• Nem Talált Eredményt

Csilla Kállay,* John Spencer,* and George E. Kostakis*

N/A
N/A
Protected

Academic year: 2022

Ossza meg "Csilla Kállay,* John Spencer,* and George E. Kostakis*"

Copied!
11
0
0

Teljes szövegt

(1)

1

Salpyran: A Cu(II) Selective Chelator with Therapeutic Potential

2

Jack Devonport, Nikolett Bodnár, Andrew McGown, Mahmoud Bukar Maina, Louise C. Serpell,

3

Csilla Kállay,* John Spencer,* and George E. Kostakis*

Cite This:https://doi.org/10.1021/acs.inorgchem.1c01912 Read Online

ACCESS

Metrics & More Article Recommendations

*

Supporting Information 4ABSTRACT: We report the rational design of a tunable Cu(II)

5chelating scaffold, 2-(((2-((pyridin-2-ylmethyl)amino)ethyl)-

6amino)methyl)phenol,Salpyran. This tetradentate (3N,1O) ligand

7is predicated to have suitable permeation, has an extremely high

8affinity for Cu compared to clioquniol (pCu7.4= 10.65 vs 5.91), and

9exhibits excellent selectivity for Cu(II) over Zn(II) in aqueous

10media. Solid and solution studies corroborate the formation of a

11stable [Cu(II)(3N,1O)]+ monocationic species at physiological pH

12values (7.4). Its action as an antioxidant was tested in ascorbate, tau,

13and human prion protein assays, which reveal thatSalpyranprevents

14the formation of reactive oxygen species from the binary Cu(II)/

15H2O2system, demonstrating its potential use as a therapeutic small

16molecule metal chelator.

17

INTRODUCTION

18The dysregulation and accumulation of biometals is a common

19pathological hallmark of many neurodegenerative disorders,

20such as Alzheimer’s (AD), Parkinson’s (PD) and prion

21diseases.1−9 AD is the most prevalent adult neurogenerative

22disorder and the most significant cause of dementia.10,11

23Currently, 24 million people suffer globally, and, with an aging

24population, this figure may double by 2040.12,13 AD is

25characterized by intracellular accumulation of neurofibrillary

26tangles formed of misfolded tau proteins and the extracellular

27deposition of fibrillar amyloid-β(Aβ) peptides. However, AD

28is a multiparameter disease, and other factors contribute to its

29etiology such as mitochondrial dysfunction, genetics, and

30age.14At present, a large body of research suggests that metal

31ion dyshomeostasis plays a role in AD’s pathology; therefore,

32the restoration of biometal homeostasis offers a new clinical

33target when developing AD therapies.6,15−22

34 Recent trends show that drug development into disease-

35modifying therapies (DMTs) for AD is broadening its scope

36beyond the classical primary targets of Aβ and tau

37aggregation.23,24 A paucity of new treatments for AD, for

38almost two decades, and the low success rate of drugs in

39clinical trials have furthered the need to widen the scope of

40both targets and approaches in curbing disease progres-

41sion.25,26Recently, thefirst DMT (aducanumab) was approved

42by the Food and Drug Adminstration (FDA) for the treatment

43of AD patients. By targeting the production and aggregation of

44Aβ, this novel therapy was found to reduce senile plaques,

45although there still remains some uncertainty in its clinical

46benefits.

Metal ions can affect the self-assembly of amyloid proteins;47

for example, Aβhas a picomolar affinity for Cu(II) binding via 48

histidine binding.27,28Cu(II) imbalances exist in AD affected49

brains, and Cu(II) can be found either upregulated or 50

downregulated depending on the locality of the tissue.6,2951

Due to its redox potential when bound to Aβ, Cu(II) 52

contributes to the generation of reactive oxygen species 53

(ROS), leading to oxidative neuronal damage.3032 54

In the past decade, there has been an increasing interest in 55

designing Cu-specific small molecule metal chelators 56

(SMMCs) aiming to reduce Cu(II)-Aβ induced oxidative57

s tre ss a n d th e r es ul ti ng pa th oge ni c co nse q ue n- 58

ces.6,15,34−39,16−22,33 Chelation therapy aims to disrupt 59

potential toxic interactions of metal ions and biomolecules 60

by targeting specific metal ions and promoting redistribution 61

or excretion. When designing a Cu-specific SMMC, both the 62

thermodynamic properties of the metal chelate and the 63

pharmacological properties of the ligand must be considered. 64

The key criteria for Cu(II) targeting AD therapeutic are 65

denticity, metal/ligand stoichiometry, and the coordination 66

environment and geometry of the complex at physiological pH 67

values. Ideally, the given ligand would coordinate to Cu(II) in 68

a 1:1 stoichiometry, as ligands of this type exhibit a higher 69

copper affinity than similar 1:2 complexes due to the70

Received: June 24, 2021

Article pubs.acs.org/IC

© XXXX American Chemical Society A

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX

(2)

71chelation.40The increased shielding observed in 1:1 complexes

72protects the metal ion from the physiological environment,

73preventing further biological interactions such as the formation

74of [Aβ(Cu)L] ternary species.39,41 Also, to be an effective

75therapeutic, both the ligand and the formed metal complex

76must be metabolically stable, nontoxic, and possess suitable

77aqueous solubility. Moreover, to be effective in AD, the

78SMMC should be able to pass through the blood−brain barrier

79(BBB) to reach the site of Cu(II) accumulation. For passive

80diffusion, this requires a SMMC that is suitably hydrophobic to

81passively pass through the membrane yet hydrophilic enough

82to stay soluble in physiological environments.42,43

f1 83 Clioquinol (CQ, Figure 1) was investigated in phase II

84clinical trials for targeting metal homeostasis as an AD

85treatment. CQ is a bidentate ligand that forms a [Cu(II)L2]

86complex with an 2N,2O coordination environment. By

87targeting both Cu(II) and Zn(II)binding, CQ showed some

88improvements in the cognition of the patients trialled.44

89However, due to neurotoxic side effects, the clinical progress of

90CQ was ultimately abandoned.45This led to the design of a

91s e c o n d - g e n e r a t i o n t r i d e n t a t e 5 , 7 - d i c h l o r o - 2 -

92((dimethylamino)methyl)quinolin-8-ol (PBT2, Figure 1),

93which completed Phase II clinical trials.46,47 Introduction of

94a dimethylamino unit at the C2 position introduced a new

95binding site, but still [Cu(II)L2] complexes are formed.48 A

96lack of reduction in amyloid plaque levels in the brains of AD

97patients and only mild cognitive benefits mean thatPBT2has

98not progressed into more extensive clinical studies. The poor

99metal selectivity is a possible reason for the clinical failure of

100CQ, as interactions with other biometals or metalloproteins/

101substrates in vivo are conceivable. The formation of the

102[Cu(II)L2] species, in both CQ and PBT2 cases, speculates

103the likely in vivo formation of ternary L(Cu)Aβ species that

104can contribute to increased ROS production.49

Due to the clinical potential demonstrated by CQ and 105

PBT2, several tetradentate ligands based on similar scaffolds 106

have been developed to increase Cu(II) selectivity and 107

minimize unwanted biological interactions.38,40,48,50,51 108

This incremental design led to the state-of-art Cu(II) chelator, 109

TDMQ-20 (Figure 1).52 TDMQ-20 is an 8-aminoquinoline110

derivative that offers a 4N coordination environment and 111

shows exceptional selectivity for Cu(II) ions. Recently, 112

TMDQ20 has been studied as an AD therapeutic in early 113

stage nontransgenic mouse models and late-stage transgenic 114

models.52 Oral treatment offered significant improvements in 115

both the behavioral and cognitive impairments observed in 116

each model, while also reducing oxidative stress in the mouse 117

cortices. This efficacy paves the way for future pharmacological 118

evaluation of SMMCs; thus, most research heavily focuses on 119

chelators based around either 8-hydroxy/8-amino quinoline 120

backbones. Having the chemical criteria and fall-outs from 121

previous studies in mind,39 and aiming to develop new122

chelators not derived from 8-hydroxy/8-amino quinoline cores, 123

we hypothesized that the scaffold 2-(((2-((pyridin-2- 124

ylmethyl)amino)ethyl)amino)methyl)phenol, Salpyran (Fig- 125

ure 1) would be an ideal therapeutic Cu(II) targeting SMMC. 126

Herein, we report the criteria considered in designing 127

Salpyran, its synthesis and characterization, solid-state and 128

solution studies, and ROS inhibition. 129

RESULTS AND DISCUSSION 130

Scaffold Development.Several organic ligands, exclusive131

of hydroxy and aminoquinolines frameworks, have been 132

investigated as potential Cu(II) SMMCs. A recent review by 133

Hureau et al. highlights the pros and cons of these structures.39 134

Among them, tetradentate bis(pyridine), ENDIP, competes135

for both copper and zinc in Aβaggregates, preventing their 136

formation and solubilizing amyloid precipitates.53 Tetrahy- 137

drosalen (Salan) ligands are strong metal binders and offer 138

Figure 1.Previous and current SMMCs 2-(((2-((pyridin-2-ylmethyl)amino)ethyl)amino)methyl)phenol,Salpyran.

Figure 2.Development ofSalpyranby combining structures ofENDIPandSalan.

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX B

(3)

139antioxidant properties.54Storr et al. designed multifunctional

140carbohydrate ligands based around an N-methylated salan core

f2 141(Salan-1, Figure 2).55 The pendant glucose arm facilitates

142access to the brain and passes through the BBB via glucose

143transporters. Both ligands were found to have significant

144antioxidant properties in vitro. The O-glycosylation of the

145Salan ligand was also investigated as a prochelator strategy,

146where the glucose moiety effectively masks the coordination

147pocket until hydrolysis occursin vivo.56,57It was confirmed that

148the enzyme Agrobacterium sp. β-glucosidase could effectively

149cleave the C−O bond of the glucose moiety releasing the N-

150methylated Salan scaffold as the active chelator at the site.

151Other attempts to improve the pharmacological profile of the

152core Salan scaffold have involved the sulfonation of the

153phenolic groups (Salan-2), which significantly improves

t1 154solubility (Table 1).58 However, it is expected that the

155presence of an ionizable sulfonate group will result in poor

156BBB permeability, making it unsuitable for AD treatments.

157 Having all these in mind and building on our recent work in

158nonsymmetric salan ligands,59 we envisaged that the

159combination of theSalan and Endipmoieties should yield a

160nonsymmetric ligand,Salpyran(Figure 2). By breaking theC2

161symmetry, a new 3N,O coordination environment is formed

162that may partially fulfill the coordination environment of the

163Cu(II) center. Salpyran offers the same number of

164heteroatoms as TDMQ-20. Pearson’s acid−base principle

165predicts that the addition of the pyridine will increase the

166Cu(II) affinity and selectivity versus theSalanscaffold; this is

167observed in the trend of pCu values observed for more

168nitrogen-rich coordination pockets (Table 1). Also, compared

169to theSalan(cLogP = 2.26,Table 1) scaffold replacement of a

170phenol with a pyridine entity improves the aqueous solubility

171by reducing the lipophilicity of the scaffold (cLogP = 1.73,

172Table 1). However, the phenolic moiety provides the scaffold

173with radical scavenging capabilities to act as an antioxidant

during AD treatments.54Our approach introduces an entirely 174

different scaffold for use in AD treatment, contrasting the more 175

classical approach of modifying known metal coordinating 176

scaffolds.54,56−58,60−63 177

Thermodynamic and Physiochemical Properties 178

Compared to Other Cu Chelators. The selectivity of the 179

ligand for Cu(II) over other metal ions is a critical factor in 180

designing Cu(II) targeting SMMC. The chelator in question 181

should have high selectivity toward copper to minimize 182

competition with other essential metal ions and interactions 183

with other metalloproteins. The stability constant (log β) of 184

the metal complex (ML) is used to assess the affinity of a 185

ligand for a specific metal (eq 1). Therefore, in designing AD186

therapeutics, it is beneficial to compare the stability constants 187

for Cu(II) and Zn(II) due to the high concentration of Zn(II) 188

in AD brains.64,65The variability in the method and conditions 189

used to measure the metal/ligand affinities has led to the use of 190

the pM (eq 2) value when comparing and assessing the 191

chelation capability of copper targeting SMMCs. The pM value 192

is calculated at physiological pH and micromolar metal and 193

ligand concentrations. Consequently, this offers added benefit 194

by comparing chelators regardless of denticity or metal/ligand 195

stoichiometry. 196

β

+ + ↔ [ ]

= [ ]

[ ] [ ] [ ] mM lL hH M L H log log M L H

M L H

m l h

m l h

m l h

MLH

i

kjjjjj y

{zzzzz

(1) 197

= − [ ]

pM log Mfree (2) 198

Synthesis of Salpyran.Salpyrancan be synthesized via a 199

stepwise protecting group strategy in which consecutive 200

reductive aminations using salicylaldehyde and 2-formylpyr- 201 202 s1

idine take place across an ethylenediamine backbone (Scheme

203 s1

1, Figures S1−S10). First, the reductive amination of either Table 1. Thermodynamic and Calculated Pharmacological Relevant Properties of Chelators Targeting Cu(II) Homeostasis in Alzheimer’s Disease*

*Constants are for the form Aβ1-x.apM =log[M]free; [M] = [L] = 10μM, pH = 7.4.bCalculated from conditional anity value.cCalculated from apparent anity value at pH = 7.4.dCu/Zn selectivity calculated by pCupZn.eCoordination environment in solid state; equatorial (eq) and apical (ap) sites.fCalculated using the SwissADME free to use webtool; both logPand logSand the consensus values.72

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX C

(4)

204aldehyde with N-Boc-ethylenediamine and subsequent depro-

205tection give the amine precursors (1) and (2) (Scheme 1). A

206second reductive amination, this time in the presence of

207stoichiometric base (NEt3), yields Salpyran. It is possible to

208modify Salpyranvia variation in the aromatic substitution of

209either aldehyde or by replacement of the diamine linker unit.

210Functionalization is also possible at either of the amine’s

211groups, makingSalpyrana highly tunable scaffold compared to

212similar symmetric structures. For this three-step synthesis, the

213total yield of Salpyran via route A is 49% and significantly

214drops to 19% for route B. The fact that there are two simple

215synthetic routes demonstrates the synthetic accessibility

216toward Salpyran, which offers flexibility in analogue design

217in further medicinal chemistry pursuits. In the development of

218drugs targeting neurodegenerative disorders, there has been a

219trend in the design of multifunctional drugs that contain

220structural moieties aiming to target multiple pathological

221features at once or the addition of bioisosteres or isosteres to

222modify the pharmacokinetic properties.66 This has led to an

223interest in multifunctional drugs containing a metal-binding

224unit;67−69 therefore, the high synthetic accessibility and

225tunability of Salpyranmay offer future opportunities for use

226in multifunctional drugs.

227 Complexation Behavior with Cu(II) and Zn(II). The

228protonation constants (Table S1) of Salpyran were

229determined by pH-metric titrations. Using these data, the

230stability constants of the Cu(II) and Zn(II) complexes were

t2 231calculated (Table 2). At low pH (<4) values, the dicationic

232[CuLH]2+ is the dominant species. At the same time, the

233phenolic hydroxyl group remains protonated and uncoordi-

234nated. Across the physiological pH values (7.4), the

monocationic [CuL]+ is the dominant species. In contrast, at235

high pH values (>11), a further deprotonation process occurs, 236

forming a neutral [CuLH−1] species likely via the deprotona- 237 238 f3

tion of a coordinated water molecule (Figure 3A,B). The aqueous solution behavior is alike for Zn(II); however, no 239

protonated complex is formed. At pH 5, 50% of Zn(II) is 240

found unbound (Figure 3C). In all, the stability of the formed241

Zn(II) species is lower than that of the corresponding Cu(II) 242

species, demonstrating the Cu(II) selectivity of Salpyran 243

(Table 1). The species distribution plots of the Cu(II) 244

complexes formed in equimolar metal to ligand solutions are 245

shown inFigure 3. Further solution studies with Cu(II) were 246

performed in a mixture of DMSO:H2O (70:30), as it is a247

common practice for biological studies. Notably, the ligand 248

behavior changes drastically, corroborated by UV−vis studies 249

(Figure 3D,E), showcasing the formation of other species and250

indicating that speciation is highly dependent on the solvent 251

system (Table S2). In the less polar DMSO-containing solvent 252

mixture, the positively charged [CuL]+ species is dominant in253

both systems in the physiological pH range (Figure 3) but is 254

present in a narrower pH range, and the formation of the 255

neutral [CuLH−1] species is favorable. Based on this evidence, 256

we considered that solution studies in DMSO solution would 257

add no value to our conclusion. 258

The complex formation ofSalpyranwith Cu(II) and Zn(II)259

ions was studied at 1:2 and 1:1 ligand to metal ion ratios in the 260

pH range 3−11 (Figure S11). Comparison of the UV−vis 261

spectra of the Cu(II)-Salpyransystem at 1:2 and 1:1 metal to 262

ligand ratios (Figure S12) shows that similar spectra are 263

obtained. These studies indicate that irrespectively of the metal 264

to ligand ratio, only the 1:1 complex forms at pH values 265

ranging from 3 to 11. Therefore, we assume that duringin vivo 266

studies, the 1:1 species is dominant, reducing the possibility of 267

interactions with endogenous metalloproteins 268

The thermodynamic properties and drug-likeness of 269

Salpyran and other chelators discussed in this work are 270

summarized inTable 1. The affinity of the ligands for Cu(II) 271

and Zn(II) is measured using pCu and pZn values calculated 272

from the reported conditional (logβcon) or apparent (logβapp) 273

stability constants using [M] = [L] = 10 μM, p.H = 7.4. This 274

was achieved using the Hyperquad simulation and speciation 275

(HySS) software.71 Copper/zinc selectivity is given as pCu/276

Scheme 1. Two Alternate Synthetic Routes Towards Salypyran Starting from N-Boc-ethylenediamine and Using Either (A) Salicylaldehyde or (B) 2-Formylpyridine

Table 2. Stability Constants (logβ) for Salpyran complexes with Cu(II) and Zn(II) in Aqueous Solution Calculated Using the SUPERQUAD software (ref 70)*

*I = 0.2 mol ×dm−3 KCl,T = 298 K, standard deviations are in parentheses.

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX D

(5)

pZn; the larger the value, the greater selectivity toward Cu(II) 277

over Zn(II). Also included inTable 1is the stoichiometry and 278

coordination environment of the copper complexes according 279

to the reported solid-state structures. The drug-likeness of the 280

ligands has been predicted using the SwissADME web tool, 281

and the calculated physicochemical properties and predicted 282

BBB permeation and gastrointestinal absorption are also 283

given.72Ideally, any SMMC would follow the ‘ Lipinski rule 284

of 5’73and have a topological polar surface not exceeding 140 285

Å2 (Veber rule).74 In all, the complexation behavior of 286

Salpyran supports its potential use as a Cu(II) targeting 287

SMMC. It has an exceptional affinity for Cu(II) (pCu = 10.65) 288

and good selectivity for Cu(II) over Zn(II) (Cu/Zn = 4.60) 289

(Table 1). Salpyran acts as a tridentate or tetradentate, 290

dependent on the pH and only forms the 1:1 complex with 291

Cu(II). (The characteristic bands of the Cu(II) complexes are 292

summarized in Table S3.)Salpyran has a higher affinity and293

selectivity for copper when compared to its C2 symmetric 294

analogues,ENDIPandSalanand comparable affinity but with 295

lower selectivity when compared toTMDQ-20(Table 1). 296

FromTable 1, it is evident thatSalpyranoffers both high 297

affinity and selectivity for Cu(II) (pCu = 10.65, Cu/Zn = 4.6).298

Compared to both “parent” ligands, EDNIP and Salan, 299

Salpyranoutperforms, and its values are close to the state of 300

the artTDMQ-20(pCu = 10.75, Cu/Zn = 5.06).Salpyranhas301

good solubility, and its calculated log P value suggests that 302

good BBB permeation could be expected, although the number 303

of hydrogen bond donors (HBD = 3) may be deleterious to 304

BBB influx and may need to be factored into future drug 305

design (e.g., masked HBDs, rigidification).80,81 306

Salpyran Copper Crystal Structure. To better under-307

stand the complexation behavior ofSalpyranwith Cu(II), we 308

carried out several complexation reactions in protic or aprotic 309

solvents. The reflux of an equimolar solution of Salpyran, 310

CuCl2, and NEt3for 1 h in methanol yielded a viscous, green 311

oil, which upon dissolving in DMF, followed by vapor diffusion 312

of diethyl ether over 1 week, yielded blue crystals suitable for 313

single X-ray diffraction in low yield (13%,Tables S4 and S5). 314 315 f4

The solid-state structure is shown in Figure 4. Upon complexation with CuCl2, Salpyran yields an asymmetric 316

Cu(II)-dimer consisting of two different (CuCl2HL) units, and 317

Cl2serves as a bridge of these two entities. The coordination318

geometry of the two Cu centers varies; Cu1 adopts a 3N,2Cl 319

coordination environment (square pyramidal), while Cu2 320

adopts a 3N,3Cl environment (distorted octahedron) (Figure321

4); notably, both phenol moieties remain protonated. This 322

observation is in line with the potentiometric studies, which 323

suggest that at low pH values (pH < 4), the [CuHL]2+species 324

is dominant. The crystal structure confirms that the ligands 325

exhibit twofive-membered chelated rings via coordination of326

the three nitrogen donor atoms (NH, NH, Npy), which may327

account for the high stability of [CuHL]2+ species (Table 1). 328

Moreover, a close inspection of bond lengths and angles 329

(Table S3) reveals three different Cu−Cl bond types: Cl2 and 330

Cl3 strongly bind to Cu1 and Cu2, respectively, [2.2780(14) Å 331

and 2.2649(15) Å], the Cu1−Cl1 [2.6466(15) Å] and Cu2− 332

Cl4 [2.7294(15) Å] are weakened bonds, while the value of 333

the Cu2−Cl2 bond is 3.0454(15) Å, which is indicative of a334

secondary, very weak interaction.82 335

Further attempts to isolate crystals of the complex with the 336

deprotonated ligand were unsuccessful. HRMS of the isolated 337

crystals and viscous green oil is provided in the Supporting 338

Information (Figures S13 and S14) and is in line with the 339

Figure 3.(A, B) Species distribution and UVvis data of the Cu(II)- Salpyrancomplexes formed in the equimolar solutions as a function of pH in H2O. (C) Species distribution of the Zn(II)-Salpyran complexes formed in the equimolar solutions as a function of pH. (D, E) Species distribution and UVvis data of the Cu(II)-Salpyran complexes formed in the equimolar solutions as a function of pH in mixture DMSO:H2O (70:30).

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX E

(6)

340[CuL]+ and [CuHL]2+ structures, respectively. In all, taking

341into account that (a) differentiation in Cu−Cl bonding is due

342to the weakly binding character of the Cl anion, (b) solution

343studies were carried out using CuCl2stock solutions, (c) UV−

344vis studies suggest the existence of a Cu,3N (low pH value)

345and Cu,3N,O (physiological pH values) chromophores, and

346(d) ESI-MS studies corroborate the existence of monomeric,

347not dimeric species, in methanolic or aqueous solution, we can

348correlate the solid and solution phases and confirm the

349dominance of the [CuL]+ species at physiological pH values.

350 Antioxidant Properties.Redox-active Cu(II) is known to

351induce ROS formation and oxidative stress accumulation.83

352Therefore, potential therapeutic SMMCs must be capable of

353effectively inhibiting Cu(II)-induced ROS formation. As a

354starting point, we adopted a recently reported protocol83and

355investigated the ability of Salpyran to arrest the ROS

356production by monitoring ascorbate consumption under

357three different conditions (open air, Ar, and sealed cuvette).

358The ascorbate consumption is plotted as a function of time in

f5 359seconds (Figure 5 and Figures S15−S20). The ascorbate

consumption withoutSalpyranwas followed for 2 h, while in 360

the presence ofSalpyran, the samples were monitored for 3 h. 361

Samples were prepared in situ from stock solutions in 100 mM 362

HEPES buffer at pH 7.1, and the pH was adjusted with 0.2 M 363

HCl. The components were added in the following order: 364

HEPES, HCl, water, ascorbate, CuCl2, andSalpyran(if any).365

The assay was carried out under anaerobic and aerobic 366

conditions. In the anaerobic studies, the ascorbate con- 367

sumption was not completed even after 2 h, while under 368

aerobic conditions, the ascorbate is fully consumed in 1.5 h. 369 370 t3

The calculated rate constants (from 5000 to 10,000 s,Table 3) for the samples containingSalpyranare under argon, 1.07 ×371

10−9Ms−1(=1.07 nMs−1), in open air, 1.37×10−9Ms−1(1.37372

nMs−1), and in a sealed cuvette, 1.36 × 10−9 Ms−1 (=1.36373

nMs−1). The rate constants were calculated by dividing the 374

slope by the extinction coefficient of ascorbate,ε= 14,500 M−1 375

cm−1. Any difference in rates with the reported protocol83may 376

be attributed to the stirring rate (300 rpm over 800 rpm) and 377

ligand framework. These studies clearly show Salpyran slows378

Figure 4.Solid-state structure of protonatedSalpyran−copper complex.

Figure 5.Kinetics of ascorbate consumption with(out)Salpyranin different conditions (open air, Ar, and sealed cuvette). The reactantsSalpyran (if any)/CuCl2/ascorbate (12μM/10μM/100μM) ratio.

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX F

(7)

379the ascorbate consumption, thus demonstrating its capability

380to prevent ROS production.

381 Also, we investigated Salpyran’s oxidation ability in the

f6 382presence of H2O2 (Figure 6 and Figures S21 and S22). The

383reaction mixtures containing 1.0 mM Salpyran at metal to

384ligand molar ratio 1:1 were incubated at 25 °C for different

385time periods in the presence of H2O2at ligand to H2O2molar

386ratio 1:4. The pH was adjusted to 7.4. The reaction was

387initiated by the addition of a freshly prepared 1% H2O2

388solution. The reaction was stopped by the addition of

389Na2EDTA at ligand to Na2EDTA ratio 1:5. The reaction

390process was monitored by analytical RP-HPLC using a Jasco

391instrument, equipped with a Jasco MD-2010 plus a multi-

wavelength detector. From these data, it is evident that 392

oxidation does not occur in the sample containing equivalent 393

amount of Cu(II) andSalpyraneven after 2 days (Figure S21, 394

upper). While in the sample containing 4-fold excess 1% H2O2, 395

some oxidation occurs in thefirst 4 h (Figure S21, lower). 396

Then, we assessed the ability of Salpyran in preventing 397

Cu(II)-catalyzed oxidation in two different protein fragment 398

assays at physiological pH values. It has previously been shown 399

that a fragment of the human prion protein (HuPrP(103− 400

112), dMKHM) (Figure S23) undergoes oxidation in the 401

presence of radicals formed from the Cu(II)/H2O2system.84 402

The oxidation occurs only at the methionine residues, yielding 403

three main products: two singly oxidized products (dMKHM + 404

O, orange) and a doubly oxidized product (dMKHM + 2O, 405

yellow). Both methionine residues at position 7 (Met109) or/ 406

and at position 10 (Met112) can be oxidized. However, only 407

methionine sulfoxides are produced and not the corresponding 408

sulfones. The oxidation was initiated by adding H2O2 to an409

equimolar Cu(II)-dMKHM-Salpyran solution, and the reac- 410 411 f7

tion was monitored by HPLC for 1 day (Figure 7). After 1 h, almost 60% of HuPrP(103−112) remains intact, and no412

oxidation occurs in any methionine group, three times higher 413

than the blank experiment. In contrast, after 2 h, the 414

Table 3. Calculated Rate Constants*for Kinetics of Ascorbate Consumption in Different Conditions with Ratio Salpyran/CuCl2/Ascorbate (12μM/10μM/100μM)

*In nMs−1.

Figure 6.(upper) Ratio of the Cu(II)/H2O2oxidized prion protein fragment, HuPr(103112) (dMKHM), formed products with and without Salpyran. (lower) An HPLC chromatograph of the oxidation process 0 min, 10 min, 60 min, 120 min, and 1 day. Teknokroma Europa Protein C18 (250×4.6 mm, 300 Å, 5μm) column at aow rate of 1 mL·min−1, monitoring the absorbance at 222 nm. Mobile phases were water (A) and acetonitrile (B) containing 0.1% TFA.

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX G

(8)

415percentage of dMKHM is still high (40%, doubled compared

416to that of the blank), while unreacted dMKHM parts are still

417evident after 1 day (HPLC,Figure 6). These data demonstrate

418Salpyran’sefficiency in hindering the oxidation of the peptide,

419possibly by protecting the Cu(II) ions and inhibiting the ROS

420formation from the binary Cu(II)/H2O2system. The lack of

421total inhibition of peptide oxidation was not observed,

422potentially due to an excess of peroxide used in the experiment.

423These results demonstrate the potential of Salpyran in

424targeting Cu(II) dyshomeostasis and reducing the oxidative

425stress associated with neuronal death.

426 One known product of oxidation induced by Cu(II) is

427dityrosine cross-links on proteins, such as Aβ.85 Dityrosine

428(DiY) formation, whereby closely spaced tyrosines covalently

429cross-link by ortho−ortho coupling at C3 of their benzene

430rings, has been used as a marker of oxidative stress, and DiY

431has been shown to form under Cu(I/II)/H2O2 oxidative

432conditions for Aβand tauin vitro86−89and within AD amyloid

433plaques in vivo.87 In the presence of H2O2, Cu(II) induces

434dityrosine cross-linking more efficiently, serving as an excellent

435marker of oxidation.89Also, Cu(II) is known to bind tau and

436induce tau oxidation, dimerization, and aggregation.90,91

437Recently, it was demonstrated that Cu(II) alone or in the

438presence of H2O2 induces oxidation and dityrosine cross-

439linking of a tau297−391 fragment which contains one tyrosine

440at position 310.89,92 To further demonstrate the antioxidant

441ability of Salpyran, we performed a series of reactions using

442tau297−391 and Cu(II) (1:10 ratio) in combination with

443H2O2 to induce oxidation and dityrosine formation, which

444were quenched after 1 h with the addition of EDTA. The

445appearance of the dityrosine species was observed by

446monitoring the intensity of the peak at 410 nm (Figure 7).

447Unlike the reactions with just Cu(II) or more so in

448combination with H2O2, which showed robust induction of

449dityrosine to approximately 1% and 7% dityrosine levels

450(Figure S24), similar reactions mixed with Salpyran showed

451no dityrosine cross-linking alongside the controls (below 0.5%)

452(Figure 7). This suggests that Salpyran effectively prevents

453dityrosine formation and thus oxidation of dGAE via binding

to Cu(II). Combined with the aforementioned antioxidant 454

studies, these results indicate that Salpyran can reduce ROS 455

production in both Cu(II)/H2O2 and Cu(II)/O2/reductant 456

systems. 457

CONCLUSION 458459

We rationally designed and synthesized a highly modifiable copper chelating scaffold, Salpyran. This tetradentate ligand460

offers a 3N,O coordination environment and possesses good 461

drug-likeness.Salpyranexhibits an extremely high affinity for 462

Cu and excellent Cu(II) selectivity over Zn(II), comparable to 463

the state of the art components. Solid and solution studies 464

corroborate variation in coordination behavior at different pH465

values, but confirm the existence of only one dominant species 466

at physiological pH values in aqueous solutions. Under 467

physiological pH values and unaerobic conditions, the 468

[Cu(II)(3N,1O)]+ complex remains intact for at least 2 469

days, while in the presence of H2O2, an oxidation procedure 470

occurs. Further studies showcase that Salpyran slows the471

ascorbate consumption, thus preventing ROS production. 472

Finally, two different protein fragment assays that investigate 473

antioxidant properties revealedSalpyran’sexcellent efficacy to 474

prevent the formation of ROS from Cu(II)/H2O2. Due to its 475

drug-likeness, desirable coordination behavior, antioxidant 476

properties, and tunability, Salpyran is an alternative scaffold 477

to 8-hydroxy/aminoquinolines for further pharmaceutical 478

development of Cu(II) targeting drugs in neurodegenerative 479

disorders such as AD. 480

ASSOCIATED CONTENT 481

* Supporting Information 482

The Supporting Information is available free of charge at 483

https://pubs.acs.org/doi/10.1021/acs.inorgchem.1c01912. 484

Copies of1H,13C NMR, HRMS, and LCMS data for the 485

ligand (PDF) 486

Accession Codes 487

CCDC 2090343 contains the supplementary crystallographic 488

data for this paper. These data can be obtained free of charge 489

Figure 7.Fluorescence monitoring of the formation of dityrosine bridges from Cu(II)/H2O2oxidation of the tau dGAE fragment. Reactions were prepared usingμM dGAE mixed with Cu(II) at a 1:10 ratio or in combination with 2.5 mM H2O2to induce oxidation and dityrosine cross-linking.

A separate dGAE reaction was prepared with Salpyran at a 1:10 ratio orSalpyran in combination with Cu(II) at a 1:1 ratio alone and in combination with 2.5 mM H2O2. The reactions were quenched after 1 h with the addition of 2 mM EDTA.

https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX H

(9)

490via www.ccdc.cam.ac.uk/data_request/cif, or by emailing

491data_request@ccdc.cam.ac.uk, or by contacting The Cam-

492bridge Crystallographic Data Centre, 12 Union Road,

493Cambridge CB2 1EZ, UK; fax: +44 1223 336033.

494

AUTHOR INFORMATION

495Corresponding Authors

496 George E. Kostakis−Department of Chemistry, School of Life

497 Sciences, University of Sussex, Brighton BN1 9QJ, United

498 Kingdom; orcid.org/0000-0002-4316-4369;

499 Email:G.Kostakis@sussex.ac.uk

500 John Spencer−Department of Chemistry, School of Life

501 Sciences, University of Sussex, Brighton BN1 9QJ, United

502 Kingdom; orcid.org/0000-0001-5231-8836;

503 Email:j.spencer@sussex.ac.uk

504 Csilla Kállay−Department of Inorganic and Analytical

505 Chemistry, University of Debrecen, H-4032 Debrecen,

506 Hungary; Email:kallay.csilla@science.unideb.hu

507Authors

508 Jack Devonport −Department of Chemistry, School of Life

509 Sciences, University of Sussex, Brighton BN1 9QJ, United

510 Kingdom

511 Nikolett Bodnár−Department of Inorganic and Analytical

512 Chemistry, University of Debrecen, H-4032 Debrecen,

513 Hungary

514 Andrew McGown−Department of Chemistry, School of Life

515 Sciences, University of Sussex, Brighton BN1 9QJ, United

516 Kingdom

517 Mahmoud Bukar Maina−Sussex Neuroscience, School of Life

518 Sciences, University of Sussex, Brighton BN1 9QJ, United

519 Kingdom; College of Medical Sciences, Yobe State University,

520 PMB 1144 Damaturu, Yobe State, Nigeria

521 Louise C. Serpell −Sussex Neuroscience, School of Life

522 Sciences, University of Sussex, Brighton BN1 9QJ, United

523 Kingdom; orcid.org/0000-0001-9335-7751

524Complete contact information is available at:

525https://pubs.acs.org/10.1021/acs.inorgchem.1c01912

526Author Contributions

527All authors contributed to writing the manuscript and

528approved itsfinal version. J.D. devised the project with critical

529input and comments from G.E.K, J.S., and C.K. J.D. designed,

530synthesized, and characterized the ligand and performed and

531evaluated, with G.E.K., the crystallographic data. C.K. and N.B.

532performed and evaluated the potentiometry, UV−vis, and

533human prion fragment studies. L.S. and M.B.M. performed and

534evaluated the dityrosine studies. A.M. provided valuable

535feedback and comments.

536Notes

537The authors declare no competingfinancial interest.

538

ACKNOWLEDGMENTS

539G.E.K. and J.S. received funding from the School of Life

540Sciences, the University of Sussex (J.D. Ph.D. fellowship). C.K.

541and N.B. thank the Hungarian Scientific Research Fund

542(NKFI-115480 and NKFI- 124983) for its financial support.

543The research was also supported by the János Bolyai Research

544Scholarship of the Hungarian Academy of Sciences and by the

545ÚNKP-20-5 New National Excellence Program of the Ministry

546for Innovation and Technology from the source of the

547National Research, Development, and Innovation Fund. M.M.

is funded by Alzheimer’s Society (AS-PG-16b-010). L.C.S. is 548

supported by funding from BBSRC (BB/S003657/1). 549

(1)REFERENCESToni, M.; Massimino, M. L.; De Mario, A.; Angiulli, E.; Spisni, E. 550551

Metal Dyshomeostasis and Their Pathological Role in Prion and 552

Prion-like Diseases: The Basis for a Nutritional Approach. Front.553

Neurosci.2017,11(JAN), 3. 554

(2)Sayre, L. M.; Moreira, P. I.; Smith, M. A.; Perry, G. Metal Ions 555

and Oxidative Protein Modification in Neurological Disease.Ann. Ist.556

Super. Sanita2005,41(2), 143−164. 557

(3)Bonda, D. J.; Lee, H. G.; Blair, J. A.; Zhu, X.; Perry, G.; Smith, 558

M. A. Role of Metal Dyshomeostasis in Alzheimer’s Disease. 559

Metallomics2011,3(3), 267−270. 560

(4)Bolognin, S.; Messori, L.; Zatta, P. Metal Ion Physiopathology in 561

Neurodegenerative Disorders. NeuroMol. Med. 2009, 11 (4), 223− 562

238. 563

(5) Zhou, C.; Huang, Y.; Przedborski, S. Oxidative Stress in 564

Parkinsons Disease: A Mechanism of Pathogenic and Therapeutic 565

Significance.Ann. N. Y. Acad. Sci.2008,1147, 93−104. 566

(6) Ayton, S.; Lei, P.; Bush, A. I. Metallostasis in Alzheimer’s567

Disease.Free Radical Biol. Med.2013,62, 76−89. 568

(7) Barnham, K. J.; Masters, C. L.; Bush, A. I. Neurodegenerative 569

Diseases and Oxidative Stress.Nat. Rev. Drug Discovery2004,3(3), 570

205−214. 571

(8) Bush, A. I. Metals and Neuroscience. Curr. Opin. Chem. Biol. 572

2000,4(2), 184191. 573

(9)Greenough, M. A.; Camakaris, J.; Bush, A. I. Metal Dyshomeo- 574

stasis and Oxidative Stress in Alzheimers Disease. Neurochem. Int. 575

2013,62(5), 540−555. 576

(10)Lane, C. A.; Hardy, J.; Schott, J. M. Alzheimer’s Disease.Eur. J. 577

Neurol.2018,25(1), 59−70. 578

(11) World Alzheimer Report 2018; Alzheimer’s Disease Interna- 579

tional: London, 2018. 580

(12)Mayeux, R.; Stern, Y. Epidemiology of Alzheimer Disease.Cold 581

Spring Harbor Perspect. Med.2012,2(8), a006239. 582

(13)Ferri, C. P.; Prince, M.; Brayne, C.; Brodaty, H.; Fratiglioni, L.;583

Ganguli, M.; Hall, K.; Hasegawa, K.; Hendrie, H.; Huang, Y.; Jorm, 584

A.; Mathers, C.; Menezes, P. R.; Rimmer, E.; Scazufca, M. Global 585

Prevalence of Dementia: A Delphi Consensus Study. Lancet 2005, 586

366(9503), 2112−2117. 587

(14)Fratiglioni, L. Epidemiology of Alzheimer’s Disease. Issues of 588

Etiology and Validity.Acta Neurol. Scand. Suppl.1993,145, 1−70. 589

(15)Bush, A. I.; Pettingell, W. H.; Multhaup, G.; Paradis, M. D.; 590

Vonsattel, J. P.; Gusella, J. F.; Beyreuther, K.; Masters, C. L.; Tanzi, R. 591

E. Rapid Induction of Alzheimer Aβ Amyloid Formation by Zinc. 592

Science1994,265(5177), 14641467. 593

(16) Huang, X.; Atwood, C. S.; Hartshorn, M. A.; Multhaup, G.;594

Goldstein, L. E.; Scarpa, R. C.; Cuajungco, M. P.; Gray, D. N.; Lim, J.; 595

Moir, R. D.; Tanzi, R. E.; Bush, A. I. The AβPeptide of Alzheimer’s596

Disease Directly Produces Hydrogen Peroxide through Metal Ion 597

Reduction.Biochemistry1999,38(24), 7609−7616. 598

(17)Atwood, C. S.; Huang, X.; Moir, R. D.; Tanzi, R. E.; Bush, A. I.599

Role of Free Radicals and Metal Ions in the Pathogenesis of 600

Alzheimers Disease.Met. Ions Biol. Syst.1999,36, 309364. 601

(18)Bush, A. I. The Metallobiology of Alzheimer’s Disease.Trends 602

Neurosci.2003,26(4), 207214. 603

(19)Maynard, C. J.; Bush, A. I.; Masters, C. L.; Cappai, R.; Li, Q.-X. 604

Metals and Amyloid-β in Alzheimers Disease. Int. J. Exp. Pathol. 605

2005,86(3), 147−159. 606

(20)Schrag, M.; Mueller, C.; Oyoyo, U.; Smith, M. A.; Kirsch, W.607

M. Iron, Zinc and Copper in the Alzheimer’s Disease Brain: A 608

Quantitative Meta-Analysis. Some Insight on the Influence of Citation 609

Bias on Scientific Opinion.Prog. Neurobiol.2011,94(3), 296−306. 610

(21)Cilliers, K. Trace Element Alterations in Alzheimer’s Disease: A 611

Review.Clin. Anat.2021,34(5), 766773. 612

(22)Kabir, M. T.; Uddin, M. S.; Zaman, S.; Begum, Y.; Ashraf, G.613

M.; Bin-Jumah, M. N.; Bungau, S. G.; Mousa, S. A.; Abdel-Daim, M. 614 https://doi.org/10.1021/acs.inorgchem.1c01912 Inorg. Chem.XXXX, XXX, XXX−XXX I

Ábra

Figure 2. Development of Salpyran by combining structures of ENDIP and Salan.
f2 141 (Salan-1, Figure 2). 55 The pendant glucose arm facilitates
Table 2. Stability Constants (log β ) for Salpyran complexes with Cu(II) and Zn(II) in Aqueous Solution Calculated Using the SUPERQUAD software (ref 70) *
Figure 5. Kinetics of ascorbate consumption with(out) Salpyran in different conditions (open air, Ar, and sealed cuvette)
+3

Hivatkozások

KAPCSOLÓDÓ DOKUMENTUMOK

For all other alcohols, including EtOH, 1-PrOH, 2-PrOH and 2-BuOH, the selectivity to carbonyl compounds in the liquid phase oxidation reactions were either similar or higher than

Carbonate species arising from surface reaction of CO 2 with water adsorbed on Pt (where H 2 O acts as a source of oxygen for the oxidation reaction [58,63]) are reported to

The formation rate of methane during CO 2 hydrogenation reaction in the case of (a) pure, Pt-loaded and Cu-loaded catalysts prepared by using different milling speed and

Experimental results were confronted with calculated single-particle cross sections ( σ sp ) and momentum distri- butions of neutron removal from p 1=2 , p 3=2 , f 5=2

Resident mouse peritoneal macrophages synthesize and release large amounts of PGE2 when exposed in culture to various in- flammatory stimuli such as zymosan, phorbol myristate

methohexital is stable for 6 weeks.. unconsciousness in 60 sec with a peak effect in 1 to 2 min with duration of anesthesia between 5 min to 10 min. If benzodiazepines, opiates, or α

&#34;Diastereoselective [2+2]-Cycloaddition Reactions of Unsymmetrical Cyclic Ketenes with Imines: Synthesis of Modified Prolines and Theoretical Study of the

Vagy egyszerűen, túl- erőben voltak, többen lehettek, mint azok heten, és arra ment a harc, hogy kifosszák őket, ami nyilván sikerült is nekik, mert különben jóval több