pharmaceutical-grade pre-mir-29 purification using an agmatine monolithic support

10
Journal of Chromatography A, 1368 (2014) 173–182 Contents lists available at ScienceDirect Journal of Chromatography A j o ur na l ho me page: www.elsevier.com/locate/chroma Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support Patrícia Pereira a , Ângela Sousa a , João A. Queiroz a , Ana Figueiras a,b , Fani Sousa a,a CICS Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal b CNC Center of Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal a r t i c l e i n f o Article history: Received 18 June 2014 Received in revised form 14 September 2014 Accepted 27 September 2014 Available online 7 October 2014 Keywords: Affinity chromatography Agmatine monolith pre-miR-29 MicroRNA purification Small RNA a b s t r a c t MicroRNA-based therapeutic applications have fostered a growing interest in the development of microR- NAs purification processes in order to obtain the final product with high purity degree, good quality and biologically active. The pre-miR-29 deficiency or overexpression has been associated to a number of clini- cally important diseases, and its therapeutic application can be considered. Monolithic columns emerged as a new class of chromatographic supports used in the plasmid DNA purification platforms, being an interesting alternative to the conventional particle-based columns. Thus, the current work describes, for the first time, a new affinity chromatography method that combines the high selectivity of agmatine lig- ands with the versatility of monoliths to specifically and efficiently purify pre-miR-29 from other small RNA species and Rhodovulum sulfidophilum impurities. The effect of different flow rates on pre-miR-29 separation was also evaluated. Moreover, breakthrough experiments were designed to study the effect of different RNA concentrations on the modified monolithic support binding capacity, being verified that the dynamic binding capacity for RNA molecules is dependent of the feed concentration. In order to achieve higher efficiency and selectivity, three different binding and elution strategies based on increased sodium chloride (1.75–3 M) or arginine (100 mM) and decreased ammonium sulfate (2.4–0 M) stepwise gradients are described to purify pre-miR-29. As a matter of fact, by employing elution strategies using sodium chloride or arginine, an improvement in the final pre-miR-29 yields (97.33 and 94.88%, respec- tively) as well as purity (75.21 and 90.11%, respectively) were obtained. Moreover, the quality control analysis revealed that the level of impurities (proteins, endotoxins, sRNA) in the final pre-miR-29 sample was negligible. In fact, this new monolithic support arises as a powerful instrument on the microRNA purification to be used in further clinical applications, providing a more rapid and economical purification platform. © 2014 Elsevier B.V. All rights reserved. 1. Introduction In the past decade, countless species of non-coding RNA, namely ribosomal RNA (rRNA), transfer RNA (tRNA) and a range of smaller like microRNA (miRNA) molecules, have been discovered and deeply studied. Of these, miRNAs molecules attract particular inter- est due to their essential roles in most cellular processes, as potential biomarkers and drug targets [1,2]. For this reason, RNA represents an important target of a wide range of laboratory analy- sis, being particularly relevant in the diagnosis of several disorders, as well as in basic and applied research. Hence, RNA quality and Corresponding author at: Centro de Investigac ¸ ão em Ciências da Saúde, Univer- sidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal. Tel.: +351 275 329 074; fax: +351 275 329 099. E-mail address: [email protected] (F. Sousa). purity are prerequisites of a multitude of molecular biology and therapeutic applications [3]. Thereby, RNA purification is impor- tant to achieve pure, stable and intact RNA, free from contaminants, including genomic DNA, proteins and organic solvents, once these impurities greatly affect the pharmaceutical and clinical applica- tions [3,4]. This fact greatly contributes to the need to develop novel methods for the rapid and inexpensive isolation and purification of microRNAs. RNA is an unstable molecule and has a very short half-life once extracted due to the ubiquitous presence of RNA-degrading enzymes (RNases) which are present in biological samples, aque- ous buffers, on labware and can be introduced via human handling [5]. To overcome the problems associated with RNA isolation, sev- eral strategies are available to isolate and purify miRNA molecules chemically synthesized or derived from various biological sources [3,6]. The purification of RNA molecules is already reported by using preparative denaturing polyacrylamide gel electrophoresis (PAGE), http://dx.doi.org/10.1016/j.chroma.2014.09.080 0021-9673/© 2014 Elsevier B.V. All rights reserved.

Upload: fani

Post on 10-Apr-2017

214 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

Pm

Pa

b

a

ARR1AA

KAApMS

1

rldeprsa

sT

h0

Journal of Chromatography A, 1368 (2014) 173–182

Contents lists available at ScienceDirect

Journal of Chromatography A

j o ur na l ho me page: www.elsev ier .com/ locate /chroma

harmaceutical-grade pre-miR-29 purification using an agmatineonolithic support

atrícia Pereiraa, Ângela Sousaa, João A. Queiroza, Ana Figueirasa,b, Fani Sousaa,∗

CICS – Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, PortugalCNC – Center of Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal

r t i c l e i n f o

rticle history:eceived 18 June 2014eceived in revised form4 September 2014ccepted 27 September 2014vailable online 7 October 2014

eywords:ffinity chromatographygmatine monolithre-miR-29icroRNA purification

mall RNA

a b s t r a c t

MicroRNA-based therapeutic applications have fostered a growing interest in the development of microR-NAs purification processes in order to obtain the final product with high purity degree, good quality andbiologically active. The pre-miR-29 deficiency or overexpression has been associated to a number of clini-cally important diseases, and its therapeutic application can be considered. Monolithic columns emergedas a new class of chromatographic supports used in the plasmid DNA purification platforms, being aninteresting alternative to the conventional particle-based columns. Thus, the current work describes, forthe first time, a new affinity chromatography method that combines the high selectivity of agmatine lig-ands with the versatility of monoliths to specifically and efficiently purify pre-miR-29 from other smallRNA species and Rhodovulum sulfidophilum impurities. The effect of different flow rates on pre-miR-29separation was also evaluated. Moreover, breakthrough experiments were designed to study the effectof different RNA concentrations on the modified monolithic support binding capacity, being verifiedthat the dynamic binding capacity for RNA molecules is dependent of the feed concentration. In order toachieve higher efficiency and selectivity, three different binding and elution strategies based on increasedsodium chloride (1.75–3 M) or arginine (100 mM) and decreased ammonium sulfate (2.4–0 M) stepwisegradients are described to purify pre-miR-29. As a matter of fact, by employing elution strategies usingsodium chloride or arginine, an improvement in the final pre-miR-29 yields (97.33 and 94.88%, respec-

tively) as well as purity (75.21 and 90.11%, respectively) were obtained. Moreover, the quality controlanalysis revealed that the level of impurities (proteins, endotoxins, sRNA) in the final pre-miR-29 samplewas negligible. In fact, this new monolithic support arises as a powerful instrument on the microRNApurification to be used in further clinical applications, providing a more rapid and economical purificationplatform.

. Introduction

In the past decade, countless species of non-coding RNA, namelyibosomal RNA (rRNA), transfer RNA (tRNA) and a range of smallerike microRNA (miRNA) molecules, have been discovered andeeply studied. Of these, miRNAs molecules attract particular inter-st due to their essential roles in most cellular processes, asotential biomarkers and drug targets [1,2]. For this reason, RNA

epresents an important target of a wide range of laboratory analy-is, being particularly relevant in the diagnosis of several disorders,s well as in basic and applied research. Hence, RNA quality and

∗ Corresponding author at: Centro de Investigac ão em Ciências da Saúde, Univer-idade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal.el.: +351 275 329 074; fax: +351 275 329 099.

E-mail address: [email protected] (F. Sousa).

ttp://dx.doi.org/10.1016/j.chroma.2014.09.080021-9673/© 2014 Elsevier B.V. All rights reserved.

© 2014 Elsevier B.V. All rights reserved.

purity are prerequisites of a multitude of molecular biology andtherapeutic applications [3]. Thereby, RNA purification is impor-tant to achieve pure, stable and intact RNA, free from contaminants,including genomic DNA, proteins and organic solvents, once theseimpurities greatly affect the pharmaceutical and clinical applica-tions [3,4]. This fact greatly contributes to the need to develop novelmethods for the rapid and inexpensive isolation and purification ofmicroRNAs.

RNA is an unstable molecule and has a very short half-lifeonce extracted due to the ubiquitous presence of RNA-degradingenzymes (RNases) which are present in biological samples, aque-ous buffers, on labware and can be introduced via human handling[5]. To overcome the problems associated with RNA isolation, sev-

eral strategies are available to isolate and purify miRNA moleculeschemically synthesized or derived from various biological sources[3,6]. The purification of RNA molecules is already reported by usingpreparative denaturing polyacrylamide gel electrophoresis (PAGE),
Page 2: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

1 atogr.

acsoIc[vtaieo

crsttthppapohlwlbrldg

amtsioldobfigpepbmibeo

2

2

fd

74 P. Pereira et al. / J. Chrom

ffinity tag-based purification, anion-exchange and size exclusionhromatography [7–11]. These methods make use of enzymes andtructural modifications in the RNA molecule by the introductionf tags sequences, which can affect the overall topology of the RNA.n addition, the application of this methodology and RNA modifi-ations is typically limited to sRNAs, smaller than 40 nucleotides4,12,13]. Overall, and although these purification methods can beery efficient for the recovery of RNA with high quality and quan-ity, they still are expensive on large scale, labor intensive and onlyllow the separation of small RNAs (sRNAs) from rRNA. In addition,n most cases the isolation of pure RNA is achieved by a secondarynrichment, either through enzymatic removal of DNA or by a sec-nd set of columns specific to miRNA [14].

In the last years, our research group developed a new affinityhromatography approach, named amino acid-affinity chromatog-aphy to efficiently purify different RNA species (total RNA, rRNA,RNA and 6S RNA) [15–17]. This powerful technique is based onhe application of amino acids as specific ligands to purify RNA onhe basis of their biological function or individual chemical struc-ure [18]. More recently, lysine, tyrosine and arginine amino acidsave been successfully applied as affinity ligands for microRNAsurification, particularly in pre-miR-29 isolation, in conventionalarticle-based columns [19–21]. Amino acid ligands describedbove showed high selectivity, however a faster and more robusturification method is required due to the structural characteristicsf the RNA molecule, including their stability. Monolithic supportsave been largely explored in recent years for the separation of

arge biomolecules owing to their structural properties comparedith conventional columns, namely high binding capacity, excel-

ent mass transfer properties and a huge quantity of accessibleinding sites [22,23]. Moreover, monoliths allow a very fast sepa-ation and purification with high reproducibility both at small andarge scales, support higher flow rates and reduced biomoleculesegradation due to the shorter contact times with the chromato-raphic matrix [24].

This work has the purpose of exploring the possibility of usinggmatine amino acid derivative as ligand, combined with theulti-functionality of the monolithic support [25–27] to selec-

ively isolate the pre-miRNA from the recombinant Rhodovulumulfidophilum host. Knowing that an agarose based matrix withmmobilized arginine amino acid allows an efficient separationf pre-miR-29 [21], it becomes interesting to study a new mono-ith with agmatine immobilized. Agmatine is a neurotransmittererived from the decarboxylation of arginine and plays the rolef agonist or antagonist of different enzymes involved in severaliological mechanisms [28–30]. This study demonstrates for therst time, the use of this amino acid derivative as a chromato-raphic ligand to purify microRNA. In this way, the aim of theresent study is to develop new methodologies for RNA isolation,nabling the pre-miRNA-29 purification with high integrity andurity using monoliths, in view of the application in moleculariology or therapeutic procedures in a short time. Additional chro-atographic characterization based on breakthrough experiments

s also designed to study the dynamic binding capacity for RNA. Theinding behavior of pre-miR-29 under the influence of differentnvironmental conditions, such as the elution buffer composition,r using different flow-rates is also investigated.

. Materials and methods

.1. Materials

All buffers used for the chromatographic experiments werereshly prepared with sterilized water pre-treated with 0.05%iethyl pyrocarbonate (DEPC; Sigma–Aldrich, St Louis, MO, USA),

A 1368 (2014) 173–182

filtered through a 0.20 �m pore size membrane (Schleicher Schuell,Dassel, Germany) and degassed ultrasonically. The ammonium sul-fate and sodium chloride salts used in these buffers were purchasedto Panreac (Barcelona, Spain), tris(hydroxymethyl) aminomethane(Tris) to Merck (Darmstadt, Germany) and agmatine sulfate wasfrom Sigma–Aldrich (St. Louis, MO, USA). Chromatographic exper-iments were carried out in the 0.34 mL bed volume (average poresize of 1500 nm in diameter) carbonyldiimidazole (CDI) monolithmodified with agmatine amino acid derivative, kindly providedby BIA Separations (Ajdovscina, Slovenia). The guanidinium saltand all the chemicals used in the lysis buffer were obtained fromSigma–Aldrich (St Louis, MO, USA). All the materials used in theexperiments were RNase-free. The DNA molecular weight marker,Hyper Ladder I, was obtained from Bioline (London, UK) and Green-Safe Premium was purchased to NZYTech (Lisbon, Portugal).

2.2. Pre-miR-29 production and isolation

The pre-miR-29 was obtained from the culture of Rhodovu-lum sulfidophilum DSM 1374 strain (BCCM/LMG, Belgium) modifiedwith the plasmid pBHSR1-RM containing the sequence of pre-miR-29 [31]. Growth was carried out in shaker flasks withcapacity of 500 mL containing 100 mL of Nutrient Broth medium(1 g/L beef extract; 2 g/L yeast extract; 5 g/L peptone and 5 g/Lsodium chloride) supplemented with 30 �g/mL kanamycin, ina rotary shaker at 30 ◦C and 250 rpm under dark-aerobic con-ditions. The small RNA fraction was extracted from bacterialpellets of Rhodovulum sulfidophilum by a modified acid guani-dinium thiocyanate–phenol–chloroform extraction method, asdescribed by Pereira and co-workers [21]. Briefly, cells were lysedand the small RNA fraction obtained was precipitated with iso-propanol. Precipitated molecules were recovered by centrifugationat 15,000 × g for 20 min at 4 ◦C. After centrifuging, the small RNApellet was washed with 75% ethanol and incubated at room temper-ature for 10 min, followed by a 5 min centrifugation at 15,000 × g(4 ◦C). The air-dried small RNA pellet was solubilized in 1 mL ofDEPC-treated water. Finally, 260 and 280 nm absorbance of thesamples was measured using a nanophotometer in order to assesssmall RNA quantity and purity.

2.3. Chromatographic experiments

The chromatographic experiments were performed in an ÄKTAAvant system with UNICORN 6 software (GE Healthcare, Sweden).For the experiments, agmatine monolithic disk was equilibratedwith appropriate loading buffer, as described below, at a flow rateof 1 mL/min. The pre-miR-29 was purified by exploiting three dif-ferent elution strategies, namely by using sodium chloride (NaCl),ammonium sulfate ((NH4)2SO4) and arginine as competition agent.In experiments with ammonium sulfate, the monolith was equili-brated with 2.4 M of (NH4)2SO4 in 10 mM Tris–HCl buffer (pH 8).After the elution of unbound species, the ionic strength of the bufferwas stepwise decreased to reach the 0 M (NH4)2SO4 in 10 mMTris–HCl buffer (pH 8), to elute of pre-miR-29. On the other hand, inexperiments with sodium chloride, after washing out the unboundmaterial with 1.75 M NaCl in 10 mM Tris–HCl buffer (pH 9.5), theionic strength of the buffer was increased to 3 M NaCl in 10 mMTris–HCl buffer (pH 9.5). The experiments performed with argi-nine as a competition agent to purify pre-miR-29 molecules wereinitiated with the equilibration of the monolith with 1.75 M NaClin 10 mM Tris–HCl buffer (pH 9.5) to promote the total retentionof pre-miR-29 and then the elution was accomplished by chang-

ing to 1.75 M NaCl supplemented with 100 mM arginine in 10 mMTris–HCl buffer (pH 9.5). The most retained species were finallyeluted with 3 M NaCl. In the three sets of experiments, sRNAsextracts (30 �g), containing 6S RNA, pre-miR-29 and other sRNA,
Page 3: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

atogr.

i1apiaww2a

2

twc0btbbmadsvetrp51Fts

2

iAp90wgaw

2

tbSisoam

2

t

P. Pereira et al. / J. Chrom

ncluding transfer RNAs, were injected onto the column using a00 �L loop at the same flow-rate. The absorbance of the elu-te was continuously monitored at 260 nm. All experiments wereerformed at room temperature. Fractions were pooled accord-

ng to the chromatograms obtained, and following concentrationnd desalting with Vivaspin concentrators (Vivascience), the poolsere kept for quantification and further analysis. A posterior studyas performed to analyze the effect of different flow-rates (0.5, 1,

and 3 mL/min) in the pre-miR-29 purification efficiency using thegmatine monolith.

.4. Dynamic binding capacity measurement for sRNA

Agmatine monolithic disk was used for the determination ofhe dynamic binding capacity (DBC) for RNA. These experimentsere performed with 1 mL/min of flow rate using different con-

entrations of the feedstock (0.025, 0.05, 0.075, 0.1, 0.15, 0.20 and.25 mg/mL). The support was equilibrated with 10 mM Tris–HCluffer (pH 8) and thereafter, it was overloaded with the RNA solu-ion under the same equilibrium conditions. DBC was determinedy the breakthrough area integration method [32]. Briefly, eachreakthrough experiment was derived from a 100% of saturatedonolith. Then, the sample volume corresponding to the adsorbed

mount of RNA was calculated by numerical integration of theetector response. The area obtained from the filled monolith wasubtracted from that for the empty monolith. In this step, the voidolume was discounted from the DBC determination. This area isquivalent to the sample volume, which was required to saturatehe monolithic support, and can be related with RNA mass thatemained bound per milliliter of the support, reflecting the sup-ort capacity. Normally, the capacity values are represented at 10,0 and 100% of the breakthrough that corresponds to 10, 50 and00% of the column saturation, being calculated in the same way.inally, the elution of the bound RNA was achieved by increasinghe sodium chloride concentration in the mobile phase to 3 M in atepwise manner.

.5. Polyacrylamide electrophoresis

Fractions recovered from the sRNA chromatographic exper-ments were analyzed by vertical electrophoresis using anmersham Biosciences system (GE Healthcare, Sweden) with 10%olyacrylamide gel. Electrophoresis was carried out at 125 V for0 min with TBE buffer (0.84 M Tris base, 0.89 M boric acid and.01 M EDTA, pH 8.3). sRNA samples were previously denaturedith 97.5% formamide and denatured conditions were kept in the

el owing to the presence of 8 M urea. sRNA molecules were visu-lized in the gel by using the Vilber Lourmat system after stainingith ethidium bromide (0.5 mg/mL).

.6. Protein analysis

Proteins contamination in pre-miR-29 samples collected fromhe purification with agmatine monolithic support, was measuredy using the micro-BCA (bicinchoninic acid) assay (Thermo Fishercientific Inc., Rockford, IL, USA), according to manufacturer’snstructions. Briefly, the calibration curve was prepared using BSAtandards (0.01–0.25 mg/mL). A total of 25 �L of each standardr pre-miR-29 samples was added to 200 �L of BCA reagent in

microplate and incubated for 30 min at 60 ◦C. Absorbance waseasured at 570 nm in a microplate reader.

.7. Pre-miR-29 identification analysis

Pre-miR-29 identification was confirmed using reverse-ranscriptase polymerase chain reaction (RT-PCR). Thus, cDNA

A 1368 (2014) 173–182 175

synthesis was performed using RevertAid First Strand cDNA Syn-thesis Kit (Fermentas, Thermo Fisher Scientific Inc.), according tothe manufacturer’s instructions. A total of 0.5 �g of RNA sam-ples collected after the chromatographic purification process withagmatine monolith was used to initiate cDNA synthesis, whichwas denatured for 5 min at 65 ◦C with 20 pmol of gene-specificprimers (5′-GACAGC GGT ATG ATC CCC CAA-3′). Then, PCR reac-tions were carried out using 1 �L of synthesized cDNA in a 25 �Lreaction containing 0.125 U of Supreme DNA polymerase (NZYtech,Lisbon, Portugal), 50 mM of magnesium chloride (NZYtech, Lisbon,Portugal) and 150 nM of each primer. Sense (5′-GGA AGCTGG TTTCAT ATG GTG-3′) and antisense (5′-CCC CCA AGA ACA CTG ATTTC-3′) primers were used to amplify a fragment of 145 bp. ThePCR program was carried out as follows: denaturation at 95 ◦C for5 min, followed by 40 cycles at 95 ◦C for 30 s, 63 ◦C for 30 s and 72 ◦Cfor 15 s, and a final elongation step at 72 ◦C for 5 min. To confirmthe presence and purity of amplicons, PCR products were analyzedusing 1% agarose gel [21].

3. Results and discussion

The purification methods developed to purify microRNAsenvisioning therapeutic applications require the use of a chro-matographic support able to eliminate impurities, maintainingthe structural integrity of the biomolecule. Hence, the presentstudy aims to explore and characterize the interactions occur-ring between pre-miR-29 and agmatine amino acid derivativeimmobilized into a monolithic disk, combining, for the first time,the selectivity, specificity and biorecognition of agmatine ligandswith the structural versatility and capacity provided by mono-lithic supports. These supports offer several potential advantagesover conventional supports, including higher selectivity and repro-ducibility and good capacity [17,26,33,34]. The miR-29 target waschosen because it belongs to one of the most interesting miRNAfamilies in humans to date, once this miRNA is involved in severalregulatory pathways associated with neurodegenerative diseasesand also presenting tumor-suppressing and immune-modulatingproperties [35–37]. Therefore, the purpose of this work is todescribe an alternative method to purify pre-miRNA from a com-plex mixture of total RNA by affinity chromatography, exploitingthe multiple biological interactions which occur between the pre-miR-29 and the agmatine ligand immobilized in monoliths.

3.1. Agmatine monolithic disk

Non-grafted CDI monolith was properly modified with agmatineligand by BIA Separation (Ajdovscina, Slovenia), and the suitableagmatine ligand immobilization was confirmed through a compar-ison of the different chromatographic profiles of the new supportand the non-grafted CDI monolith (data not shown). Recently, ourresearch group has demonstrated the possibility of using the non-grafted CDI monolith to purify plasmid DNA by using a decreasingstepwise gradient of ammonium sulfate but not using sodiumchloride (NaCl) [25,26]. Thereby, the two monolithic disks wereequilibrated with 10 mM Tris–HCl buffer (pH 8) at 1 mL/min, andafter the injection of 30 �g of sRNA sample, a 15-min linear gradientup to 3 M of NaCl in 10 mM Tris–HCl buffer (pH 8) was established.In the non-grafted CDI monolith, a single peak was rapidly attainedin the flow through due to the elution of species with lower affin-ity. Afterwards, the NaCl linear gradient was established but nospecies were eluted (data not shown). These results indicated that

the non-grafted CDI monolith did not interact with sRNA moleculesunder the conditions used. The same experiment was conducted inthe monolith modified with agmatine ligand. The total retention ofsRNA was obtained at 10 mM Tris–HCl buffer (pH 8) and the total
Page 4: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

176 P. Pereira et al. / J. Chromatogr. A 1368 (2014) 173–182

Table 1Effect of sRNA concentration on 10%, 50% and total dynamic binding capacity ofagmatine monolithic disk. The breakthrough experiments were performed on a sin-gle monolithic disk, at flow rate of 1 mL/min. The loading sample was sRNA dissolvedin 10 mM Tris–HCl buffer (pH 8).

sRNA concentration (mg/mL) DBC (mg/mL)

10% 50% Total

0.025 0.24 1.04 3.510.05 0.71 1.37 4.740.075 0.88 1.46 5.900.10 1.28 1.63 6.920.15 1.98 2.71 7.09

eNTdii

3s

dtfw(tsiitbcclDpwisftsvfmiecOpa

tf(ditat

0.20 2.38 3.07 7.590.25 2.91 3.22 8.08

lution was verified during the linear gradient by increasing theaCl concentration in the buffer from 0 to 3 M (data not shown).hese results confirmed that the agmatine monolith is chemicallyifferent from the non-grafted CDI disk and the agmatine ligands

mmobilized on the monolith are the ones responsible for sRNAnteraction with the support.

.2. Effect of feed concentration on dynamic binding capacity ofRNA

For a complete characterization of the agmatine monolithicisk, the dynamic binding capacity (DBC) was evaluated, oncehis parameter is a critical factor for the chromatographic per-ormance. For this purpose, several breakthrough experimentsere performed at 1 mL/min, with different sRNA concentrations

0.025–0.25 mg/mL). The results summarized in Table 1 indicatehat the total capacity of the agmatine monolithic disk to bindRNA was higher for the feed concentration of 0.25 mg/mL, wheret was found a total capacity of 8.08 mg/mL. By analyzing the datan Table 1, it appears that at 10, 50 and 100% of the breakthrough,he DBC increases with increasing RNA concentrations, thus, it cane argued that the binding capacity is dependent of the RNA con-entration. The profile obtained with agmatine monolithic disk is inoncordance with other RNA binding profiles described for mono-iths, such as CIM IDA monolithic column [38]. Furthermore, theseBC results found for agmatine monolith are very good when com-ared with other values described for purified baker’s yeast RNA,here the CIM IDA monolithic column presented a binding capac-

ty of 1.20 mg/mL, using 1 mg/mL of feed RNA [38]. This comparisonuggests that the immobilized agmatine ligand can be responsibleor the improvement on the DBC of the monolithic support throughhe enhancement of the interaction ligand–sRNAs, although theRNA concentrations used are significantly lower than in the pre-ious study (0.5–2 mg/mL) [38]. These findings can be due to theact that RNA has a size of 100 nm to over 300 nm in diameter, and

onoliths present a larger channel size of 1000–5000 nm allow-ng full availability of the ligands on the chromatographic supportven at low feed concentration of sRNA. Thus, these results indi-ate that the DBC is dependent of sRNA feed concentration [39].verall, these results suggest that a very efficient chromatographicerformance for RNA purification using monolithic columns can bechieved if using low RNA feed concentration.

Through the analysis of the isotherm profile, it can be seenhat, in the sRNA concentration range studied, the DBC increasesrom 3 mg/mL to a plateau region, as the RNA concentrationCRNA) increases. Considering the equilibrium binding isothermata, Fig. 1, it is possible to define a linear and a plateau region. Thus,

t is suggested that our data follow the Langmuir model, assuminghat the adsorbed molecules have a fixed number of sites on thedsorbent where interactions can occur, and that every adsorp-ion site is energetically equivalent and accepts only one molecule

Fig. 1. Adsorption isotherm of sRNA on agmatine monolith.

[40]. At low concentrations (0.025–0.075 mg/mL), RNA is well dis-tributed at the agmatine ligands and the orientation is determinedby agmatine ligand–RNA interactions, resulting in a linear shapedcurve between RNA adsorbed amounts and mobile phase concen-trations. At high concentrations (0.1–0.25 mg/mL), the adsorptionsites may become saturated, leading to a curvature of the isotherminto an asymptote. Concentration values of sRNA absorbed above7 mg/mL are considered in the overloaded zone of the isotherm.

In addition, the dissociation constant (Kd) from adsorptionisotherm, which represents the interaction between the sRNAand agmatine monolithic support was also calculated by frontalanalysis chromatography, according to what is described byRuta and co-workers [41]. Through the values at 50% of break-through experiments obtained with agmatine monolith at differentsRNA concentrations, the Kd value was 2.6 × 10−7 M. A Kd valuebetween 10−4 and 10−8 M indicates that the risks of irreversiblebiomolecules adsorption and denaturation are minimized [40]. So,the Kd value obtained, reveals a good affinity interaction betweenthe ligand and RNA, which indicates that agmatine monolith is agood affinity support.

3.3. Effect of elution buffer composition and pH manipulation onpre-miR-29 binding

In affinity chromatography the elution of a target biomoleculecan be performed either specifically, through addition of a compet-ing agent in the elution buffer or non-specifically, through changesin the elution buffer composition, namely in type of salt and ionicstrength, and pH manipulation depending on the matrix used andthe chemical characteristics of target biomolecule [18]. As previ-ously mentioned, in this study the chromatographic behavior of theagmatine monolith interacting with RNA, was evaluated accordingto the type and salt concentration and pH manipulation. Before-hand, several binding/elution studies were performed in order todetermine the best salt concentration range to achieve the bindingand elution of the pre-miR-29 (data not shown). Linear and step-wise gradients were tested, and it was determined that the stepwiseelution is more suitable to obtain the pre-miR-29 separation, oncethis strategy allows greater selectivity between the biomoleculein study and contaminants. Moreover, it was verified the possibil-ity to establish an increased NaCl gradient to bind and recover thepre-miR-29. After this preliminary characterization of the pre-miR-29 retention behavior on the agmatine monolith it was evaluatedthe pH effect on pre-miR-29 stability, in order to determine which

pH value should be used in the purification process. In addition,the pH could also significantly influence pre-miR-29 interactionswith the immobilized agmatine, given the versatility of this ligand.Thus, several chromatographic experiments, based on ionic-based
Page 5: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

P. Pereira et al. / J. Chromatogr.

Table 2Effect of pH of mobile phase in the tRNA and pre-miR-29 retention, using 10 mMTris buffer as loading condition.

pH of mobile phase tRNA retention pre-miR-29 retention

6.5 ++ ++8.0 + ++

F

ebs[

atrosgenbdaetatsmosRc[RTin

tt

Recently, our research group demonstrated the possibility to

Fio

9.5 − ++

rom (−) no retention to (++) total retention.

lution conditions, were designed to analyze how the pH rangingetween 6.5 and 9.5 affects the retention of pre-miR-29 and other R.ulfidophilum sRNA species, considering the pKa of 13 of this ligand42].

Table 2 summarizes several chromatographic runs performedt different loading conditions (pH and NaCl manipulation), andhe relative effect of these conditions on tRNA and pre-miR-29etention ((−) no retention to (++) total retention). As a matterf fact, the results showed an increased retention of all sRNAspecies when the pH of the mobile phase is lower (pH 6.5), sug-esting that the decreased pH favors the establishment of strongerlectrostatic interactions but also the involvement of multipleon-covalent interactions, namely cation–� interactions, hydrogenonds, hydrogen � interactions and water mediated bonds. Theseifferent types of interaction established between the pre-miR-29nd agmatine ligands can be related with the positive charge (theffective surface charge) of agmatine, considering the fact that athe working pH (between 6.5.and 9.5), agmatine is protonated. Inddition, the agmatine is a derivative of arginine amino acid, con-aining basic guanidinium group and the lack of the carboxyl group,o that both amino acids have very similar structures. Thus, theultiplicity of interactions can also occur because agmatine has

ne polar center with which RNA can strongly associate. It is rea-onable to suppose that the retention of all functional classes ofNA in agmatine monolith is due to the length of agmatine sidehain and its ability to produce good hydrogen bond geometries43,44], which can promote multicontact with RNA backbone orNA bases, according to RNA folding (RNA conformations) [45–47].he cation–� interactions can be due to interactions between pos-tive guanidinium group of agmatine and aromatic rings of theitrogen bases of RNA [17].

Moreover, for the highest pH studied (pH 9.5), it was observedhat the pre-miR-29 was more retained than other species fromRNA, because some interactions described above are less favored

ig. 2. (A) Chromatographic profile of the pre-miR-29 purification from a sRNA mixture usncreasing NaCl concentration in the eluent from 1.75, 2.5 and 3 M, as represented by theutlet. Fractions corresponding to peaks (1)–(3) are shown in lanes 1–3, respectively. Lan

A 1368 (2014) 173–182 177

and therefore only some RNA species bound to the support, as pre-miR-29, whereas other species do not bind. Thus, the pH used forthe study, using sodium chloride as elution strategy, was pH 9.5,due to a higher retention of pre-miR-29 and a more pronounceddifference between the pre-miR-29 retention and the binding ofother RNA species, which allowed exploring the selectivity of theagmatine ligand.

To investigate the retention behavior of the target miRNA inthe agmatine monolith, after binding, the pre-miR-29 elution wasachieved with the application of a stepwise gradient increasingthe NaCl concentration up to 3 M in 10 mM Tris–HCl buffer (pH9.5). The chromatographic profile of the pre-miR-29 purificationfrom a complex mixture of R. sulfidophilum sRNA, using the agma-tine monolithic disk, is presented in Fig. 2A. The chromatographicrun was initiated with an ionic strength of 1.75 M NaCl in 10 mMTris–HCl buffer (pH 9.5). After injection of the complex mixture ofsRNA, a first peak was obtained with the same salt concentrationof the equilibrium buffer, resulting from the elution of unboundspecies. As shown in Fig. 2A, the ionic strength of elution bufferwas increased to 2.5 M of NaCl to elute the pre-miR-29 in a secondpeak. The elution of highly bound species, mostly tRNAs, was thenachieved by increasing the ionic strength of the buffer to 3 M NaCl(peak 3). In this way, this result demonstrates that the agmatine lig-ands distinguished and differentially interacted with various RNAmolecules, suggesting a specific recognition for the pre-miR-29.Thereby, in order to establish a correlation between the differ-ent RNAs species present in the mixture, and the peaks in thechromatogram, a polyacrylamide gel electrophoresis (Fig. 2B) wasperformed, thus the lines 1, 2 and 3 presented in the electrophoreticprofile correspond to the samples pooled from the respective peaksin the chromatogram. Hence, electrophoretic analysis revealed thatthe first peak of unbound species corresponds to the elution of tRNAspecies (Fig. 2B, lane 1), at lower ionic strength. On the other hand,the elution of the pre-miR-29 only occurs with the increase of ionicstrength in the second peak (Fig. 2B, lane 2). Finally, the 6S RNA,pre-miR-29 and other tRNAs species were eluted with 3 M NaCl,as it was observed in the third chromatographic peak (peak 3 andlane 3). These findings suggest that pre-miR-29 presents a strongerinteraction with the agmatine monolith than the majority of tRNAspecies.

purify pre-miR-29 using stepwise gradients of NaCl with thearginine–agarose matrix [21] being verified the pre-miR-29 elu-tion at 360 mM of NaCl, while in this study the agmatine monolith

ing the agmatine monolithic disk. Elution was performed at 1.0 mL/min by stepwise arrows. (B) Polyacrylamide gel electrophoresis of samples collected at the columne S, impure sRNA preparation injected onto the column.

Page 6: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

178 P. Pereira et al. / J. Chromatogr. A 1368 (2014) 173–182

Fig. 3. (A) Chromatogram showing the purification of pre-miR-29 from sRNA population by using the agmatine monolithic disk. Elution was performed at 1 mL/min bystepwise decreasing (NH4)2SO4 concentration in the eluent from 2.4 to 0 M (NH4)2SO4 in 10 mM Tris buffer (pH 8.0). (B) Polyacrylamide gel electrophoresis analysis ofs ) are sc

aNittct�wgoctiaaolbtsc(bttcbo(hiwctttateiec

m

amples collected at the column outlet. Fractions corresponding to peaks (1) and (2olumn.

llowed the recovery and purification of pre-miR-29 with 2.5 M ofaCl. In general, this comparative study evidences that the bind-

ng mechanism inherent to the biorecognition of pre-miR-29 byhe agmatine amino acid derivative is stronger than the interac-ions established with the arginine amino acid. This phenomenonan result from the combination of multiple elementary interac-ions such as electrostatic interactions, hydrogen bonds, hydrogen

interactions, water mediated bonds and cation–� interactions,hich both ligands may engage through the terminal guanidine

roup. However, in the agmatine monolith a stronger interactionccurred since the recovery of pre-miR-29 required a higher NaCloncentration. The difference between the ligands is the absence ofhe carboxyl group in agmatine, which can have two positive effectsn the RNA retention. It can prevent repulsion of charged moleculesnd enables the establishment of additional hydrophobic inter-ctions with the carbon backbone of the ligand. Thus, dependingn the environmental conditions established and the amino acidigands used, some interactions can be more favored than other,ecoming more evident under these conditions. In order to provehis hypothesis and mainly exploit hydrophobic interactions, atepwise gradient by decreasing ammonium sulfate ((NH4)2SO4)oncentration between 2.4 M and 0 M in 10 mM Tris–HCl bufferpH 8) was applied. In this case, the agmatine monolith was equili-rated with 2.4 M (NH4)2SO4 in 10 mM Tris–HCl buffer (pH 8). Afterhe binding of the sample to the disk, a first elution step designedo elute the tRNA species with lower affinity to the support wasarried out with the same salt concentration of the equilibriumuffer, promoting total pre-miR-29 retention (Fig. 3A). The elutionf pre-miR-29 was then achieved with a second step by using 0 MNH4)2SO4 in 10 mM Tris–HCl buffer (pH 8) (Fig. 3A). In fact, theigh salt concentration plays a key role on the pre-miR-29 bind-

ng to agmatine ligand. Again, a polyacrylamide gel electrophoresisas used to detect and identify different species eluting in each

hromatographic peak (Fig. 3B). The electrophoretic analysis ofhe fractions eluting from the agmatine monolith (Fig. 3A) provedhat the first peak of unbound material corresponds to the elu-ion of tRNA (Fig. 3B, lane 1), whereas the second peak was mainlyttributed to the elution of pre-miR-29 (Fig. 3B, lane 2). The interac-ions that favor the recognition of pre-miR-29, under hydrophobiclution conditions, can be van der Waals forces and hydrophobicnteractions while the main responsible group for the interactions

stablished between pre-miR-29 and agmatine ligand can be thearbon chain of the spacer arm.

The functionality of agmatine ligand to biorecognize the pre-iR-29 under hydrophobic- and ionic-based elution conditions

hown in lanes 1 and 2, respectively. Lane S, impure sRNA sample injected onto the

shows the applicability and versatility of this ligand to developseveral pre-miR-29 purification strategies due to the multipleinteractions involved for each condition. Apart from the struc-tural characteristics of the agmatine ligand, also the pre-miR-29structural features seem to be relevant on its distinct interactionbehavior with the ligand. The main explanation for the specificinteractions occurring between the pre-miR-29 and the agmatineamino acid derivative is the single-stranded nature of RNA, whichis normally involved in RNA recognition, due to the high base expo-sure and availability for interactions. Additionally, the negativecharge conferred by the phosphate groups in the RNA backbone isimportant for their interaction with the agmatine monolith, sug-gesting them to have a crucial role in RNA retention. Likewise,pre-miR-29 is a sRNA molecule with the shape of a stem-loopor hairpin consisting of two long irregular double-stranded stemregions, which are interrupted by a largely single-stranded internalloop [21]. This particular structure may also explain the multiplenon-covalent interactions established which are involved in thebiomolecular recognition of pre-miR-29 by the agmatine ligand.

In turn, to obtain an elution strategy more selective and biospe-cific as well as to achieve higher elution efficiency, it was employeda new approach for pre-miR-29 purification using arginine as com-petitive agent. This agent can bind either to the retained pre-miR-29or to the immobilized agmatine ligand depending on their char-acteristics, allowing thus to predict the interactions that can beinvolved once agmatine is derived of arginine. The competitivestudies were performed by adding 250 mM of arginine to theelution buffer in stepwise gradient to exploit specific elution ofpre-miR-29 from the agmatine monolith and therefore to evalu-ate the possibility of reaching higher purification factors. Elutiongradients applied were adapted to ensure that the RNA sample ison ideal conditions to bind to the support and be eluted only dueto the competing agent. Agmatine monolith was first equilibratedwith 1.75 M NaCl in 10 mM Tris–HCl buffer (pH 9.5) to promote thetotal retention of pre-miR-29. After sample application, a first peakof unbound species was obtained and then the ionic strength wasincreased upon arginine addition. In this step, 1.75 M of NaCl sup-plemented with 100 mM arginine in 10 mM Tris–HCl buffer (pH 9.5)was used for the elution of pre-miR-29 in a second peak (Fig. 4A).

These findings are in accordance with previous published resultswhich show that this competition strategy is able to elute bound

pre-miR-29 from arginine–agarose column [21]. It is suggestedthat the major mechanism from which pre-miR-29 is eluted fromthe agmatine support is the preferential binding of free arginineto the pre-miR-29, an interaction that we believe to be stronger
Page 7: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

P. Pereira et al. / J. Chromatogr. A 1368 (2014) 173–182 179

F erfor1 lysis oc ample

ttifmldttetwppsitoasis

aa

cagoowtematwwpedbN

ig. 4. (A) Separation of pre-miR-29 using agmatine monolithic disk. Elution was p00 mM as represented by the arrows. (B) Polyacrylamide gel electrophoresis anaorresponding to peaks (1)–(3) are shown in lanes 1–3, respectively. Lane S, sRNA s

han the interaction pre-miR-29–agmatine support. In fact, sincehe arginine is positively charged at the pH in study, electrostaticnteractions can possible occur between the pre-miR-29 and theree arginine. However, another mechanism that can explain pre-

iR-29 elution is the interaction of free arginine with the agmatineigand in the immobilized support [48], that could also promote theisplacement of bound pre-miR-29. The last peak corresponded tohe elution of tRNAs and some residual pre-miR-29 by increasinghe ionic strength to 3 M of NaCl. The results of polyacrylamide gellectrophoresis (Fig. 4B) showed that the tRNAs were recovered inhe first peak (lane 1), being separated from the pre-miR-29 thatas attained in the second peak (lane 2), while other tRNAs andre-miR-29 were obtained in the third peak (lane 3). As judged byolyacrylamide gel electrophoresis, pre-miR-29 was isolated anduitably purified in the second chromatographic step with highntegrity. However, even if some arginine remains in the sample,he structure of pre-miR-29 is not changed, as we have previ-usly studied by circular dichroism (data not shown). Actually, thepplication of arginine can also be strongly associated with the pre-erved integrity observed in RNA samples since arginine, owing tots multiplicity for interactions, has been largely associated withtabilizing effects on RNA conformation [17,21].

In a general way, the competition study has shown that the freerginine could promote the pre-miR-29 biospecific elution of thegmatine monolith.

Beyond the potential application of agmatine-based affinityhromatography to purify pre-miR-29 described here, this studylso demonstrates the possibility of using three different strate-ies to achieve the pre-miRNA purification. In fact, the purificationf pre-miR-29 was accomplished either using an increased NaClr decreased (NH4)2SO4 gradients or using a competition strategyith arginine supplemented buffer. In addition, this work proves

he relevance of the establishment of well-defined binding andlution conditions to enhance the pre-miR-29 purification perfor-ance, resulting in an improvement of the final pre-miRNA yields,

s this could represent an important impact on therapeutic applica-ions of the purified pre-miR-29. Comparing the elution strategiesith NaCl or arginine to elute bound pre-miR-29 biomolecules, itas found that the competition strategy favors the selectivity forre-miR-29 because the arginine concentration needed to promote

lution is lower than the NaCl concentrations. This fact is in accor-ance with what has been previously described by other authors,ecause as arginine is monovalent at the pH used, it is equivalent toaCl in terms of valency; however the conductivity is much lower

med at 1 mL/min by stepwise increasing of arginine concentration in the eluent tof each peak in both experiments is represented in the respective figure. Fractions

injected onto the column.

than for NaCl at identical concentrations [21,49]. This study alsoallowed the purification of pre-miR-29 in 20 min at a flow-rate of1 mL/min.

3.4. Effect of flow rate on pre-miR-29 purification

The effect of flow rate on purification of pre-miR-29 was investi-gated, although no change is expected on the separation selectivityonce some studies proved that the molecules separation is flow-independent due to the physical and chemical constitution ofmonoliths [26,50]. Therefore, to verify the impact of flow rate onpurification of pre-miR-29, the same elution gradient previouslyestablished for the ideal separation was used. Fig. 5A shows theresulting chromatograms for the separation of pre-miR-29 at thedifferent flow rates from 0.5 to 3 mL/min. The purity of pre-miR-29separated with different flow rates was followed by electrophore-sis as shown in Fig. 5B. Evaluating the chromatograms and therespective electrophoresis, it is clearly evident that no changes haveoccurred in the separation efficiency of pre-miR-29 for the differentflow rates under study, but a significant reduction on the chromato-graphic run time was verified when higher flow rates were used(Fig. 5A). It is also possible to observe that the chromatograms over-lap each other even at the highest applied flow rate of 3 mL/min. Inthis case the separation was completed in 9 min, which is in accor-dance with the advantages presented by the monolithic supports,where the chromatographic run was 4 times faster that the timeused in conventional matrix of arginine [21].

3.5. Pre-miR-29 characterization

To ensure the success of the pre-miR-29 purification method-ologies here described for therapeutic applications it is essentialto guarantee the total elimination of impurities such as proteins,assessed by the micro-BCA method and endotoxins evaluatedby LAL assay as well as to isolate the pre-miR-29 from othersRNAs species, which may be assured by polyacrylamide gel elec-trophoresis [5,51]. Moreover, RNA preparations should maintainthe structural integrity, free from enzymatic inhibitors for RT andPCR reactions and free from nucleases [52].

The concentration and quality of the pre-miR-29 preparations

isolated by the agmatine monolith were evaluated by Phoretix1D software (Nonlinear Dynamics, Newcastle, United Kingdom)through the evaluation of the intensity of electrophoretic bands.The quantitative analysis of pre-miR-29 throughout the isolation
Page 8: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

180 P. Pereira et al. / J. Chromatogr. A 1368 (2014) 173–182

F ormed in the same salt and gradient conditions applied for the pre-miR-29 purification( ined in the chromatographic runs at different flow rates were identified by polyacrylamideg

pdotgspiafopobs

cttggs2tuntabatot

Fig. 6. pre-miR-29 identification by reverse-transcription PCR. The agarose-gelelectrophoretic analysis of PCR products shows amplification of pre-miR-29 cDNAfragments. The negative control had no band intensification. Lane M, DNA molec-ular weight marker; lane 1, negative control; lanes 2, 3 and 4, second peaks of the

TQ

TD

ig. 5. (A) Effect of flow rate on resolution of pre-miR-29. Experiments were perfpresented in Fig. 2) at different flow rates (0.5, 1, 2 and 3 mL/min). (B) The peaks obtael electrophoresis.

rocedure is summarized in Table 3, which presents the purityegree, purification factor and recovery yield of pre-miR-29 samplebtained with the agmatine monolith. In a general point of view,he strategy with NaCl enabled a high recovery (97.33%) of the tar-et miRNA but the purity degree only reached 75.21%, whereas thetrategy with (NH4)2SO4 allows the recovery of the partially purifyre-miR-29 (35.51% of purity) when the salt concentration is signif-

cantly reduced. On the other hand, the competition studies usingrginine in the elution buffer, demonstrated the possibility of puri-ying pre-miR-29 with higher purity (90.11% of purity and 94.88%f yield) in comparison to the other strategies, even though somere-miRNA is lost in the final elution step. Thus, if larger amountsf pure pre-miRNA are desired, the strategy that employs NaCl cane used, while if higher purity levels are necessary, the competitiontrategy with arginine is more appropriate [21].

The protein quantification in the purified pre-miR-29 fractionsollected from the chromatographic peaks was performed usinghe micro-BCA assay (Table 3). The analysis of the protein con-ent in the fractions collected along the pre-miR-29 purificationradient revealed that proteins tend to elute more in the firstradient step (data not shown). Therefore, the chromatographictrategies used in these new pre-miRNA isolation methods (peak

in Figs. 2A, 3A and 4A) also provide a reduction of protein con-amination in RNA samples, specifically in the competition strategysing arginine in the elution buffer, where the protein contami-ation level was negligible (2.774 ± 0.005 ng/�L), as required forherapeutic applications (Table 3). Endotoxins contamination wasssessed using the ToxinSensorTM Chromogenic Limulus Amoe-ocyte Lysate (LAL) Endotoxin Assay Kit (GenScript, USA, Inc.)

ccording to the manufacturer’s instructions. The endotoxins con-ent in the final pre-miR-29 sample indicates a significant reductionf the endotoxins level throughout the chromatographic step withhe agmatine monolith, with a purification factor of pre-miR-29

able 3uantitative analysis of purity and recovery yield of pre-miR-29 isolated by agmatine mo

Elution strategy sRNA sample (�g) Pre-miR-29 pu

NaCl 20 75.21

(NH4)2SO4 20 35.51

Arginine 20 90.11

he correlation coefficients of protein calibration curves was 0.9998.ata are presented as means with SD (N = 3).

three strategies under study (sodium chloride, ammonium sulfate and arginine),respectively.

relatively to LPS of 50 times (data not shown). A more accurateidentification of pre-miR-29 molecules purified from the sRNApopulations with the agmatine monolith was performed by PCR.As it can be observed from the electrophoretic analysis of the PCR

products (Fig. 6), by using specific primers for pre-miR-29 cDNA,the PCR reaction allowed the amplification of pre-miR-29. No fur-ther amplification was seen in the control (lane 1). The negative

nolithic disk.

rity (%) Pre-miR-29 yield (%) Proteins (ng/�L)

97.33 3.258 ± 0.00199.40 5.516 ± 0.00694.88 2.774 ± 0.005

Page 9: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

atogr.

cfo

4

poMt8KpaTpcaopceerWaam2actteut

A

SBPp“(aaTvtla

R

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

P. Pereira et al. / J. Chrom

ontrol was made of PCR reaction solutions without cDNA. There-ore, it was verified the identity of the sRNA isolated from theriginal population as pre-miR-29.

. Conclusions

This work combines, for the first time, the structural versatilityrovided by monolithic supports with the specificity and selectivityf agmatine ligand, as a promising strategy for miRNA purification.oreover, the characterization of the modified monolith revealed

hat a maximum binding capacity at 100% breakthrough was.08 mg/mL for a RNA concentration of 0.25 mg/mL. The obtainedd value, 2.6 × 10−7 M, confirms that the agmatine monolith sup-ort develops a good affinity interaction with RNA, showing thatgmatine monolith is a good affinity chromatographic support.he implementation of agmatine monolithic chromatography forre-miR-29 purification was based on the development of spe-ific interactions between the pre-miR-29 and agmatine ligands,llowing the removal of other RNA species, as well as the reductionf proteins and endotoxins contaminants, obtaining highly purere-miR-29 for therapeutic applications. The type and salt con-entration and pH manipulation in the loading buffer allows thestablishment of different interactions and consequently differentlution strategies. The exploitation of these affinity interactions,esulting from multiple intermolecular forces, namely van der

aals forces, hydrogen bonds, electrostatic and hydrophobic inter-ctions, can trigger new insights not only in isolation strategies butlso in many other RNA research fields owing to its implication inolecular recognition phenomena. Additionally, for the pre-miR-

9 purification this monolithic support represents an advantageouslternative to conventional supports due to fast separation andonsequent short contact time, ensuring structural stability of thearget molecule. In conclusion, our approach revealed an efficientechnique to obtain pharmaceutical-grade miRNA with high recov-ry yield, purity and good integrity, which may in a near future besed in RNA structural and functional studies as well as in geneherapy.

cknowledgments

This work was supported by the Portuguese Foundation forcience and Technology (PTDC/EBB-BIO/114320/2009, EXPL/BBB-IO/1056/2012, COMPETE: FCOMP-01-0124-FEDER-027560 andest-C/SAU/UI0709/2011). The authors also acknowledge theroject with Reference CENTRO-07-ST24-FEDER-002014, fromPrograma Operacional Regional do Centro 2007-2013 QREN”“Mais Centro” program). Patrícia Pereira and Ângela Sousacknowledge fellowships (Ph.D. Grant, Ref SFRH/BD/81914/2011nd Postdoctoral Grant, Ref SFRH/BPD/79106/2011, respectively).he authors would like to thank Prof. Yo Kikuchi (Toyohashi Uni-ersity of Technology) for kindly provide pBHSR1-RM plasmid ando BIA Separations for having kindly provided the agmatine mono-ithic disk and U. Cernigoj for having made the immobilization ofgmatine ligand.

eferences

[1] D.P. Bartel, MicroRNAs: genomics, biogenesis, mechanism, and function, Cell116 (2004) 281–297.

[2] J.A. Broderick, P.D. Zamore, MicroRNA therapeutics, Gene Ther. 18 (2011)1104–1110.

[3] I. Vomelova, Z. Vanickova, A. Sedo, Methods of RNA purification. All ways

(should) lead to Rome, Folia Biol. (Praha) 55 (2009) 243–251.

[4] S.C. Tan, B.C. Yiap, DNA RNA, and protein extraction: the past and the present,J. Biomed. Biotechnol. 2009 (2009) 574398.

[5] F. Re, RNA Methodologies – Laboratory Guide for Isolation and Characterization,Academic Press, York, USA, 2005.

[

A 1368 (2014) 173–182 181

[6] M.A. Dineva, L. MahiLum-Tapay, H. Lee, Sample preparation: a challenge in thedevelopment of point-of-care nucleic acid-based assays for resource-limitedsettings, Analyst 132 (2007) 1193–1199.

[7] L.E. Easton, Y. Shibata, P.J. Lukavsky, Rapid, nondenaturing RNA purificationusing weak anion-exchange fast performance liquid chromatography, RNA 16(2010) 647–653.

[8] N. Nonne, M. Ameyar-Zazoua, M. Souidi, A. Harel-Bellan, Tandem affinity purifi-cation of miRNA target mRNAs (TAP-Tar), Nucleic Acids Res. 38 (2010) e20.

[9] S.M. McCarthy, M. Gilar, J. Gebler, Reversed-phase ion-pair liquid chromatog-raphy analysis and purification of small interfering RNA, Anal. Biochem. 390(2009) 181–188.

10] J.S. Kieft, R.T. Batey, A general method for rapid and nondenaturing purificationof RNAs, RNA 10 (2004) 988–995.

11] R. Martins, J.A. Queiroz, F. Sousa, Ribonucleic acid purification, J. Chromatogr.A 1355 (2014) 1–14.

12] L. Ponchon, F. Dardel, Large scale expression and purification of recombinantRNA in Escherichia coli, Methods 54 (2011) 267–273.

13] S.C. Walker, F.H. Scott, C. Srisawat, D.R. Engelke, RNA affinity tags for the rapidpurification and investigation of RNAs and RNA–protein complexes, MethodsMol. Biol. 488 (2008) 23–40.

14] J. Chen, J. Lozach, E.W. Garcia, B. Barnes, S. Luo, I. Mikoulitch, L. Zhou, G. Schroth,J.B. Fan, Highly sensitive and specific microRNA expression profiling usingBeadArray technology, Nucleic Acids Res. 36 (2008) e87.

15] R. Martins, J.A. Queiroz, F. Sousa, Histidine affinity chromatography-basedmethodology for the simultaneous isolation of Escherichia coli small and ribo-somal RNA, Biomed. Chromatogr. 26 (2011) 781–788.

16] R. Martins, J.A. Queiroz, F. Sousa, A new affinity approach to isolateEscherichia coli 6S RNA with histidine-chromatography, J. Mol. Recognit. 23(2010) 519–524.

17] R. Martins, C.J. Maia, J.A. Queiroz, F. Sousa, A new strategy for RNA isolation fromeukaryotic cells using arginine affinity chromatography, J. Sep. Sci. 35 (2012)3217–3226.

18] F. Sousa, D.M. Prazeres, J.A. Queiroz, Affinity chromatography approaches toovercome the challenges of purifying plasmid DNA, Trends Biotechnol. 26(2008) 518–525.

19] P. Pereira, A. Sousa, J. Queiroz, A. Figueiras, F. Sousa, New approach for purifi-cation of pre-miR-29 using lysine-affinity chromatography, J. Chromatogr. A1331 (2014) 129–132.

20] A. Afonso, P. Pereira, J.A. Queiroz, A. Sousa, F. Sousa, Purification of pre-miR-29by a new O-phospho-l-tyrosine affinity chromatographic strategy optimizedusing design of experiments, J. Chromatogr. A 1343 (2014) 119–127.

21] P. Pereira, A. Sousa, J. Queiroz, I. Correia, A. Figueiras, F. Sousa, Purificationof pre-miR-29 by arginine-affinity chromatography, J. Chromatogr. B: Analyt.Technol. Biomed. Life Sci. 951–952 (2014) 16–23.

22] A. Strancar, A. Podgornik, M. Barut, R. Necina, Short monolithic columns asstationary phases for biochromatography, Adv. Biochem. Eng. Biotechnol. 76(2002) 49–85.

23] A. Podgornik, S. Yamamoto, M. Peterka, N.L. Krajnc, Fast separation of largebiomolecules using short monolithic columns, J. Chromatogr. B: Analyt. Tech-nol. Biomed. Life Sci. 927 (2013) 80–89.

24] A. Sousa, F. Sousa, J.A. Queiroz, Advances in chromatographic supportsfor pharmaceutical-grade plasmid DNA purification, J. Sep. Sci. 35 (2012)3046–3058.

25] A. Sousa, C.T. Tomaz, F. Sousa, J.A. Queiroz, Successful application of monolithicinnovative technology using a carbonyldiimidazole disk to purify supercoiledplasmid DNA suitable for pharmaceutical applications, J. Chromatogr. A 1218(2011) 8333–8343.

26] A. Sousa, D. Bicho, C.T. Tomaz, F. Sousa, J.A. Queiroz, Performance of a non-grafted monolithic support for purification of supercoiled plasmid DNA, J.Chromatogr. A 1218 (2011) 1701–1706.

27] A. Sousa, A.M. Almeida, U. Cernigoj, F. Sousa, J.A. Queiroz, Histamine mono-lith versatility to purify supercoiled plasmid deoxyribonucleic acid fromEscherichia coli lysate, J. Chromatogr. A 1355 (2014) 125–133.

28] D. El-Agamy, M. Sharawy, E. Ammar, Agmatine attenuates silica-induced pul-monary fibrosis, Hum. Exp. Toxicol. 33 (2014) 650–660.

29] N. Kitanaka, J. Kitanaka, F.S. Hall, G.R. Uhl, K. Watabe, H. Kubo, H. Takahashi, K.Tanaka, N. Nishiyama, M. Takemura, Agmatine attenuates methamphetamine-induced hyperlocomotion and stereotyped behavior in mice, Behav. Pharmacol.25 (2014) 158–165.

30] N.R. Kotagale, S. Walke, G.P. Shelkar, D.M. Kokare, M.J. Umekar, B.G. Taksande,Agmatine attenuates nicotine induced conditioned place preference in micethrough modulation of neuropeptide Y system, Behav. Brain Res. 262 (2014)118–124.

31] H. Suzuki, S. Umekage, T. Tanaka, Y. Kikuchi, Artificial RNA aptamer produc-tion by the marine bacterium Rhodovulum sulfidophilum: improvement of theaptamer yield using a mutated transcriptional promoter, J. Biosci. Bioeng. 112(2011) 458–461.

32] A. Jungbauer, O. Kaltenbrunner, Fundamental questions in optimizing ion-exchange chromatography of proteins using computer-aided process design,Biotechnol. Bioeng. 52 (1996) 223–236.

33] A. Soares, J.A. Queiroz, F. Sousa, A. Sousa, Purification of human papillomavirus

16 E6/E7 plasmid deoxyribonucleic acid-based vaccine using an arginine mod-ified monolithic support, J. Chromatogr. A 1320 (2013) 72–79.

34] F. Sousa, T. Matos, D.M. Prazeres, J.A. Queiroz, Specific recognition of super-coiled plasmid DNA in arginine affinity chromatography, Anal. Biochem. 374(2008) 432–434.

Page 10: Pharmaceutical-grade pre-miR-29 purification using an agmatine monolithic support

1 atogr.

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

matogr. A 1188 (2008) 227–233.

82 P. Pereira et al. / J. Chrom

35] M.V. Latronico, D. Catalucci, G. Condorelli, MicroRNA and cardiac pathologies,Physiol. Genomics 34 (2008) 239–242.

36] P.T. Nelson, W.X. Wang, B.W. Rajeev, MicroRNAs (miRNAs) in neurodegenera-tive diseases, Brain Pathol. 18 (2008) 130–138.

37] L. Du, A. Pertsemlidis, Cancer and neurodegenerative disorders: pathogenicconvergence through microRNA regulation, J. Mol. Cell Biol. 3 (2011) 176–180.

38] M.J. Shin, L. Tan, M.H. Jeong, J.H. Kim, W.S. Choe, Monolith-based immobilizedmetal affinity chromatography increases production efficiency for plasmid DNApurification, J. Chromatogr. A 1218 (2011) 5273–5278.

39] A. Jungbauer, Chromatographic media for bioseparation, J. Chromatogr. A 1065(2005) 3–12.

40] R.E. Boto, P. Almeida, J.A. Queiroz, Thiacarbocyanine as ligand in dye-affinitychromatography for protein purification, Biomed. Chromatogr. 22 (2008)278–288.

41] J. Ruta, C. Ravelet, J. Desire, J.L. Decout, E. Peyrin, Covalently bonded DNAaptamer chiral stationary phase for the chromatographic resolution of adeno-sine, Anal. Bioanal. Chem. 390 (2008) 1051–1057.

42] D. Grundemann, C. Hahne, R. Berkels, E. Schomig, Agmatine is efficiently trans-

ported by non-neuronal monoamine transporters extraneuronal monoaminetransporter (EMT) and organic cation transporter 2 (OCT2), J. Pharmacol. Exp.Ther. 304 (2003) 810–817.

43] B.J. Calnan, B. Tidor, S. Biancalana, D. Hudson, A.D. Frankel, Arginine-mediatedRNA recognition: the arginine fork, Science 252 (1991) 1167–1171.

[

[

A 1368 (2014) 173–182

44] T. Janas, J.J. Widmann, R. Knight, M. Yarus, Simple, recurring RNA binding sitesfor l-arginine, RNA 16 (2010) 805–816.

45] F. Jiang, A. Gorin, W. Hu, A. Majumdar, S. Baskerville, W. Xu, A. Elling-ton, D.J. Patel, Anchoring an extended HTLV-1 Rex peptide within an RNAmajor groove containing junctional base triples, Structure 7 (1999) 1461–1472.

46] J.D. Puglisi, J.R. Wyatt, I. Tinoco Jr., Solution conformation of an RNA hairpinloop, Biochemistry 29 (1990) 4215–4226.

47] M. Yarus, J.J. Widmann, R. Knight, RNA–amino acid binding: a stereochemicalera for the genetic code, J. Mol. Evol. 69 (2009) 406–429.

48] R. Mallik, D.S. Hage, Affinity monolith chromatography, J. Sep. Sci. 29 (2006)1686–1704.

49] T. Arakawa, D. Ejima, K. Tsumoto, M. Ishibashi, M. Tokunaga, Improved perfor-mance of column chromatography by arginine: dye-affinity chromatography,Protein Exp. Purif. 52 (2007) 410–414.

50] M.K. Danquah, G.M. Forde, Large-volume methacrylate monolith for plasmidpurification. Process engineering approach to synthesis and application, J. Chro-

51] L.V. Madabusi, G.J. Latham, B.F. Andruss, RNA extraction for arrays, MethodsEnzymol. 411 (2006) 1–14.

52] S.A. Bustin, T. Nolan, Pitfalls of quantitative real-time reverse-transcriptionpolymerase chain reaction, J. Biomol. Tech. 15 (2004) 155–166.