ACS Chem. Neurosci 1

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 38

Subscriber access provided by YORK UNIV

Article
Power of Tyrosine Assembly in Microtubule Stabilization
and Neuroprotection Fuelled by Phenol Appendages
Surajit Barman, Gaurav Das, Prasenjit Mondal, Krishnangsu Pradhan,
Debmalya Bhunia, Juhee Khan, Chirantan Kar, and Surajit Ghosh
ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.8b00497 • Publication Date (Web): 19 Dec 2018
Downloaded from http://pubs.acs.org on December 20, 2018

Just Accepted

“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted
online prior to technical editing, formatting for publication and author proofing. The American Chemical
Society provides “Just Accepted” as a service to the research community to expedite the dissemination
of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in
full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully
peer reviewed, but should not be considered the official version of record. They are citable by the
Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore,
the “Just Accepted” Web site may not include all articles that will be published in the journal. After
a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web
site and published as an ASAP article. Note that technical editing may introduce minor changes
to the manuscript text and/or graphics which could affect content, and all legal disclaimers and
ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or
consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W.,


Washington, DC 20036
Published by American Chemical Society. Copyright © American Chemical Society.
However, no copyright claim is made to original U.S. Government works, or works
produced by employees of any Commonwealth realm Crown government in the course
of their duties.
Page 1 of 37 ACS Chemical Neuroscience

1
2
3
4
5
Power of Tyrosine Assembly in Microtubule
6
7
8
9
Stabilization and Neuroprotection Fuelled by Phenol
10
11
12
13
Appendages
14
15
16
17 Surajit Barman,1 Gaurav Das,1, 2 Prasenjit Mondal, 1, 2 Krishnangsu Pradhan,1 Debmalya Bhunia,1
18
19 Juhee Khan,1 Chirantan Kar,1 Surajit Ghosh1, 2 *
20
21
22 1. Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja
23
24
25 S. C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India. Fax: +91-33-2473-
26
27 5197/0284; Tel: +91-33-2499-5872
28
29 2. Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical
30
31
Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India.
32
33
34
35
36 ABSTRACT: Microtubules play crucial role in maintenance of structure, function, axonal
37
38 extensions, cargo transport and polarity of neurons. During neurodegenerative diseases,
39
40
41
microtubule structure and function gets severely damaged due to destabilization of its major
42
43 structural proteins. Therefore, design and development of molecules that stabilize these
44
45 microtubule networks have always been an important strategy for development of potential
46
47
neurotherapeutic candidates. Towards this venture, we designed and developed a tyrosine rich
48
49
50 tri-substituted triazine molecule (TY3) that stabilizes microtubules through close interaction with
51
52 the taxol binding site. Detailed structural investigations revealed that the phenolic protons are the
53
54 key interacting partners of tubulin. Interestingly, we found that this molecule is non-cytotoxic in
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 2 of 37

1
2
3 PC12 derived neurons, stabilized microtubules against nocodazole induced depolymerization and
4
5
6 increases expression of acetylated tubulin (K-40), an important marker of tubulin stability.
7
8 Further, results show that TY3 significantly induces neurite sprouting as compared to the
9
10 untreated control as well as the two other analogues (TS3 and TF3). It also possesses anti-Aβ
11
12
13
fibrillation property as confirmed by ThT assay, which leads to its neuro-protective effect against
14
15 amyloidogenic induced toxicity caused through NGF deprivation in PC12 derived neurons.
16
17 Remarkably, our result reveals that it reduces the expression of pY490 TrkA associated with
18
19
NGF deprived amyloidogenesis, which further proves that it is a potent amyloid beta inhibitor.
20
21
22 Moreover, it promoted the health of the rat primary cortical neurons through higher expression of
23
24 key neuronal markers such as MAP2 and Tuj1. Finally, we observed that it has good serum
25
26 stability and has the ability to cross the blood brain barrier (BBB). Overall, our work indicates
27
28
29 the importance of Phenolic –OH in promoting neuroprotection and its importance could be
30
31 implemented in the development of future neuro-therapeutics.
32
33
34 KEYWORDS:
35
36 Triazine, Microtubule, FRET, STD NMR, TR-NOESY, Neuroprotection, Blood Brain-Barrier
37
38
39
40
1. INTRODUCTION
41
42
43 The embellished shape of neuron and its polarity has been a centre point of interest for wide
44
45 range of researchers in the field of neurobiology for the last several decades.1-3 To maintain its
46
47 incredible structure and polarity several factors play crucial role and microtubules are one of the
48
49
50
most important players.4-7 Microtubules (MT) are dynamic polymers of α, β-tubulin heterodimers
51
52 that along with actin and intermediate filaments forms the key cytoskeletal components of the
53
54 eukaryotic cells including neurons.1-7 Along with other eukaryotic cells, microtubules have
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 3 of 37 ACS Chemical Neuroscience

1
2
3 special importance in neurons where apart from providing them the much-needed shape and
4
5
6 structure, they also have important functional aspects.8 In neurons, microtubules are involved in a
7
8 variety of tasks like cargo transport through gigantic axonal projections with the aid of the
9
10 associated molecular motors.9 This role of axonal transport in neurons is so significant that even
11
12
13
slightest perturbations in the microtubule structure or functions create severe damage to the
14
15 normal neuronal physiology as observed in several neurodegenerative diseases.10 In the axons,
16
17 the linear arrays of microtubules comprising of two ends- plus ends towards synapse and minus
18
19
ends towards cell body provides directionality and structural support to the neurons.11, 12 Many
20
21
22 studies have indicated that neurodegenerative diseases like Alzheimer’s Disease (AD) have been
23
24 caused due to defects in axonal microtubules and this has given rise to the importance of MT-
25
26 stabilizing molecules.13-19 During AD progression, microtubule networks in neurons is severely
27
28
29 damaged due to disintegration of microtubule associated protein Tau, which further aggregates
30
31 and generates neurofibrillary tangles (NFT). Moreover, it has been also reported that Aβ
32
33 aggregation damages the microtubule network. Many microtubule stabilizing molecules have
34
35
36
been so far reported to have shown promising effects in restoration of microtubules in axonal
37
38 transport as well as improving cognition in animal models. As a result, such compounds may be
39
40 considered as potential candidates for the treatment of AD and related taupathies. Microtubule
41
42
stabilising agents (MSA) accelerates the equilibrium from tubulin to the polymeric microtubule
43
44
45 and promotes tubulin polymerization. MSA’s are already well established chemotherapeutic
46
47 agents like taxol but the action of these neuro-protective microtubule stabilizing agents are
48
49 somewhat different as their order of stabilization is not so pronounced as to arrest the cell
50
51
52 division and cause cell death.20-22 Though a number of neuroprotective MT-stabilizing agents
53
54 have been reported, most of them so far have produced clinically insignificant results. Moreover,
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 4 of 37

1
2
3 many naturally occurring and synthesized molecules with phenolic -OH moiety like in
4
5
6 neurotransmitter molecules such as dopamine and Ferulic acid have been reported to possess
7
8 neuroprotective effects23-27 but none of the studies directly or indirectly have explored the
9
10 importance of phenolic -OH in neuroprotection and their possible cross talk with tubulin or
11
12
13
microtubule. Talking about phenolic –OH, several groups have already reported tyrosine
14
15 analogues with effective biological activity and is one of the hot arenas in current research
16
17 among the chemical biologists. 28-30 To address this fundamental aspects and to provide more
18
19
detailed understanding in this manuscript, we have designed novel tripodal molecules based on
20
21
22 1, 3, 5-triazine core with tri-substitution by tyrosine (Y), phenylalanine (F) and serine (S) named
23
24 as TY3, TF3 and TS3 respectively. Among the three molecules, TY3 and TF3 binds close to the
25
26 taxol binding pocket of the tubulin. Next, the identification of pharmacophore of TY3 for the
27
28
29 interaction with tubulin was investigated through STD NMR and TR-NOESY experiment.
30
31 Further, it was observed that TY3 confers significant microtubule stabilization in PC12 derived
32
33 neurons against nocodazole induced depolymerization, increases the expression of acetylated
34
35
36
tubulin (K-40) and facilitates neurite outgrowth, while TF3 and TS3 does not show any activity
37
38 in neurons. Interestingly, we found that TY3 also inhibits Aβ fibrillation and confers significant
39
40 neuroprotection against Aβ mediated toxicity in PC12 derived neurons. It has been reported
41
42
earlier that NGF deprivation is associated with an increase in the expression of a particular
43
44
45 phosphorylation of TrkA (pY490).31 We found that TY3 reduces this TrkA associated
46
47 phosphorylation, thus reinstating the fact that it has anti-amyloidogenic activity. Moreover, we
48
49 observed that TY3 crosses the blood-brain barrier, shows significant serum stability and
50
51
52 promotes healthy morphology of the cultured primary rat cortical neurons by upregulation of
53
54 neuronal markers (Tuj1 and MAP2). Overall, results reveal that our newly designed molecules
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 5 of 37 ACS Chemical Neuroscience

1
2
3 confers significant microtubule stabilization and neuroprotection, where spatially positioned
4
5
6 phenolic-OH group interacts close to the taxol binding pocket of tubulin and plays an important
7
8 role. We strongly believe that this novel tripodal molecule with tyrosine substitution has
9
10 significant potential to serve as a unique neurotherapeutics template.
11
12
13
14
15
16 2. RESULTS AND DISCUSSION
17
18
19 2.1. Synthesis of triazine based small molecules.
20
21
22 We have synthesized three triazine based compounds (TS3, TF3 and TY3) and characterized
23
24 them by ESI-mass spectroscopy, 1H-NMR, 13C-NMR, DEPT-135 and HPLC (Figure 1A and S1-
25
26
27
10). The fluorescein attached TF3 and TY3 were synthesized by solid phase peptide synthesis
28
29 (SPPS) method using CEM, purified them by HPLC and characterized by MALDI-TOF mass
30
31 spectroscopy (Figure S11, 12).
32
33
34 2.2 Microtubule assembly assay.
35
36
37
To check whether our compounds have any interaction with tubulin, we have performed
38
39
40 microtubule assembly assay using the change in fluorescence intensity of 4', 6-diamidino-2-
41
42 phenylindole (DAPI) as an indicator during tubulin polymerization.32, 33 Remarkably, we found
43
44 that fluorescence intensity of DAPI increased in case of TF3, TY3 as compared to untreated
45
46
47 control (tubulin alone). But, there was no significant change in fluorescence intensity in case of
48
49 TS3. Next, we performed microtubule assembly assay of the TF3 and TY3 in dose dependent
50
51 manner (Figure 1B, D and S13). These results clearly shows that TF3 and TY3 are promoting the
52
53
54
tubulin polymerization.
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 6 of 37

1
2
3 2.3 Determination of binding constant by Surface Plasmon Resonance (SPR) study.
4
5
6
7
Above results encouraged us to further explore the affinity of TF3 and TY3 with tubulin and
8
9 their association and dissociation kinetics. For this, we performed Surface Plasmon Resonance
10
11 (SPR)34 for TF3 and TY3. The SPR studies suggest that the association constant (Ka) for TF3
12
13 and TY3 are 1.42 × 103 M-1, 1.05 ×103 M-1 respectively and the dissociation constant of TF3 and
14
15
16 TY3 are 158 μM and 16.4 μM respectively (Figures 1 C, E).
17
18
19 2.4 Determination of binding affinity using tryptophan fluorescence quenching study.
20
21
22 Next, we have performed the tryptophan fluorescence quenching study to determine the binding
23
24 affinity with tubulin. Binding affinity of TF3 and TY3 were determined using modified Stern-
25
26
27
Volmer equation. The binding constants of TF3 and TY3 are 2.68 × 103 M-1 and 7.75 × 103 M-1
28
29 respectively (Figure S14). These results are in agreement with the association constant measured
30
31 through previous SPR experiments.
32
33
34 2.5 Förster Resonance Energy Transfer (FRET) study to determine the binding location in
35
36
37 tubulin.
38
39
40 To know more in details about the binding location of tubulin, we performed Förster Resonance
41
42 Energy Transfer (FRET) experiment using these two compounds (TF3 and TY3). Here, we used
43
44 three florescent probes (colchicine, 8-anilinonaphthalene-1-sulfonic acid (ANS) and TAMRA-
45
46
47 E3NAP) as FRET partner with fluorescein tagged TF3 and TY3. The binding sites of colchicine,
48
49 ANS and TAMRA-E3NAP in tubulin are well understood and the Förster distance (R0) between
50
51 tubulin bound colchicine and fluorescein dye attached peptide is 29.5 ± 1 Å, tubulin bound ANS
52
53
54
and fluorescein-peptide is 50.3 ± 2 Å and tubulin bound TAMRA-peptide with fluorescein-
55
56 peptide is 62.9 ± 2 Å.35, 36 We performed the FRET experiment with the above FRET partners
57
58
59
60 ACS Paragon Plus Environment
Page 7 of 37 ACS Chemical Neuroscience

1
2
3 with TY3 compound resulting in a distance of 34.64 ± 1 Å, 53.4 ± 1 Å and no FRET with
4
5
6 TAMRA-E3NAP complex (Figure 2A-C). Similarly, when we performed FRET experiments
7
8 with TF3 and the above FRET partners, we found similar result like TY3 (34 ± 1 Å from
9
10 colchicine site, 54 ± 1 Å from ANS site and no FRET between F-TF3 with TAMRA-E3NAP
11
12
13
complex) (Figure 2D-F). Then we calculated the distance obtained from the FRET results that
14
15 revealed TY3 and TF3 binds close to the taxane binding site of tubulin (Figure 2G).
16
17
18 2.6 Conformational analysis and subsequent docking study.
19
20
21 We performed molecular docking to know the binding affinity and interacting partners of TY3
22
23
and TF3 at taxol pocket using DFT minimized conformers of TY3 and TF3 molecules (Figure
24
25
26 S15-25). The docking results reveal that the interacing partners of TY3 are ARG278, ARG284,
27
28 PRO274, ARG320 and GLY370 with binding affinity of -7.7 kcal/mol and for TF3 are GLY370,
29
30 PRO274, THR276 and LEU371 with binding affinity of -7.9 kcal/mol (Figure 2H, I and S26).
31
32
33 TY3 and TF3 are shown in docking image bound to their respective binding sites in tubulin
34
35 which are shown in Figure 2J.
36
37
38 2.7 STD NMR and TR-NOESY experiments.
39
40
41 In order to obtain more minute details of their molecular recognition, STD and TR-NOESY were
42
43
44
performed to derive a tubulin-bound conformer of TY3. First, we have assigned different protons
45
46 of TY3 using 1D NMR (1H-NMR, 13C-NMR and DEPT study) and 2D NMR experiments
47
48 (COSY, HSQC, HMBC and NOESY) (Figure S27-30). To study more in details, we have
49
50 performed saturation transfer difference (STD) and transfer nuclear overhauser spectroscopy
51
52
53 (TR-NOESY) experiment. These studies are useful for the epitope mapping of a molecule with a
54
55 protein. In STD-NMR, the bound ligand only shows the STD signals and higher STD signals
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 8 of 37

1
2
3 indicate the close proximity of the epitope to the protein. The STD results revealed that aromatic
4
5
6 protons of ligands TY3 are showing the highest STD effect while less signal for the aliphatic
7
8 group was observed (Figure 3A-C). From this result, we can conclude that aryl moiety is
9
10 essential for its interaction with tubulin.
11
12
13 In TR-NOESY experiment, the negative cross peaks were detected in the presence of tubulin.
14
15
16 We have deduced the tubulin bound conformation of TY3 where two aromatic protons of TY3
17
18 (ArH6.67 and ArH6.97) are in close proximity to the –OMe,-CH2 and -CH protons (Figure 4A).
19
20 Further, we deduced the plausible conformation of tubulin bound TY3. For this, we performed
21
22
23 the molecular docking study with TY3 using DFT energy minimized conformer of TY3. The
24
25 results of molecular docking of TY3 are in agreement with the TR-NOESY data (Figure 4B, C
26
27 and S31). The STD NMR and TR-NOESY experiments clearly shows that the phenyl moiety is
28
29
30
crucial for its interaction with tubulin.
31
32
33 2.8 Cell viability assay
34
35
36 So far we have performed several in vitro assays that indicate the microtubule stabilizing nature
37
38 of TY3 and TF3. In order to perform further cell based screening, it is pertinent to confirm their
39
40 cyto-toxic nature. Hence, we performed the cell viabilty assay using differentiated PC12 cell
41
42
43 lines upto 200uM concentrations of both TY3 and TF3. The results revealed that all the
44
45 compounds are non-cyto-toxic to the differentiated neurons (Figure 5A, B).
46
47
48 2.9 Nocodazole induced depolymerization and expression of Acetylated Tubulin (AcK-40)
49
50
51 Previous in vitro studies like SPR and microtubule assembly assay using DAPI have proven TY3
52
53
54
and TF3 both to be microtubule polymerizing compound. Now, it was to be tested, whether it
55
56 shows similar results in neurons. After reversibly depolymerizing microtubules of the PC12
57
58
59
60 ACS Paragon Plus Environment
Page 9 of 37 ACS Chemical Neuroscience

1
2
3 derived neurons by 4 h treatment with 10 µM nocodazole, the cells were washed free of the
4
5
6 nocodazole and divided into three groups one being control replaced with only fresh medium
7
8 while the other two with TY3 and TF3. After 2 h of treatment, the cells were fixed and
9
10 immunostained with primary antibodies against alpha-tubulin. The cells treated with TY3
11
12
13
showed healthy microtubule network completely recovered from the nocodazole treatment while
14
15 the control cells and TF3 treated cells still suffered from the depolymerizing effect of nocodazole
16
17 (Figure 5C and S32). This proves that our cellular data is in line with our in vitro observation and
18
19
TY3 is indeed a microtubule stabilizer. The treated cells also showed a higher expression of
20
21
22 acetylated tubulin (AcK 40), a known marker of tubulin stabilization compared to the untreated
23
24 control (Figure 5D, E and S33).
25
26
27 2.10 TY3 promoted significant neurite outgrowth among the other screened molecules.
28
29
30 Many microtubule stabilizing drugs like Methyl 3, 4-dihydroxybenzoate and GIT 1 have also
31
32
33 shown to promote neurite outgrowth.37, 38 All these factors prompted us to check the neurite
34
35 outgrowth potential of our microtubule stabilizing compounds TY3 and TF3. When these
36
37 compounds were incubated with undifferentiated PC12 cells, TY3 showed neurite outgrowth
38
39
40
after 24 h of incubation while the others (TF3 and TS3) failed to do so (Figure 6A, B). This
41
42 above results motivated us to conduct more studies for TY3 in neurons. It was also seen that TY3
43
44 promoted the healthy morphology and architecture of microtubule network (Figure 6 C, D). It is
45
46
important to note that although TY3 binds close to the taxol binding site, yet it does not cause
47
48
49 toxicity due to the fact that it binds at the taxol binding site 3428 fold times weaker and also
50
51 dissociates in a faster rate (Kd=16.4 μM) compared to taxol.39 Due to this reasons, TY3 can
52
53 provide microtubule stabilization through weak binding as reported before.34
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 10 of 37

1
2
3 2.11 ThT assay indicates that TY3 inhibits Aβ (1-42) aggregation
4
5
6
7
We initially performed ThT assay with TY3 and incubated it with Aβ (1-42). To our
8
9 astonishment, TY3 inhibited Aβ (1-42) aggregation (Figure 7A) and this proves that TY3 would
10
11 be able to show significant neuroprotective effect when tested in some cellular model of
12
13 neurotoxicity. For this, it was very important to first ascertain the fact that our compound TY3 is
14
15
16 non-toxic to the neurons. We have already screened TY3 on PC12 derived neurons with a wide
17
18 range of concentrations and it was observed that TY3 is completely non-toxic to the neurons at
19
20 even very high concentrations which paved way for the future studies in cell.
21
22
23
2.12 TY3 confers neuroprotection in NGF deprived PC12 derived neurons by reducing the
24
25
26 phosphorylation of TrkA.
27
28
29 It is now well documented that NGF deprivation in PC12 derived neurons leads to
30
31 amyloidogenesis40 and that may cause an unexpected phosphorylation of TrkA at Y490, thus
32
33
34
activating phospholipase C  (PLC) associated cell death pathway causing severe neuro-
35
36 toxicity. This phosphorylation is usually connected with two kinases namely Src and CDK5
37
38 which are known to be involved in amyloidogenesis and  and  mediated p75 processing. Our
39
40
41 in vitro ThT assay data encouraged us to pursue the neuroprotective potential of TY3 in this
42
43 NGF deprived model of PC12 derived neurons. We observed that TY3 confers neuroprotection
44
45 upto 1.5 µM concentrations (Figure 7B-E) and causes inhibition of the neuronal death inducing
46
47
48
TrkA phosphorylation (pY490). But no change in the expression of TrkA was observed (Figure
49
50 7F, G and S34), which denotes that TY3 specifically inhibits amyloidogenesis. Here, we would
51
52 like to point out that TF3 and TS3 does not show any neuroprotection ability although TF3
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 11 of 37 ACS Chemical Neuroscience

1
2
3 contains phenyl group and TS3 contains -OH group. This fact clearly indicates that combination
4
5
6 of both phenyl and -OH group that is phenol group is essential to confer neuroprotection.
7
8
9 2.13 TY3 promotes the healthy morphology of cultured primary cortical neurons and upregulates
10
11 several neuronal markers.
12
13
14 All the cellular studies concerning TY3 have been so far studied in PC12 derived neurons but
15
16
17
what about its effect on primary neurons. On treating the rat primary cortical neurons with TY3
18
19 for 2 days, we observed that TY3 was well tolerated by them and also promoted the healthy
20
21 morphology of the neurons (Figure 8A-C) when compared with the untreated control. Moreover,
22
23
we observed significant increase in the expression of important neuronal markers MAP2 and
24
25
26 Tuj1 in TY3 treated neurons as revealed from the immunoblots (Figure 8D, E and S35).
27
28
29 2.14 Serum stability of TY3 performed by high-performance liquid chromatography (HPLC).
30
31
32 For any candidate to qaulify as a neuroprotective drug, it must cross the blood brain barrier
33
34 (BBB). But before performing the blood brain barrier crossing, we checked the stability of TY3
35
36
37 in serum. TY3 was incubated with serum up to 24 h and monitored using HPLC. It was observed
38
39 that more than 40% TY3 remained stable after 24 h of incubation with serum. This result
40
41 suggests significant serum stability of TY3 (Figure S36).
42
43
44 2.15 Blood-Brain Barrier (BBB) crossing experiment.
45
46
47
Since TY3 showed serum stability, we finally needed to check whether it can cross the blood
48
49
50 brain barrier (BBB). To address this, we have performed the MALDI-Mass spectroscopy of brain
51
52 extract obtained from mice treated with TY3. The MALDI-Mass spectrum showed molecular
53
54 peaks at (M=661 Da) and (M+K+ =699 Da). This data indicated the presence of TY3 in the brain
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 12 of 37

1
2
3 extract, which confirmed its potential to cross the blood-brain barrier (BBB) (Figure 8F and S37,
4
5
6 38). Hence we can now consider this as a promising neurotherapeutic candidate.
7
8
9 3. Conclusion
10
11
12 In summary, we have shown in this paper the power of phenolic –OH appendages in
13
14 neuroprotection and neurite outgrowth through its interaction with microtubule. Our results
15
16
17
reveal that tyrosine substitution on a tripodal platform plays an important role where this phenol
18
19 group acts like a key player. Moreover, we found that phenolic –OH group interacts with tubulin
20
21 close to the taxol binding pocket by FRET and NMR where the aromatic substituent binds in the
22
23
hydrophobic pocket. Interestingly, we found that TF3 and TS3 did not have any significant effect
24
25
26 on microtubules and that further indicated that the phenolic –OH moieties are the key partners
27
28 for interaction with tubulin. The cellular assays in PC12 derived neurons and primary rat cortical
29
30 neurons also reciprocated similar results. Moreover, it also showed serum stability and crossed
31
32
33 blood brain barrier. Finally, the tripodal core substituted with the phenolic –OH group in TY3
34
35 could serve as a potential candidate for the development of future neuro-therapeutics.
36
37
38 4. Experimental Section
39
40
41 4.1 Chemicals
42
43
44 Cyanuric chloride, Tyrosine methyl ester, Phenyl alanine methyl ester and Serine methyl ester,
45
46
47 Thiazolyl Blue Tetrazolium Bromide (MTT), Kanamycin sulfate, Dulbecco’s Modified Eagle’s
48
49 Medium (DMEM), Guanosine 5′-triphosphate sodium salt hydrate (GTP), 4, 6-diamidino-2-
50
51 phenylindole (DAPI), Trypsin-EDTA solution, Colchicine, 5(6)-carboxyfluorescein, PIPES, Eth-
52
53
54
ylene glyolbi(2-aminoethyl ether)-N,N,N′,N′-tetra acetic acid (EGTA), N,N′-
55
56 Diisopropylcarbodiimide (DIC), 8-anilino-1-naphthalenesulfonic acid ammonium salt (ANS) and
57
58
59
60 ACS Paragon Plus Environment
Page 13 of 37 ACS Chemical Neuroscience

1
2
3 Cell culture grade DMSO were purchased from Sigma-Aldrich. Sodium chloride, Sodium
4
5
6 hydrogen carbonate, Di-sodium hydrogen phosphate dihydrate, Potassium hydroxide,
7
8 Magnesium chloride hexahydrate, Trifluoroacetic acid (TFA) and Potassium dihydrogen
9
10 phosphate were purchased from Merck. Potassium chloride and Agarose were purchased from
11
12
13
Fisher Scientific. Dimethyl sulfoxide, N, N’-Diisopropylethylamine (DIPEA), Ethyl acetate,
14
15 Chloroform and Hexane were purchased from Spectrochem. Triton-X-100 was purchased from
16
17 SRL. Amyloid-beta 1-42 (Aβ42) was purchased from AnaSpec. NGF-β (7S) was purchased from
18
19
Sigma (St. Louis, MO, USA). Neurobasal media, B27, glutaMAX and Pen/strep were bought
20
21
22 from Gibco, Life technologies. 2-[4-(2-Hydroxyethyl) piperazin-1-yl] ethanesulfonic acid
23
24 (HEPES) and Horse Serum were purchased from Himedia. Penicillin-Streptomycin and Fetal
25
26 bovine serum were purchased from Invitrogen. Anti-alpha Tubulin clone EP 1332Y, Anti-
27
28
29 Tubulin Anti-body, Beta III isoform (Tuj 1) were purchased from Merck Millipore. Anti-TrkA
30
31 (pY490), Anti-TrkA and Anti-alpha Tubulin (acetyl K-40) were purchased from Abcam.
32
33 Bisbenzimide H 33258 (hoechst) was purchased from Calbiochem. MAP2 Monoclonal Antibody
34
35
36
(M13) was purchased from Thermofisher scientific. HPLC-grade H2O and ACN were purchased
37
38 from J.T. Baker. All the NMR solvents were purchased from Cambridge isotope. All compounds
39
40 were used without further purification.
41
42
43 4.2 Cell culture
44
45
46
Pheochromocytoma of the rat adrenal medulla (PC12) were purchased from NCCS, Pune (India).
47
48
49 The PC12 cells were cultured in complete DMEM with 10% horse serum and 5% FBS. The
50
51 differentiation of PC12 cells into neurons was achieved by culturing the cells in serum free
52
53 medium with 1% horse serum and 100 ng/mL NGF (Nerve Growth Factor) for 5 days.
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 14 of 37

1
2
3 4.3 Protein purification
4
5
6
7
After the isolation of tubulin from goat brain, we purified it by two cycle’s polymerization and
8
9 depolymerization procedure followed by storing it in liquid nitrogen cryochamber using 10%
10
11 glycerol.41
12
13
14 4.4 Synthesis, characterization of TY3, TS3 and TF3 and its fluorescein derivatives.
15
16
17 In 100mL round bottom flask, methyl ester of amino acids (4 equivalents) and cyanuric chloride
18
19
20 (1 equivalent) in dry acetonitrile was stirred at room temperature. Then N, N′-
21
22 Diisopropylethylamine (DIPEA) was added to the reaction mixture. The reaction mixtures were
23
24 refluxed overnight under inert atmosphere. Finally, the compounds were purified by column
25
26
27
chromatography and characterized by ESI-mass spectroscopy, 1H-NMR, 13C-NMR, and DEPT-
28
29 135. Fluorescein tagged triazine derivatives (F-TY3 and F-TF3) were synthesized using solid
30
31 phase peptide synthesis (SPPS) method in a microwave-based (35 watt) automated peptide
32
33
synthesizer (CEM, Liberty 1). All the synthesized fluorescein tagged small molecules were
34
35
36 purified through C-18 reverse phase HPLC column at 210 nm and characterized by MALDI-
37
38 TOF in acetonitrile and water (1:1) mixture.
39
40
41 4.5 Conformational Analysis.
42
43
44 We deduced the energy minimized conformers of the TY3 and TF3 molecules using
45
46
47 Spartan’1642-45 and Gaussian ’09 program46. Geometry optimizations of each molecules were
48
49 performed in multistep.
50
51
52 In first step 1000 most probable conformers were chosen and a conformational search was
53
54 performed to predict their corresponding energies using molecular mechanics force field
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 15 of 37 ACS Chemical Neuroscience

1
2
3 (MMFF). In the next step 100 lowest energy conformers chosen from the MMFF calculation
4
5
6 were further refined by semi-empirical calculation at PM6 level. Next, the 20 lowest energy
7
8 conformers taken after the semi-empirical calculation were optimized applying Hartree-Fock
9
10 method at 3-21G level.
11
12
13 4.6 Docking study
14
15
16
17
We performed blind docking study using the software Autodock-Vina version 1.1.2.47 98×60×64
18
19 affinity grids were centered on the tubulin [PDB ID: 1Z2B]48 for docking TY3 and TF3. All the
20
21 images were visualized and developed in PyMOL (The PyMOL Molecular Graphics System,
22
23
Version 1.7.4 Schrödinger, LLC.)
24
25
26
27
4.7 Microtubule assembly assay
28
29
30 A stock solution of 10 μM DAPI in BRB80 buffer containing 100 μM tubulin, 10 mM GTP and
31
32 different concentration of TF3 and TY3 were mixed. Emission spectra of solutions were
33
34 recorded in region ranging from 400 nm to 600 nm wavelength at 37 0C after excitation at 355
35
36
37 nm.
38
39
40 4.8 Surface Plasmon Resonance (SPR) experiment
41
42
43 The SPR experiment was performed using NTA biosensor chip, Ni2+ and streptavidin-His6 were
44
45 immobilized on to the NTA biosensor chip. Next, we washed the surface with BRB80 buffer and
46
47
flowed biotinylated tubulin (20 μg/mL) followed by incubation for 15 min. The surface was
48
49
50 washed and desired compounds were added at various concentrations as analytes. The recorded
51
52 data were analyzed by plotting the curve with a local fitting.
53
54
55 4.9 Tryptophan fluorescence quenching experiment
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 16 of 37

1
2
3 Tubulin was mixed with different concentrations of TF3 and TY3 separately in BRB80 and these
4
5
6 mixtures were incubated for 40 min at 25 °C. Then, the intrinsic tryptophan fluorescence data
7
8 was recorded and calculated by modified Stern-Volmer equation.
9
10
11 4.10 Förster resonance energy transfer (FRET) experiment
12
13
14 We performed the FRET study to know the binding pocket of TF3 and TY3 in the tubulin.
15
16
17
Tubulin-colchicine, tubulin-ANS and tubulin-TAMRA-E3NAP complexes were used to find out
18
19 the distance of fluorescein attached small molecules from their specific binding pocket in tubulin
20
21 following previously described method. The excitation and emission ranges were set at 355 nm
22
23
and 450 to 650 nm respectively.
24
25
26
27
4.11 STD NMR sample preparation and experiments49, 50
28
29
30 The samples of the ligands were prepared with a 300 μM concentration of the TY3 and 10 μM of
31
32 tubulin in D2O, 10 mM NaPi, 0.1 mM GTP. Small amount of D6-DMSO were used to solubilize
33
34 the TY3 ligand . We recorded the STD-NMR by Bruker AVANCE 600 MHz spectrometer with
35
36
37 saturation time 2s. Percentage of relative STD effects was calculated by the following equation
38
39
40 ASTD = (I0 - Isat)/I0 = ISTD/I0 where intensity of the signals in the STD NMR spectrum (ISTD)
41
42 with signal intensities of a reference spectrum (I0). The maximum STD signal was considered as
43
44 100% STD effect and others were calculated relatively.
45
46
47
4.12 TR-NOESY experiments
48
49
50
51 Strong negative NOE cross peaks were detected by TR-NOESY experiments (mixing times of
52
53 200 ms) in presence of tubulin receptor compared to the free state of the TY3 ligand. All the
54
55 datas were calculated using Mnova software.
56
57
58
59
60 ACS Paragon Plus Environment
Page 17 of 37 ACS Chemical Neuroscience

1
2
3 4.13 Neurite outgrowth experiment
4
5
6
7
For neurite outgrowth experiment, all the three triazine derivatives were treated with the
8
9 undifferentiated PC12 cells and incubated for 24h at 370C.Then the treated cells were imaged
10
11 under a microscope at DIC mode. Evaluation of neurite length was carried out using the CellSens
12
13 software.
14
15
16
17
4.14 Effect of TF3 and TY3 on neuronal microtubule
18
19
20 PC12 cells (density~3000-5000) were grown in a confocal dish on the day before the treatment.
21
22 We treated TY3 for 24 h. Next day, cells were fixed using 4% paraformaldehyde and incubated
23
24 with 5% BSA and 0.2% triton-X in PBS for 1 h. Then we washed the fixed cells with PBS and
25
26
27
treated with primary antibody polyclonal anti-α-tubulin IgG antibody. After 2 h, we treated
28
29 secondary antibody (Cy3.5 pre-absorbed goat anti-rabbit IgG) after washing the cells with PBS
30
31 and kept for 2 h. On the day of the imaging of the cells, cells were washed with PBS and treated
32
33
with Hoechst 33258 (1 μg/mL) for half an hour. Images of various zone of culture dish were
34
35
36 captured using microscope.
37
38
39 4.15 Thioflavin T (ThT) assay
40
41
42 Thioflavin-T (ThT) is used to identify the amyloid fibrils both in vitro and in vivo. There is an
43
44 enhancement of its fluorescence intensity upon binding with the fibrils. To monitor the Aβ
45
46
47 peptide aggregation, we performed the following experiment.51 Different concentrations of the
48
49 TY3 were mixed with 10 μM Aβ42 peptide and incubated for 48h. Then, we added ThT solution
50
51 to the incubated solution and recorded the fluorescence in PTI QM-40 spectrofluorimeter
52
53
54
(excitation: 435 nm and emission: 460-650 nm).
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 18 of 37

1
2
3 4.16 Neuroprotection study using differentiated PC12 cells
4
5
6
7
For the event of differentiation, PC12 cells were cultured for 24 h and treated with serum free
8
9 DMEM containing 1% horse serum and 100 ng/mL NGF and incubated for 5 days.
10
11 Differentiated PC12 cells were seeded in 96-well plates (1 × 104 cells/mL) and then cells were
12
13 treated with anti-NGF alone and along with different concentrations of TY3 for 24 h. MTT
14
15
16 solution were added to each well and incubated at 37oC for 4 h. Then the MTT solutions were
17
18 replaced by 1:1 DMSO-MeOH. Then the percentage of cell viability was analyzed using
19
20 microplate ELISA reader (Thermo; Multiskan GO Microplate Spectrophotometer) at 550 nm
21
22
23 wavelength.
24
25
26 4.17 Effect of TY3 on primary cortical neuron culture
27
28
29 Primary cortical neurons were cultured by previously described protocol.52 In brief, we have
30
31 received timed-pregnant Sprague Dawley rat. Then we isolated the brain from the dissected E18
32
33
embryos. After the micro-dissection of the brain, cortices were dissected out in the MEM
34
35
36 medium containing 10% horse serum and glucose (0.6% wt/vol). ~3-5 × 105/mL cells were
37
38 plated on confocal dishes coated with poly-D lysine and was incubated in CO2 incubator for
39
40 about 4 h. Then this culture medium was changed with neurobasal media supplemented with
41
42
43 B27, Pen/Strep and GlutaMAX and cultured for another 14 days untill the neurons matured. We
44
45 treated these neurons with TY3 and assessed its effect on primary cortical neurons.
46
47
48 4.18 Serum stability
49
50
51 Stability of any small/large molecules is important for their biological aspects.53 For this
52
53
54
purpose, we checked the stability of TY3 in horse serum. Quantitative stability of the compounds
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 19 of 37 ACS Chemical Neuroscience

1
2
3 was monitored by C18 reverse phase HPLC system upto 24 h. It was found that almost 40% of
4
5
6 the TY3 remained intact after 24 h.
7
8
9 4.19 Blood-Brain Barrier (BBB) crossing experiment. 54, 55
10
11
12 C57BL/6J female mice of around 20 g have been used for our study. Animals were divided into
13
14 two groups (3 mice /group) and the mice were injected intraperitoneally with TY3 dissolved in
15
16
17
saline solution at the concentration of 50 µg/Kg body weight of mice. For control, mice were
18
19 treated with only saline solution (2nd group). After 6 h of treatment, animals were deeply
20
21 anaesthetized with ketamine (i.p) and were sacrificed by transcardial perfusion. The mice brains
22
23
were isolated and directly transferred into liquid nitrogen, crushed using a mortar and pestle.
24
25
26 Acetonitrile and water mixture (1:1) was added into the crushed brain. Mixture was centrifuged
27
28 to separate out the soluble part. HPLC and mass analysis were performed to analyze the data and
29
30 for the identification of TY3 mass.
31
32
33
ASSOCIATED CONTENT
34
35
36 Supporting Information
37
38 This materials are available free of charge via the internet at http://pubs.acs.org.
39
40 Synthesis and characterization of TS3, TF3, TY3, F-TF3 and F-TY3 using 1H-NMR, 13C-NMR,
41 DEPT-135 and mass spectroscopy. HPLC chromatograms, Microtubule assembly assay,
42
43
Quenching of Tryptophan fluorescence experiment, DFT study of tri-substituted triazine
44 derivatives. 2D NMR (COSY, NOESY, HSQC and HMBC) of TY3 and TF3. Control
45 experiments for TR-NOESY and BBB experiment. Serum stability of TY3. Immunoblots of
46
47
AcK-40, phospho-TrkA (pY490), TrkA, MAP2 and Tuj1. (PDF)
48
49 AUTHOR INFORMATION:
50
51 Corresponding Author
52
53 Dr. Surajit Ghosh
54
55
56 Principal Scientist
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 20 of 37

1
2
3 Organic and Medicinal Chemistry Division
4
5
6 CSIR-IICB, Jadavpur, Kolkata-700 032, India
7
8 Tel: +91-33-2499-5872
9
10 Fax: +91-33-2473-5197/0284
11
12
13
E-mail: sghosh@iicb.res.in
14
15 ORCID ID
16
17 Surajit Barman: 0000-0003-3584-4716
18
19
Gaurav Das: 0000-0002-8432-5384
20
21
22 Prasenjit Mondal: 0000-0003-0767-449X
23
24 Surajit Ghosh: 0000-0002-8203-8613
25
26
27
28
29 Author contributions
30
31 SB performed synthesis, characterization, major experiments and analyzed the data. GD
32
33 performed various cell based assays like neurite outgrowth, microtubule associated
34
35
36
depolymerization and the immunoblots. PM and KP performed FRET, computational experiment
37
38 and in vitro inhibition of Aβ aggregation. DB helped GD and JK in performing the BBB
39
40 experiment. JK and GD performed the primary neuron culture. CK performed the DFT
41
42
calculations .SG conceived the idea, designed and monitored all the experiments, analyzed the
43
44
45 data, prepared figures and supervised GD and SB in writing the manuscript.
46
47 Notes
48
49 The authors declare no competing financial interest
50
51
52 ACKNOWLEDGMENT
53
54 We thank Ms. Varsha Gupta for critically reading the manuscript. We also want to thank Dr. E.
55
56 Padmanban for helping in STD and TR-NOESY experiments and Mr. K Suresh Kumar for SPR
57
58
59
60 ACS Paragon Plus Environment
Page 21 of 37 ACS Chemical Neuroscience

1
2
3 experiments. SB, KP thanks UGC, PM thanks CSIR, DB, JK thanks DST and GD thanks ICMR,
4
5
6 India for their fellowships. CK thanks SERB for his postdoctoral fellowship. SG kindly
7
8 acknowledges DST, India for providing support (EMR/2015/002230) and CSIR-IICB for
9
10 infrastructure. Animal experiments were performed following IICB ethical guidelines.
11
12
13 Reference:
14
15 1. Craig, A. M., and Banker, G. (1994) Neuronal polarity. Annu. Rev. Neurosci., 17, 267-310.
16
17 2. Arimura, N., and Kaibuchi, K. (2007) Neuronal polarity: from extracellular signals to
18
19 intracellular mechanisms. Nature Rev. Neurosci., 8, 194-205.
20
21
3. Conde, C., and Cáceres A. (2009) Microtubule assembly, organization and dynamics in axons
22
23
and dendrites. Nat Rev Neurosci., 5, 319-32.
24
25
26 4. Baas, P. W. (2002) Neuronal polarity: microtubules strike back. Nature Cell Biol., 4, 194-195.
27
28 5. Witte, H., and Bradke, F. (2008) The role of the cytoskeleton during neuronal polarization. Curr.
29
30 Opin. Neurobiol., 18, 1-9.
31
32 6. Desai, A., and Mitchison, T. (1997) Microtubule polymerization dynamics. Annu. Rev. Cell Dev.
33
34 Biol., 13, 83-117.
35
36
7. Nogales, E. (2001) Structural insights into microtubule function. Annu. Rev. Biophys. Biomol.
37
38
39 Struct., 30, 397-420.
40
41 8. Akhmanova, A., and Hoogenraad, C.C. (2015) Microtubule minus-end-targeting proteins. Curr.
42
43 Biol., 25, 162-171.
44
45 9. Baas, P. W., and Ahmad, F. J. (2001) Force generation by cytoskeletal motor proteins as a
46
47 regulator of axonal elongation and retraction. Trends Cell Biol., 11, 244-249.
48
49 10. Baas, P. W., and Mozgova, O. I. (2012) A novel role for retrograde transport of
50
51
52 microtubules in the axon. Cytoskeleton, 69, 416-425.
53
54 11. Baas, P. W., and Lin, S. (2011) Hooks and comets: The story of microtubule polarity orientation
55
56 in the neuron. Dev Neurobiol., 71, 403-418.
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 22 of 37

1
2
3 12. Lasser, M., Tiber, J., and Lowery, L. A. (2018) The Role of the Microtubule Cytoskeleton in
4
5
Neurodevelopmental Disorders. Front Cell Neurosci., 12, 165.
6
7
8 13. Ballatore, C., Smith, A. B., Lee, V. MY., Trojanowski, J. Q., and Brunden, K. R. (2016)
9
10 Microtubule-Stabilizing Agents for Alzheimer’s and Other Tauopathies. In: Wolfe M. (eds).
11
12 Alzheimer’s Disease II. Topics in Medicinal Chemistry, 24. Springer, Cham
13
14 14. Ballatore, J. C., Brunden, K. R., Huryn, D. M., Trojanowski, J. Q., Lee, V. M.-Y. and Smith, A.
15
16 B. (2012) Microtubule Stabilizing Agents as Potential Treatment for Alzheimer’s Disease and
17
18 Related Neurodegenerative Tauopathies. J. Med. Chem., 55, 8979-8996.
19
20
15. Varidaki, A., Hong, Y., and Coffey, E. T. (2018) Repositioning Microtubule Stabilizing Drugs
21
22
23
for Brain Disorders. Front Cell Neurosci., 12, 226.
24
25 16. Michaelis, M. L., Chen, Y., Hill, S., Reiff, E., Georg, G., Rice, A., and Audus, K. (2002)
26
27 Amyloid peptide toxicity and microtubule-stabilizing drugs. J. Mol. Neurosci., 19, 101-105.
28
29 17. Roy, S., Zhang, B., Lee, V. M., and Trojanowski, J. Q. (2005) Axonal transport defects: a
30
31 common theme in neurodegenerative diseases. Acta Neuropathol., 109, 5-13
32
33 18. Liu, X., Rizzo, V., and Puthanveettil, S. V. (2012) Pathologies of axonal transPort in
34
35
neurodegenerative diseases. Transl Neurosci., 3, 355-372.
36
37
38 19. Fanara, P., Banerjee, J., Hueck, R. V., Harper, M. R., Awada, M., Turner, H., Husted, K. H.,
39
40 Brandt, R., and Hellerstein, M. K. (2007) Stabilization of hyperdynamic microtubules is
41
42 neuroprotective in amyotrophic lateral sclerosis. J Biol Chem., 282, 23465-23472.
43
44 20. Mukhtar, E., Adhami, V. M., and Mukhtar, H. (2014) Targeting Microtubules by Natural Agents
45
46 for Cancer Therapy. Mol Cancer Ther., 13, 275-284.
47
48 21. Weaver, B. A. (2014) How Taxol/paclitaxel kills cancer cells. Mol Biol Cell., 25, 2677-2681.
49
50
51
22. Dumontet, C., and Jordan, M. A. (2010) Microtubule-binding agents: a dynamic field of cancer
52
53 therapeutics. Nat Rev Drug Discov., 9, 790-803.
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 23 of 37 ACS Chemical Neuroscience

1
2
3 23. Sarkar, S., Raymick, J., and Imam, S. (2016) Neuroprotective and Therapeutic Strategies against
4
5
Parkinson’s disease: Recent Perspectives. Int. J. Mol. Sci., 17, 904
6
7
8 24. Spencer, G. E., Klumperman, J., and Syed, N. I. (1998) Neurotransmitters and
9
10 neurodevelopment. Role of dopamine in neurite outgrowth, target selection and specific synapse
11
12 formation. Perspect Dev Neurobiol., 5, 451-67.
13
14 25. Spencer, G. E., Lukowiak, K., and Syed, N. I. (1996) Dopamine regulation of neurite outgrowth
15
16 from identified Lymnaea neurons in culture. Cell Mol Neurobiol., 16, 577-589.
17
18 26. Ojha, S., Javed, H., Azimullah, S., Abul Khair, S. B., and Haque, M. E. (2015) Neuroprotective
19
20
potential of ferulic acid in the rotenone model of Parkinson's disease. Drug Des Devel Ther., 9,
21
22
23
5499-510.
24
25 27. Ren, Z., Zhang, R., Li, Y., Li, Y., Yang, Z., and Yang, H. (2017) Ferulic acid exerts
26
27 neuroprotective effects against cerebral ischemia/reperfusion-induced injury via antioxidant and
28
29 anti-apoptotic mechanisms in vitro and in vivo. Int J Mol Med., 40, 1444-1456.
30
31 28. Neidhardt, M. M., Schmitt, K., Baro, A., Schneider, C., Bilitewski, U. and Laschat, S. (2018)
32
33 Self-assembly and biological activities of ionic liquid crystals derived from aromatic amino
34
35 acids. Phys Chem Chem Phys., 20, 20371-20381.
36
37
38
29. Augustyn, E., Finke, K., Zur, A. A., Hansen, L., Heeren, N., Chien, H. C., Lin, L., Giacomini, K.
39
40 M., Colas, C., Schlessinger, A. and Thomas, A. A. (2016) LAT-1 activity of meta-substituted
41
42 phenylalanine and tyrosine analogs. Bioorg Med Chem Lett., 26, 2616-2621.
43
44 30. Bader, K., Neidhardt, M. M., Wöhrle, T., Forschner, R., Baro, A., Giesselmann, F. and Laschat,
45
46 S. (2017) Amino acid/crown ether hybrid materials: how charge affects liquid crystalline self-
47
48 assembly. Soft Matter, 13, 8379-8391.
49
50 31. Matrone, C., Maroldaa, R., Ciafre`a, S., Ciottia, M. T. Mercantia, D., and Calissanoa, P. (2009)
51
52
Tyrosine kinase nerve growth factor receptor switches from prosurvival to proapoptotic activity
53
54
55 via Abeta-mediated phosphorylation, Proc Natl Acad Sci U S A., 27, 11358-11363.
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 24 of 37

1
2
3 32. Bonne, D., Heusele, C., Simon, C., and Pantaloni, D. (1985) 4', 6-Diamidino-2-phenylindole, a
4
5
Fluorescent Probe for Tubulin and Microtubules. J. Biol. Chem., 260, 2819-2825.
6
7
8 33. Ghosh, J. G., Houck, S. A., and Clark, J. I. (2007) Interactive Domains in the Molecular
9
10 Chaperone Human αB Crystallin Modulate Microtubule Assembly and Disassembly. PLoS
11
12 ONE, 2, e498.
13
14 34. Mondal, P., Das, G., Khan, J., Pradhan, K., and Ghosh, S. (2018) Crafting of Neuroprotective
15
16 Octapeptide from Taxol-Binding Pocket of β-Tubulin. ACS Chem. Neurosci., 9, 615-625.
17
18 35. Jana, B., Mohapatra, S., Mondal, P., Barman, S., Pradhan, K., Saha, A., and Ghosh, S. (2016) α-
19
20
Cyclodextrin Interacts Close to Vinblas-tine Site of Tubulin and Delivers Curcumin
21
22
23
Preferentially to the Tubulin Surface of Cancer Cell. ACS Appl. Mater. Interfaces, 8, 13793-
24
25 13803.
26
27 36. Adak, A., Mohapatra, S., Mondal, P., Jana, B., and Ghosh, S. (2016) Design of a novel
28
29 microtubule targeted peptide vesicle for delivering different anticancer drugs. Chem. Commun.,
30
31 52, 7549-7552.
32
33 37. Zhang, Z., Zhou, X., Zhou, X., Zhou, X., X, Xiao., Liao, M., Yan, L., Lv, R., and Luo, H. (2012)
34
35 Methyl 3,4-dihydroxybenzoate promotes neurite outgrowth of cortical neurons cultured in vitro.
36
37
Neural Regen Res., 7, 971-977.
38
39
40 38. Li, Y., Qin, L., Liu, J., Xia, W., Li, J., Shen, H., and Gao, W. (2016) GIT1 enhances neurite
41
42 outgrowth by stimulating microtubule assembly. Neural Regen Res., 11, 427-434.
43
44 39. Ross, J. L., Santangelo, C. D., Makrides, V., and Fygenson, D. K. (2004) Tau induces
45
46 cooperative Taxol binding to microtubules. Proc Natl Acad Sci U S A., 101, 12910-12915.
47
48
49 40. Carmelaa, M., Annaa, D. L., Giovannib, M., Simonac, D., Cinziaa, S., Teresaa, C. M.,
50
51 Antoninob, C., and Pietroa, C. (2008) Activation of the Amyloidogenic Route by NGF
52
53 Deprivation Induces Apoptotic Death in PC12 Cells. J Alzheimers Dis., 13, 81-96.
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 25 of 37 ACS Chemical Neuroscience

1
2
3 41. Hyman, A., Drechsel, D., Kellogg, D., Salser, S., Sawin, K., Steffen, P., Wordeman, L., and
4
5
Mitchison, T. (1991) Preparation of Modified Tubulins. Methods Enzymol 196, 478-485.
6
7
8 42. Becke, A. D. (1993) Density‐functional thermochemistry. III. The role of exact exchange. J.
9
10 Chem. Phys. 98, 5648-5652.
11
12 43. Lee, C., Yang, W., and Parr, R. G. (1988) Development of the Colle Salvetti correlation-energy
13
14 formula into a functional of the electron density. Phys. Rev. B, 37, 785-789.
15
16 44. Andrae, D., Haeussermann, U., Dolg, M., Stoll, H., and Preuss, H. (1990) Energy-adjustedab
17
18 initio pseudopotentials for the second and third row transition elements. Theor. Chim. Acta, 77,
19
20
123-141.
21
22
23 45. Gaussian 09, Revision D.01, M. J. Frisch et al Gaussian, Inc., Wallingford CT, 2013.
24
25 46. Kong, J., White, C. A., Krylov, A. I., Sherrill, D., Adamson, R. D., Furlani, T. R., Lee, M. S.,
26
27 Lee, A. M., Gwaltney, S. R., Adams, T. R., Ochsenfeld, C., Gilbert, A. T. B., Kedziora, G. S.,
28
29 Rassolov, V. A., Maurice, D. R., Nair, N., Shao, Y., Besley, N. A., Maslen, P. E., Dombroski, J.
30
31 P., Daschel, H., Zhang, W., Korambath, P. P., Baker, J., Byrd, E. F. C., Van Voorhis, T., Oumi,
32
33 M., Hirata, S., Hsu, C.P., Ishikawa, N., Florian, J., Warshel, A., Johnson, B. G., Gill, P. M. W.,
34
35
Head-Gordon, M., and Pople, J. A. (2000) Q‐Chem 2.0: a high‐performance ab initio electronic
36
37
38
structure program package. J. Comput. Chem., 21, 1532-1548.
39
40 47. Trott, O., and Olson, A. J. (2010) AutoDock Vina: Improving the Speed and Accuracy of
41
42 Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J. Comput.
43
44 Chem., 31, 455-461.
45
46 48. Gigant, B., Wang, C., Ravelli, R.B., Roussi, F., Steinmetz, M.O., Curmi, P.A., Sobel, A., and
47
48 Knossow, M. (2005) Structural basis for the regulation of tubulin by vinblastine. Nature, 435,
49
50 519-522.
51
52
53
49. Kotar, A., Tomašič, T., Živković, M. L., Jug, G., Plavec, J., and Anderluh, M. (2016) STD NMR
54
55 and molecular modelling insights into interaction of novel mannose-based ligands with DC-
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 26 of 37

1
2
3 SIGN. Org. Biomol. Chem., 14, 862-875.
4
5
50. Canales, A., Salarichs, J. R., Trigili, C., Nieto, L., Coderch, C., Andreu, J. M., Paterson, I.,
6
7
8 Barbero, J. J., and Díaz, J. F. (2011) Insights into the Interaction of Discodermolide and
9
10 Docetaxel with Tubulin. Mapping the Binding Sites of Microtubule-Stabilizing Agents by Using
11
12 an Integrated NMR and Computational Approach. ACS Chem. Biol., 6, 789-799.
13
14 51. Biswas, A., Kurkute, P., Saleem, S., Jana, B., Mohapatra, S., Mondal, P., Adak, A., Ghosh, S.,
15
16 Saha, A., Bhunia, D., Biswas, S. C. and Ghosh, S. (2015) Novel hexapeptide interacts with
17
18 tubulin andmicrotubules, inhibits Aβ fibrillation, and shows significant neuroprotection. ACS
19
20 Chem. Neurosci., 6, 1309-1316.
21
22
23
52. Xu, S. Y., Wu, Y. M., Ji, Z., Gao, X. Y., and Pan. S. Y. (2012) A Modified Technique for
24
25 Culturing Primary Fetal Rat Cortical Neurons. J. Biomed. Biotechnol., 2012, 803930.
26
27 53. Bhunia, D., Mondal, P., Das, G., Saha, A., Sengupta, P., Jana, J., Mohapatra, S., Chatterjee, S.,
28
29 and Ghosh, S. (2018) Spatial Position Regulates Power of Tryptophan: Discovery of a Major-
30
31 Groove-Specific Nuclear-Localizing, Cell Penetrating Tetrapeptide. J. Am. Chem. Soc., 140,
32
33 1697-1714.
34
35 54. Gynther, M., Petsalo, A., Hansen, S. H., Bunch, L., and Pickering DS (2015) Blood-brain barrier
36
37 permeability and brain uptake mechanism of kainic acid and dihydrokainic acid. Neurochem
38
39
Res., 40, 542-549.
40
41
42 55. Alonso, E., Vieira, A. C., Rodriguez, I., Alvariño, R., Gegunde, S., Fuwa, H., Suga, Y., Sasaki,
43
44 M., Alfonso, A., Cifuentes, J. M., and Botana, L. M. (2017) Tetracyclic Truncated Analogue of
45
46 the Marine Toxin Gambierol Modifies NMDA, Tau, and Amyloid β Expression in Mice Brains:
47
48 Implications in AD Pathology. ACS Chem. Neurosci., 8, 1358-1367.
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 27 of 37 ACS Chemical Neuroscience

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28 Figure 1: (A) Synthesis scheme and structure of three triazine based compound. Reagents and
29 conditions: (a) Methyl ester of amino acids, DIPEA, Dry THF, Reflux for overnight. Microtubule
30 polymerization assay using DAPI of TY3 (B) and TF3 (D). SPR sensogram for TY3 (C) and TF3 (E) in
31 presence of tubulin.
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 28 of 37

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30 Figure 2: Energy transfer graph between (A) Tub-Colchicine complex and F-TY3; (B) Tub-ANS
31
complex and F-TY3; (C) Tub-TAMRA E3NAP and F-TY3 complex; (D) Tub-Colchicine complex and F-
32
33 TF3; (E) Tub-ANS complex and F-TF3; (F) Tub-TAMRA E3NAP and F-TF3 complex. (G) Diagram for
34 the distance calculation from FRET experiments. Molecular docking image showing interaction of (H)
35 TY3 with PRO 274, ARG 278, ARG 284, ARG 320 and GLY 370 and (I) TF3 with PRO 274, THR 276,
36 GLY 370 and LEU 371. (N) Binding position of TF3 (shown in grey colored sphere model) and TY3
37
38 (shown in cyan colored sphere model) in dimeric tubulin.
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 29 of 37 ACS Chemical Neuroscience

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 30 of 37

1
2
3
4
5
6 Figure 3: (A) Off-resonance NMR spectrum (600MHz) of TY3 (upper line) STD experiment (lower line)
7 with TY3 in presence of tubulin. Zoom spectrum for aromatic region (Inset). Aromatic region shown in
8 zoom spectrum (Inset). (B) Epitope mapping of TY3 (C). (D) % STD of different proton of TY3.
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44 Figure 4: (A) TR-NOESY spectrum (mixing time: 300ms) of presence of TY3. (B)Tubulin bound
45 conformation of TY3 and (C) its interacting partners.
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 31 of 37 ACS Chemical Neuroscience

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 32 of 37

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 33 of 37 ACS Chemical Neuroscience

1
2
3
4
5
6
7 Figure 5: Cell viability assay of (A) TY3 and (B) TF3 in PC12 cell lines. (C) Nocodazole induced
8 depolymerization Experiment. (D) Immunoblot depicts the increase of acetylated tubulin (AcK-40) as
9 compared to control with TY3 treatments. (E) Bar diagram reveals the relative expressions of acetylated
10
11
tubulin normalized with loading control. All experiments have been performed in triplicate. The error bar
12 corresponds to the standard deviation of the value. (*p < 0.05, performing two tailed student’s t-test).
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37 Figure 6: (A) Neurite outgrowth of PC12 derived neurons after treatment with triazine derivatives (TF3,
38
TY3 and TS3). (B) Quantative analysis for neurite outgrowth of PC12 derived neurons. Microscopic
39
40 images of microtubule networks of PC12 cells for control (C) and after treatment with TY3 (D) at merged
41 images. All experiments have been performed in triplicate. The error bar corresponds to the standard
42 deviation of the value. (*p < 0.05, performing two tailed student’s t-test).
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 34 of 37

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
Figure 7: (A) ThT assay of TY3. (B) Bar diagram analysis represents cell viability of differentiated PC12
46
47 derived neurons by NGF assay in absence or presence of various concentrations of TY3 and Anti-NGF.
48 Microscopic images of the (C) control PC12 cells, (D) Anti-NGF treated cells and (E) TY3 treated cells.
49 (F) Western blot of phospho-TrkA (pY490) (pTrkA) and full-length TrkA (TrkA) protein levels in PC12
50 derived neurons exposed to NGF (+NGF) or deprived of NGF for 24 h (-NGF). The expression of pTrkA
51
increases due to 24 h of NGF deprivation, while decreases on treatment with TY3. On the other hand the
52
53 expression of full length TrkA remains the same. (G) Bar diagram represents the quantitative analysis of
54 the immunoblots. All experiments have been performed in triplicate. The error bar corresponds to the
55 standard deviation of the value. (*p < 0.05, performing two tailed student’s t-test).
56
57
58
59
60 ACS Paragon Plus Environment
Page 35 of 37 ACS Chemical Neuroscience

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47 Figure 8: Microscopic images of primary cortical neurons after treatment with TY3 at (A) bright field,
48 (B) 561 nm channel and (C) merged channel. (D) Immunoblotting experiment showing higher activation
49 of MAP2 and β-III tubulin in primary rat cortical neurons after TY3 treatment as compared to Control and
50
(E) their quantitative analysis. (F) Blood Brain Barrier crossing experiment. All experiments have been
51
52 performed in triplicate. The error bar corresponds to the standard deviation of the value. (*p < 0.05 and
53 **p < 0.03, performing two tailed student’s t-test).
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Chemical Neuroscience Page 36 of 37

1
2
3 Table of content
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 37 of 37 ACS Chemical Neuroscience

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37 A tyrosine rich tripodal neuroprotective molecule has been designed, where phenol group plays crucial role
38 in interacting with microtubule and confers neuroprotection. Results reveal that molecule has significant
39 serum stability and capability to cross blood-brain barrier.
40
417x394mm (300 x 300 DPI)
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment

You might also like