, that can be produced by its sense (poly(Gly-Ala), poly Reprinted from Cell Reports, VOLUME 42, ISSUE 8, Jinyoung Park, Yanwei Wu,
(Gly-Pro), and poly(Gly-Arg)) [102,103] as well as antisense trans­ Wei Shao, Tania F. Gendron, Sophie J. F. van der Spek, Grigorii Sulta­
lation (poly(Pro-Ala), poly(Pro-Arg), and poly(Pro-Gly)) [104]. All six nakhmetov, Avik Basu, Paula Castellanos Otero, Caroline J Jones, Karen
DPR species are produced from the expanded microsatellite region via Jansen-Wes1, Lillian M. Daughrity, Sadhna Phanse, Giulia Del Rosso, Jimei
repeat-associated non-ATG (RAN) translation [104] and can be found in Tong, Monica Castanedes-Casey, Lulu Jiang, Jenna Libera, Björn Oskarsson,
Dennis W. Dickson, David W. Sanders, Clifford P. Brangwynne, Andrew Emili,
CNS tissue from the ALS and FTLD patients [102–104]. Since the length
Benjamin Wolozin, Leonard Petrucelli, Yong-Jie Zhang. Poly(GR) interacts with
of the pathogenic expanded repeat region of C9orf72 can range from 45
key stress granule factors promoting its assembly into cytoplasmic inclusions,
to several thousand units [101], the resulting DPR polypeptides can be 112,822 [119]. Copyright (2023), with permission from Elsevier.
very long. It was indicated that the highly charged arginine-rich poly­
peptides poly(Gly-Arg) and poly(ProArg) are the most toxic species in
processing and ribosome biogenesis, and interact with RNA-binding
Drosophila, yeast, and mammalian primary neurons [105–109].
proteins [108,109,114,117,120–124].
Furthermore, it was shown that both sense and anti-sense RNA foci from
C9orf72 expansions can be found in the same cell indicating that mul­
4. Importance of arginine in post-translational modifications of
tiple DPRs can be translated simultaneously [110,111] and therefore can
proteins
have the complex biological interactions originating from simultaneous
expression of multiple DPR variants [112].
Post-translational modifications (PTMs) “affect localization, inter­
In cellular model studies, the arginine-rich DPRs were found in
action state, stability, and turnover of proteins” [125]. PTMs are
nucleolus causing impaired rRNA synthesis and ribosome biogenesis
extensively involved in regulation of biological activities of proteins and
[113,114]. Furthermore, the majority of the binding partners involved
are part of the mechanisms by which signal transduction takes place
the overlapping interactomes of the poly(Gly-Arg) and poly(ProArg) and
[126,127]. For example, in moonlighting function, the PTM may be
were reported as proteins with low-complexity domains (LCDs) and ri­
absent or different. PTMs are also involved in redox homeostasis [128].
bosomal proteins [115–117]. Based on these and similar observations,
IDPs and IDRs are more prone to PTMs than ordered proteins and do­
Hana M. Odeh and Shorter J. Shorter concluded that one of the impor­
mains [23,26,36,39,125–127,129–131].
tant mechanisms of high toxicity of the arginine-rich DPRs can be
Arginine, being a disorder-promoting amino acid, is also targeted by a
associated with the capability of these polypeptides to cause aberrations
few biologically important PTMs [132]. However, an essential conse­
in cellular LLPS processes [118]. Furthermore, poly(Gly-Arg) was shown
quence of replacing amino group in the side chain (of lysine) with the
to form cytoplasmic inclusions that sequester RNA and RNA-binding
guanidino group (in arginine) is the drastic reduction in the post-
proteins including stress granule (SG) proteins, including the key
translational modifications of the side chain. Lysine is known to undergo
driver of the SG assembly, Ras GTPase-activating protein-binding pro­
many different kinds of acylation of the side chain amino groups in pro­
tein 1 (G3BP1), as well as YTH domain-containing family (YTHDF)
teins. In fact, >100,000 sites of Lys modifications in over 10,000 proteins
proteins capable of binding to the N6-methyladenosine (m6A)-modified
have been mapped [133], and the list of the lysine-centric PTMs includes
mRNAs [119]. Fig. 3 represents this pathological mechanism, showing
acetylation, methylation, ubiquitination, SUMOylation, NEEDylation,
the importance of the interplay between poly(Gly-Arg), RNA, RNA-
propionylation, butylation, crotonylation, malonylation, succinylation,
binding proteins (RBPs), G3BP1, YTHDF, and m6A-mRNA in the
glutarylation, β-hydroxybutylation, 2-hydroxyisobutyryation, lactylation,
development of the aforementioned cytoplasmic biomolecular conden­
and benzoylation [134]. This wide range of reversible lysine-based PTMs
sates [119]. Therefore, the capability of arginine-rich DPR proteins to
are known to regulate enzyme activities, chromatin structure, protein-
alter the LLPS and biogenesis of various MLOs represents an important
protein interactions, protein stability, and cellular localization. In fact,
mechanism contributing to the potential of these polypeptides to pro­
acetylation alone regulates a large set of biological functions, such as
mote nucleolar toxicity, inhibit protein synthesis, impair ribosomal RNA

5
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

epigenetics, homeostasis, metabolism, signal transduction, cell cycle, DNA mechanism of cellular metabolism. In fact, protein methylation is a PTM
repair, transcription, development, and aging [135]. The biological involved in a vast number of processes [144] and “1% of the functional
importance of the PTMs of lysine residues is borne out by the fact that genome encodes for the enzymes catalyzing protein methylation”
there are enzymes which are called writers, which introduce these PTMs, [144,145]. Methylation of proteins involves the transfer of a methyl
erasers, which reverse these PTMs, and readers, which are responsible for group (CH3) onto either an arginine or lysine residue, with the arginine
the downstream outputs of these PTMs [134]. methylation being far more common on the proteomic scale than lysine
On the other hand, until quite recently, only two kinds of PTMs were methylation [146,147]. Both mono- and di-methylarginines are formed,
reported for arginine, methylation and citrullination, which are cata­ the latter in either asymmetric or symmetric forms. Arginine methyl­
lyzed by arginine methyltransferases and arginine deiminases, respec­ ation is catalyzed by protein arginine methyltransferases (PRMTs)
tively [132]. Citrullination is involved in several important, biologically [148], which are “ubiquitously expressed both at the cellular compart­
relevant processes as well as is related to the pathogenesis of various mentalization, and tissue expression levels” [144]. This family of pro­
diseases, and hence continues to attract attention of the researchers teins includes “three types of that catalyze this reaction, each
[136–138]. The enzymatic citrullination is irreversible and its target can responsible for a different ArgMe end-product: Type I PRMTs (PRMT1,
be numerous proteins in cytoplasm, membranes, nucleus, and mito­ -2, -3, -4, -6 and -8) lead to asymmetric dimethylarginine (ADMA); Type
chondria as a part of apoptosis. A defect in the process can lead to the II PRMTs (PRMT5 and -9) produce symmetric dimethylarginine
leakage of the enzyme and citrullinated peptides, which induce auto­ (SDMA); and Type III PRMT (PRMT7) forms monomethyl arginine
immune responses [139].Citrullination of several proteins leads to the (MMA) only” [144] (see Fig. 5). ADMA and SDMA are both reported to
production of auto-antibodies in rheumatoid pathogenesis [140]. In inhibit nitric oxide synthase [132].
2020, a team led by Dr. Anna L. Vagstad of Prof. Dr. Jörn Piel reported Samuel et al. (2021) have discussed inhibitors of protein methyl­
that the non-proteinogenic ornithine residues can be generated post- transferases as they have shown promise as new therapies for certain
translationally via the hydrolysis of arginine residues incorporated in types of brain cancers [144]. Fig. 6 outlines how arginine methylation is
a peptide chain by a recently discovered enzyme family of peptide ar­ involved in oncogenesis. Arginine methyltransferases, apart from being
ginases (see Fig. 4) [141,142]. The enzymes are associated with the inhibited by synthetic small molecular weight inhibitors, are also
biosynthetic pathways of ribosomally synthesized and posttranslation­ controlled by alternative splicing, PTMs, miRNA, and via interactions
ally modified peptides (RiPP), being able to modify a broad range of with other proteins [144].
peptide sequences including peptides containing polyarginine repeats The importance of arginine methylation in renal transplants has
[142]. Recent detailed structural comparison revealed that although the attracted considerable attention [149–152], as circulatory ADMA,
binuclear metal cluster and the active-site environments in the con­ which serves as the endogenous nitric oxide (NO) synthase inhibitor is
ventional arginases and the peptide arginase strongly resemble each involved in progression of kidney disease, being associated with mor­
other, their quaternary structures diverge [143]. The discovery of the tality in renal transplant recipients (RTR) and increased risk of end-stage
peptide arginases that can install ornithine residues in the ribosomally renal disease in chronic kidney disease (CKD) populations [153].
synthesized peptides opens new routes in the development of the diverse Furthermore, hypertension was also shown to be associated with
ornithine peptides through RiPP technology [142]. elevated circulating ADMA concentrations [154], whereas ADMA and
Both lysine and arginine undergo methylation. Methylation of his­ SDMA are considered as cardiovascular risk factors and have emerged as
tones and other regulatory proteins involved in RNA binding, tran­ predictors of cardiovascular events and death in a range of pathologies
scription, translation, chaperone activity, etc. is a key regulatory [150–152,155,156]. Recently, arginine methylation has been shown to
be useful as a biomarker of cardiovascular diseases [157].

5. Crosslinks of arginines and their biological importance

Apart from the aforementioned PTMs of arginine, its participation in


the crosslink formation and implications of such crosslinks have been a
focus of attention for a number of years now. Crosslinking occurs both in
vivo and in vitro [158]. Although chemical crosslinking often links
amino group of the lysine or -SH group of the cysteine, the crosslinks
involving arginine have also been reported to form in vitro [159]. Collier
et al. (2016) described formation of four types of lysine-arginine cross­
links in collagen present in bones, tendons, ligaments, and dermis as a
part of the non-enzymatic glycation [160]. These advanced glycation
end products (AGEs) are formed in diabetes and other age related dis­
eases [160]. Because type I collagen has long half-life, which can be up
to 200 years in tendons [161], this protein is particularly prone to AGE
cross-linking in a number of different tissues [160]. Depending on the
reactive dicarbonyl donors, such as glucose, deoxyglucosone, methyl­
glyoxal, and glyoxal, four lysine-arginine AGE cross-links, glucosepane,
3-deoxyglucosone-derived imidazolium crosslink (DOGDIC),
methylglyoxal-derived imidazolium crosslink (MODIC), and glyoxal-
derived imidazolium crosslink (GODIC), are commonly formed [160].
It was also reported that these AGE cross-links can occur in human lens
protein, where they can be found at concentrations of 132.3–241.7
Fig. 4. Schematic representation of the peptide arginase action catalyzing
pmol/mg of protein for glucosepane, 1.3–8.0 pmol/mg of protein for
arginine to ornithine conversion during the RiPP biosynthesis.
Reprinted from Angewandte Chemie, International Edition (2020), 59 (48), DOGDIC, 40.7–97.2 pmol/mg of protein for MODIC and concentrations
21,442–21,447, Silja Mordhorst, Brandon I Morinaka, Anna L Vagstad, Jörn below the quantifiable level of the instrument for GODIC [162].
Piel. Post-translationally Acting Arginases Provide a Ribosomal Route to Non- Photosensitized crosslinking of cornea has many clinical applications
proteinogenic Ornithine Residues in Diverse Peptide Sequences [142]. Copy­ that include stromal stiffening for treatment of ectatic diseases, such as
right (2020), with permission from John Wiley and Sons. keratoconus, photobonding of LASIK flaps to the corneal stroma, and

6
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

Fig. 5. Scheme showing the different enzymatic activities of type I, II and III protein arginine N-methyltransferases (PRMTs). Type I, II and III PRMT enzymes
catalyze the transfer of a methyl group from S-adenosyl-l-methionine (SAM) to the side chain nitrogen on arginine residues on the target protein to produce
monomethylarginine (MMA). Type I enzymes then catalyze the transfer of a further methyl group, asymmetrically onto the same nitrogen atom that results in
asymmetric dimethylarginine (ADMA). Type II enzymes catalyze the transfer of a second methyl group symmetrically, onto the opposite nitrogen on the side chain of
arginine (symmetric dimethylarginine, SDMA).
Reproduced from [144] under CC Attribution license 4.0.

sealing wounds and lacerations [163–167]. While riboflavin had been protein were synthesized as a probe to identify binding regions in such
used earlier for the photosensitized crosslinking, its replacement by rose proteins including histones [174]. It should be mentioned here that the
bengal has several advantages [168], including the important capability chemical modification of guanidino group has been mostly restricted to
of rose bengal molecules to remain in a ~ 100 μm layer of stroma near the reaction with diketones, such as phenylglyoxal, 1,2-cyclohexane­
the epithelial surface rather than diffusing throughout as occurs for dione and a trimer of 2,3-butanedione [175–177].
riboflavin [169,170]. The protocols involving presence of oxygen or its Jones et al. (2019) have described the synthesis of two crosslinkers,
absence have been described [168]. Wertheimer et al. (2020) reported one homobifunctional (based upon aromatic glyoxal moieties) for
that arginine acting as an electron donor promoted the corneal photo­ forming arginine-arginine crosslinks; and another heterobifunctioal
sensitized crosslinking by rose bengal even in the absence of oxygen (based upon diketone and NHS moieties) for forming lysine-arginine
[168]. crosslinks. In these designs, attention was paid to typical distances
A collagen-chitosan 3D-hybrid scaffold was reported to be “cross- found in protein-protein interactions involving lysine and arginine
linked by arginine” to improve stability of this scaffold for tissue engi­ groups [3]. This was factored in deciding the span between the two
neering [171]. No explanation of how arginine acts as a cross-linker was reactive ends of the cross-linkers [158].
given; even more intriguing was the statement that crosslinking could Because arginine was shown to act as a very useful bioactive
also be carried out in the absence of arginine. The protocol for prepa­ component due to its excellent biosafety, antimicrobial properties, and
ration of the scaffold involved freeze-drying. It is likely that arginine therapeutic effects on wound healing, and because it can also be used for
helped the scaffold structure during freezing or drying stage [171]. treatment of specific pathological conditions, such as diabetes and
An unusual catalytic reaction was recently described for one of the trauma/hemorrhagic shock, there are multiple forms of arginine-based
radical S-adenosylmethionine (RaS) enzymes (a diverse protein super­ therapy [178]. The usefulness of arginine for wound healing is known
family capable of catalyzing chemically difficult transformations), for decades, and there are multiple arginine-based systems for the
which led to the formation of a crosslink between arginine and tyrosine application in wound healing that can be classified as direct supple­
during the biosynthesis of ribosomally synthesized and post- mental approaches of free arginine and indirect approaches based on the
translationally modified peptides (RiPPs that represent natural prod­ arginine derivatives, where modified arginine can be released after
ucts with diverse structures and functions) [172]. This arginine-tyrosine biodegradation, e.g., from wound-healing dressings [178]. Among
crosslinking resulted in the “installation of a macrocyclic carbon–carbon various means for arginine incorporation into the wound-dressing ma­
bond that links the unactivated δ-carbon of an arginine side chain to the terial are electrostatic attachment to high-molecular-weight hyaluronic
ortho-position of a tyrosine-phenol” thereby generating a unique mac­ acid (HA, which is one of the most common extracellular matrix bio­
rocyclization motif [172]. macromolecule) [179], or to the lignin nanofibrils [180], or to silicon
DNA-binding proteins frequently have arginine residues that are and inositol to form arginine silicate inositol (ASI) complex [181].
used for interaction with DNA [173]. This observation was used to Arginine can also be covalently attached to scaffold molecular chains via
construct reactive DNA probes for the proximity labeling of DNA- imine-type bonds or conjugate to polymeric skeletons [178], with the
binding [174]. For example, 1,3-diketone-modified nucleotides and characteristic examples given by the composite hydrogels of poly(vinyl
DNA capable of cross-linking with arginine-containing peptides and alcohol) (PVA) and oxidized polysaccharides [182], arginine-crafted

7
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

Fig. 6. Schematic representation of the mechanisms by which methylated arginine (ArgMe) might lead to oncogenesis. PRMTs and their alternatively spliced
isoforms have diverse roles in transcriptional regulation, splicing and DNA damage repair, through MMA, SDMA and ADMA on specific targets. PRMTs methylate a
combination of histone and non-histone targets. For example, PRMT2 methylates H3R8, inducing gene expression. PRMT1 methylates double strand break repair
protein meiotic recombination 11 homolog (Mre11), anchoring it to the double strand break. PRMT5 methylates Sm ribonucleosomal proteins, promoting uridine-
rich small nuclear ribonuclear protein (UsnRNPs) and survival motor neuron (SMN) spliceosomal complex assembly. Methylation of these targets results in the
increased expression of oncogenic genes and the attenuation of tumor suppressive genes, either through promoter activation or repression by epigenetic regulation,
alterations in splicing patterns or through an increase in genomic instability. PRMT activity has also been shown to promote tumor stem cell characteristics.
Reproduced along with the legend from [144] under CC Attribution license 4.0.

chitosan [183], and arginine-based poly(ester amides) (Arg-PEAs), important active site arginine is given by the argininosuccinate syn­
which represent a family of biodegradable and biocompatible synthetic thase, a key enzyme in urea synthesis, deficiency of which is associated
polymers consisting of three nontoxic building blocks, L-arginine, diols, with hyperammonemia [188]. The participation of this arginine in the
and dicarboxylic acids [184], ATP binding during the catalysis was also identified by chemical
modification studies [189].
6. Arginine in the active sites of enzymes It is important to emphasize here that the role of arginine in the
enzyme active sites is not limited to binding of the substrates/ligands, as
Given the positive charge on its side chain, which enable both this residue is also known to have a catalytic role in several cases. For
electrostatic and cation-π interactions, it is not surprising that arginine example, arginine has been shown to act as a general acid catalyst in
plays an important role in the active site of enzymes, quite often directly DNA cleavage by a site specific serine recombinase [190]. In xanthine
participating in the binding of a substrate. Cotton et al. (1977) had dehydrogenase also, arginine actively participates in both binding and
studied complex formation between gunidine hydrochloride and p- catalytic steps [191]. Ypt/Rab proteins are monomeric GTPases and
nitrophenyl phosphate dianion and suggested that gunidino side chain were found to have 5 invariant arginine residues (dubbed as arginine
of arginine orients the phosphate group correctly during enzymatic finger) in their catalytic domain, with substitution of only one of them
hydrolysis of phosphate compounds [185]. The involvement of arginine rendering the GAPs almost completely inactive [192].
in binding of iodine in the active site of horse radish peroxidase was Sulphotranferases have broad specificity toward -OH containing
established by chemical modification studies with phenylglyoxal [186]. substrates. Arginine residues were found to be critical for binding of the
Even much earlier, chemical modification with diones was used for coenzyme in sulphation of simple phenols by human phenol sulpho­
identifying arginine as an important active site residue of the avian liver transferase, an important detoxification enzyme with broad substrate
phosphoenolpyruvate carboxykinase, an enzyme which is a part of the specificity and lack of endogenous substrates [193]. Recently, a volume
central metabolic pathway associated with the gluconeogenesis [187]. on the sulfurtransferases represented studies focused on the use of the
The kinetic data indicated involvement of the active site arginine in the site directed mutagenesis to establish the role of arginine residue in the
carbon dioxide binding and activation [187]. Another example with catalysis by thiosulphate sulfurtransferases (TST) containing R-K-G-V-T-

8
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

A motif [194]. TSTs, which are also known as rhodanases, find wide introduction to molecular tweezers as a class supramolecular systems,
applications in medicine and biotechnology. TST also produces which now have footprints in diverse areas [207]. Two interacting
hydrogen sulfide which has emerged as an important player in both moieties connected by a span shape the tweezers. Rigidity or flexibility
intra- and intercellular signaling [194]. of the span decides how specific the tweezers would be in recognizing
Human type I D-myo-Inositol 1,4,5-trisphosphate 5-phosphatase, an the molecules. Smart tweezers, which are sensitive to the ionic, light,
enzyme involved in generating calcium signal, which in turn regulates and redox stimuli are also known [207].
several cellular process, has two reactive arginine residues in its active Kaiser et al. (2018) have investigated the evolution of amino acyl
site that play crucial role in the enzymatic activity of this protein [195]. tRNA synthtases since the pre-biotic era [208]. These enzymes have two
These arginine residues are part of the 10 amino acid-long sequence M- classes. In both, activation of the amino acid is the most conserved
N-T-R-C-P-A-W-C-D-R-I-L, which is conserved and is involved in sub­ feature. The authors described how a conserved pair of arginine residues
strate recognition [195]. Analogously, the triad of arginine residues in form the functional ends of tweezers to hold the adenosine phosphate in
the anionic binding pocket of the ArsC arsenate reductase of plasmid class II enzymes (see Fig. 7) and also established that this structural
R773 that catalyzes reduction of arsenate in Escherichia coli, was shown motif (together with the Backbone Brackets motif found in the class I
to be in arsenate binding and transition-state stabilization [196]. In the enzymes) can be traced back to the primordial forms of Protozymes of
same vein, in sulfite oxidizing enzymes, the active site arginine has been the aminoacyl tRNA synthetases and their more efficient successors, the
shown to be critical for electron transfer from Mo to heme redox center Urzymes [208].
[197]. This arginine residue is conserved in these Mo-containing en­ In yet another powerful application of arginine-based molecular
zymes and is associated with a clinical mutation leading to sulfite oxi­ tweezers, these were shown to interfere with the pathogenic aggregation
dase deficiency [197]. of abnormal Aβ, α-synuclein, and TTR to redirect this process to the
An interesting role of arginine has been observed in the peroxisomal formation of non-toxic amorphous aggregates that could be degraded
enzyme human D-amino acid oxidase (hDAAO) [198]. The enzyme is and cleared [209]. A good review of emerging importance of molecular
involved in the degradation of D-serine, which is the main co-agonist of tweezers in general in the drug discovery programs is available [210].
N-methyl D-aspartate receptors in brain and hence is involved in brain An important review focusing on applications of tweezers based upon
function and some of its diseases. An arginine residue present on the basic amino acids lysine and arginine and their applications as drugs for
monomer-monomer interface and located 20 Å from the assumed second inhibiting toxic aggregates is also available [211].
ligand-binding site was shown to be responsible for FAD binding. The
mutation of this arginine resulted in increasing innate mistargetting of 8.2. Sprocket arginine residues in histones
the enzyme to the nucleus [198]. Heme nitrite reductase produces NO
and ammonium ion and is a key enzyme of nitrogen cycle. Recently, an Hodges et al. (2015) discussed another interesting arginine-based
arginine residue was reported to assist substrate binding and donate a functionality, the DNA minor groove binding of the histone octamer in
proton during the catalysis [199]. a sprocket-like manner [212]. As the authors explained, “the intermo­
lecular contacts that bind DNA to the histone octamer is the series of
7. Metabolic importance of arginine histone arginine residues that insert into the DNA minor groove at each
superhelical location where the minor groove faces the histone
Arginine is involved in production of urea, creatine phosphate, octamer…” [212]. These arginine residues that insert into the DNA
polyamines, nitric oxide, ornithine and citrulline. Most of the physio­ minor groove are referred to as “sprocket” arginines, “since they engage
logical effects of arginine result from its production of NO by nitric oxide the DNA ‘chain’ like the teeth of a bicycle sprocket wheel” [212]. They
synthase [200,201]. Arginine is a conditional essential amino acid and are highly conserved among eukaryotic species and have important roles
lack of its adequate bioavailability plays “pivotal role in the pathogen­ in DNA binding. In fact, the nucleosome core particle is formed via the
esis of a growing number of varied diseases, including sickle cell disease, high affinity binding of the histone octamer containing two copies each
thalassemia, malaria, acute asthma, cystic fibrosis, pulmonary hyper­ of histones H2A, H2B, H3, and H4 to ~147 bp of DNA. The corre­
tension, cardiovascular disease, certain cancers, and trauma, among sponding histone-DNA binding is facilitated by >100 direct interactions
others” [202]. of the histone main-chain and side-chain groups with the DNA sugar-
Hristina et al. (2014) have reviewed the role of arginine in energy phosphate backbone and a similar number of indirect water-mediated
metabolism [203]. These authors emphasized that despite the possibility interactions [213,214]. In the nucleosome structure, the DNA minor
of the body to synthesize L-arginine, burns, infections, insufficient cir­ groove faces the histone octamer at 14 locations [215], each containing
culation, intensive physical activity, severe wounds, or sterility require a “sprocket” arginine, the side chain of which extends into the DNA
exogenous supplementation of this amino acid [203]. Its exogenous minor groove and makes extensive contacts with the DNA backbone
supplementation under intensive sport activity helps in generation of [212], constituting a significant fraction of the solvent accessible surface
ATP via AMP kinase pathway [203]. In a view of arginine involvement area that is buried upon histone-DNA binding [214]. In Saccharomyces
in energy production, it is not surprising that its metabolism is also cerevisiae (budding yeast), mutations of the individual “sprocket” argi­
affected during bacterial infection and cancer, with the two key enzymes nine residues, which are histone H4 R45A, H3 R63A, H3 R83A, H2A
in this context being arginine deiminase and arginase [204]. Although R43A, H2B R36A, H2A R78A, and H3 R49A, compromised repair of UV-
arginine is necessary for function of neuronal cells, it also causes neu­ induced DNA lesions and affected gene expression and cryptic tran­
roinflammation and NO production. A consequence of latter is that scription [212].
arginine depletion actually helps spinal cord regeneration after an injury
[205]. Furthermore, recent metabonomic-based analysis identified 8.3. Rings, strings, and stacks made of the arginine residues at protein-
arginine in plasma as a key biomarker among the 85 differentials me­ protein interfaces
tabolites identified between the chronic obstructive pulmonary disease
(COPD) patients and healthy people [206]. Although the most obvious role of positively charged arginines
(which are overrepresented at the oligomerization interfaces) in protein-
8. Arginine in protein interactions protein interactions is the formation electrostatic contacts with oppo­
sitely charged residues, in a solvated environment, arginines are capable
8.1. Arginine tweezers of formation of stable, short-range interactions via their guanidinium
groups, with water molecules stabilizing the positively charged guani­
Jeanne Leblond and Anne Petitjean (2011) provided a good dinium groups and posessing a bridging effect on the arginines pairs

9
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

Fig. 7. Arginine Tweezers. Based on the analysis of 972 protein 3D structures (448 protein chains for Class I and 524 chains for Class II), Backbone Brackets and
Arginine Tweezers were identified as structural motifs distinctive for binding to adenosine phosphate.
Reproduced from Kaiser, F.; Bittrich, S.; Salentin, S.; Leberecht, C.; Haupt, V.J.; Krautwurst, S.; Schroeder, M.; Labudde, D. Backbone Brackets and Arginine Tweezers
delineate Class I and Class II aminoacyl tRNA synthetases. PLoS Comput Biol 2018, 14, e1006101 [208] under CC Attribution license 4.0, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author and source are credited.

[216]. Based on the analysis of over 70,000 protein structures in PDB, observed. Ashlesha S. Raut and Devendra S. Kalonia have given a good
Neves et al. (2012) have found that”Clusters of four to eight arginine overview of the theoretical framework concerning formation of this
residues with Cζ-Cζ distances <5 Å, organized as rings with 4 to 8 cloudy appearance [241]. While pH, ionic strength, temperature obvi­
members, stacks of two arginines, and strings of stacked arginines, are ously are important parameters, a useful survey of excipients which
commonly located at the interfaces of oligomeric proteins” [217]. The prevent this phenomenon has been provided.
authors also revealed the presence of the planar stacking of guanidinium Oki et al. reported that of the various excipients tested, arginine was
groups of arginines, bridged by hydrogen bonds and interactions with the most effective in preventing formation of opalescence and LLPS
water molecules and showed that “guanidinium groups are commonly [242]. Banks and Cordia screened sucrose, proline, sorbitol, glycerol,
involved in 5 hydrogen bonds with water molecules and acceptor groups arginine and urea for preventing phase separation at different concen­
from surrounding amino acids” [217]. These are important observations trations [243]. They observed that “…. all six were found to be prefer­
indicating the prevalence of arginine clusters “that are exposed to entially excluded from the native state monomer by vapor pressure
interact with and potentially be controlled or switched by charged me­ osmometry, and no apparent correlations to the excipient dependence of
tabolites, membrane lipids, nucleic acids or side chains of other pro­ mAb-B melting temperatures were observed. These results and those of
teins” [217]. the effects of solution pH, addition of salt, and impact of a small number
of charge mutations were most consistent with a mechanism of local
9. Arginine applications in biotechnology excipient accumulation, to an extent dependent on their type, with the
specific residues that mediate mAb-B electrostatic protein–protein in­
So far, we have focused on arginine as a part of proteins. Its diverse teractions” [243].
applications as a free amino acid are also very important. There are some Pyne and Mitra tracked the hydration of lysozyme by attenuated
other applications as well, wherein arginine is a part of peptides. We total reflection Fourier transform infrared spectroscopy (ATR-FTIR) in
briefly review such applications in different contexts in biotechnology. the presence of sucrose, arginine, BSA, and ubiquitin [244]. They found
that hydration level changes can be correlated with the extent of LLPS.
Sucrose promoted LLPS; BSA diminished it. Arginine had a “subtle” ef­
9.1. Arginine as an excipient in drug formulations fect, while the effect of ubiquitin was concentration dependent [244].
It is notable that arginine has been an excipient in the FDA approved
Excipients are added to enhance protein stability, ensure tonicity and drugs TNKase® (tenecteplase) and Activase® (alteplase), which are
enable efficient drug delivery. In a series of reports, Wang has formulations of the biosynthetic forms of human tissue-type plasmin­
comprehensively reviewed the use of excipients for oral drug delivery ogen activator produced by Genentech [2].
and in liquid and solid formulations [218–220]. Arginine is among the
few amino acids, which have been used as excipients. While it has been
used in solid drug formulations, its usefulness in protein drug injectable 9.2. Arg as a suppressor of protein aggregation during protein refolding
formulations is even more interesting.
Administration by injections, by far, has been the most frequently Among amino acids, interactions of free arginine with proteins are
utilized route of administration of protein drugs. Several recent more complex than often realized. It has attracted significant attention
comprehensive reviews on proteins as injectables are available from researchers in the areas of protein refolding. Arakawa’s group
[221–223]. One major problem is the formation of opalescence in pro­ [2,245,246] pointed out that arginine helps in protein refolding by
tein solutions as injectables. Opalescence in protein solutions is gener­ reducing aggregation but has no effect in increasing the protein stability
ally associated with aggregation and often precedes phase separation. In under thermal stress. At the concentration range of 0.5–2 M, arginine
recent years, phase separation (discussed earlier) in cells has attracted could help in extracting the active proteins from the inclusion bodies,
considerable attention [80,91,224–236]. The biomolecular condensates thereby indicating that it enhances protein solubility. These researchers
formed as a result of this liquid-liquid phase separation (LLPS) are suggested that arginine prevents aggregation because of π electron-
known as membrane-less organelles or biomolecular condensates. Weak cation interaction between the guanidino side chain of arginine and
interactions of all kinds, such as electrostatic, hydrophobic, π-π, and tryptophan residues of the proteins [2,245,246].
cation-π are implicated in LLPS. Proteins with intrinsic disorder and Johannes Buchner and Rainer Rudolph (1991) had described a pro­
prions are more prone to participate in LLPS [237–240]. tocol for refolding of the recombinant Fab fragments of the murine
In the case of antibodies (including monoclonal antibodies), high antibody chains from the inclusion antibodies produced in E. coli [247].
concentrations (about 100 mg/ml) are commonly used in therapeutic Addition of 0.4 M Arg in the renaturation buffer led to “preferential
formulations. Under these conditions, aggregation leading to cloudy destabilization of incorrectly folded or aggregated species” and
appearance (opalescence) followed by crystallization or LLPS has been enhanced the yield of correctly refolded fragment [247]. Another

10
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

important work which showed the suppression of aggregation by Arg hydrophobic amino acids like leucine, isoleucine, and valine [255].
was reported by Shiraki et al. (2002) [248]. Eight proteins varying in Arginine also binds to peptide bonds. This is the complex nature of the
size, structural organization, and isoelectric point were used in the ex­ arginine binding to the protein surface. This overcomes the surface
periments to find means for preventing protein aggregation at two types tension effects and exclusion from the protein surface as per preferential
of conditions, thermal unfolding-induced aggregation and dilution- interaction theory. This leads to arginine suppressing protein aggrega­
induced aggregation from denatured state [248]. Of the 15 amino tion but not destabilizing proteins either [255].
acids tried, Arg was the most effective in suppressing aggregation [248]. A major insight into arginine function was provided by Das et al.
Inoue et al. (2014) described the use of 1 M Arg in reducing the high (2007), and their results deserve more attention [256]. Working with
viscosity of concentrated solutions of bovine and human gamma glob­ the Aβ1–42 peptide, these authors observed that it is the cluster of argi­
ulins for use in subcutaneous injections [249]. nines, which bind to the peptide and suppresses its aggregation [256].
Baynes et al. (2005) suggested a different and more complex hy­ Light scattering increased with arginine concentration but reached a
pothesis about prevention of aggregation by arginine [250]. They plateau beyond 0.5 M. They believe that this clustering aligns methylene
worked with association of insulin and its antibody and with refolding of groups of arginine to create a hydrophobic surface, thereby suggesting
carbonic anhydrase. According to these workers, Arg acts as a neutral that the hydrophobic interactions are the cause of the aggregation
crowder. Their coining this term is an unfortunate description [250]. suppression in the amyloid peptide [256].
Protein crowders with large diversity of structures have been described, While highlighting the involvement of hydrophobic interaction per
including those which carry no charge; PEG is one such example and has se is not novel, the molecular size of arginine has been an important
been frequently used as a protein crowder [251]. Unlike these conven­ consideration in the discussion of preferential exclusion in the incisive
tional crowders, which generally facilitate association, a “neutral analysis by Arakawa et al. [255]. Clustering by arginines, if taken into
crowder” as described by Baynes et al. (2005) “slows protein association consideration, would make a considerable difference to that quantitative
and may either speed or slow dissociation” [250]. Arginine as per their analysis.
observations slowed down association and speeded up dissociation; on Finally, mention should be made of the caution by Kim et al. (2016)
the other hand, guanidium salt had no effect on association, but accel­ that arginine under oxidizing conditions can produce oxides of nitrogen
erates dissociation of protein-protein complexes [250]. The authors which can “fragment” the proteins [257]. All in all, one needs to test an
explain these effects in terms of the transition state theory in the context additive like arginine with the specific pharmaceutical protein under all
of protein-protein interactions [250]. possible exposure conditions before using it.
Working with refolding of recombinant consensus interferon, Liu
et al. (2007) observed that arginine prevented the formation of insoluble 9.3. Arginine containing cationic cell-penetrating peptides in drug design
aggregates but soluble oligomers were still formed [252]. These authors
suggested that arginine stabilized unfolded structures giving them more Cell-penetrating peptides (CCPs) are short peptides which can cross
time to refold correctly. At the same time, arginine beyond 0.5 M con­ cell membranes. The CCPs also enable other molecules (bound or linked
centration stabilized these unfolded species so much that they did not to them) to enter into cells and hence have shown considerable promise
fold correctly but oligomerize (explaining why refolding yield decreased in drug design and even possibility of engineering of mammalian
when arginine concentration was increased beyond 0.5 M). While their genome [258–260]. Cationic CCPs include arginine-based peptides of
results about concentration dependence of the yield of refolded inter­ essentially three kinds: polyarginine (having 6 or 8 consecutive arginine
feron are interesting, the explanations are far from convincing [252]. residues); Tat peptides (such as Tat 49–57 derived from Tat protein of
The same group, investigating the effect of arginine on refolding on HIV, in which 6 arginine residues are present), and four arginine resi­
different proteins, later observed that while carbonic anhydrase (with no dues are linked on both ends of 6-amniohexanoic acid. Recent work
cysteine) could be refolded to 100 % yield; GFP (with 2 cysteine resi­ using variants of TAT-based CPP variants suggest that hydrophobicity
dues) formed soluble oligomers [253]. Recombinant human colony correlates positively with the cell-penetrating ability of these CPPs
stimulating factor (with 5 cysteines) formed significant amount of [261].
insoluble aggregates. It is known that correct pairing of cysteines to form
disulfide bonds requires a separate strategy necessitating chemicals or 9.4. Arginine in ion-exchange, hydrophobic interaction, and affinity
enzymatic interventions [254]. Therefore, mixing up the role of arginine chromatography
with disulfide bonds was not a good idea.
Around the same time, Arakawa’s group in collaboration with Tim­ Arakawa et al. (2007) presented a useful review covering usefulness
sheff further refined their ideas on how arginine suppresses protein- of arginine in protein purification with various separation strategies [2].
protein interactions [255]. Their discussion involved comparing argi­ It was found that the presence of arginine decreases the non-specific
nine with GdnHCl (a denaturant) and betaine (an osmolyte known to protein binding to the media during gel filtration. In ion-exchange
stabilize proteins). They noted that protein stabilizers like betaine are chromatography of antibodies and human interleukin-6, protein re­
excluded from the protein surface (preferential exclusion) allowing covery was better if arginine was present in the loading buffer. In case of
proteins to get preferentially hydrated, whereas the denaturants bind to hydrophobic interaction chromatography (HIC), arginine presence in
the protein surface [255]. Arginine was shown to behave in a more eluting buffer improved recovery of proteins. It also led to better re­
complex way, which is in between the behavior of the denaturants and covery of antibodies from Protein A column [262]. More recent work
stabilizers. It binds to proteins weakly requiring high concentration of showed that arginine in elution buffer reduced the presence of host
around 2 M to elute antibodies from protein A columns. Unlike what is proteins and soluble aggregates while purifying bi-specific antibodies
expected from an aggregation suppressor, arginine increases the surface and mAb on HIC columns [263].
tension of water, in fact more strongly than even GdnHCl. At the same The positive charge of arginine enables its interaction with nucleic
time, it is chaotropic (breaks the water structure including in the hy­ acids. Arginine as a ligand has been quite extensively used for purifi­
dration shell of water); i.e., not forming H-bonds with water and directly cation of nucleic acids. Notable are the protocols for purifying super­
binding to the protein surface. As chaotopes increase the solubility of coiled DNA plasmids [264] and DNA-based vaccine [265]. Meanwhile,
hydrophobic moieties, they are aggregate suppressors for proteins but 3-D printed chromatographic supports linked to arginine have been
can unfold the proteins (by interacting with hydrophobic side chains in described which bind to pDNA [266].
proteins); this property is dependent on temperature and concentration Affinity chromatography is currently mostly used by employing an
[255]. It was also observed that arginine interacts well with tyrosine and affinity matrix, which can selectively bind to a recombinant fusion
tryptophan, moderately with hydrophilic amino acids and least with protein consisting of a fusion tag attached to the target protein

11
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

[267–269]. An “Argi” system has been described in which the affinity review & editing. Vladimir N. Uversky: Conceptualization, Data
matrix is a biotinylated DNA aptamer linked to streptavidine-agarose curation, Formal analysis, Investigation, Validation, Writing – original
support [270]. The matrix could selectively bind fusion proteins with draft, Writing – review & editing.
fusion tags of cationic CPPs consisting of either polyarginine or
Tat49–57 peptide. The bound fusion proteins could be eluted in fairly
pure state by incorporating around 300 mM GuHCl; the yield was >90 % Declaration of competing interest
[270].
The authors declare that they have no known competing financial
10. Conclusions and future perspective interests or personal relationships that could have appeared to influence
the work reported in this paper.
The physical chemistry of arginine (a guanidino group and 3 meth­
ylene groups attached to the alpha carbon atom) leads to many inter­ Data availability
esting and intriguing functional consequences and a wide range of
applications. For example, arginine is among the most abundant No data was used for the research described in the article.
osmolyte in case of mollusks and crustaceans in shallow waters [271]. Its
protective behavior during freezing and freeze-drying, as well its use­ References
fulness in protein folding have both led to considerable debate. But its
usefulness in preventing opalescence in protein injectibles, which is [1] C.A. Fitch, G. Platzer, M. Okon, B.E. Garcia-Moreno, L.P. McIntosh, Arginine: its
likely to see much more work, is unquestionable. The finding that pKa value revisited, Protein Sci. 24 (5) (2015) 752–761.
[2] T. Arakawa, K. Tsumoto, Y. Kita, B. Chang, D. Ejima, Biotechnology applications
arginine self-assembles in aqueous medium in the form of clusters may of amino acids in protein purification and formulations, Amino Acids 33 (4)
possibly lead to some rethink about the exact mechanisms in its roles. (2007) 587–605.
Recent work indicates that the design of biosensors based on arginine [3] A.X. Jones, Y. Cao, Y.L. Tang, J.H. Wang, Y.H. Ding, H. Tan, Z.L. Chen, R.Q. Fang,
J. Yin, R.C. Chen, X. Zhu, Y. She, N. Huang, F. Shao, K. Ye, R.X. Sun, S.M. He,
and for its determination is also likely to see more work in near future X. Lei, M.Q. Dong, Improving mass spectrometry analysis of protein structures
[272–275]. Another interesting futuristic direction is obtaining arginine with arginine-selective chemical cross-linkers, Nat. Commun. 10 (1) (2019) 3911.
from waste via production of arginine-rich proteins. Waste valorization [4] V. Vacic, V.N. Uversky, A.K. Dunker, S. Lonardi, Composition profiler: a tool for
discovery and visualization of amino acid composition differences, BMC
is now is a part of the development of bioeconomy that shows immense
Bioinformatics 8 (2007) 211.
potential in crafting sustainable technology [276]. Li et al. (2018) have [5] A. Bairoch, R. Apweiler, C.H. Wu, W.C. Barker, B. Boeckmann, S. Ferro,
mentioned how arginine indirectly obtained from waste can prove E. Gasteiger, H. Huang, R. Lopez, M. Magrane, M.J. Martin, D.A. Natale,
useful in this context [277]. Many other recent articles highlight this as C. O’Donovan, N. Redaschi, L.S. Yeh, The universal protein resource (UniProt),
Nucleic Acids Res. 33 (Database issue) (2005) D154–D159.
an emerging strategy [278–280]. A comprehensive techno-economic [6] H.M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T.N. Bhat, H. Weissig, I.
analysis of this approach (though not just limited to arginine) has also N. Shindyalov, P.E. Bourne, The protein data bank, Nucleic Acids Res. 28 (1)
appeared recently [280]. (2000) 235–242.
[7] S. Khan, M. Vihinen, Spectrum of disease-causing mutations in protein secondary
As a disorder promoting amino acid, it continues to find novel ap­ structures, BMC Struct. Biol. 7 (2007) 56.
plications. A cluster of five arginine residues engineered into Fab frag­ [8] G.A. Petsko, D. Ringe, Protein Structure and Function, New Science Press, 2004.
ment of an antibody against hen egg while lysozyme enhanced the [9] L. Stryer, Biochemistry, 3rd ed., W H Freeman, New York, 1988.
[10] M.J. Harms, J.L. Schlessman, G.R. Sue, B. Garcia-Moreno, Arginine residues at
association rate of Ag-Fab complex formation by increasing the internal positions in a protein are always charged, Proc. Natl. Acad. Sci. U. S. A.
conformational diversity of the antibody fragment [281]. As an affinity 108 (47) (2011) 18954–18959.
maturation strategy, this has tremendous value in therapeutics. Both [11] K.V. Brinda, S. Vishveshwara, A network representation of protein structures:
implications for protein stability, Biophys. J. 89 (6) (2005) 4159–4170.
protein injectables and antibodies are seeing an upsurge in drug dis­ [12] H. Chen, F. Gu, Z. Huang, Improved Chou-Fasman method for protein secondary
covery and design sector. structure prediction, BMC Bioinformatics 7 (Suppl. 4) (2006), S14.
Molecular tweezers based upon arginine is another area where we [13] S. Kumar, M. Bansal, Geometrical and sequence characteristics of alpha-helices in
globular proteins, Biophys. J. 75 (4) (1998) 1935–1944.
are likely to see more work and more exciting applications. Same goes
[14] C.N. Pace, J.M. Scholtz, A helix propensity scale based on experimental studies of
for CPPs based upon arginine in drug targeting. In general, while its role peptides and proteins, Biophys. J. 75 (1) (1998) 422–427.
in defining protein structure, especially via cation-π interactions, would [15] H. Meuzelaar, J. Vreede, S. Woutersen, Influence of Glu/Arg, Asp/Arg, and Glu/
continue to be interesting; it is its function in IDPs/IDRs (such as being Lys salt bridges on alpha-helical stability and folding kinetics, Biophys. J. 110
(11) (2016) 2328–2341.
part of hot spots in protein-protein interactions involving regulatory [16] B.M. Huyghues-Despointes, J.M. Scholtz, R.L. Baldwin, Helical peptides with
proteins), which is more likely to see more innovative work and novel three pairs of Asp-Arg and Glu-Arg residues in different orientations and spacings,
insights. Arginine can still throw up surprises as we further look at it in Protein Sci. 2 (1) (1993) 80–85.
[17] V. Munoz, L. Serrano, Elucidating the folding problem of helical peptides using
the context of structure-disorder-function continuum model [282]. empirical parameters, Nat. Struct. Biol. 1 (6) (1994) 399–409.
[18] H. Meuzelaar, M. Tros, A. Huerta-Viga, C.N. van Dijk, J. Vreede, S. Woutersen,
Funding Solvent-exposed salt bridges influence the kinetics of alpha-Helix folding and
unfolding, J. Phys. Chem. Lett. 5 (5) (2014) 900–904.
[19] M.S. Cortese, V.N. Uversky, A. Keith Dunker, Intrinsic disorder in scaffold
This research received no external funding. proteins: getting more from less, Prog. Biophys. Mol. Biol. 98 (1) (2008) 85–106.
[20] A.K. Dunker, C.J. Brown, J.D. Lawson, L.M. Iakoucheva, Z. Obradovic, Intrinsic
disorder and protein function, Biochemistry 41 (21) (2002) 6573–6582.
Institutional review board statement [21] A.K. Dunker, C.J. Brown, Z. Obradovic, Identification and functions of usefully
disordered proteins, Adv. Protein Chem. 62 (2002) 25–49.
Not applicable. [22] A.K. Dunker, M.S. Cortese, P. Romero, L.M. Iakoucheva, V.N. Uversky, Flexible
nets: the roles of intrinsic disorder in protein interaction networks, FEBS J. 272
(20) (2005) 5129–5148.
Informed consent statement [23] A.K. Dunker, J.D. Lawson, C.J. Brown, R.M. Williams, P. Romero, J.S. Oh, C.
J. Oldfield, A.M. Campen, C.M. Ratliff, K.W. Hipps, J. Ausio, M.S. Nissen,
R. Reeves, C. Kang, C.R. Kissinger, R.W. Bailey, M.D. Griswold, W. Chiu, E.
Not applicable.
C. Garner, Z. Obradovic, Intrinsically disordered protein, J. Mol. Graph. Model.
19 (1) (2001) 26–59.
CRediT authorship contribution statement [24] A.K. Dunker, Z. Obradovic, The protein trinity—linking function and disorder,
Nat. Biotechnol. 19 (9) (2001) 805–806.
[25] A.K. Dunker, C.J. Oldfield, J. Meng, P. Romero, J.Y. Yang, J.W. Chen, V. Vacic,
Munishwar Nath Gupta: Conceptualization, Data curation, Formal Z. Obradovic, V.N. Uversky, The unfoldomics decade: an update on intrinsically
analysis, Investigation, Validation, Writing – original draft, Writing – disordered proteins, BMC Genomics 9 (Suppl. 2) (2008), S1.

12
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

[26] A.K. Dunker, I. Silman, V.N. Uversky, J.L. Sussman, Function and structure of [59] A. Mohan, W.J. Sullivan Jr., P. Radivojac, A.K. Dunker, V.N. Uversky, Intrinsic
inherently disordered proteins, Curr. Opin. Struct. Biol. 18 (6) (2008) 756–764. disorder in pathogenic and non-pathogenic microbes: discovering and analyzing
[27] H.J. Dyson, P.E. Wright, Intrinsically unstructured proteins and their functions, the unfoldomes of early-branching eukaryotes, Mol. BioSyst. 4 (4) (2008)
Nat. Rev. Mol. Cell Biol. 6 (3) (2005) 197–208. 328–340.
[28] M.N. Gupta, J. Mukherjee, Enzymology: some paradigm shifts over the years, [60] F. Huang, C. Oldfield, J. Meng, W.L. Hsu, B. Xue, V.N. Uversky, P. Romero, A.
Curr. Sci. (2013) 1178–1186. K. Dunker, Subclassifying disordered proteins by the CH-CDF plot method, Pac.
[29] P. Radivojac, L.M. Iakoucheva, C.J. Oldfield, Z. Obradovic, V.N. Uversky, A. Symp. Biocomput. (2012) 128–139.
K. Dunker, Intrinsic disorder and functional proteomics, Biophys. J. 92 (5) (2007) [61] G.W. Dayhoff 2nd, V.N. Uversky, Rapid prediction and analysis of protein
1439–1456. intrinsic disorder, Protein Sci. 31 (12) (2022), e4496.
[30] P. Tompa, Intrinsically unstructured proteins, Trends Biochem. Sci. 27 (10) [62] K. Rajagopalan, S.M. Mooney, N. Parekh, R.H. Getzenberg, P. Kulkarni,
(2002) 527–533. A majority of the cancer/testis antigens are intrinsically disordered proteins,
[31] P. Tompa, The interplay between structure and function in intrinsically J. Cell. Biochem. 112 (11) (2011) 3256–3267.
unstructured proteins, FEBS Lett. 579 (15) (2005) 3346–3354. [63] V.N. Uversky, Analyzing IDPs in Interactomes, Intrinsically Disordered Proteins:
[32] V.N. Uversky, What does it mean to be natively unfolded? Eur. J. Biochem. 269 Methods and Protocols, 2020, pp. 895–945.
(1) (2002) 2–12. [64] R. Balhorn, M. Cosman, K. Thornton, V. Krishnan, M. Corzett, G. Bench,
[33] V.N. Uversky, Natively unfolded proteins: a point where biology waits for physics, C. Kramer, N. Hud, M. Allen, M. Prieto, Protamine Mediated Condensation of
Protein Sci. 11 (4) (2002) 739–756. DNA in Mammalian Sperm, Cache River Press, 1999.
[34] V.N. Uversky, A.K. Dunker, Understanding protein non-folding, Biochim. [65] G.S. Bench, A.M. Friz, M.H. Corzett, D.H. Morse, R. Balhorn, DNA and total
Biophys. Acta 1804 (6) (2010) 1231–1264. protamine masses in individual sperm from fertile mammalian subjects,
[35] V.N. Uversky, J.R. Gillespie, A.L. Fink, Why are “natively unfolded” proteins Cytometry 23 (4) (1996) 263–271.
unstructured under physiologic conditions? Proteins 41 (3) (2000) 415–427. [66] M.S. Marshall, R.P. Steele, K.S. Thanthiriwatte, C.D. Sherrill, Potential energy
[36] V.N. Uversky, C.J. Oldfield, A.K. Dunker, Showing your ID: intrinsic disorder as curves for cation-pi interactions: off-axis configurations are also attractive,
an ID for recognition, regulation and cell signaling, J. Mol. Recognit. 18 (5) J. Phys. Chem. A 113 (48) (2009) 13628–13632.
(2005) 343–384. [67] G. Kim, S.E. Lee, S. Jeong, J. Lee, D. Park, S. Chang, Multivalent electrostatic pi-
[37] S. Vucetic, H. Xie, L.M. Iakoucheva, C.J. Oldfield, A.K. Dunker, Z. Obradovic, V. cation interaction between synaptophysin and synapsin is responsible for the
N. Uversky, Functional anthology of intrinsic disorder. 2. Cellular components, coacervation, Mol. Brain 14 (1) (2021) 137.
domains, technical terms, developmental processes, and coding sequence [68] M. Harel, I. Schalk, L. Ehret-Sabatier, F. Bouet, M. Goeldner, C. Hirth, P.
diversities correlated with long disordered regions, J. Proteome Res. 6 (5) (2007) H. Axelsen, I. Silman, J.L. Sussman, Quaternary ligand binding to aromatic
1899–1916. residues in the active-site gorge of acetylcholinesterase, Proc. Natl. Acad. Sci. U.
[38] P.E. Wright, H.J. Dyson, Intrinsically unstructured proteins: re-assessing the S. A. 90 (19) (1993) 9031–9035.
protein structure-function paradigm, J. Mol. Biol. 293 (2) (1999) 321–331. [69] L.A. Michael, J.A. Chenault, B.R. Miller 3rd, A.M. Knolhoff, M.C. Nagan, Water,
[39] H. Xie, S. Vucetic, L.M. Iakoucheva, C.J. Oldfield, A.K. Dunker, Z. Obradovic, V. shape recognition, salt bridges, and cation-pi interactions differentiate peptide
N. Uversky, Functional anthology of intrinsic disorder. 3. Ligands, post- recognition of the HIV rev-responsive element, J. Mol. Biol. 392 (3) (2009)
translational modifications, and diseases associated with intrinsically disordered 774–786.
proteins, J. Proteome Res. 6 (5) (2007) 1917–1932. [70] W. Hohlweg, G.E. Wagner, H.F. Hofbauer, F. Sarkleti, M. Setz, N. Gubensak,
[40] L.M. Iakoucheva, C.J. Brown, J.D. Lawson, Z. Obradovic, A.K. Dunker, Intrinsic S. Lichtenegger, S.F. Falsone, H. Wolinski, S. Kosol, C. Oostenbrink, S.
disorder in cell-signaling and cancer-associated proteins, J. Mol. Biol. 323 (3) D. Kohlwein, K. Zangger, A cation-pi interaction in a transmembrane helix of
(2002) 573–584. vacuolar ATPase retains the proton-transporting arginine in a hydrophobic
[41] V.D. Manyilov, N.S. Ilyinsky, S.V. Nesterov, B. Saqr, G.W. Dayhoff 2nd, E. environment, J. Biol. Chem. 293 (49) (2018) 18977–18988.
V. Zinovev, S.S. Matrenok, A.V. Fonin, I.M. Kuznetsova, K.K. Turoverov, [71] J.P. Gallivan, D.A. Dougherty, Cation-pi interactions in structural biology, Proc.
V. Ivanovich, V.N. Uversky, Chaotic aging: intrinsically disordered proteins in Natl. Acad. Sci. U. S. A. 96 (17) (1999) 9459–9464.
aging-related processes, Cell. Mol. Life Sci. 80 (9) (2023) 269. [72] K. Kumar, S.M. Woo, T. Siu, W.A. Cortopassi, F. Duarte, R.S. Paton, Cation-pi
[42] V.N. Uversky, Protein intrinsic disorder and structure-function continuum, Prog. interactions in protein-ligand binding: theory and data-mining reveal different
Mol. Biol. Transl. Sci. 166 (2019) 1–17. roles for lysine and arginine, Chem. Sci. 9 (10) (2018) 2655–2665.
[43] V.N. Uversky, Dancing protein clouds: the strange biology and chaotic physics of [73] S. Bergsma, E. Poulios, N. Charalampogiannis, O. Andraws, S. Achinas, Cation-pi
intrinsically disordered proteins, J. Biol. Chem. 291 (13) (2016) 6681–6688. Interaction as a Key Player in Healthcare: A Mini-review, Digital Medicine and
[44] R. van der Lee, M. Buljan, B. Lang, R.J. Weatheritt, G.W. Daughdrill, A.K. Dunker, Healthcare Technology, 2022.
M. Fuxreiter, J. Gough, J. Gsponer, D.T. Jones, P.M. Kim, R.W. Kriwacki, C. [74] D.M. Kruger, S. Neubacher, T.N. Grossmann, Protein-RNA interactions: structural
J. Oldfield, R.V. Pappu, P. Tompa, V.N. Uversky, P.E. Wright, M.M. Babu, characteristics and hotspot amino acids, RNA 24 (11) (2018) 1457–1465.
Classification of intrinsically disordered regions and proteins, Chem. Rev. 114 [75] A. Barik, N. C, S.P. Pilla, R.P. Bahadur, Molecular architecture of protein-RNA
(13) (2014) 6589–6631. recognition sites, J. Biomol. Struct. Dyn. 33 (12) (2015) 2738–2751.
[45] J. Habchi, P. Tompa, S. Longhi, V.N. Uversky, Introducing protein intrinsic [76] B.J. Calnan, B. Tidor, S. Biancalana, D. Hudson, A.D. Frankel, Arginine-mediated
disorder, Chem. Rev. 114 (13) (2014) 6561–6588. RNA recognition: the arginine fork, Science 252 (5009) (1991) 1167–1171.
[46] C.J. Oldfield, A.K. Dunker, Intrinsically disordered proteins and intrinsically [77] S.S. Chavali, C.E. Cavender, D.H. Mathews, J.E. Wedekind, Arginine forks are a
disordered protein regions, Annu. Rev. Biochem. 83 (2014) 553–584. widespread motif to recognize phosphate backbones and guanine nucleobases in
[47] T.L. Blundell, M.N. Gupta, S.E. Hasnain, Intrinsic disorder in proteins: relevance the RNA major groove, J. Am. Chem. Soc. 142 (47) (2020) 19835–19839.
to protein assemblies, drug design and host-pathogen interactions, Prog. Biophys. [78] F. Leclerc, M. Karplus, MCSS-based predictions of RNA binding sites, Theor.
Mol. Biol. 156 (2020) 34–42. Chem. Accounts 101 (1999) 131–137.
[48] M.N. Gupta, V.N. Uversky, Structure and disorder: protein functions depend on [79] I.A. Belashov, D.W. Crawford, C.E. Cavender, P. Dai, P.C. Beardslee, D.
this new binary transforming lock-and-key into structure-function continuum, in: H. Mathews, B.L. Pentelute, B.R. McNaughton, J.E. Wedekind, Structure of HIV
Structure and Intrinsic Disorder in Enzymology, Elsevier, 2023, pp. 127–148. TAR in complex with a lab-evolved RRM provides insight into duplex RNA
[49] V.N. Uversky, Intrinsically disordered proteins and their “mysterious”(meta) recognition and synthesis of a constrained peptide that impairs transcription,
physics, Front. Phys. 7 (2019) 10. Nucleic Acids Res. 46 (13) (2018) 6401–6415.
[50] R.M. Williams, Z. Obradovic, V. Mathura, W. Braun, E.C. Garner, J. Young, [80] T.J. Nott, E. Petsalaki, P. Farber, D. Jervis, E. Fussner, A. Plochowietz, T.
S. Takayama, C.J. Brown, A.K. Dunker, The protein non-folding problem: amino D. Craggs, D.P. Bazett-Jones, T. Pawson, J.D. Forman-Kay, A.J. Baldwin, Phase
acid determinants of intrinsic order and disorder, Pac. Symp. Biocomput. (2001) transition of a disordered nuage protein generates environmentally responsive
89–100. membraneless organelles, Mol. Cell 57 (5) (2015) 936–947.
[51] J. Bhowmick, S. Chandra, R. Varadarajan, Deep mutational scanning to probe [81] Y. Lin, S.L. Currie, M.K. Rosen, Intrinsically disordered sequences enable
specificity determinants in proteins, in: Structure and Intrinsic Disorder in modulation of protein phase separation through distributed tyrosine motifs,
Enzymology, Elsevier, 2023, pp. 31–71. J. Biol. Chem. 292 (46) (2017) 19110–19120.
[52] C.A. Smith, L. Chen, A.D. Frankel, Using peptides as models of RNA-protein [82] J. Wang, J.M. Choi, A.S. Holehouse, H.O. Lee, X. Zhang, M. Jahnel, S. Maharana,
interactions, Methods Enzymol. 318 (2000) 423–438. R. Lemaitre, A. Pozniakovsky, D. Drechsel, I. Poser, R.V. Pappu, S. Alberti, A.
[53] R. Oliva, Protamines and male infertility, Hum. Reprod. Update 12 (4) (2006) A. Hyman, A molecular grammar governing the driving forces for phase
417–435. separation of prion-like RNA binding proteins, Cell 174 (3) (2018) 688–699 e16.
[54] F. Miescher, Das Protamin, eine neue organische Base aus den Samenfäden des [83] T.C. Sudhof, F. Lottspeich, P. Greengard, E. Mehl, R. Jahn, A synaptic vesicle
Rheinlachses, Ber. Dtsch. Chem. Ges. 7 (1) (1874) 376–379. protein with a novel cytoplasmic domain and four transmembrane regions,
[55] R. Balhorn, The protamine family of sperm nuclear proteins, Genome Biol. 8 (9) Science 238 (4830) (1987) 1142–1144.
(2007) 227. [84] D. Park, Y. Wu, S.E. Lee, G. Kim, S. Jeong, D. Milovanovic, P. De Camilli,
[56] N.V. Hud, F.P. Milanovich, R. Balhorn, Evidence of novel secondary structure in S. Chang, Cooperative function of synaptophysin and synapsin in the generation
DNA-bound protamine is revealed by Raman spectroscopy, Biochemistry 33 (24) of synaptic vesicle-like clusters in non-neuronal cells, Nat. Commun. 12 (1)
(1994) 7528–7535. (2021) 263.
[57] R. Oliva, G.H. Dixon, Vertebrate protamine genes and the histone-to-protamine [85] T. Ueda, P. Greengard, Adenosine 3′:5′-monophosphate-regulated phosphoprotein
replacement reaction, Prog. Nucleic Acid Res. Mol. Biol. 40 (1991) 25–94. system of neuronal membranes. I. Solubilization, purification, and some
[58] C.J. Oldfield, Y. Cheng, M.S. Cortese, C.J. Brown, V.N. Uversky, A.K. Dunker, properties of an endogenous phosphoprotein, J. Biol. Chem. 252 (14) (1977)
Comparing and combining predictors of mostly disordered proteins, Biochemistry 5155–5163.
44 (6) (2005) 1989–2000.

13
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

[86] L. Ferrari, R. Stucchi, K. Konstantoulea, G. van de Kamp, R. Kos, W.J.C. Geerts, L. Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form
S. van Bezouwen, F.G. Forster, M. Altelaar, C.C. Hoogenraad, S.G.D. Rudiger, nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/
Arginine pi-stacking drives binding to fibrils of the Alzheimer protein tau, Nat. ALS, Acta Neuropathol. 126 (6) (2013) 829–844.
Commun. 11 (1) (2020) 571. [105] K.Y. Shi, E. Mori, Z.F. Nizami, Y. Lin, M. Kato, S. Xiang, L.C. Wu, M. Ding, Y. Yu, J.
[87] Y. Hong, S. Najafi, T. Casey, J.E. Shea, S.I. Han, D.S. Hwang, Hydrophobicity of G. Gall, S.L. McKnight, Toxic PR(n) poly-dipeptides encoded by the C9orf72
arginine leads to reentrant liquid-liquid phase separation behaviors of arginine- repeat expansion block nuclear import and export, Proc. Natl. Acad. Sci. U. S. A.
rich proteins, Nat. Commun. 13 (1) (2022) 7326. 114 (7) (2017) E1111–E1117.
[88] S. Das, Y.H. Lin, R.M. Vernon, J.D. Forman-Kay, H.S. Chan, Comparative roles of [106] H. Tran, S. Almeida, J. Moore, T.F. Gendron, U. Chalasani, Y. Lu, X. Du, J.
charge, pi, and hydrophobic interactions in sequence-dependent phase separation A. Nickerson, L. Petrucelli, Z. Weng, F.B. Gao, Differential toxicity of nuclear RNA
of intrinsically disordered proteins, Proc. Natl. Acad. Sci. U. S. A. 117 (46) (2020) foci versus dipeptide repeat proteins in a Drosophila model of C9ORF72 FTD/ALS,
28795–28805. Neuron 87 (6) (2015) 1207–1214.
[89] B.S. Schuster, G.L. Dignon, W.S. Tang, F.M. Kelley, A.K. Ranganath, C.N. Jahnke, [107] A. Jovicic, J. Mertens, S. Boeynaems, E. Bogaert, N. Chai, S.B. Yamada, J.
A.G. Simpkins, R.M. Regy, D.A. Hammer, M.C. Good, J. Mittal, Identifying W. Paul 3rd, S. Sun, J.R. Herdy, G. Bieri, N.J. Kramer, F.H. Gage, L. Van Den
sequence perturbations to an intrinsically disordered protein that determine its Bosch, W. Robberecht, A.D. Gitler, Modifiers of C9orf72 dipeptide repeat toxicity
phase-separation behavior, Proc. Natl. Acad. Sci. U. S. A. 117 (21) (2020) connect nucleocytoplasmic transport defects to FTD/ALS, Nat. Neurosci. 18 (9)
11421–11431. (2015) 1226–1229.
[90] J.A. Greig, T.A. Nguyen, M. Lee, A.S. Holehouse, A.E. Posey, R.V. Pappu, G. Jedd, [108] X. Wen, W. Tan, T. Westergard, K. Krishnamurthy, S.S. Markandaiah, Y. Shi,
Arginine-enriched mixed-charge domains provide cohesion for nuclear speckle S. Lin, N.A. Shneider, J. Monaghan, U.B. Pandey, P. Pasinelli, J.K. Ichida,
condensation, Mol. Cell 77 (6) (2020) 1237–1250 e4. D. Trotti, Antisense proline-arginine RAN dipeptides linked to C9ORF72-ALS/FTD
[91] S. Boeynaems, S. Alberti, N.L. Fawzi, T. Mittag, M. Polymenidou, F. Rousseau, form toxic nuclear aggregates that initiate in vitro and in vivo neuronal death,
J. Schymkowitz, J. Shorter, B. Wolozin, L. Van Den Bosch, P. Tompa, Neuron 84 (6) (2014) 1213–1225.
M. Fuxreiter, Protein phase separation: a new phase in cell biology, Trends Cell [109] S. Mizielinska, S. Gronke, T. Niccoli, C.E. Ridler, E.L. Clayton, A. Devoy, T. Moens,
Biol. 28 (6) (2018) 420–435. F.E. Norona, I.O.C. Woollacott, J. Pietrzyk, K. Cleverley, A.J. Nicoll, S. Pickering-
[92] R.M. Vernon, P.A. Chong, B. Tsang, T.H. Kim, A. Bah, P. Farber, H. Lin, J. Brown, J. Dols, M. Cabecinha, O. Hendrich, P. Fratta, E.M.C. Fisher, L. Partridge,
D. Forman-Kay, Pi-Pi contacts are an overlooked protein feature relevant to phase A.M. Isaacs, C9orf72 repeat expansions cause neurodegeneration in Drosophila
separation, Elife 7 (2018). through arginine-rich proteins, Science 345 (6201) (2014) 1192–1194.
[93] G. Krainer, T.J. Welsh, J.A. Joseph, J.R. Espinosa, S. Wittmann, E. de Csillery, [110] M. Yamakawa, D. Ito, T. Honda, K. Kubo, M. Noda, K. Nakajima, N. Suzuki,
A. Sridhar, Z. Toprakcioglu, G. Gudiskyte, M.A. Czekalska, W.E. Arter, J. Guillen- Characterization of the dipeptide repeat protein in the molecular pathogenesis of
Boixet, T.M. Franzmann, S. Qamar, P.S. George-Hyslop, A.A. Hyman, c9FTD/ALS, Hum. Mol. Genet. 24 (6) (2015) 1630–1645.
R. Collepardo-Guevara, S. Alberti, T.P.J. Knowles, Reentrant liquid condensate [111] S. Mizielinska, T. Lashley, F.E. Norona, E.L. Clayton, C.E. Ridler, P. Fratta, A.
phase of proteins is stabilized by hydrophobic and non-ionic interactions, Nat. M. Isaacs, C9orf72 frontotemporal lobar degeneration is characterised by frequent
Commun. 12 (1) (2021) 1085. neuronal sense and antisense RNA foci, Acta Neuropathol. 126 (6) (2013)
[94] L.J. Prather, G.M. Weerasekare, M. Sima, C. Quinn, R.J. Stewart, Aqueous liquid- 845–857.
liquid phase separation of natural and synthetic polyguanidiniums, Polymers [112] A.L. Darling, L. Breydo, E.G. Rivas, N.T. Gebru, D. Zheng, J.D. Baker, L.J. Blair, C.
(Basel) 11 (4) (2019). A. Dickey, J. Koren 3rd, V.N. Uversky, Repeated repeat problems: combinatorial
[95] M. Vazdar, J. Heyda, P.E. Mason, G. Tesei, C. Allolio, M. Lund, P. Jungwirth, effect of C9orf72-derived dipeptide repeat proteins, Int. J. Biol. Macromol. 127
Arginine “magic”: guanidinium like-charge ion pairing from aqueous salts to cell (2019) 136–145.
penetrating peptides, Acc. Chem. Res. 51 (6) (2018) 1455–1464. [113] Z. Tao, H. Wang, Q. Xia, K. Li, K. Li, X. Jiang, G. Xu, G. Wang, Z. Ying, Nucleolar
[96] B. Gao, T. Wyttenbach, M.T. Bowers, Protonated arginine and protonated lysine: stress and impaired stress granule formation contribute to C9orf72 RAN
hydration and its effect on the stability of salt-bridge structures, J. Phys. Chem. B translation-induced cytotoxicity, Hum. Mol. Genet. 24 (9) (2015) 2426–2441.
113 (29) (2009) 9995–10000. [114] I. Kwon, S. Xiang, M. Kato, L. Wu, P. Theodoropoulos, T. Wang, J. Kim, J. Yun,
[97] J. Vondrasek, P.E. Mason, J. Heyda, K.D. Collins, P. Jungwirth, The molecular Y. Xie, S.L. McKnight, Poly-dipeptides encoded by the C9orf72 repeats bind
origin of like-charge arginine-arginine pairing in water, J. Phys. Chem. B 113 (27) nucleoli, impede RNA biogenesis, and kill cells, Science 345 (6201) (2014)
(2009) 9041–9045. 1139–1145.
[98] H. Daoud, H. Suhail, M. Sabbagh, V. Belzil, A. Szuto, A. Dionne-Laporte, J. Khoris, [115] T.G. Moens, T. Niccoli, K.M. Wilson, M.L. Atilano, N. Birsa, L.M. Gittings, B.
W. Camu, F. Salachas, V. Meininger, J. Mathieu, M. Strong, P.A. Dion, G. V. Holbling, M.C. Dyson, A. Thoeng, J. Neeves, I. Glaria, L. Yu, J. Bussmann,
A. Rouleau, C9orf72 hexanucleotide repeat expansions as the causative mutation E. Storkebaum, M. Pardo, J.S. Choudhary, P. Fratta, L. Partridge, A.M. Isaacs,
for chromosome 9p21-linked amyotrophic lateral sclerosis and frontotemporal C9orf72 arginine-rich dipeptide proteins interact with ribosomal proteins in vivo
dementia, Arch. Neurol. 69 (9) (2012) 1159–1163. to induce a toxic translational arrest that is rescued by eIF1A, Acta Neuropathol.
[99] A.E. Renton, E. Majounie, A. Waite, J. Simon-Sanchez, S. Rollinson, J.R. Gibbs, J. 137 (3) (2019) 487–500.
C. Schymick, H. Laaksovirta, J.C. van Swieten, L. Myllykangas, H. Kalimo, [116] H. Hartmann, D. Hornburg, M. Czuppa, J. Bader, M. Michaelsen, D. Farny,
A. Paetau, Y. Abramzon, A.M. Remes, A. Kaganovich, S.W. Scholz, J. Duckworth, T. Arzberger, M. Mann, F. Meissner, D. Edbauer, Proteomics and C9orf72
J. Ding, D.W. Harmer, D.G. Hernandez, J.O. Johnson, K. Mok, M. Ryten, neuropathology identify ribosomes as poly-GR/PR interactors driving toxicity,
D. Trabzuni, R.J. Guerreiro, R.W. Orrell, J. Neal, A. Murray, J. Pearson, I. Life Sci Alliance 1 (2) (2018), e201800070.
E. Jansen, D. Sondervan, H. Seelaar, D. Blake, K. Young, N. Halliwell, J. [117] K.H. Lee, P. Zhang, H.J. Kim, D.M. Mitrea, M. Sarkar, B.D. Freibaum, J. Cika,
B. Callister, G. Toulson, A. Richardson, A. Gerhard, J. Snowden, D. Mann, M. Coughlin, J. Messing, A. Molliex, B.A. Maxwell, N.C. Kim, J. Temirov,
D. Neary, M.A. Nalls, T. Peuralinna, L. Jansson, V.M. Isoviita, A.L. Kaivorinne, J. Moore, R.M. Kolaitis, T.I. Shaw, B. Bai, J. Peng, R.W. Kriwacki, J.P. Taylor,
M. Holtta-Vuori, E. Ikonen, R. Sulkava, M. Benatar, J. Wuu, A. Chio, G. Restagno, C9orf72 dipeptide repeats impair the assembly, dynamics, and function of
G. Borghero, M. Sabatelli, I. Consortium, D. Heckerman, E. Rogaeva, L. Zinman, J. membrane-less organelles, Cell 167 (3) (2016) 774–788 e17.
D. Rothstein, M. Sendtner, C. Drepper, E.E. Eichler, C. Alkan, Z. Abdullaev, S. [118] H.M. Odeh, J. Shorter, Arginine-rich dipeptide-repeat proteins as phase disruptors
D. Pack, A. Dutra, E. Pak, J. Hardy, A. Singleton, N.M. Williams, P. Heutink, in C9-ALS/FTD, Emerg. Top. Life Sci. 4 (3) (2020) 293–305.
S. Pickering-Brown, H.R. Morris, P.J. Tienari, B.J. Traynor, A hexanucleotide [119] J. Park, Y. Wu, W. Shao, T.F. Gendron, S.J.F. van der Spek, G. Sultanakhmetov,
repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD, A. Basu, P. Castellanos Otero, C.J. Jones, K. Jansen-West, L.M. Daughrity,
Neuron 72 (2) (2011) 257–268. S. Phanse, G. Del Rosso, J. Tong, M. Castanedes-Casey, L. Jiang, J. Libera,
[100] M. DeJesus-Hernandez, I.R. Mackenzie, B.F. Boeve, A.L. Boxer, M. Baker, N. B. Oskarsson, D.W. Dickson, D.W. Sanders, C.P. Brangwynne, A. Emili,
J. Rutherford, A.M. Nicholson, N.A. Finch, H. Flynn, J. Adamson, N. Kouri, B. Wolozin, L. Petrucelli, Y.J. Zhang, Poly(GR) interacts with key stress granule
A. Wojtas, P. Sengdy, G.Y. Hsiung, A. Karydas, W.W. Seeley, K.A. Josephs, factors promoting its assembly into cytoplasmic inclusions, Cell Rep. 42 (8)
G. Coppola, D.H. Geschwind, Z.K. Wszolek, H. Feldman, D.S. Knopman, R. (2023), 112822.
C. Petersen, B.L. Miller, D.W. Dickson, K.B. Boylan, N.R. Graff-Radford, [120] C.N. Cook, Y. Wu, H.M. Odeh, T.F. Gendron, K. Jansen-West, G. Del Rosso,
R. Rademakers, Expanded GGGGCC hexanucleotide repeat in noncoding region of M. Yue, P. Jiang, E. Gomes, J. Tong, L.M. Daughrity, N.M. Avendano,
C9ORF72 causes chromosome 9p-linked FTD and ALS, Neuron 72 (2) (2011) M. Castanedes-Casey, W. Shao, B. Oskarsson, G.S. Tomassy, A. McCampbell,
245–256. F. Rigo, D.W. Dickson, J. Shorter, Y.J. Zhang, L. Petrucelli, C9orf72 poly(GR)
[101] I. Gijselinck, M. Cruts, C. Van Broeckhoven, The genetics of C9orf72 expansions, aggregation induces TDP-43 proteinopathy, Sci. Transl. Med. 12 (559) (2020).
Cold Spring Harb. Perspect. Med. 8 (4) (2018). [121] M.R. White, D.M. Mitrea, P. Zhang, C.B. Stanley, D.E. Cassidy, A. Nourse, A.
[102] P.E. Ash, K.F. Bieniek, T.F. Gendron, T. Caulfield, W.L. Lin, M. Dejesus- H. Phillips, M. Tolbert, J.P. Taylor, R.W. Kriwacki, C9orf72 poly(PR) dipeptide
Hernandez, M.M. van Blitterswijk, K. Jansen-West, J.W. Paul 3rd, R. Rademakers, repeats disturb biomolecular phase separation and disrupt nucleolar function,
K.B. Boylan, D.W. Dickson, L. Petrucelli, Unconventional translation of C9ORF72 Mol. Cell 74 (4) (2019) 713–728 e6.
GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS, [122] Y.J. Zhang, L. Guo, P.K. Gonzales, T.F. Gendron, Y. Wu, K. Jansen-West, A.
Neuron 77 (4) (2013) 639–646. D. O’Raw, S.R. Pickles, M. Prudencio, Y. Carlomagno, M.A. Gachechiladze,
[103] K. Mori, S.M. Weng, T. Arzberger, S. May, K. Rentzsch, E. Kremmer, B. Schmid, H. C. Ludwig, R. Tian, J. Chew, M. DeTure, W.L. Lin, J. Tong, L.M. Daughrity,
A. Kretzschmar, M. Cruts, C. Van Broeckhoven, C. Haass, D. Edbauer, The C9orf72 M. Yue, Y. Song, J.W. Andersen, M. Castanedes-Casey, A. Kurti, A. Datta,
GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ G. Antognetti, A. McCampbell, R. Rademakers, B. Oskarsson, D.W. Dickson,
ALS, Science 339 (6125) (2013) 1335–1338. M. Kampmann, M.E. Ward, J.D. Fryer, C.D. Link, J. Shorter, L. Petrucelli,
[104] T.F. Gendron, K.F. Bieniek, Y.J. Zhang, K. Jansen-West, P.E. Ash, T. Caulfield, Heterochromatin anomalies and double-stranded RNA accumulation underlie
L. Daughrity, J.H. Dunmore, M. Castanedes-Casey, J. Chew, D.M. Cosio, M. van C9orf72 poly(PR) toxicity, Science 363 (6428) (2019).
Blitterswijk, W.C. Lee, R. Rademakers, K.B. Boylan, D.W. Dickson, L. Petrucelli,

14
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

[123] Y.J. Zhang, T.F. Gendron, M.T.W. Ebbert, A.D. O’Raw, M. Yue, K. Jansen-West, [149] A. Post, A. Bollenbach, S.J.L. Bakker, D. Tsikas, Whole-body arginine
X. Zhang, M. Prudencio, J. Chew, C.N. Cook, L.M. Daughrity, J. Tong, Y. Song, S. dimethylation is associated with all-cause mortality in adult renal transplant
R. Pickles, M. Castanedes-Casey, A. Kurti, R. Rademakers, B. Oskarsson, D. recipients, Amino Acids 53 (4) (2021) 541–554.
W. Dickson, W. Hu, A.D. Gitler, J.D. Fryer, L. Petrucelli, Poly(GR) impairs protein [150] M.Y. Said, A. Bollenbach, I. Minovic, M. van Londen, A.R. Frenay, M.H. de Borst,
translation and stress granule dynamics in C9orf72-associated frontotemporal E. van den Berg, A.A. Kayacelebi, D. Tsikas, H. van Goor, G. Navis, S.J.L. Bakker,
dementia and amyotrophic lateral sclerosis, Nat. Med. 24 (8) (2018) 1136–1142. Plasma ADMA, urinary ADMA excretion, and late mortality in renal transplant
[124] S. Boeynaems, E. Bogaert, D. Kovacs, A. Konijnenberg, E. Timmerman, A. Volkov, recipients, Amino Acids 51 (6) (2019) 913–927.
M. Guharoy, M. De Decker, T. Jaspers, V.H. Ryan, A.M. Janke, P. Baatsen, [151] M.Y. Said, R.M. Douwes, M. van Londen, I. Minovic, A.R. Frenay, M.H. de Borst,
T. Vercruysse, R.M. Kolaitis, D. Daelemans, J.P. Taylor, N. Kedersha, P. Anderson, E. van den Berg, M.R. Heiner-Fokkema, A.A. Kayacelebi, A. Bollenbach, H. van
F. Impens, F. Sobott, J. Schymkowitz, F. Rousseau, N.L. Fawzi, W. Robberecht, Goor, G. Navis, D. Tsikas, S.J.L. Bakker, Effect of renal function on homeostasis of
P. Van Damme, P. Tompa, L. Van Den Bosch, Phase separation of C9orf72 asymmetric dimethylarginine (ADMA): studies in donors and recipients of renal
dipeptide repeats perturbs stress granule dynamics, Mol. Cell 65 (6) (2017) transplants, Amino Acids 51 (3) (2019) 565–575.
1044–1055 e5. [152] A.R. Frenay, E. van den Berg, M.H. de Borst, B. Beckmann, D. Tsikas, M. Feelisch,
[125] P.M. Harrison, Intrinsic disorder and posttranslational modification: an G. Navis, S.J. Bakker, H. van Goor, Plasma ADMA associates with all-cause
evolutionary perspective, in: Structure and Intrinsic Disorder in Enzymology, mortality in renal transplant recipients, Amino Acids 47 (9) (2015) 1941–1949.
Elsevier, 2023, pp. 377–396. [153] D. Fliser, F. Kronenberg, J.T. Kielstein, C. Morath, S.M. Bode-Boger, H. Haller,
[126] V.N. Uversky, Posttranslational modifications, in: S. Maloy, K. Hughes (Eds.), E. Ritz, Asymmetric dimethylarginine and progression of chronic kidney disease:
Brenner’s Encyclopedia of Genetics, Academic Press, London, UK; Walham, MA, the mild to moderate kidney disease study, J. Am. Soc. Nephrol. 16 (8) (2005)
USA; San Diego, CA, USA, 2013, pp. 425–430. 2456–2461.
[127] A.L. Darling, V.N. Uversky, Intrinsic disorder and posttranslational modifications: [154] V. Achan, M. Broadhead, M. Malaki, G. Whitley, J. Leiper, R. MacAllister,
the darker side of the biological dark matter, Front. Genet. 9 (2018) 158. P. Vallance, Asymmetric dimethylarginine causes hypertension and cardiac
[128] M.N. Gupta, V.N. Uversky, Moonlighting enzymes: when cellular context defines dysfunction in humans and is actively metabolized by dimethylarginine
specificity, Cell. Mol. Life Sci. 80 (5) (2023) 130. dimethylaminohydrolase, Arterioscler. Thromb. Vasc. Biol. 23 (8) (2003)
[129] V. Pejaver, W.L. Hsu, F. Xin, A.K. Dunker, V.N. Uversky, P. Radivojac, The 1455–1459.
structural and functional signatures of proteins that undergo multiple events of [155] Y.L. Tain, C.N. Hsu, Toxic Dimethylarginines: asymmetric Dimethylarginine
post-translational modification, Protein Sci. 23 (8) (2014) 1077–1093. (ADMA) and symmetric Dimethylarginine (SDMA), Toxins (Basel) 9 (3) (2017).
[130] V.N. Uversky, Intrinsic disorder-based protein interactions and their modulators, [156] M. Busch, C. Fleck, G. Wolf, G. Stein, Asymmetrical (ADMA) and symmetrical
Curr. Pharm. Des. 19 (23) (2013) 4191–4213. dimethylarginine (SDMA) as potential risk factors for cardiovascular and renal
[131] L.M. Iakoucheva, P. Radivojac, C.J. Brown, T.R. O’Connor, J.G. Sikes, outcome in chronic kidney disease - possible candidates for paradoxical
Z. Obradovic, A.K. Dunker, The importance of intrinsic disorder for protein epidemiology? Amino Acids 30 (3) (2006) 225–232.
phosphorylation, Nucleic Acids Res. 32 (3) (2004) 1037–1049. [157] A.J. Marsden, D.R.J. Riley, S. Birkett, Q. Rodriguez-Barucg, B.A. Guinn, S. Carroll,
[132] D. Tsikas, Post-translational modifications (PTM): analytical approaches, L. Ingle, T. Sathyapalan, P. Beltran-Alvarez, Love is in the hair: arginine
signaling, physiology and pathophysiology-part I, Amino Acids 53 (4) (2021) methylation of human hair proteins as novel cardiovascular biomarkers, Amino
485–487. Acids 54 (4) (2022) 591–600.
[133] P.V. Hornbeck, J.M. Kornhauser, S. Tkachev, B. Zhang, E. Skrzypek, B. Murray, [158] M.N. Gupta, M. Perwez, M. Sardar, Protein crosslinking: uses in chemistry,
V. Latham, M. Sullivan, PhosphoSitePlus: a comprehensive resource for biology and biotechnology, Biocatal. Biotransformation 38 (3) (2020) 178–201.
investigating the structure and function of experimentally determined post- [159] J. Mukherjee, A.B. Majumder, M.N. Gupta, Adding an appropriate amino acid
translational modifications in man and mouse, Nucleic Acids Res. 40 (Database during crosslinking results in more stable crosslinked enzyme aggregates, Anal.
issue) (2012) D261–D270. Biochem. 507 (2016) 27–32.
[134] Z.A. Wang, P.A. Cole, The chemical biology of reversible lysine Post-translational [160] T.A. Collier, A. Nash, H.L. Birch, N.H. de Leeuw, Intra-molecular lysine-arginine
modifications, Cell Chem. Biol. 27 (8) (2020) 953–969. derived advanced glycation end-product cross-linking in type I collagen: a
[135] I. Ali, R.J. Conrad, E. Verdin, M. Ott, Lysine acetylation Goes global: from molecular dynamics simulation study, Biophys. Chem. 218 (2016) 42–46.
epigenetics to metabolism and therapeutics, Chem. Rev. 118 (3) (2018) [161] C.T. Thorpe, I. Streeter, G.L. Pinchbeck, A.E. Goodship, P.D. Clegg, H.L. Birch,
1216–1252. Aspartic acid racemization and collagen degradation markers reveal an
[136] C. Marondedze, G. Elia, L. Thomas, A. Wong, C. Gehring, Citrullination of accumulation of damage in tendon collagen that is enhanced with aging, J. Biol.
proteins as a specific response mechanism in plants, Front. Plant Sci. 12 (2021), Chem. 285 (21) (2010) 15674–15681.
638392. [162] K.M. Biemel, D.A. Friedl, M.O. Lederer, Identification and quantification of major
[137] S. Mondal, S. Wang, Y. Zheng, S. Sen, A. Chatterjee, P.R. Thompson, Site-specific maillard cross-links in human serum albumin and lens protein. Evidence for
incorporation of citrulline into proteins in mammalian cells, Nat. Commun. 12 (1) glucosepane as the dominant compound, J. Biol. Chem. 277 (28) (2002)
(2021) 45. 24907–24915.
[138] M. Beato, P. Sharma, Peptidyl arginine deiminase 2 (PADI2)-mediated arginine [163] E. Lorenzo-Martin, P. Gallego-Munoz, L. Ibares-Frias, S. Marcos, P. Perez-Merino,
Citrullination modulates transcription in Cancer, Int. J. Mol. Sci. 21 (4) (2020). I. Fernandez, I.E. Kochevar, M.C. Martinez-Garcia, Rose Bengal and green light
[139] M. Alghamdi, D. Alasmari, A. Assiri, E. Mattar, A.A. Aljaddawi, S.G. Alattas, E. versus riboflavin-UVA cross-linking: corneal wound repair response, Invest.
M. Redwan, An overview of the intrinsic role of Citrullination in autoimmune Ophthalmol. Vis. Sci. 59 (12) (2018) 4821–4830.
disorders, J Immunol Res 2019 (2019), 7592851. [164] T.G. Seiler, M. Engler, E. Beck, R. Birngruber, I.E. Kochevar, Interface bonding
[140] A. Catrina, A. Krishnamurthy, B. Rethi, Current view on the pathogenic role of with corneal crosslinking (CXL) after LASIK ex vivo, Invest. Ophthalmol. Vis. Sci.
anti-citrullinated protein antibodies in rheumatoid arthritis, RMD Open 7 (1) 58 (14) (2017) 6292–6298.
(2021). [165] P. Gallego-Munoz, L. Ibares-Frias, E. Lorenzo, S. Marcos, P. Perez-Merino,
[141] N.M. Bosch, M. Borsa, U. Greczmiel, B.I. Morinaka, M. Gugger, A. Oxenius, A. N. Bekesi, I.E. Kochevar, M.C. Martinez-Garcia, Corneal wound repair after rose
L. Vagstad, J. Piel, Landornamides: antiviral ornithine-containing ribosomal bengal and green light crosslinking: clinical and histologic study, Invest.
peptides discovered through genome mining, Angew. Chem. Int. Ed. Eng. 59 (29) Ophthalmol. Vis. Sci. 58 (9) (2017) 3471–3480.
(2020) 11763–11768. [166] H. Zhu, C. Alt, R.H. Webb, S. Melki, I.E. Kochevar, Corneal crosslinking with rose
[142] S. Mordhorst, B.I. Morinaka, A.L. Vagstad, J. Piel, Posttranslationally acting bengal and green light: efficacy and safety evaluation, Cornea 35 (9) (2016)
arginases provide a ribosomal route to non-proteinogenic ornithine residues in 1234–1241.
diverse peptide sequences, Angew. Chem. Int. Ed. Eng. 59 (48) (2020) [167] N. Bekesi, I.E. Kochevar, S. Marcos, Corneal biomechanical response following
21442–21447. collagen cross-linking with rose bengal-green light and riboflavin-UVA, Invest.
[143] S. Mordhorst, T. Badmann, N.M. Bosch, B.I. Morinaka, H. Rauch, J. Piel, M. Groll, Ophthalmol. Vis. Sci. 57 (3) (2016) 992–1001.
A.L. Vagstad, Structural and biochemical insights into post-translational arginine- [168] C.M. Wertheimer, B. Mendes, Q. Pei, K. Brandt, I.E. Kochevar, Arginine as an
to-ornithine peptide modifications by an atypical arginase, ACS Chem. Biol. 18 enhancer in Rose Bengal photosensitized corneal crosslinking, Transl. Vis. Sci.
(3) (2023) 528–536. Technol. 9 (8) (2020) 24.
[144] S.F. Samuel, A. Barry, J. Greenman, P. Beltran-Alvarez, Arginine methylation: the [169] C.M. Wertheimer, C. Elhardt, S.M. Kaminsky, L. Pham, Q. Pei, B. Mendes,
promise of a ’silver bullet’ for brain tumours? Amino Acids 53 (4) (2021) S. Afshar, I.E. Kochevar, Enhancing Rose Bengal-photosensitized protein
489–506. crosslinking in the cornea, Invest. Ophthalmol. Vis. Sci. 60 (6) (2019) 1845–1852.
[145] J.E. Katz, M. Dlakic, S. Clarke, Automated identification of putative [170] D. Cherfan, E.E. Verter, S. Melki, T.E. Gisel, F.J. Doyle Jr., G. Scarcelli, S.H. Yun,
methyltransferases from genomic open reading frames, Mol. Cell. Proteomics 2 R.W. Redmond, I.E. Kochevar, Collagen cross-linking using rose bengal and green
(8) (2003) 525–540. light to increase corneal stiffness, Invest. Ophthalmol. Vis. Sci. 54 (5) (2013)
[146] A. Guo, H. Gu, J. Zhou, D. Mulhern, Y. Wang, K.A. Lee, V. Yang, M. Aguiar, 3426–3433.
J. Kornhauser, X. Jia, J. Ren, S.A. Beausoleil, J.C. Silva, V. Vemulapalli, M. [171] S. Udhayakumar, K.G. Shankar, S. Sowndarya, S. Venkatesh, C. Muralidharan,
T. Bedford, M.J. Comb, Immunoaffinity enrichment and mass spectrometry C. Rose, L-arginine intercedes bio-crosslinking of a collagen–chitosan 3D-hybrid
analysis of protein methylation, Mol. Cell. Proteomics 13 (1) (2014) 372–387. scaffold for tissue engineering and regeneration: in silico, in vitro, and in vivo
[147] P.V. Hornbeck, B. Zhang, B. Murray, J.M. Kornhauser, V. Latham, E. Skrzypek, studies, RSC Adv. 7 (40) (2017) 25070–25088.
PhosphoSitePlus, 2014: mutations, PTMs and recalibrations, Nucleic Acids Res. [172] A. Caruso, R.J. Martinie, L.B. Bushin, M.R. Seyedsayamdost, Macrocyclization via
43 (Database issue) (2015) D512–D520. an arginine-tyrosine crosslink broadens the reaction scope of radical S-
[148] Y. Yang, M.T. Bedford, Protein arginine methyltransferases and cancer, Nat. Rev. adenosylmethionine enzymes, J. Am. Chem. Soc. 141 (42) (2019) 16610–16614.
Cancer 13 (1) (2013) 37–50. [173] J.F. Riordan, Arginyl residues and anion binding sites in proteins, Mol. Cell.
Biochem. 26 (2) (1979) 71–92.

15
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

[174] D.L. Leone, M. Hubalek, R. Pohl, V. Sykorova, M. Hocek, 1,3-Diketone-modified [204] L. Xiong, J.L. Teng, M.G. Botelho, R.C. Lo, S.K. Lau, P.C. Woo, Arginine
nucleotides and DNA for cross-linking with arginine-containing peptides and metabolism in bacterial pathogenesis and cancer therapy, Int. J. Mol. Sci. 17 (3)
proteins, Angew. Chem. Int. Ed. Eng. 60 (32) (2021) 17383–17387. (2016) 363.
[175] G. Means, R. Feeney, Chemical Modification of Proteins, Hoiden-Day Inc., San [205] C. Erens, J. Van Broeckhoven, A. Bronckaers, S. Lemmens, S. Hendrix, The dark
Francisco, CA, 1971. side of an essential amino acid: L-arginine in spinal cord injury, J. Neurotrauma
[176] R. Tyagi, M.N. Gupta, Chemical modification and chemical crosslinking for 40 (9–10) (2023) 820–832.
enhancing thermostability of enzymes, in: M.N. Gupta (Ed.), Thermostability of [206] C. Ma, K. Liao, J. Wang, T. Li, L. Liu, L-arginine, as an essential amino acid, is a
Enzymes, Springer-Verlag, Berlin, 1993, pp. 146–160. potential substitute for treating COPD via regulation of ROS/NLRP3/NF-kappaB
[177] R. Tyagi, M.N. Gupta, Chemical modification and chemical crosslinking of signaling pathway, Cell Biosci. 13 (1) (2023) 152.
proteins/enzyme stabilization, Biochem. Mosc. 63 (1998) 395–407. [207] J. Leblond, A. Petitjean, Molecular tweezers: concepts and applications,
[178] Y. Zhou, G. Liu, H. Huang, J. Wu, Advances and impact of arginine-based Chemphyschem 12 (6) (2011) 1043–1051.
materials in wound healing, J. Mater. Chem. B 9 (34) (2021) 6738–6750. [208] F. Kaiser, S. Bittrich, S. Salentin, C. Leberecht, V.J. Haupt, S. Krautwurst,
[179] Y. Matsumoto, Y. Kuroyanagi, Development of a wound dressing composed of M. Schroeder, D. Labudde, Backbone brackets and arginine tweezers delineate
hyaluronic acid sponge containing arginine and epidermal growth factor, class I and class II aminoacyl tRNA synthetases, PLoS Comput. Biol. 14 (4) (2018),
J. Biomater. Sci. Polym. Ed. 21 (6–7) (2010) 715–726. e1006101.
[180] F. Reesi, M. Minaiyan, A. Taheri, A novel lignin-based nanofibrous dressing [209] T. Schrader, G. Bitan, F.G. Klarner, Molecular tweezers for lysine and arginine -
containing arginine for wound-healing applications, Drug Deliv. Transl. Res. 8 (1) powerful inhibitors of pathologic protein aggregation, Chem. Commun. (Camb.)
(2018) 111–122. 52 (76) (2016) 11318–11334.
[181] A.S. Durmus, M. Tuzcu, O. Ozdemir, C. Orhan, N. Sahin, I.H. Ozercan, J. [210] A. Mbarek, G. Moussa, J.L. Chain, Pharmaceutical applications of molecular
R. Komorowski, S. Ali, K. Sahin, Arginine silicate inositol complex accelerates tweezers, clefts and clips, Molecules 24 (9) (2019).
cutaneous wound healing, Biol. Trace Elem. Res. 177 (1) (2017) 122–131. [211] I. Hadrovic, P. Rebmann, F.G. Klarner, G. Bitan, T. Schrader, Molecular lysine
[182] R.I. Baron, M.E. Culica, G. Biliuta, M. Bercea, S. Gherman, D. Zavastin, L. Ochiuz, tweezers counteract aberrant protein aggregation, Front. Chem. 7 (2019) 657.
M. Avadanei, S. Coseri, Physical hydrogels of oxidized polysaccharides and poly [212] A.J. Hodges, I.J. Gallegos, M.F. Laughery, R. Meas, L. Tran, J.J. Wyrick, Histone
(vinyl alcohol) for wound dressing applications, Materials (Basel) 12 (9) (2019). sprocket arginine residues are important for gene expression, DNA repair, and cell
[183] B.P. Antunes, A.F. Moreira, V.M. Gaspar, I.J. Correia, Chitosan/arginine-chitosan viability in Saccharomyces cerevisiae, Genetics 200 (3) (2015) 795–806.
polymer blends for assembly of nanofibrous membranes for wound regeneration, [213] U.M. Muthurajan, Y.J. Park, R.S. Edayathumangalam, R.K. Suto, S. Chakravarthy,
Carbohydr. Polym. 130 (2015) 104–112. P.N. Dyer, K. Luger, Structure and dynamics of nucleosomal DNA, Biopolymers 68
[184] J. Wu, M.A. Mutschler, C.C. Chu, Synthesis and characterization of ionic charged (4) (2003) 547–556.
water soluble arginine-based poly(ester amide), J. Mater. Sci. Mater. Med. 22 (3) [214] C.A. Davey, D.F. Sargent, K. Luger, A.W. Maeder, T.J. Richmond, Solvent
(2011) 469–479. mediated interactions in the structure of the nucleosome core particle at 1.9 a
[185] F.A. Cotton, T. la Cour, E.E. Hazen Jr., M.J. Legg, The role of arginine residues at resolution, J. Mol. Biol. 319 (5) (2002) 1097–1113.
enzyme active sites. The interaction between guanidinium ions and p-nitro- [215] K. Luger, A.W. Mader, R.K. Richmond, D.F. Sargent, T.J. Richmond, Crystal
phenyl phosphate and its effect on the rate of hydrolysis of the ester, Biochim structure of the nucleosome core particle at 2.8 A resolution, Nature 389 (6648)
Biophys Acta 481 (1) (1977) 1–5. (1997) 251–260.
[186] S. Adak, D. Bandyopadhyay, U. Bandyopadhyay, R.K. Banerjee, An essential role [216] A. Magalhaes, B. Maigret, J. Hoflack, J.N. Gomes, H.A. Scheraga, Contribution of
of active site arginine residue in iodide binding and histidine residue in electron unusual arginine-arginine short-range interactions to stabilization and
transfer for iodide oxidation by horseradish peroxidase, Mol. Cell. Biochem. 218 recognition in proteins, J. Protein Chem. 13 (2) (1994) 195–215.
(1–2) (2001) 1–11. [217] M.A. Neves, M. Yeager, R. Abagyan, Unusual arginine formations in protein
[187] K.C. Cheng, T. Nowak, Arginine residues at the active site of avian liver function and assembly: rings, strings, and stacks, J. Phys. Chem. B 116 (23)
phosphoenolpyruvate carboxykinase, J. Biol. Chem. 264 (6) (1989) 3317–3324. (2012) 7006–7013.
[188] V. Walker, Ammonia metabolism and hyperammonemic disorders, Adv. Clin. [218] W. Wang, Oral protein drug delivery, J. Drug Target. 4 (4) (1996) 195–232.
Chem. 67 (2014) 73–150. [219] W. Wang, Lyophilization and development of solid protein pharmaceuticals, Int.
[189] S. Kumar, J. Lennane, S. Ratner, Argininosuccinate synthetase: essential role of J. Pharm. 203 (1–2) (2000) 1–60.
cysteine and arginine residues in relation to structure and mechanism of ATP [220] W. Wang, Instability, stabilization, and formulation of liquid protein
activation, Proc. Natl. Acad. Sci. U. S. A. 82 (20) (1985) 6745–6749. pharmaceuticals, Int. J. Pharm. 185 (2) (1999) 129–188.
[190] R.A. Keenholtz, K.W. Mouw, M.R. Boocock, N.S. Li, J.A. Piccirilli, P.A. Rice, [221] C. Garcia Garcia, S.S. Patkar, B. Wang, R. Abouomar, K.L. Kiick, Recombinant
Arginine as a general acid catalyst in serine recombinase-mediated DNA cleavage, protein-based injectable materials for biomedical applications, Adv. Drug Deliv.
J. Biol. Chem. 288 (40) (2013) 29206–29214. Rev. 193 (2023), 114673.
[191] J.M. Pauff, C.F. Hemann, N. Junemann, S. Leimkuhler, R. Hille, The role of [222] S. Ryan, K. Shortall, M. Dully, A. Djehedar, D. Murray, J. Butler, J. Neilan,
arginine 310 in catalysis and substrate specificity in xanthine dehydrogenase from T. Soulimane, S.P. Hudson, Long acting injectables for therapeutic proteins,
Rhodobacter capsulatus, J. Biol. Chem. 282 (17) (2007) 12785–12790. Colloids Surf. B: Biointerfaces 217 (2022), 112644.
[192] S. Albert, E. Will, D. Gallwitz, Identification of the catalytic domains and their [223] N. Ibeanu, R. Egbu, L. Onyekuru, H. Javaheri, P.T. Khaw, G.R. Williams,
functionally critical arginine residues of two yeast GTPase-activating proteins S. Brocchini, S. Awwad, Injectables and depots to prolong drug action of proteins
specific for Ypt/Rab transport GTPases, EMBO J. 18 (19) (1999) 5216–5225. and peptides, Pharmaceutics 12 (10) (2020).
[193] G. Chen, X. Chen, Arginine residues in the active site of human phenol [224] C.P. Brangwynne, C.R. Eckmann, D.S. Courson, A. Rybarska, C. Hoege,
sulfotransferase (SULT1A1), J. Biol. Chem. 278 (38) (2003) 36358–36364. J. Gharakhani, F. Julicher, A.A. Hyman, Germline P granules are liquid droplets
[194] N. Nagahara, Sulfurtransferases: essential enzymes for life, in: M.N. Gupta (Ed.), that localize by controlled dissolution/condensation, Science 324 (5935) (2009)
Foundations and Frontiers of Enzymology Elsevier, San Diego, CA, 2023. 1729–1732.
[195] D. Communi, R. Lecocq, C. Erneux, Arginine 343 and 350 are two active residues [225] C.P. Brangwynne, Phase transitions and size scaling of membrane-less organelles,
involved in substrate binding by human type I D-myo-inositol 1,4,5,-trisphos­ J. Cell Biol. 203 (6) (2013) 875–881.
phate 5-phosphatase, J. Biol. Chem. 271 (20) (1996) 11676–11683. [226] C.P. Brangwynne, P. Tompa, R.V. Pappu, Polymer physics of intracellular phase
[196] J. Shi, R. Mukhopadhyay, B.P. Rosen, Identification of a triad of arginine residues transitions, Nat. Phys. 11 (2015) 899–904.
in the active site of the ArsC arsenate reductase of plasmid R773, FEMS Microbiol. [227] V.N. Uversky, I.M. Kuznetsova, K.K. Turoverov, B. Zaslavsky, Intrinsically
Lett. 227 (2) (2003) 295–301. disordered proteins as crucial constituents of cellular aqueous two phase systems
[197] J.C. Hsiao, A.P. McGrath, L. Kielmann, P. Kalimuthu, F. Darain, P.V. Bernhardt, and coacervates, FEBS Lett. 589 (1) (2015) 15–22.
J. Harmer, M. Lee, K. Meyers, M.J. Maher, U. Kappler, The central active site [228] M. Dundr, T. Misteli, Biogenesis of nuclear bodies, Cold Spring Harb. Perspect.
arginine in sulfite oxidizing enzymes alters kinetic properties by controlling Biol. 2 (12) (2010), a000711.
electron transfer and redox interactions, Biochim. Biophys. Acta Bioenerg. 1859 [229] L. Zhu, C.P. Brangwynne, Nuclear bodies: the emerging biophysics of
(1) (2018) 19–27. nucleoplasmic phases, Curr. Opin. Cell Biol. 34 (2015) 23–30.
[198] G. Murtas, S. Sacchi, L. Pollegioni, Substitution of arginine 120 in human D-amino [230] V.N. Uversky, Intrinsically disordered proteins in overcrowded milieu:
acid oxidase favors FAD-binding and nuclear Mistargeting, Front. Mol. Biosci. 6 membrane-less organelles, phase separation, and intrinsic disorder, Curr. Opin.
(2019) 125. Struct. Biol. 44 (2017) 18–30.
[199] A. Sarkar, S. Bhakta, S. Chattopadhyay, A. Dey, Role of distal arginine residue in [231] V.N. Uversky, Protein intrinsic disorder-based liquid-liquid phase transitions in
the mechanism of heme nitrite reductases, Chem. Sci. 14 (29) (2023) 7875–7886. biological systems: complex coacervates and membrane-less organelles, Adv.
[200] S.M. Morris Jr., Arginine metabolism: boundaries of our knowledge, J. Nutr. 137 Colloid Interf. Sci. 239 (2017) 97–114.
(6 Suppl 2) (2007) 1602S–1609S. [232] M. Feric, N. Vaidya, T.S. Harmon, D.M. Mitrea, L. Zhu, T.M. Richardson, R.
[201] B.C. Tong, A. Barbul, Cellular and physiological effects of arginine, Mini-Rev. W. Kriwacki, R.V. Pappu, C.P. Brangwynne, Coexisting liquid phases underlie
Med. Chem. 4 (8) (2004) 823–832. nucleolar subcompartments, Cell 165 (7) (2016) 1686–1697.
[202] C.R. Morris, J. Hamilton-Reeves, R.G. Martindale, M. Sarav, J.B. Ochoa Gautier, [233] D.M. Mitrea, R.W. Kriwacki, Phase separation in biology; functional organization
Acquired amino acid deficiencies: a focus on arginine and glutamine, Nutr. Clin. of a higher order, Cell Commun. Signal 14 (2016) 1.
Pract. 32 (1_suppl) (2017) 30S–47S. [234] A. Molliex, J. Temirov, J. Lee, M. Coughlin, A.P. Kanagaraj, H.J. Kim, T. Mittag, J.
[203] K. Hristina, T. Langerholc, M. Trapecar, Novel metabolic roles of L-arginine in P. Taylor, Phase separation by low complexity domains promotes stress granule
body energy metabolism and possible clinical applications, J. Nutr. Health Aging assembly and drives pathological fibrillization, Cell 163 (1) (2015) 123–133.
18 (2) (2014) 213–218. [235] H. Jiang, S. Wang, Y. Huang, X. He, H. Cui, X. Zhu, Y. Zheng, Phase transition of
spindle-associated protein regulate spindle apparatus assembly, Cell 163 (1)
(2015) 108–122.

16
M.N. Gupta and V.N. Uversky International Journal of Biological Macromolecules 257 (2024) 128646

[236] I.A. Antifeeva, A.V. Fonin, A.S. Fefilova, O.V. Stepanenko, O.I. Povarova, S. [259] F. Wang, Y. Wang, X. Zhang, W. Zhang, S. Guo, F. Jin, Recent progress of cell-
A. Silonov, I.M. Kuznetsova, V.N. Uversky, K.K. Turoverov, Liquid-liquid phase penetrating peptides as new carriers for intracellular cargo delivery, J. Control.
separation as an organizing principle of intracellular space: overview of the Release 174 (2014) 126–136.
evolution of the cell compartmentalization concept, Cell. Mol. Life Sci. 79 (5) [260] M. Peitz, K. Pfannkuche, K. Rajewsky, F. Edenhofer, Ability of the hydrophobic
(2022) 251. FGF and basic TAT peptides to promote cellular uptake of recombinant Cre
[237] A.L. Darling, Y. Liu, C.J. Oldfield, V.N. Uversky, Intrinsically disordered proteome recombinase: a tool for efficient genetic engineering of mammalian genomes,
of human membrane-less organelles, Proteomics 18 (5–6) (2018), e1700193. Proc. Natl. Acad. Sci. U. S. A. 99 (7) (2002) 4489–4494.
[238] H.X. Zhou, V. Nguemaha, K. Mazarakos, S. Qin, Why do disordered and structured [261] J. Allen, J.P. Pellois, Hydrophobicity is a key determinant in the activity of
proteins behave differently in phase separation? Trends Biochem. Sci. 43 (7) arginine-rich cell penetrating peptides, Sci. Rep. 12 (1) (2022), 15981.
(2018) 499–516. [262] T. Arakawa, K. Tsumoto, K. Nagase, D. Ejima, The effects of arginine on protein
[239] K.K. Turoverov, I.M. Kuznetsova, A.V. Fonin, A.L. Darling, B.Y. Zaslavsky, V. binding and elution in hydrophobic interaction and ion-exchange
N. Uversky, Stochasticity of biological soft matter: emerging concepts in chromatography, Protein Expr. Purif. 54 (1) (2007) 110–116.
intrinsically disordered proteins and biological phase separation, Trends [263] T. Hall, G.M. Kelly, W.R. Emery, Use of mobile phase additives for the elution of
Biochem. Sci. 44 (8) (2019) 716–728. bispecific and monoclonal antibodies from phenyl based hydrophobic interaction
[240] A.V. Fonin, I.A. Antifeeva, I.M. Kuznetsova, K.K. Turoverov, B.Y. Zaslavsky, chromatography resins, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 1096
P. Kulkarni, V.N. Uversky, Biological soft matter: intrinsically disordered proteins (2018) 20–30.
in liquid-liquid phase separation and biomolecular condensates, Essays Biochem. [264] J.F.A. Valente, A. Sousa, G.A. Azevedo, J.A. Queiroz, F. Sousa, Purification of
66 (7) (2022) 831–847. supercoiled p53-encoding plasmid using an arginine-modified macroporous
[241] A.S. Raut, D.S. Kalonia, Opalescence in monoclonal antibody solutions and its support, J. Chromatogr. A 1618 (2020), 460890.
correlation with intermolecular interactions in dilute and concentrated solutions, [265] A. Soares, J.A. Queiroz, F. Sousa, A. Sousa, Purification of human papillomavirus
J. Pharm. Sci. 104 (4) (2015) 1263–1274. 16 E6/E7 plasmid deoxyribonucleic acid-based vaccine using an arginine
[242] S. Oki, S. Nishinami, K. Shiraki, Arginine suppresses opalescence and liquid-liquid modified monolithic support, J. Chromatogr. A 1320 (2013) 72–79.
phase separation in IgG solutions, Int. J. Biol. Macromol. 118 (Pt B) (2018) [266] J.F.A. Valente, T.S. Carreira, J.R. Dias, F. Sousa, N. Alves, Arginine-modified 3D-
1708–1712. printed chromatographic supports, Pharmaceutics 14 (11) (2022).
[243] D.D. Banks, J.F. Cordia, Suppression of electrostatic mediated antibody liquid- [267] M.N. Gupta, Methods for Affinity-Based Separations of Enzymes and Proteins,
liquid phase separation by charged and noncharged preferentially excluded Springer Science & Business Media, 2002.
excipients, Mol. Pharm. 18 (3) (2021) 1285–1292. [268] I. Roy, M.N. Gupta, Current trends in affinity-based separations of proteins/
[244] P. Pyne, R.K. Mitra, Excipients do regulate phase separation in lysozyme and thus enzymes, Curr. Sci. (2000) 587–591.
also its hydration, J. Phys. Chem. Lett. 13 (3) (2022) 931–938. [269] K. Mondal, M.N. Gupta, The affinity concept in bioseparation: evolving paradigms
[245] K. Tsumoto, M. Umetsu, I. Kumagai, D. Ejima, J.S. Philo, T. Arakawa, Role of and expanding range of applications, Biomol. Eng. 23 (2–3) (2006) 59–76.
arginine in protein refolding, solubilization, and purification, Biotechnol. Prog. [270] F. Bartnicki, P. Bonarek, E. Kowalska, W. Strzalka, The Argi system: one-step
20 (5) (2004) 1301–1308. purification of proteins tagged with arginine-rich cell-penetrating peptides, Sci.
[246] T. Arakawa, K. Tsumoto, The effects of arginine on refolding of aggregated Rep. 7 (1) (2017) 2619.
proteins: not facilitate refolding, but suppress aggregation, Biochem. Biophys. [271] M.B. Burg, J.D. Ferraris, Intracellular organic osmolytes: function and regulation,
Res. Commun. 304 (1) (2003) 148–152. J. Biol. Chem. 283 (12) (2008) 7309–7313.
[247] J. Buchner, R. Rudolph, Renaturation, purification and characterization of [272] M. Xu, P. Liu, J. Chen, A. Peng, T. Yang, X. Zhang, Z. Xu, Z. Rao, Development of a
recombinant fab-fragments produced in Escherichia coli, Bio/technology 9 (2) novel biosensor-driven mutation and selection system via in situ growth of
(1991) 157–162. Corynebacterium crenatum for the production of L-arginine, Front. Bioeng.
[248] K. Shiraki, M. Kudou, S. Fujiwara, T. Imanaka, M. Takagi, Biophysical effect of Biotechnol. 8 (2020) 175.
amino acids on the prevention of protein aggregation, J. Biochem. 132 (4) (2002) [273] N. Stasyuk, G. Gayda, O. Demkiv, L. Darmohray, M. Gonchar, M. Nisnevitch,
591–595. Amperometric biosensors for L-arginine determination based on L-arginine
[249] N. Inoue, E. Takai, T. Arakawa, K. Shiraki, Specific decrease in solution viscosity oxidase and peroxidase-like nanozymes, Appl. Sci. 11 (15) (2021) 7024.
of antibodies by arginine for therapeutic formulations, Mol. Pharm. 11 (6) (2014) [274] K. Berketa, O. Saiapina, L. Fayura, A. Sibirny, S. Dzyadevych, O. Soldatkin, Novel
1889–1896. highly sensitive conductometric biosensor based on arginine deiminase from
[250] B.M. Baynes, D.I. Wang, B.L. Trout, Role of arginine in the stabilization of mycoplasma hominis for determination of arginine, Sensors Actuators B Chem.
proteins against aggregation, Biochemistry 44 (12) (2005) 4919–4925. 367 (2022), 132023.
[251] M.N. Gupta, V.N. Uversky, Macromolecular crowding: how it affects protein [275] M. Rahman, J. Niu, X. Cui, C. Zhou, N. Tang, H. Jin, D. Cui, Electrochemical
structure, disorder, and catalysis, in: Structure and Intrinsic Disorder in biosensor based on l-arginine and rGO-AuNSs deposited on the electrode
Enzymology, Elsevier, 2023, pp. 353–376. combined with DNA probes for ultrasensitive detection of the gastric cancer-
[252] Y.D. Liu, J.J. Li, F.W. Wang, J. Chen, P. Li, Z.G. Su, A newly proposed mechanism related PIK3CA gene of ctDNA, ACS Appl. Bio Mater. 5 (11) (2022) 5094–5103.
for arginine-assisted protein refolding—not inhibiting soluble oligomers although [276] I. Roy, M.N. Gupta, White & grey biotechnologies for shaping a sustainable
promoting a correct structure, Protein Expr. Purif. 51 (2) (2007) 235–242. future, RSC Sustainability 1 (7) (2023) 1722–1736.
[253] J. Chen, Y. Liu, Y. Wang, H. Ding, Z. Su, Different effects of l-arginine on protein [277] S.Y. Li, I.S. Ng, P.T. Chen, C.J. Chiang, Y.P. Chao, Biorefining of protein waste for
refolding: suppressing aggregates of hydrophobic interaction, not covalent production of sustainable fuels and chemicals, Biotechnol. Biofuels 11 (2018)
binding, Biotechnol. Prog. 24 (6) (2008) 1365–1372. 256.
[254] D. Fass, C. Thorpe, Chemistry and enzymology of disulfide cross-linking in [278] F. De Schouwer, L. Claes, A. Vandekerkhove, J. Verduyckt, D.E. De Vos, Protein-
proteins, Chem. Rev. 118 (3) (2018) 1169–1198. rich biomass waste as a resource for future biorefineries: state of the art,
[255] T. Arakawa, D. Ejima, K. Tsumoto, N. Obeyama, Y. Tanaka, Y. Kita, S. challenges, and opportunities, ChemSusChem 12 (7) (2019) 1272–1303.
N. Timasheff, Suppression of protein interactions by arginine: a proposed [279] M. Peydayesh, M. Bagnani, W.L. Soon, R. Mezzenga, Turning food protein waste
mechanism of the arginine effects, Biophys. Chem. 127 (1–2) (2007) 1–8. into sustainable technologies, Chem. Rev. 123 (5) (2023) 2112–2154.
[256] U. Das, G. Hariprasad, A.S. Ethayathulla, P. Manral, T.K. Das, S. Pasha, A. Mann, [280] E. Piercy, W. Verstraete, P.R. Ellis, M. Banks, J. Rockström, P. Smith, O.C. Witard,
M. Ganguli, A.K. Verma, R. Bhat, S.K. Chandrayan, S. Ahmed, S. Sharma, P. Kaur, J. Hallett, C. Hogstrand, G. Knott, A sustainable waste-to-protein system to
T.P. Singh, A. Srinivasan, Inhibition of protein aggregation: supramolecular maximise waste resource utilisation for developing food-and feed-grade protein
assemblies of arginine hold the key, PLoS One 2 (11) (2007), e1176. solutions, Green Chem. 25 (3) (2023) 808–832.
[257] N.A. Kim, S. Hada, R. Thapa, S.H. Jeong, Arginine as a protein stabilizer and [281] S. Maeta, M. Nakakido, H. Matsuura, N. Sakai, K. Hirata, D. Kuroda, A. Fukunaga,
destabilizer in liquid formulations, Int. J. Pharm. 513 (1–2) (2016) 26–37. K. Tsumoto, Arginine cluster introduction on framework region in anti-lysozyme
[258] G. Guidotti, L. Brambilla, D. Rossi, Cell-penetrating peptides: from basic research antibody improved association rate constant by changing conformational
to clinics, Trends Pharmacol. Sci. 38 (4) (2017) 406–424. diversity of CDR loops, Protein Sci. 32 (9) (2023), e4745.
[282] M.N. Gupta, V.N. Uversky, Structure and Intrinsic Disorder in Enzymology,
Elsevier, 2022.

17

You might also like