Journal Pre-proof Theranostic application of a novel G-Quadruplex forming DNA aptamer targeting glyoxylate cycle enzyme malate synthase of Mycobacterium tuberculosis Abhijeet Dhiman, Chanchal Kumar, Subodh Kumar Mishra, Kriti Sikri, IsharaDatta, Pradeep Sharma, Tej P. Singh, Sagarika Haldar, Neera Sharma, Anjali Bansal, Yusra Ahmad, Amit Kumar, Tarun Kumar Sharma, Jaya SivaswamiTyagi PII:
S2162-2531(19)30274-4
DOI:
https://doi.org/10.1016/j.omtn.2019.09.026
Reference:
OMTN 646
To appear in:
Molecular Therapy: Nucleic Acid
Received Date: 9 July 2019 Accepted Date: 27 September 2019
Please cite this article as: Dhiman A, Kumar C, Mishra SK, Sikri K, IsharaDatta Sharma P, Singh TP, Haldar S, Sharma N, Bansal A, Ahmad Y, Kumar A, Sharma TK, SivaswamiTyagi J, Theranostic application of a novel G-Quadruplex forming DNA aptamer targeting glyoxylate cycle enzyme malate synthase of Mycobacterium tuberculosis, Molecular Therapy: Nucleic Acid (2019), doi: https:// doi.org/10.1016/j.omtn.2019.09.026. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 The Author(s).
1
Theranostic application of a novel G-Quadruplex forming DNA aptamer targeting glyoxylate
2
cycle enzyme malate synthase of Mycobacterium tuberculosis
3
Abhijeet Dhiman1,8, Chanchal Kumar1, Subodh Kumar Mishra2#, Kriti Sikri1#,IsharaDatta1, Pradeep
4
Sharma3, Tej P. Singh3,Sagarika Haldar4,5, Neera Sharma6, Anjali Bansal7, Yusra Ahmad8, Amit
5
Kumar2, Tarun Kumar Sharma5* and Jaya SivaswamiTyagi1,5*
6
1
Department of Biotechnology, All India Institute of Medical Sciences, New Delhi-110029, India.
7
2
Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol,
8
Indore-453552, India.
9
3
Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India.
10
4
Department of Experimental Medicine and Biotechnology, PGIMER, Sector12, Chandigarh-160012,
11
India.
12
5
13
Institute, Faridabad, Haryana-121001 India.
14
6
Department of Biochemistry, Dr. Ram Manohar Lohia Hospital, New Delhi-110001, India.
15
7
Department of Pediatrics, Dr. Ram Manohar Lohia Hospital, New Delhi-110001, India.
16
8
Faculty of Pharmacy, Uttarakhand Technical University, Dehradun-248007, Uttarakhand, India.
17
Corresponding Authors
18
*Email: Jaya S. Tyagi,
[email protected],
[email protected], Telefax +91 011-26588491;
19
Tarun K. Sharma,
[email protected]; Telephone +91 8800931999.
20
#
Multidisciplinary Clinical and Translational Research, Translational Health Science and Technology
Contributed equally.
21
22
1
23
ABSTRACT
24
The successful management of tuberculosis (TB) requires efficient diagnosis and treatment. Further,
25
the increasing prevalence of drug resistant TB highlights the urgent need to develop novel inhibitors
26
against both drug susceptible and resistant forms of disease. Malate synthase (MS), an enzyme of the
27
glyoxylate pathway, plays a vital role in mycobacterial persistence, and therefore it is considered as an
28
attractive target for novel anti-TB drug development. Recent studies have also ascribed an adhesin
29
function to MS and established it as a potent diagnostic biomarker. In this study a panel of
30
Mycobacterium tuberculosis (Mtb) MS-specific single-stranded DNA aptamers was identified by
31
Systematic Evolution of Ligands by EXponential Enrichment (SELEX). The best-performing G-
32
quadruplex-forming 44-mer aptamer, MS10, was optimized post-SELEX to generate an 11-mer
33
aptamer, MS10-Trunc. This aptamer was characterized by various biochemical, biophysical and in-
34
silico techniques. Its theranostic activity towards Mtb was established using enzyme inhibition, host
35
cell binding and invasion assays.MS10-Trunc aptamer exhibited high affinity for MS (Kd ~19 pM) and
36
displayed robust inhibition of MS enzyme activity with IC50 of 251.1nM and Ki of 230 nM. This
37
aptamer blocked mycobacterial entry into host cells by binding to surface-associated MS. In addition,
38
we have also demonstrated its application in the detection of tuberculous meningitis (TBM) in patients
39
with sensitivity and specificity each of >97%.
40 41
42
2
43
INTRODUCTION
44
Active tuberculosis (TB) affected more than 10.4 million people in 2017, while an estimated 1.7
45
billion individuals (~one quarter of the world’s population) are estimated to harbor a latent/ persistent
46
infection with Mycobacterium tuberculosis (Mtb).1 The current treatment for TB is prolonged and
47
drug-resistant cases are increasing2, which together pose a significant threat to TB control in the
48
community.3,4 Therefore, there is an urgent need to develop new inhibitory compounds or molecules
49
that have the potential to work against both the drug susceptible and resistant TB. Metabolic pathways
50
of central metabolism have attracted attention in recent times as a source of new targets for novel TB
51
drug development.
52
The glyoxylate shunt pathway in particular is considered as a prominent target pathway owing to its
53
role in mycobacterial survival and persistence, as demonstrated in cell and animal models of Mtb
54
infection.5,6 This two-enzyme pathway enables bypass of the decarboxylation steps in the tricarboxylic
55
acid cycle and conserves carbon for subsequent gluconeogenesis. Isocitratelyase, the first enzyme of
56
this pathway, generates glyoxylate and succinate from isocitrate, and malate synthase (MS), the
57
second enzyme, catalyzes the formation of malate using glyoxylate and acetyl-CoA.5 Notably, these
58
enzymes appear to be absent in mammals5, a feature which enhances its relevance as an anti-TB drug
59
target. In addition to its enzymatic activity, MS was shown to be expressed on the cell wall of Mtb and
60
impart host adhering function to bacteria, thus establishing an additional role for it as an adhesin.7
61
Furthermore, MS is reported as a potential biomarker for TB infections as it is expressed in the early
62
stage of infection and independent of HIV co-infection.8 Thus, the presence of this antigen in
63
biological fluid is considered as a direct evidence of infection.
64
The crystal structures of both isocitratelyase and MS have been solved
65
structures have suggested MS as a more druggable target in comparison to isocitrate lyase.5 A recent
66
study demonstrated that knockdown and knock-out of MS leads to Mtb clearance inthe mouse model 3
9,10
and insights from these
67
of infection, showing the essentiality of MS for the survival of Mtb during the acute and chronic
68
phases of infection.11 Therefore, owing to the attractiveness of MS as a drug target, inhibitors of MS
69
have been reported, such as oxalate, phosphoenol pyruvate (PEP), bromopyruvate (BP) and glycolate,
70
that exhibit high inhibition constants (Ki) ranging between 60 µM and 900 µM.10 These inhibitors have
71
limitations in terms of their use in humans; oxalate is not pharmacologically suitable due to its toxic
72
nature, PEP is a key constituent of glycolysis and gluconeogenesis pathways in all organisms
73
including humans, while BP exhibits toxicity impairing mitochondrial function.12,13 Recently a series
74
of phenyl-diketo-acid (PDKA) molecules having inhibitory activity against Mtb MS enzyme were also
75
developed which are under optimization.14,15
76
A recent study from our group has established the clinical utility of polyclonal antibody to MS for the
77
diagnosis of tuberculous meningitis (TBM), the most lethal form of tuberculosis that affects central
78
nervous system.8 However, polyclonal antibody has an inherent problem of batch-to-batch variation,
79
thus it is difficult to scale up the assay to a diagnostic test level. To address this limitation associated
80
with antibody, it is imperative to develop a diagnostic reagent of uniform quality. Nucleic acid
81
aptamers have received a great deal of attention due to their enormous potential to be used as both
82
therapeutic and diagnostic agents. By acquiring a complex 2-D and 3-D structure, DNA aptamers
83
often recognize their cognate target with unmatched affinity and specificity.16
84
Here we report the development of a G-quadruplex (G4) forming aptamer MS10-Trunc against MS of
85
Mtb that can serve as a novel theranostic molecule for the management of TB.
86
4
87
RESULTS
88
In the current study, high-affinity MS-specific single-stranded (ss) DNA aptamers were identified
89
from an 80-mer random DNA library (RDL) using SELEX-based screening. The best-performing
90
aptamer candidate, MS10, was further optimized post-SELEX to generate MS10-Trunc.The
91
theranostic potential of MS10-Trunc was established by its inhibitory activity against MS enzyme,
92
interference of Mtb binding and invasion into host cells and ability to detect MS in CSF specimens for
93
the diagnosis of TBM.
94
95
Aptamer selection and screening for MS aptamers
96
Mtb MS was expressed in E.coli BL21(DE3)pLysS and purified through affinity chromatography (Ni-
97
NTA) and used as a target to generate MS-specific aptamers by the process of SELEX (Figure 1). The
98
SELEX process included a step of negative selection to exclude nitrocellulose membrane (NCM)
99
binders (step a), followed by partitioning of unbound sequences (step b) and a positive selection step
100
to obtain binders to MS coated on NCM (step c). The bound DNA was eluted and amplified (step d)
101
followed by the generation of ssDNA for the next round of SELEX. After ten iterative rounds of
102
SELEX, the archived aptamer populations from rounds 2, 4, 6, 7, 8, 9 and 10 were assessed for
103
binding to MS using Aptamer Linked Immobilized Sorbent Assay (ALISA). The enrichment of MS-
104
binding aptamers was evident over consecutive rounds of SELEX (Figure 2A). However, the aptamer
105
population of rounds 8, 9 and 10 evinced the highest binding over the random DNA library (RDL).
106
Interestingly, the round 9 aptamer population showed higher binding to MS compared to round 10
107
aptamer pool (Figure 2A). The reason for this could be the improper partitioning between specific and
108
non-specific MS binders during the 10th round of selection process or a decrease in the aptamer pool
109
complexity or a combination of both.17 The aptamers of rounds R8, R9 and R10 were individually 5
110
cloned in the TA cloning vector pTZ57R/T and clones were randomly picked and sequenced (Figure 1,
111
step e). Based on the sequencing results, representative aptamer candidates from each round were
112
subjected to CLUSTAL-W and BioEdit analysis to identify the primary sequence homology and
113
nucleotide richness (Figure S1). Based on the nucleotide richness, all the 22 aptamer candidates were
114
categorized into 2 groups G-rich and Non-G rich (Figure S1). Interestingly ~50% aptamer candidates
115
were G-rich with high G+C content (Figure S2A and S2B), suggesting the preferential binding of MS
116
to G-rich DNA aptamers. These 22 monoclonal aptamer candidates were first screened for their
117
binding to MS by ALISA (Figure 2B) and 5 aptamers, namely, MS4, MS5, MS6, MS10 and MS20
118
were selected for further study using an arbitrary cut-off of ∆O.D.450nm ≥0.6. Among them, MS4 and
119
MS20 were derived from the R8 pool, MS5 and MS6 belonged to the R9 pool while MS10 was
120
obtained from the R10 aptamer population.
121
In-silico secondary structure prediction of aptamers
122
To gain structural insights into aptamers, their secondary structure was predicted by a web-based tool,
123
RNA fold, using DNA parameters (http://rna.tbi.univie.ac.at/cgi-bin/RNAWebSuite/RNAfold.cgi).
124
MS4 and MS20 acquire a typical stem-loop structure while MS5, MS6 and MS10 form only a loop-
125
like structure (Figure S3). The selected aptamer candidates belong to the G-rich category that usually
126
have propensity to form G4 structures. Analysis of the sequences using an in-silico tool QGRS
127
Mapper
128
(http://bsbe.iiti.ac.in/bsbe/ipdb/index.php), predicted that all the queried aptamers, namely, MS4, MS5,
129
MS6, MS10 and MS20 form G4 structure and show scores comparable or higher than the known G4
130
forming aptamer sequences (anti-thrombin, anti-HIV integrase and VEGF-aptamers)18-20 (Table S1).
131
Further to validate in-silico the predictions, CD spectroscopy analysis was undertaken which revealed
132
that all the aptamers form G4structures. Each aptamer shows a negative peak near 242 nm and a
(http://bioinformatics.ramapo.edu/QGRS/index.php)
6
and
G4IPDB
133
positive peak close to 265 nm, which typically signify a parallel G4 structure formation by all of these
134
aptamers 21-23(Figure 2C).
135
Determination of dissociation constant (Kd) of aptamers by SPR
136
Purified MS was amine-coupled onto a CM5 chip through EDC-NHS chemistry, followed by injection
137
of the individual aptamers, to obtain the dissociation equilibrium constant (Kd) values for each aptamer.
138
All the aptamers exhibited tight binding with Kd in low micro to nanomolar range; the aptamers were
139
ranked in terms of decreasing affinity (Kds) as MS6 (0.28 nM) >MS4 (9.9 nM) >MS10 (12 nM) >
140
MS20 (0.91 µM) > MS5 (2.7 µM) (Table 1 and Figure S4).
141
Inhibition of MS enzyme activity by aptamers
142
Purified MS protein was pre-incubated with selected aptamers (namely MS4, MS5, MS6, MS10 and
143
MS20) for 30 min followed by the addition of glyoxylate and acetylCoA to complete the reaction. The
144
inhibition of MS enzyme activity by various aptamers was monitored by estimating free CoA as
145
described in Methods.10 Based on the extent of inhibition of MS activity, the aptamers were ranked as
146
MS10 >MS5 >MS20 >MS4 >MS6. MS10 was determined to be the most potent inhibitor; it evinced
147
inhibitory activity of ~91% against purified MS protein (Figure 3A). A control aptamer of the same
148
length (C1, Table S2) showed negligible inhibition of MS activity and suggested that MS10 aptamer-
149
mediated inhibition was sequence-specific.
150
Among all the tested aptamers, MS10 aptamer exhibited maximum inhibitory activity against MS.
151
Therefore, MS10 aptamer was taken up for further studies.
152
Post-SELEX optimization of MS10 aptamer
153
In the literature, numerous G4 forming aptamers are reported as potent inhibitors of biological
154
activity.24 MS10 was a G-rich aptamer and to establish its structure-activity relationship (SAR), its G4 7
155
forming sequence that was identified through QGRS mapper (Figure S5), was synthesized and
156
designated as MS10-Trunc (5’- GGTGGTGGTGG-3’) and assessed for its ability to bind MS using
157
ALISA. The 11-mer MS10-Trunc displayed three GGT repeats and exhibited similar binding to MS
158
compared to its 44-mer parent aptamer MS10 (Figure 3B), thus representing the smallest functional
159
unit of the parent aptamer. To establish the role of each ‘T’ nucleotide present in MS10-Trunc, the ‘T’
160
nucleotides were mutated by deleting one ‘T’ nucleotide at a time (Table S3) and the binding of
161
individual mutant aptamers (∆T3, ∆T6 and ∆T9) to MS was assessed by ALISA. In terms of aptamer
162
binding to MS, the deletion of T3 or T9 was well tolerated, whereas the deletion of T6 led to ~28 %
163
reduction in aptamer binding compared to MS10-Trunc (Figure 3B), indicating a crucial role for T6 in
164
maintaining the stable G4 aptamer structure. To further evaluate the impact of truncation on aptamer
165
binding, the Kd of MS10-Trunc was determined using SPR to be 19 pM (Figure S4), indicating a ~631
166
fold improvement in affinity in comparison to the parent aptamer, MS10 (Kd= 12 nM, Table 1).
167
Determination of inhibition kinetics of MS10 and its truncated derivative, MS10-Trunc
168
The 11-mer MS10-Trunc aptamer exhibited a similar extent of inhibitory activity (~94%) as compared
169
to the parent aptamer MS10 (~91%, Figure 3C). A comparative inhibition study was performed for
170
MS10 and MS10-Trunc, along with the known MS inhibitors, phosphoenolpyruvate (PEP) and
171
Bromopyruvate (BP).10 The IC50 of MS10, MS10-Trunc were determined to be 275.6 nM and 251.1
172
nM whereas for BP and PEP it was 114.3 µM and 374.1 µM, respectively (Figure 4A), indicating that
173
MS10-Trunc was more potent than its parent aptamer, MS10, as well as the known inhibitors, BP and
174
PEP. While the IC50 value of an inhibitor is an indicator of its functional strength (such as inhibiting
175
enzyme activity), the Ki value (the equilibrium dissociation constant of the inhibitor) is more reflective
176
of its binding affinity for the target. We further determined the Ki value for MS using linear replots of
177
Lineweaver-Burk slopes using a range of aptamer and substrate concentrations. MS10 and MS10-
178
Trunc showed inhibition constant of 270 nM and 230 nM, respectively (Figure 4B and 4C), which 8
179
further supports their robust inhibitory potential. PEP and BP are reported to exhibit Ki ~200 µM and
180
~60 µM, respectively.10 The parent aptamer exhibited ~740 and ~220 fold more potency than PEP and
181
BP whereas MS10-Trunc displayed ~870 and ~260 fold more potency than PEP and BP.
182
We further determined the inhibition mechanism of the MS10 and MS10-Trunc to evaluate the mode
183
of aptamer binding to MS that leads to such robust inhibition. There was no significant change in
184
inhibition of enzyme activity when the order of addition of aptamer or substrate to MS protein was
185
reversed. Both MS10 and MS10-Trunc inhibit MS by mixed inhibition mechanism, indicating that
186
they can inhibit catalysis regardless of whether the substrate is bound to the enzyme (Figure 4B and
187
4C).
188
Inhibition of Mycobacterium tuberculosis invasion in cells
189
Since MS is also expressed on the cell surface of Mtb and exhibits adhesin activity that contributes to
190
bacterial invasion into host cells7, we assessed the interaction of MS10-Trunc with cell surface-
191
associated MS. Flow cytometry analysis (Figure 5A) clearly demonstrated preferential binding of
192
MS10-Trunc aptamer with Mtb as compared to the control oligo C2, indicating that the former
193
aptamer interacted with cell surface-associated MS. We next assessed the ability of MS10-Trunc to
194
modulate invasion/ infection of Mtb H37Rv into human THP-1 monocytic cells. A ~40% decrease in
195
Mtb infectivity was observed in the presence of MS10-Trunc (Figure 5B; ***P <0.0001) and no such
196
inhibition was observed with 44-mer control oligos (C1 and C2), suggesting that inhibition is aptamer-
197
specific and Mtb utilizes a MS-dependent mechanism, at least in part, for host cell entry. Taken
198
together these findings demonstrate the potential application of aptamer-based inhibitors as a TB
199
therapeutic agent.
200
Diagnostic application of MS10-Trunc
9
201
The diagnostic potential of MS10-Trunc aptamer to detect MS in CSF samples was assessed next in 86
202
archived CSF specimens belonging to TBM and disease control categories. A detailed description
203
about ‘Classification of CSF samples’ is given in Supplementary Methods Section. To determine
204
diagnostic accuracy of MS10-Trunc aptamer, a ROC curve was constructed first using ∆OD450nm
205
values obtained with CSF from Definite TB (True positive) and NTIM group (True negative) samples
206
and a scatter plot was constructed for the two sample categories. The area under the curve was 1.0
207
which indicated the robustness of the assay (Figure S6A and S6B). The NTIM category comprised of
208
cases of pyogenic bacterial meningitis that included 2 culture confirmed cases of E. coli (n=1), and
209
Acinetobacter sp (n=1). The remaining 14 cases were diagnosed on the basis of response to
210
appropriate antibiotics, clinical presentation and symptoms. Based on the cut-off derived from the
211
ROC curve (0.3444), the performance of MS10-Trunc aptamer ALISA was evaluated (Figure 6). The
212
aptamer MS10-Trunc yielded a very high sensitivity and specificity (> 97%) with a highly significant
213
discrimination (***p<0.0001) between CSF from TBM and non-TBM subjects was observed. This is a
214
highly encouraging finding and requires substantiation in a larger study on geographically distinct
215
populations so that this lead can be translated in to a commercially viable diagnostic assay.
216
Structural insights into MS10-Trunc
217
Secondary structure analysis of MS10-Trunc, the most potent inhibitor among the molecules assessed
218
in the study, was predicted to have only a loop configuration, albeit a smaller one, as compared to
219
MS10, its parent aptamer, suggesting that the loop is sufficient for aptamer interaction with MS
220
(Figure S5). A recent report also suggests that the loop region is sufficient for interaction of aptamer
221
to human erythropoietin αwith its cognate target.25 CD spectra analysis revealed that MS10-Trunc
222
aptamer, like its parent aptamer (Figure 2C), exhibited a negative peak at 242 nm and a positive peak
223
at 265 nm (Figure 7A), that is typical of parallel-type G4 structure.21,22 This G4 structure was dynamic
224
and underwent a structural change in the presence of MS in an aptamer concentration-dependent 10
225
manner, as seen by changes in ellipticity (Figure 7A). The formation of G4 structure of MS10-Trunc
226
aptamer was confirmed by 1H NMR analysis. The formation of a G4 structure byMS10-Trunc aptamer
227
was confirmed by 1H NMR spectrum analysis; the aptamer exhibited a set of merged imino proton
228
resonances at 10-12 ppm with broad line widths, clearly demonstrating the formation of a predominant
229
G4 fold 26 (Figure 7B). The NMR melting temperature experiment further confirms thermostability of
230
the DNA aptamer and showing the existence of imino signals at the higher temperature (upto 65ºC).
231
These NMR findings are consistent with in-silico prediction of G4 structure by QGRS-Mapper and the
232
findings of CD spectral analysis.
233
Molecular modeling and in-silico docking studies
234
Towards identifying the binding location of MS10-Trunc on MS in accordance to structural insights
235
obtained from CD and NMR analysis, we generated a parallel inter-stranded G4 structure model of
236
MS10-Trunc aptamer (Figure 8A). This energetically refined model contains 4 chains (O, P, Q, R
237
displayed in different color code) of identical length (11mer) and it was selected as the input ligand for
238
docking studies using the crystal structure of Mtb MS (PDB ID: 2GQ3, Figure 8). The analysis of the
239
docked structure predicted interactions of MS10-Trunc aptamer with Thr528, Lys484, Thr482, and
240
Gln486 residues of MS protein (Figure 9). The G10 nucleotide of R chain forms 2 hydrogen bonds by
241
interacting with Thr482 and Gln486 residues of MS, G5 nucleotide of Q chain forms one hydrogen
242
bond with Thr528, and G7 of O chain forms one hydrogen bond with Lys484. All these residues map
243
in the TIM barrel domain of MS which forms the active site of MS at the interface of TIM barrel
244
residues (115-134 and 266-557), residues of domain II (591-727) and residues of a loop region (616-
245
633).10
246
247 11
248
DISCUSSION
249
There is a need for versatile reagents with theranostic activity that can both detect and treat
250
tuberculosis. The selection of a unique target that can meet both diagnostic and theranostic criteria
251
underlies the successful design of a useful theranostic agent. MS is the second and terminal enzyme of
252
the glyoxylate cycle that is essential for sustaining a persistent TB infection 10 of both drug susceptible
253
and drug resistant forms of TB. This enzyme is absent in humans which makes MS an attractive TB
254
drug target.15,27 Furthermore, in recent years its role as a biomarker for detecting active TB was also
255
established.7,8 Thus, taking together these attributes, MS can be considered as a potent and novel
256
theranostic target for the management of TB.
257
In recent years, nucleic acid aptamers have gained significant attention from the scientific community
258
as promising inhibitors and drug candidates due to their rapid synthesis, high stability, non-toxic
259
nature, ease in optimization and functionalization and ability to control pharmacokinetic and
260
pharmacodynamic parameters.28-30 We have developed a panel of parallel G4 structure forming
261
ssDNA aptamers that showed high-affinity binding and robust inhibition of MS catalytic activity using
262
SELEX technology. An 11mer G4 structure forming aptamer, MS10-Trunc, having high performance
263
characteristics, was developed by post-SELEX optimization of its parent aptamer, MS10. Post-SELEX
264
optimization led to ~630-fold improvement in Kd of the 11-mer for MS over its full-length 44-mer
265
counterpart. This finding is consistent with previously published reports where an improvement in
266
binding affinity is reported on aptamer truncation.31 Further, MS10-Trunc demonstrated a high
267
inhibitory potential, with low IC50 (230 nM) that was comparable to that of its parent aptamer MS10
268
(270 nM). IC50 data unequivocally demonstrated that the inhibitory activity of MS10-Trunc is
269
considerably superior (~1000 fold) to that of the known MS inhibitors, BP and PEP10 and comparable
270
to that of recently reported PDKA inhibitors which had IC50 values in the range of 0.01 µM to 0.6
271
µM.14 There are reports that G4 forming structures allow aptamers to interact strongly with their 12
18
cognate targets, such as anti-thrombin aptamer
273
aptamer.30 More recently, G4 forming inhibitory DNA aptamers against Mtb polyphosphate kinase-2
274
were also reported.4 A truncated aptamer with similar length (11-mer) is previously reported by Nadal
275
et al. (2013) for detecting ß-conglutin, an allergen.33 The high affinity binding properties of MS10-
276
Trunc to MS can be ascribed to its ability to form an 11-mer G4 structure (MS10-Trunc) that was
277
confirmed by CD spectroscopy analysis and 1H NMR studies.
278
In-silico modeling and docking studies have provided useful insights into the molecular interactions
279
between MS10-Trunc aptamer and its cognate target, MS that may contribute to its theranostic activity.
280
In-silico generated parallel G4 structure of MS10-Trunc was predicted to have 4-stranded structure
281
containing R, O, Q and P chains. This observation is in agreement in the previous reports where a 6-
282
mer anti-HIV DNA molecule forms a similar parallel G4 structure involving four chains.34 G-
283
nucleotides mapped to the R, O and Q chains of G4 structure of MS10-Trunc were predicted to have
284
H-bonding interactions with 4 amino acid residues (Thr482, Thr528, Lys484 and Gln486) that are
285
located in the TIM barrel domain of MS. Since the TIM barrel domain forms the active site interface
286
by interacting with the residues of domain II and loop region (616-663), therefore binding of MS10-
287
Trunc aptamer to residues of the TIM barrel domain may possibly lead to a conformational change
288
in/near the active site of MS that adversely influences catalytic function of MS and results in an
289
inhibition of enzymatic activity of MS in presence of MS10-Trunc. It is also established that MS acts
290
as an adhesin and is known to play a vital role in bacterial invasion into host cells.7 Our findings
291
clearly demonstrate that MS10-Trunc aptamer blocks bacterial invasion by ~40%. In addition to MS,
292
Mtb is also known to use other adhesins, including but not limited to HupB, PstS-1 and LpqH (19-kDa
293
antigen)35, during host cell invasion. A previous study has also reported aptamer-mediated inhibition
294
of Mtb entry into murine macrophages although the identity of the aptamer target was not known.36
13
, nucleolin-targeting aptamer
32
272
and anti-VEGF
295
Therefore, to achieve more robust inhibition of bacterial invasion, a combinatorial approach (a cocktail
296
of aptamers targeting various adhesins of Mtb) may possibly be used.
297
This study offers proof of concept for a TB theranostic reagent that is directed against MS. Our
298
findings indicate that MS10-Trunc aptamer has the potential to target Mtb infection. However, the
299
stability and delivery of aptamers into mycobacteria poses a challenge for their therapeutic application.
300
Although G4 based aptamers are relatively more stable in comparison to their non-G4 counterparts,
301
their stability may further be improved by chemical modification strategies.37,38 Efforts are ongoing in
302
our laboratory to suitably modify the aptamer using various functionalization and conjugation
303
strategies to achieve high stability and efficient delivery of MS10-Trunc aptamer into Mtb.
304
The future challenges relating to aptamer delivery in animal model or humans can possibly be
305
addressed by administration of aptamer in the form of an inhaled aerosol mist or by injecting the
306
aptamer into the blood. The delivery of aptamer through the inhalation route might protect the
307
truncated aptamer from the exonucleases in serum while delivery of aptamer as injectables would
308
require suitable modification such as blockage of the 3' end with PEG, modification of the backbone
309
and introduction of modified bases etc. to avoid enzymatic breakdown and rapid clearance by the
310
kidneys and liver.
311
The performance of aptamer-based TBM diagnostic test developed in the current study is superior to
312
various previously reported diagnostic modalities (Table 2). MS10-Trunc aptamer has shown a very
313
high diagnostic accuracy (>97% sensitivity and specificity) with an ability to efficiently discriminate
314
between TBM and non-TBM subjects (p-value of ***p < 0.0001). This aptamer being a synthetic
315
DNA molecule, will evince minimal/no batch-to-batch variation and can easily be adapted on to
316
various biosensing platforms for developing a rapid point of care test for TBM and possibly other
317
forms of TB. 14
318
In conclusion, we have identified a panel of G4 forming ssDNAaptamers that represents a novel class
319
of theranostic molecules against Mtb MS. The theranostic potential of the best-performing and
320
optimized G4 forming aptamer, MS10-Trunc was established through binding studies using ALISA
321
and SPR, enzyme inhibition assay, binding and inhibition of Mtb invasion into host cells and
322
demonstrating its diagnostic utility in TBM. We anticipate that MS10-Trunc holds promise for the
323
development of novel MS-targeted theranostics approach against Mtb and may complement the
324
existing diagnostic and therapeutic modalities in the near future.
325 326
MATERIAL AND METHODS
327
Reagents and Samples
328
All oligonucleotides and other primers utilized in the study were purchased from Integrated DNA
329
Technologies (IDT, USA). Other Routine reagents and chemicals were purchased from Sigma Aldrich,
330
USA. Bacterial expression plasmids of MS (GlcB), MPT51, MPT64, CFP-10 and purified proteins
331
Ag85C, HspX, GroES, LAM, ESAT-6 and culture filtrate proteins (CFPs) were provided by BEI
332
Resources, NIAID, NIH, USA, (please refer to acknowledgement for details).
333
Purification of MS
334
Plasmid pMRLB.8 encoding C-terminal hexa-histidine fusion of Mtb MS (Rv1837c) was used to
335
overexpress recombinant MS protein in E.coli BL21(DE3)pLysS. Bacterial cultures were grown in
336
2×YT medium at 37°C to an O.D595 of ~0.5 followed by an induction with 1 mMisopropyl-1-thio-β-D-
337
galactopyranoside (IPTG) for 5 hours at 25°C. MS was purified from the induced culture by Ni-NTA
338
affinity chromatography as described.8
339
Selection of DNA Aptamer against Mtb MS antigen using SELEX
15
340
A synthetic 80-mer single-stranded (ss) random DNA library (RDL) containing a random region of 44
341
nucleotides flanked by fixed primer binding sequences of 18-mer each that was used in our previous
342
study39 was employed to screen MS specific aptamers. The fixed primer binding sequences allow
343
DNA amplification by PCR using DRF (Forward- 5’ GTC TTG ACT AGT TAC GCC-3’) and DRR
344
(Reverse -5’ GAG GCG CCA ACT GAA TGA-3’) primers. The subtractive nitrocellulose membrane
345
(NCM)-based SELEX approach was used as described. For Round 1, 10 µg of MS was blotted onto
346
the NCM and allowed to dry at room temperature (RT). Mean while the RDL (1500 pmole) was
347
heated at 92°C followed by snap chilling on ice and finally brought to RT. The denatured library was
348
first incubated with NCM in a step of negative selection. The unbound sequence pool was then
349
incubated with pre-dried NCM blotted with MS in SELEX screening buffer [10 mM Tris pH 7.5
350
supplemented with 10 mM MgCl2, 50 mM KCl, 25 mM NaCl, 0.05% Tween-20 (v/v), 0.1 µg/µL yeast
351
tRNA and 0.1µg/µL BSA]. The unbound ssDNA was washed away with selection buffer (SB, 10 mM
352
Tris pH 7.5, 10 mM MgCl2, 50 mM KCl, 25 mM NaCl) supplemented with 0.5% Tween-20. The
353
bound DNA was eluted using nuclease-free water, followed by heating at 92°C for 5 min. The eluted
354
DNA binder population was used as a template for PCR amplification with the forward and reverse
355
primers for an optimized number of cycles of thermal cycling; 94°C for 30 sec, 55°C for 30 sec and
356
72°C for 30 sec with a final extension at 72°C for 5 min. In order to avoid formation of any undesired
357
high molecular weight amplicon, PCR cycle numbers were optimized. Subsequently, ssDNA was
358
prepared using alkaline treatment as described previously39,40 and used in the next round of SELEX.
359
The rigorousness of selection was intensified in every successive round of SELEX by increasing the
360
strength of Tween-20 during washing and by decreasing the interaction time with the target during
361
selection. In order to minimize cross-reactivity, counter-selection was employed by incubating the
362
binder population with other Mtb proteins, namely, purifiedHspX,CFP-10, ESAT-6, Ag85C,LAM,
363
MPT51, GroES and culture filtrate proteins (CFPs). After 10 rounds of selection, the high-affinity 16
364
pools were cloned in pTZ57R/T vector system (Thermo ScientificTMInsTAClone PCR cloning Kit)
365
and transformed in E.coli DH5α. The obtained colonies were randomly picked and subjected to DNA
366
sequencing.
367
Monitoring the rounds of SELEX
368
After ten iterative rounds of SELEX, the archived aptamer populations from rounds R2, R4, R6, R7,
369
R8, R9 andR10 were amplified through PCR using 5’ biotinylated forward primer and rA-containing
370
reverse primer, followed by strand separation using denaturing Urea-PAGE as described previously.39
371
These ssDNA pools of different rounds were checked for their binding to MS by Aptamer Linked
372
Immobilized Sorbent Assay (ALISA). For this, 500 ng of purified MS protein was coated onto a
373
NuncMaxiSorp™ 96 well-plate (Thermo Scientific)overnight at 4°C followed by blocking with 5%
374
Bovine Serum Albumin (BSA) at RT. The biotinylated aptamers pool of the archived population from
375
each round were heated at 92°C for 10 min, cooled on ice and brought to RT. One hundred ng of
376
biotinylated aptamer pool was then added to individual wells pre-coated with MS. Streptavidin-
377
Peroxidase Polymer Ultrasensitive (Sigma) was then added at1:2000 dilution followed by washing
378
with SB to remove unbound streptavidin-HRP conjugate and finally TMB (3, 3’, 5, 5’
379
Tetramethylbenzidine) (BD OptEIA™), was added. The reaction was stopped by 5% sulfuric acid
380
followed by absorbance measurement at 450 nm.
381
Screening of Aptamers via Aptamer Linked Immobilized Sorbent Assay (ALISA)
382
Briefly, 500 ng of MS was coated in 100 µL (0.1 mol/L) of sodium bicarbonate buffer (pH 9.6) on a
383
96 well-plate and incubated at 4°C overnight. The wells were blocked with 5% BSA solution for 2 hr
384
at RT. The rest of the ALISA procedure was followed as described previously.39
385
17
386
Circular dichroism (CD) spectra analysis
387
The CD spectra was obtained on Jasco J-815 Spectropolarimeter (Jasco Hachioji, Tokyo, Japan)
388
equipped with Peltier junction temperature controller with constant supply of nitrogen gas just to avoid
389
water condensation around the cuvette. The path length of the used cuvette was 0.2 cm, whereas,
390
spectral interval of each scan was set as 0.1 nm and the scanning rate was fixed at 20 nm/min. A blank
391
spectrum was recorded with SB only and subtracted from the CD spectrum of sample containing DNA
392
aptamer or DNA aptamer +MS. In each case, three scans were recorded in a range of 200-340 nm, and
393
the average of three scans was plotted to examine the target-dependent change in aptamer structure.
394 395
Surface plasmon resonance (SPR) analysis of aptamer interactions with MS.
396
The binding affinity of ssDNA aptamers to MS was investigated using surface plasmon resonance
397
(SPR) spectroscopy, where MS was immobilized on a CM5sensor chip (GE Healthcare). Briefly, the
398
carboxylic group on the sensor chip was activated with 1-ethyl-3-(3-dimethylaminopropyl)
399
carbodiimide (EDC) and N-hydroxysuccinimide (NHS) (BIAcore, Separations Scientific). Using
400
EDC-NHS chemistry, MS was coupled to the flow cell and the remaining carboxy site on the chip
401
surface was blocked using 1 M ethanolamine.41 MS specific ssDNA aptamer at different
402
concentrations (25, 50, 100, 200, 400 and 800 nM) was injected over the activated flow cells at a flow
403
rate of 10 µl/min for 5 min with a dissociation time of 5 min using Biacore 3000 instrument (GE
404
Healthcare). The blank surface of CM5sensor chip was used for background subtraction. The results
405
were obtained and analyzed using BiaEvaluation Software (Version 4.1.Biacore) to determine the
406
equilibrium dissociation constant Kd for each aptamer.
407 408 409 18
410
Aptamer-based inhibition of catalytic activity of MS
411
MS inhibition assay was performed at room temperature in a 96 well Corning® UV-Transparent
412
microplate (Corning, Inc. USA).The reaction mixture consisted of 20 mM Tricine buffer pH 7.5, 5
413
mM MgCl2, 0.8 mM EDTA, 2 mM glyoxylate, and 2 mM acetyl-CoA and 0.5 µg purified MS protein
414
was added and incubated at RT for 30 min. The reaction was stopped by the addition of 5,5'-dithiobis-
415
2-nitrobenzoic acid (DTNB) to a final concentration of 2 mM in Tris-HCl, pH-8.0. The amount of
416
CoA-SH released was measured by titrating the free thiol groups with DTNB and measuring the
417
change in absorption at 412 nm.10 Enzyme activity unit was represented as µmol/min. The screening
418
of ssDNA aptamers inhibiting MS enzyme activity was performed by using the same protocol
419
mentioned above with a slight modification. DNA aptamers (2 µM final concentration) and purified
420
MS protein (0.5 µg) was pre-incubated for 30 min in buffer containing 20 mM Tricine buffer (pH 7.5),
421
5 mM MgCl2 and 0.8 mM EDTA followed by the addition of 2 mM glyoxylate, and 2 mM acetyl-CoA.
422
The enzyme activity in the presence of aptamer was determined as stated above.
423 424
Post-SELEX optimization of aptamer MS10
425
Post-SELEX truncation and mutation studies were performed with the best-performing aptamerMS10
426
to determine the minimal functional unit within it. For this, the MS10 sequence was analyzed using a
427
web-based tool QGRS mapper (http://bioinformatics.ramapo.edu/QGRS/index.php) and G4IPDB
428
(http://bsbe.iiti.ac.in/bsbe/ipdb/index.php) to determine the high propensity G4 forming region.42,43
429
Based on the output, truncated and mutant aptamers were designed and procured from IDT.
430
Determination of IC50 and inhibition constant (Ki) of aptamer candidates
431
The inhibitory activity of aptamers was assessed by determining IC50 values for aptamer-mediated
432
inhibition of enzymatic activity of MS. In this assay, 2-10 mM glyoxylate and 2 mM acetyl-CoA was 19
433
used in the reaction. Purified MS protein (0.5 µg) was incubated with aptamers for 30 min at room
434
temperature followed by the addition of glyoxylate and acetyl-CoA and measurement of enzyme
435
activity. Also, MS protein was incubated with substrate for 30 min at room temperature followed by
436
addition of aptamer and measurement of enzyme activity. The IC50 values for individual aptamer
437
inhibition were determined by fitting the data into a sigmoidal dose-response curve in GraphPad Prism
438
software (version 5.01). All the kinetic measurements were performed at room temperature. The mode
439
of enzyme inhibition was measured as a function of substrate concentration at a fixed concentration of
440
aptamer and obtained data were plotted using double reciprocal Lineweaver-Burk (LB) plot. Ki values
441
were determined from linear replots of Lineweaver-Burk slopes versus aptamer or mentioned inhibitor
442
concentrations. Experimental data were analyzed using GraphPad Prism (version 5.01).
443
444
Aptamer binding to Mtb cell surface
445
Log phase 107 CFU of Mtb H37Rv was treated with 10 mM vitamin C for 24 hr to overexpress the
446
glyoxylate shunt enzymes44,45. Cultures were washed with SB and incubated with 0.5 µM of 5’-FAM-
447
labeled aptamers (C2 and MS10-Trunc) for 1 hr at RT. After removal of unbound aptamers, bacilli
448
were washed, fixed in 2% paraformaldehyde at 4ºC for 4 hrs and acquired on BD LSRII
449
flowcytometer, to assess aptamer binding under FITC-A channel. Overlay plots were prepared and
450
data was analyzed using FlowJoTM 10.4.1 software.
451
Inhibition of Mtb invasion into cells
452
Mtb H37Rv was cultured in DTA medium (Dubos containing 0.5% BSA, 0.75% Dextrose and 0.085%
453
NaCl plus 0.1% Tween-80) with shaking at 220 rpm at 37°C till OD595 ~0.1-0.2 and treated with 10
454
mM vitamin C for 24 hr. The bacteria were washed with SB and then exposed to 8 µg aptamer (~2.2
455
µM) for 1hr in SB. Bacteria were washed to remove excess of aptamer and infected into 5 x 105 THP20
456
1 cells at a MOI 10:1 for 2 hr. The extracellular Mtb were removed by treatment with amikacin (200
457
µg/ml) for 2 hr following which, the Mtb infected THP-1 cells were lysed with 0.025% SDS. Bacterial
458
colony forming units (CFUs) were enumeratedby plating THP-1 lysates on Middlebrook 7H11 agar
459
supplemented with 1.5% glycerol (MB agar) and the counts were finalized at 5 weeks.
460 461
Diagnostic application of MS10-Trunc
462
The diagnostic utility of MS10-Trunc aptamer was evaluated through ALISA on 86 archived
463
cerebrospinal fluid samples.8,39 The samples belonged to pediatric TBM subjects (n=38) and disease
464
controls subjects (n=48). The ALISA procedure was same as described above except the CSF samples
465
were diluted 1:20 in coating buffer. Data was plotted after background subtraction [(O.D.450 of CSF
466
sample) - (O.D.450 of mean + 3SD of control wells)].
467
Structure determination by Nuclear Magnetic Resonance (NMR)
468
To validate the aptamer structure, 1H NMR spectra were acquired on AVANCE 500 MHz BioSpin
469
International AG, Switzerland equipped with a 5 mm broadband inverse probe. All the NMR spectra
470
were acquired using solvent containing H2O/D2O at 9:1 ratio and spectral data were processed,
471
integrated and analyzed on Topspin (1.3 version) software. NMR samples were referenced with 3 -
472
(Trimethylsilyl) propionic-2, 2, 3, 3-d4 acid sodium salt (TSP).
473
Molecular modeling of MS10-Trunc and docking on MS
474
To simulate the structure and binding of MS10-Trunc aptamer (5’-GGTGGTGGTGG-3’) to MS, a
475
model of aptamer structure was first generated and used as the input for molecular docking on MS
476
[Protein Data Bank (PDB) code:2GQ3]. A well-known intra-molecular G4 formed by the SV40 repeat
477
sequence (PDB code: 1EVO), was taken as the initial input for modeling the 3D structure of MS10-
478
Trunc aptamer and further needful mutations were performed on Discovery studio 3.5. Protein 1EVO 21
479
was chosen as initial input model due the presence of a similar kind of G4 topology and close
480
sequence homology between 1EVO and MS10-Trunc aptamer. The resulting structure was subjected
481
to energy minimization and optimized using various iterations of conjugate gradient and steepest
482
descent method. Electrostatics was set as ‘particle mesh Ewald’ and apply SHAKE constraints were
483
chosen for every minimization run. The molecular docking studies were carried out using Hex 8.0.0
484
tool in which G4 DNA was treated as a rigid body. Correlation type was set as “Shape+Electro” and
485
FFT mode was set as 3D for every docking iteration. Sampling methods were set as “Rang angles” and
486
docking output was ranked according to their free energy.
487 488
AUTHORS’ CONTRIBUTIONS
489
J.S.T., T.K.S., A.K., T.P.S designed and supervised the experiment. A.D., C.K., K.S., I.D., S.M., P.S.
490
performed experiments, J.S.T., T.K.S., A.K., A.D., C.K., K.S., I.D., S.M., P.S. collected and analyzed
491
the data. J.S.T., T.K.S., A.D., C.K., K.S., S.M., and A.K. wrote the manuscript. All authors reviewed
492
the manuscript.
493
Competing financial interests: The authors declare no competing financial interests.
494
Acknowledgments
495
This work was supported through National Biodesign Alliance project by Department of
496
Biotechnology, Govt. of India awarded to J.S.T. (BT/PR14638/MED/12/483/2010) and an Innovation
497
Award and THSTI core grant to T.K.S. by the Department of Biotechnology, Govt. of India. J.S.T. is
498
thankful to the Department of Science and Technology, Govt. of India for the J. C. Bose National
499
Fellowship during part of the study. T.K.S. is thankful to the Department of Biotechnology, Govt. of
500
India for the Innovative Young Biotechnologist Award (BT/010/IYBA/2016/10). A.D. acknowledges
501
the Indian Council of Medical Research, Govt. of India for a Senior Research Fellowship. K.S. is 22
502
thankful to Department of Science and Technology, Government of India (PDF/2016/000745) for
503
funding. The aptamer sequences reported in this study are proprietary reagents and are deposited in
504
the Indian Patent Office as part of Indian Patent application no. 201611021901. We thank Dr. Priya
505
Kalra for her help during FACS experiment. The admirable technical assistance of Sanjay Kumar and
506
Jitendra Singh is duly acknowledged. We sincerely thank Sarin Prakash and Sunil Kumar Gupta for
507
their active involvement in data management and sample collection. BEI Resources, NIAID, NIH,
508
USA is acknowledged as below for providing various proteins that were utilized in this study. “The
509
following reagents were obtained through BEI Resources, NIAID, NIH: Plasmid pMRLB.8
510
Containing Gene Rv1837c (Protein GlcB), from Mycobacterium tuberculosis, NR-13281, Plasmid
511
pMRLB.38 Containing Gene Rv3803c (Protein Mpt51) from Mycobacterium tuberculosis, NR-
512
13291, Plasmid pMRLB.12 Containing Gene Rv1980c (Protein mpt64) from Mycobacterium
513
tuberculosis, NR-13273, Plasmid pMRLB.46 Containing Gene Rv3874 (Protein Cfp10)
514
from Mycobacterium tuberculosis, NR-13297 and Ag85 Complex, Purified Native Protein
515
from Mycobacterium tuberculosis, Strain H37Rv, NR-14855, HspX, Recombinant Protein Reference
516
Standard, NR-49428, GroES (Gene Rv3418c), Purified Native Protein from Mycobacterium
517
tuberculosis, Strain H37Rv, NR-14861, Mycobacterium tuberculosis, Strain H37Rv, Purified
518
Lipoarabinomannan (LAM), ESAT-6, Recombinant Protein Reference Standard, NR-49424 and
519
Mycobacterium tuberculosis, Strain H37Rv, Culture Filtrate Proteins, NR-14825."
520
ADDITIONAL INFORMATION
521
Supplementary Information.
522 523
23
524
REFERENCES
525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570
1 2 3
4 5 6
7 8
9 10 11
12 13
14
15
16 17 18
19
WHO. WHO Global TB report. (2018). WHO: Global Tuberculosis Report. (2017). Baig, I. A., Moon, J. Y., Lee, S. C., Ryoo, S. W. & Yoon, M. Y. Development of ssDNA aptamers as potent inhibitors of Mycobacterium tuberculosis acetohydroxyacid synthase. Biochimica et biophysica acta 1854, 1338-1350, doi:10.1016/j.bbapap.2015.05.003 (2015). Shum, K. T. et al. Aptamer-mediated inhibition of Mycobacterium tuberculosis polyphosphate kinase 2. Biochemistry 50, 3261-3271, doi:10.1021/bi2001455 (2011). Myler, P. J. & Stacy, R. A new drug for an old bug. Chemistry & biology 19, 1499-1500, doi:10.1016/j.chembiol.2012.12.003 (2012). Marrero, J., Rhee, K. Y., Schnappinger, D., Pethe, K. & Ehrt, S. Gluconeogenic carbon flow of tricarboxylic acid cycle intermediates is critical for Mycobacterium tuberculosis to establish and maintain infection. Proceedings of the National Academy of Sciences of the United States of America 107, 9819-9824, doi:10.1073/pnas.1000715107 (2010). Kinhikar, A. G. et al. Mycobacterium tuberculosis malate synthase is a laminin-binding adhesin. Mol Microbiol 60, 999-1013, doi:10.1111/j.1365-2958.2006.05151.x (2006). Haldar, S. et al. Detection of Mycobacterium tuberculosis GlcB or HspX Antigens or devR DNA impacts the rapid diagnosis of tuberculous meningitis in children. PloS one 7, e44630, doi:10.1371/journal.pone.0044630 (2012). Sharma, V. et al. Structure of isocitrate lyase, a persistence factor of Mycobacterium tuberculosis. Nature structural biology 7, 663-668, doi:10.1038/77964 (2000). Smith, C. V. et al. Biochemical and structural studies of malate synthase from Mycobacterium tuberculosis. J Biol Chem 278, 1735-1743, doi:10.1074/jbc.M209248200 (2003). Puckett, S. et al. Glyoxylate detoxification is an essential function of malate synthase required for carbon assimilation in Mycobacterium tuberculosis. Proceedings of the National Academy of Sciences of the United States of America 114, E2225-E2232, doi:10.1073/pnas.1617655114 (2017). Sobotka, O. et al. Impaired mitochondrial functions contribute to 3-bromopyruvate toxicity in primary rat and mouse hepatocytes. J Bioenerg Biomembr 48, 363-373, doi:10.1007/s10863-016-9674-8 (2016). Dunn, M. F., Ramirez-Trujillo, J. A. & Hernandez-Lucas, I. Major roles of isocitrate lyase and malate synthase in bacterial and fungal pathogenesis. Microbiology 155, 3166-3175, doi:10.1099/mic.0.030858-0 (2009). Krieger, I. V. et al. Structure-guided discovery of phenyl-diketo acids as potent inhibitors of M. tuberculosis malate synthase. Chemistry & biology 19, 1556-1567, doi:10.1016/j.chembiol.2012.09.018 (2012). Huang, H. L., Krieger, I. V., Parai, M. K., Gawandi, V. B. & Sacchettini, J. C. Mycobacterium tuberculosis Malate Synthase Structures with Fragments Reveal a Portal for Substrate/Product Exchange. J Biol Chem 291, 27421-27432, doi:10.1074/jbc.M116.750877 (2016). Suzy Kedzierski, M. K., G. Thomas Synthetic Antibodies: The Emerging Field of Aptamers. Bioprocessing Journal, 46-49 (2013). Hamula, C. L., Le, X. C. & Li, X. F. DNA aptamers binding to multiple prevalent M-types of Streptococcus pyogenes. Analytical chemistry 83, 3640-3647, doi:10.1021/ac200575e (2011). Nagatoishi, S., Isono, N., Tsumoto, K. & Sugimoto, N. Loop residues of thrombin-binding DNA aptamer impact G-quadruplex stability and thrombin binding. Biochimie 93, 1231-1238, doi:10.1016/j.biochi.2011.03.013 (2011). Phan, A. T. et al. An interlocked dimeric parallel-stranded DNA quadruplex: a potent inhibitor of HIV-1 integrase. Proceedings of the National Academy of Sciences of the United States of America 102, 634639, doi:10.1073/pnas.0406278102 (2005). 24
571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619
20
21 22 23
24 25
26 27
28 29 30 31
32
33
34
35
36
37 38
Platella, C., Riccardi, C., Montesarchio, D., Roviello, G. N. & Musumeci, D. G-quadruplex-based aptamers against protein targets in therapy and diagnostics. Biochimica et biophysica acta. General subjects 1861, 1429-1447, doi:10.1016/j.bbagen.2016.11.027 (2017). Sharma, T. K., Bruno, J. G. & Dhiman, A. ABCs of DNA aptamer and related assay development. Biotechnology advances 35, 275-301, doi:10.1016/j.biotechadv.2017.01.003 (2017). Kypr, J., Kejnovska, I., Renciuk, D. & Vorlickova, M. Circular dichroism and conformational polymorphism of DNA. Nucleic acids research 37, 1713-1725, doi:10.1093/nar/gkp026 (2009). Choi, E. W., Nayak, L. V. & Bates, P. J. Cancer-selective antiproliferative activity is a general property of some G-rich oligodeoxynucleotides. Nucleic acids research 38, 1623-1635, doi:10.1093/nar/gkp1088 (2010). Tucker, W. O., Shum, K. T. & Tanner, J. A. G-quadruplex DNA aptamers and their ligands: structure, function and application. Curr Pharm Des 18, 2014-2026 (2012). He, X., Guo, L., He, J., Xu, H. & Xie, J. Stepping Library-Based Post-SELEX Strategy Approaching to the Minimized Aptamer in SPR. Analytical chemistry 89, 6559-6566, doi:10.1021/acs.analchem.7b00700 (2017). Tawani, A., Amanullah, A., Mishra, A. & Kumar, A. Evidences for Piperine inhibiting cancer by targeting human G-quadruplex DNA sequences. Scientific reports 6, 39239, doi:10.1038/srep39239 (2016). Kondrashov, F. A., Koonin, E. V., Morgunov, I. G., Finogenova, T. V. & Kondrashova, M. N. Evolution of glyoxylate cycle enzymes in Metazoa: evidence of multiple horizontal transfer events and pseudogene formation. Biol Direct 1, 31, doi:10.1186/1745-6150-1-31 (2006). Keefe, A. D., Pai, S. & Ellington, A. Aptamers as therapeutics. Nature reviews. Drug discovery 9, 537550, doi:10.1038/nrd3141 (2010). Nimjee, S. M., White, R. R., Becker, R. C. & Sullenger, B. A. Aptamers as Therapeutics. Annual review of pharmacology and toxicology 57, 61-79, doi:10.1146/annurev-pharmtox-010716-104558 (2017). Zhou, J. & Rossi, J. Aptamers as targeted therapeutics: current potential and challenges. Nature reviews. Drug discovery 16, 440, doi:10.1038/nrd.2017.86 (2017). Macdonald, J., Houghton, P., Xiang, D., Duan, W. & Shigdar, S. Truncation and Mutation of a Transferrin Receptor Aptamer Enhances Binding Affinity. Nucleic Acid Ther 26, 348-354, doi:10.1089/nat.2015.0585 (2016). Bates, P. J., Laber, D. A., Miller, D. M., Thomas, S. D. & Trent, J. O. Discovery and development of the G-rich oligonucleotide AS1411 as a novel treatment for cancer. Experimental and molecular pathology 86, 151-164, doi:10.1016/j.yexmp.2009.01.004 (2009). Nadal, P., Svobodova, M., Mairal, T. & O'Sullivan, C. K. Probing high-affinity 11-mer DNA aptamer against Lup an 1 (beta-conglutin). Analytical and bioanalytical chemistry 405, 9343-9349, doi:10.1007/s00216-013-7385-0 (2013). Hotoda, H. et al. Biologically active oligodeoxyribonucleotides. 5. 5'-End-substituted d(TGGGAG) possesses anti-human immunodeficiency virus type 1 activity by forming a G-quadruplex structure. J Med Chem 41, 3655-3663, doi:10.1021/jm970658w (1998). Squeglia, F., Ruggiero, A., De Simone, A. & Berisio, R. A structural overview of mycobacterial adhesins: Key biomarkers for diagnostics and therapeutics. Protein science : a publication of the Protein Society 27, 369-380, doi:10.1002/pro.3346 (2018). Chen, F., Zhang, X., Zhou, J., Liu, S. & Liu, J. Aptamer inhibits Mycobacterium tuberculosis (H37Rv) invasion of macrophage. Molecular biology reports 39, 2157-2162, doi:10.1007/s11033-011-0963-3 (2012). Liao, J. et al. Cell-specific aptamers and their conjugation with nanomaterials for targeted drug delivery. Expert opinion on drug delivery 12, 493-506, doi:10.1517/17425247.2015.966681 (2015). AlShamaileh, H. & Veedu, R. N. Next-Generation Nucleic Acid Aptamers with Two-Base-Modified Nucleotides Have Improved Binding Affinity and Potency. Chembiochem : a European journal of chemical biology 18, 1565-1567, doi:10.1002/cbic.201700276 (2017). 25
620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652
39 40
41
42 43 44 45
46 47 48 49
50 51
52
Dhiman, A. et al. Generation and application of DNA aptamers against HspX for accurate diagnosis of tuberculous meningitis. Tuberculosis 112, 27-36, doi:10.1016/j.tube.2018.07.004 (2018). Kalra, P. et al. G-Quadruplex-Forming DNA Aptamers Inhibit the DNA-Binding Function of HupB and Mycobacterium tuberculosis Entry into Host Cells. Molecular therapy. Nucleic acids 13, 99-109, doi:10.1016/j.omtn.2018.08.011 (2018). Rotherham, L. S., Maserumule, C., Dheda, K., Theron, J. & Khati, M. Selection and application of ssDNA aptamers to detect active TB from sputum samples. PloS one 7, e46862, doi:10.1371/journal.pone.0046862 (2012). Kikin, O., D'Antonio, L. & Bagga, P. S. QGRS Mapper: a web-based server for predicting G-quadruplexes in nucleotide sequences. Nucleic acids research 34, W676-682, doi:10.1093/nar/gkl253 (2006). Mishra, S. K., Tawani, A., Mishra, A. & Kumar, A. G4IPDB: A database for G-quadruplex structure forming nucleic acid interacting proteins. Scientific reports 6, 38144, doi:10.1038/srep38144 (2016). Sikri. K et al. Multifaceted remodeling by vitamin C boosts sensitivity of Mycobacterium tuberculosis subpopulations to combination treatment by anti-tubercular drugs. Redox Biology (2018). Taneja, N. K., Dhingra, S., Mittal, A., Naresh, M. & Tyagi, J. S. Mycobacterium tuberculosis transcriptional adaptation, growth arrest and dormancy phenotype development is triggered by vitamin C. PloS one 5, e10860, doi:10.1371/journal.pone.0010860 (2010). Thakur, R., Goyal, R. & Sarma, S. Laboratory diagnosis of tuberculous meningitis - is there a scope for further improvement? Journal of laboratory physicians 2, 21-24, doi:10.4103/0974-2727.66705 (2010). Thwaites, G. et al. Tuberculous meningitis. Journal of neurology, neurosurgery, and psychiatry 68, 289299, doi:10.1136/jnnp.68.3.289 (2000). Rufai, S. B. et al. Diagnostic usefulness of Xpert MTB/RIF assay for detection of tuberculous meningitis using cerebrospinal fluid. The Journal of infection 75, 125-131, doi:10.1016/j.jinf.2017.04.010 (2017). Solomons, R. S. et al. Commercial nucleic acid amplification tests in tuberculous meningitis--a metaanalysis. Diagnostic microbiology and infectious disease 78, 398-403, doi:10.1016/j.diagmicrobio.2014.01.002 (2014). Pai, M. et al. Diagnostic accuracy of nucleic acid amplification tests for tuberculous meningitis: a systematic review and meta-analysis. The Lancet. Infectious diseases 3, 633-643 (2003). Haldar, S., Sharma, N., Gupta, V. K. & Tyagi, J. S. Efficient diagnosis of tuberculous meningitis by detection of Mycobacterium tuberculosis DNA in cerebrospinal fluid filtrates using PCR. J Med Microbiol 58, 616-624, doi:10.1099/jmm.0.006015-0 (2009). Flores, L. L. et al. Systematic review and meta-analysis of antigen detection tests for the diagnosis of tuberculosis. Clinical and vaccine immunology : CVI 18, 1616-1627, doi:10.1128/CVI.05205-11 (2011).
653 654
26
655
FIGURE LEGENDS
656
Figure 1. Systematic enrichment of aptamers against MS. Negative selection was performed by
657
incubating the random ssDNA library (RDL) with nitrocellulose membrane (NCM) (step
658
a);partitioning of bound and unbound sequences (step b); the unbound DNAs were collected and then
659
incubated with immobilized MS for positive selection (step c); after washing, the bound DNAs were
660
eluted and amplified by PCR for the next round of SELEX (step d); after enrichment, the DNA
661
population was cloned and sequenced to identify the sequences of individual monoclonal aptamers
662
(step e).
663
Figure 2. (A) Relative binding of various aptamer pools (Round 2, 4, 6, 7, 8, 9, 10 and RDL) to target
664
MS as determined by Aptamer Linked Immobilized Sorbent Assay (ALISA). (B) Relative binding of
665
various monoclonal aptamers with MS. The aptamers highlighted in blue color were selected for
666
further characterization based on an arbitrary cut-off O.D.450nm ≥0.6. (C) CD spectra of MS aptamer
667
candidates.
668
Figure 3. (A) Effect of aptamers on the catalytic activity of Mtb MS at a fixed aptamer concentration
669
(2 µM). (B) Binding ability of truncated and different mutant versions of MS10 aptamer to target MS.
670
The binding of individual mutant aptamers to MS was compared with MS10-Trunc binding to MS
671
using ALISA. Individual base deletions are represented as cyan color bars in the graph. (C) Effect of
672
MS10 and MS10-Trunc aptamers on catalytic activity of Mtb-MS. C1 is the control aptamer (sequence
673
given in Table S2, used in panels A and C).
674
Figure 4. (A) Determination of IC50of MS10and MS10-Truncaptamers. The relative activity of the
675
Mtb-MS as a function of the various concentrations of inhibitory aptamers MS10 and its truncated
676
form MS10-Trunc. Ki value and inhibition mechanism of aptamer MS-10 and its derivative MS10-
677
Trunc was determined using double-reciprocal Line weaver Burk (LB) plot (B and C).
678
27
679
Figure 5. Inhibition of Mtb infection with MS10-Trunc in THP-1 cell infection model. (A) Binding of
680
MS aptamer to Mtb H37Rv determined by flow cytometry using 5’-FAM labeled aptamers. C2 was
681
used as control aptamer (sequence given in Table S2, used in panels A). (B) Aptamer treated Mtb was
682
infected in THP-1 monocytes and intracellular bacteria plated on MB agar post-infection for CFU
683
determination. The graph shows the percentage infectivity calculated with respect to the untreated
684
(UT, no aptamer treatment) control. ***P <0.0001 (calculated using one way ANOVA). C1 and C2
685
were used as control aptamer (sequence given in Table S2, used in panel B). The difference in
686
infectivity between UT, C1 and C2 was found to be non-significant.
687
Figure 6. MS10-Trunc aptamer-based detection of MS antigen by ALISA in archived CSF samples
688
categorized as TBM patients (n=38) and Non-TBM patients (n=48).
689
Figure 7. (A) Circular Dichroism titration spectrum for MS10-Truncaptamer (Red) and aptamer in
690
the presence of increasing concentration of MS upto Protein (P):Aptamer(A) ratio = 2.0. (B)
691
Temperature dependent 1H NMR spectra for MS10-Trunc showing imino and base region.
692
Figure 8. (A) A model of G4 structure formed by MS10-Trunc aptamer and (B) Hydrogen Bonds in
693
each G and T quartet of G4structure.
694
Figure 9. The interaction of PEP (red) and MS10-Trunc (multicolor) with MS. The interactions
695
between residues in MS protein and bases in aptamer are shown in cartoon representation and the
696
regions in the box are shown enlarged in ball and stick model. The interactions are shown as blue
697
dashes.
698 699 700 701 28
702 703
Table 1: Surface Plasmon Response (SPR) binding kinetics data of aptamers.
704
Aptamer
Kon(1/Ms)
Koff(1/s)
KA (1/M)
KD (M)
MS4
2.4 x 105
2.4 x 10-3
1 x 108
9.9 x 10-9
MS5
3.7
1 x 10-5
3.7 x 105
2.7 x 10-6
MS6
3.6 x 104
1 x 10-5
3.6 x 109
2.8 x 10-10
MS10
2.8 x 105
3.2 x 10-3
8.7 x 107
1.2 x 10-8
MS20
11
1 x 10-5
1.1 x 106
9.1 x 10-7
MS10-Trunc
5.2 x 105
1.6 x 10-5
5.2 x 1010
1.9 x 10-11
R0 (Naïve library)
35
1 x 10-5
3.4 x 106
2.9 x 10-7
705
29
706 707
Table 2: Performance of various tests in diagnosis of TBM Diagnostic Assays for TBM Diagnosis
Comments
References
Sensitivity (%)
Specificity (%)
TB culture
27
100
Poor sensitivity, long turnaround time.
Smear microscopy
10-20
100
Poor sensitivity.
47
Moderate sensitivity, but an excellent rule-in test.
48
It requires DNA isolation and amplification steps, timeconsuming with Suboptimal sensitivity.
49
It requires DNA isolation and amplification steps and timeconsuming.
8
46
DNA-based Tests Xpert MTB/RIF assay
55.1
94.8
Commercial NAATs metaanalysis
64
98
Nucleic acid commercial amplification tests meta-analysis
56
98
100 (definite TBM) 98 (probable and possible TBM) 87.6 (filtrate) 53.1(sediment)
96-97 (definite TBM) 98 (probable and possible TBM)
MS (current study)
97.44
97.96
HspX (Aptamer-based ALISA)
100
91
100 (definite TBM)
96-97 (definite TBM)
92-95 (probable and possible TBM)
93-96 (probable and possible TBM)
In-house qPCR
In-house qPCR
50
51
92
Antigen Detection Tests
GlcB, HspX& PstS1 (Antibody-based ELISA)
GlcB, HspX& MPT51 (Antibody-based ELISA) LAM, Ag85 complex, 65 kDa& ESAT-6 (Antibody-based ELISA)
87
Excellent diagnostic tool to detect TBM with high accuracy. Excellent diagnostic accuracy, laboratory-based format.
-
Excellent diagnostic accuracy, laboratory-based format, timeconsuming, uses antibody which might have batch to batch variation.
8
Laboratory-based format, timeconsuming, uses antibody which might have batch to batch variation.
84
708
30
39
8
52