Next Article in Journal
Repurposing Dimethyl Fumarate Targeting Nrf2 to Slow Down the Growth of Areas of Geographic Atrophy
Previous Article in Journal
Immunothrombosis in Sepsis: Cellular Crosstalk, Molecular Triggers, and Therapeutic Opportunities—A Review
Previous Article in Special Issue
Recent Advances in the Search for Effective Anti-Alzheimer’s Drugs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel Au(I)- and Ag(I)-NHC Complexes with N-Boc-Protected Proline as Potential Candidates for Neurodegenerative Disorders

by
Jessica Ceramella
1,†,
Assunta D’Amato
2,†,
Francesca Procopio
3,
Annaluisa Mariconda
4,
Daniel Chavarria
5,*,
Domenico Iacopetta
1,*,
Francesco Ortuso
3,
Pasquale Longo
2,
Fernanda Borges
6,7,8,‡ and
Maria Stefania Sinicropi
1,‡
1
Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende, Italy
2
Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
3
Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
4
Department of Basic and Applied Sciences (DISBA), University of Basilicata, Via Dell’Ateneo Lucano 10, 85100 Potenza, Italy
5
CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
6
MedInUP, Center for Drug Discovery and Innovative Medicines, University of Porto, 4200-319 Porto, Portugal
7
Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
8
Department of Biomedicine-Pharmacology and Therapeutics Unit, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Co-senior authors.
Int. J. Mol. Sci. 2025, 26(13), 6116; https://doi.org/10.3390/ijms26136116
Submission received: 16 May 2025 / Revised: 20 June 2025 / Accepted: 23 June 2025 / Published: 25 June 2025

Abstract

Neurodegenerative diseases (NDDs), including Alzheimer’s disease (AD) and Parkinson’s disease (PD), are characterized by progressive neuronal dysfunction and loss and represent a significant global health challenge. Oxidative stress, neuroinflammation, and neurotransmitter dysregulation, particularly affecting acetylcholine (ACh) and monoamines, are key hallmarks of these conditions. The current therapeutic strategies targeting cholinergic and monoaminergic systems have some limitations, highlighting the need for novel approaches. Metallodrugs, especially ruthenium and platinum complexes, are gaining attention for their therapeutic use. Among metal complexes, gold(I) and silver(I) N-heterocyclic carbene (NHC) complexes exhibit several biological activities, but their application in NDDs, particularly as monoamine oxidase (MAO) inhibitors, remains largely unexplored. To advance the understanding of this field, we designed, synthesized, and evaluated the biological activity of a new series of Au(I) and Ag(I) complexes stabilized by NHC ligands and bearing a carboxylate salt of tert-butyloxycarbonyl (Boc)-N-protected proline as an anionic ligand. Through in silico and in vitro studies, we assessed their potential as acetylcholinesterase (AChE) and MAO inhibitors, as well as their antioxidant and anti-inflammatory properties, aiming to contribute to the development of potential novel therapeutic agents for NDD management.

1. Introduction

Neurodegenerative diseases (NDDs), given their substantial impact on the ageing population, are among the leading causes of disability and morbidity worldwide. These diseases are defined by a continuous decline in neuronal function, culminating in brain atrophy and significant functional limitations [1].
The most commonly known NDDs, Alzheimer’s and Parkinson’s diseases (AD and PD, respectively), are progressive and multifactorial forms of neurodegeneration. These diseases share several key characteristics, involving a complex interplay of genetic, environmental, and lifestyle factors, which lead to a progressive decline in physical, cognitive, and emotional functions. The underlying mechanisms include the degeneration of brain cells, accompanied with chronic neuroinflammation, neuronal death, and a gradual loss of brain tissue [1].
The “switch on” of the inflammatory cascade triggers multiple pathways and responses, like the production of reactive oxygen species (ROS) and cytokines, resulting in oxidative stress. Excessive ROS generation damages cellular components like proteins, lipids, and DNA, leading to neuronal dysfunction and death and contributing to the progressive decline observed in both AD and PD [2].
Together with oxidative stress and neuroinflammation, the deregulation of neurotransmitter systems represents an important hallmark that significantly contributes to the observed cognitive and motor deficits [3].
Particularly, the homeostasis of neurotransmitters including acetylcholine (ACh), butyrylcholine (BCh), and monoamines (such as dopamine, serotonin, and norepinephrine) plays a crucial role in various neurological and psychiatric conditions.
In AD, the loss of cholinergic neurons leads to reduced acetylcholine (ACh) levels, contributing to cognitive decline. Similarly, imbalances in monoamine neurotransmitters are linked to a range of disorders (e.g., PD, depression, anxiety, panic disorder, and attention-deficit/hyperactivity disorders).
The current therapeutic strategies, such as the inhibition of acetylcholinesterase (AChE), aim to increase the ACh availability to prevent cognitive decline and memory loss [4]. AChE is the main enzyme that hydrolyzes ACh; however, the effectiveness of acetylcholinesterase inhibitors (AChEIs) in treating ongoing AD may be limited by significant neuronal damage and the substantial, often irreversible, loss of AChE activity (>90%). Conversely, butyrylcholinesterase (BChE) levels can increase to up to 120% above physiological levels in ongoing AD. This BChE upregulation suggests the potential compensatory role of BChE inhibitors in disease progression [5].
Moreover, the development of monoamine oxidase inhibitors (MAOIs) in medicinal chemistry constitutes a critical area of investigation, driven by the need for safer and more effective treatments for various neurological and psychiatric disorders.
MAOIs are a class of drugs that inhibit the enzymes monoamine oxidase A (MAO-A) and/or B (MAO-B). MAO-A primarily metabolizes serotonin and norepinephrine; thus, its inhibition is therapeutically exploited to treat depression, anxiety, and social phobia. On the other hand, MAO-B inhibitors are employed in PD since this enzyme predominantly metabolizes dopamine [6].
In addition, elevated MAO expression and activity have been associated with increased oxidative stress, since the metabolism of monoamines generates their corresponding aldehydes, with the concomitant formation of H2O2 and ammonia as byproducts. In particular, H2O2 induces mitochondrial damage and neuronal apoptosis [7].
Recently, metallodrugs have garnered considerable attention within the scientific community as a potential therapeutic approach for treating NDDs [8,9]. Among the diverse complexes, ruthenium- and platinum-based metal complexes have emerged as prominent candidates for NDD therapy. The current research primarily focuses on AD, with an emphasis on Aβ-peptide targeting and AChE/BChE inhibition. However, the potential of metal complexes to modulate tau protein aggregation and MAO levels remains an area of significant research interest and needs further investigation [10].
Among metal complexes, N-heterocyclic carbene (NHC) complexes, particularly those bearing gold(I) and silver(I), exhibit a wide range of properties, including potent antimicrobial, anticancer, and anti-inflammatory effects [11,12,13,14].
The versatility of NHC ligands allows for the accurate regulation of the properties of these metal complexes, and the current research continues to explore their potential as promising candidates for the treatment of NDDs. However, to date, little literature data address this field of research, and, to the best of our knowledge, no information concerning the inhibition of MAOs by NHC metal complexes has been reported. Conversely, some results on the ability of these complexes to inhibit AChE have been published. For example, the in silico and in vitro cholinesterase (ChE) inhibitory activity of different benzimidazole- and imidazolidine-based Ag-NHC complexes has been studied, revealing IC50 values in the nanomolar range and binding mainly to the catalytic anionic sites of AChE and BChE [15,16,17].
Alongside the study of metal complexes in NDDs, experimental and clinical results have drawn attention to proline-rich polypeptides as potential therapeutic agents for treating AD and other neurodegenerative conditions [18,19]. While promising, further research is crucial to validate these findings and develop effective proline-based treatments.
To deepen the understanding of the protective role of both NHC complexes and proline in neurodegeneration, herein, we designed, synthesized, and biologically characterized, through in silico and in vitro studies, a new series of gold(I) and silver(I) complexes stabilized by NHC ligands and bearing a carboxylate salt of tert-butyloxycarbonyl (Boc)-N-protected proline as an anionic ligand (Figure 1, complexes 1aP4aP and 1bP4bP) as promising AChEIs and MAOIs. Their antioxidant and anti-inflammatory properties were also evaluated.

2. Results and Discussion

2.1. Chemistry

The synthetic approach to synthesize the new complexes 1aP4aP and 1bP4bP is illustrated in Scheme 1.
They were obtained by starting with the known anticancer active halide complexes 1a4a and 1b4b, previously reported by our research group (see Scheme 1A) [20].
The halide ions of the complexes were replaced by N-Boc-prolinate-protected anions through a metathesis reaction with N-Boc-prolinate-protected silver. Silver halide precipitates and the desired complex were formed according to procedures from the literature (see Scheme 1B) [11,21,22,23].
The co-assembled L-proline complexes were characterized by 1H-, 13C-NMR, and HRMS. In particular, the appearance of signals in the aliphatic region of the 1H-NMR spectra related to the tert-butoxycarbonyl protecting group and the methylene groups of the pyrrolidine ring indicated the presence of an amino acid. Moreover, as observed in the literature [23], an upfield shift of about 5 ppm in the carbenic carbons was detected by 13C-NMR, when compared to the parent halogenide complexes. This is evidence of the presence of a stronger σ-donating counteranion, such as a carboxylate group, instead of the halide, which was precipitated as silver iodide during the exchange process.

2.2. Cholinesterase-Inhibitory Activity

To assess the potential therapeutic value of the synthesized Ag (1aP4aP) and Au (1bP4bP) complexes, we first measured their ability to inhibit electric eel acetylcholinesterase (eeAChE) and equine butyrylcholinesterase (eqBChE). This evaluation followed the established Ellman method [24], using Donepezil as the reference ChE inhibitor. Table 1, reporting the IC50 values and SI, shows that the gold complex 4bP, which had a fused benzene ring at the carbenic ring, was the most potent ChE inhibitor, being more selective toward eqBChE. In particular, complex 4bP showed an eeAChE IC50 value of 3.72 ± 0.44 µM, which was higher than that of Donepezil (IC50 = (2.30 ± 0.53) × 10−2 µM). In contrast, complex 4bP was more active than Donepezil against eqBChE, showing an IC50 value about five times lower (0.45 ± 0.06 and 2.13 ± 0.12 µM for complex 4bP and Donepezil, respectively). Complex 2bP was less active against both enzymes than complex 4bP, exhibiting IC50 values of 4.68 ± 0.47 µM for eeAChE and 3.14 ± 0.33 µM for eqBChE. Thus, the presence of two chlorine atoms at the carbene of complex 2bP made it less effective than complex 4bP. Conversely, the gold complex 3bP, which bore two phenyl groups at the carbene, possessed slightly lower activity than 4bP and 2bP, but it was selective toward AChE. Indeed, it inhibited the activity of eeAChE with an IC50 value of 5.89 ± 0.56, while no inhibitory activity was recorded for eqBChE, at least until reaching a concentration of 25 µM. The absence of substituents at the carbenic ring, as in complex 1bP, led to a drastic loss of inhibitory activity against eeAChE compared with that of the other gold-based complexes (IC50 = 18.37 ± 1.43 µM).
All the Ag-based complexes were less active when compared with Au-based analogues. The most active Ag-based complex was 4aP, in which the carbene ring was fused with a benzene scaffold, with IC50 values of 13.95 ± 0.72 µM for eeAChE and 5.19 ± 0.95 µM for eqBChE. Complexes 2aP and 3aP, with two chloride groups or two phenyl groups at the carbene ring, acted as selective eqBChE inhibitors with IC50 values of 18.24 ± 2.4 µM and 17.38 ± 0.98 µM, respectively. Finally, the absence of substituents at the carbenic ring, as in complex 1aP, resulted in a total loss of inhibitory activity against both eeAChE and eqBChE at concentrations of up to 25 µM. Collectively, these outcomes suggest a higher selectivity of the Ag-based complexes 1aP4aP toward eqBChE, with IC50 values different to those of other NHC-Ag complexes reported in the literature showing higher selectivity toward AChE [17] or BChE [25] or that were non-selective [26], probably due to the presence of different and bulkier substituents at the carbene ring and the lack of a ProBoc scaffold. Conversely, to the best of our knowledge, less is still known about NHC-Au complexes.
In conclusion, gold-based complexes were better eeAChE and eqBChE inhibitors than silver-based complexes. The chemical structures of complexes 4bP and 3bP may represent a good starting point to develop selective AChE and BChE inhibitors.

2.3. MAO-A/B-Inhibitory Activity

Neurodegenerative diseases are often accompanied by a decline in multiple neurotransmitter systems, leading to cognitive and motor deficits. Both ChEs and MAOs are enzymes involved in neurotransmitter metabolism; therefore, targeting both enzymes can have a synergistic effect addressing multiple aspects of these diseases.
Altered levels of MAO-A/B are associated with the degeneration of neurotransmitter biogenic amines such as serotonin, noradrenaline, and dopamine and are implicated in several diseases, such as depression, cancer, PD, and AD [27]. With this in mind, we explored the potential of these complexes in inhibiting human MAO (hMAO) isoforms. Clorgyline and (R)-(–)-Deprenyl were used as reference inhibitors for hMAO-A and B, respectively. The obtained results are presented in Table 2.
All the gold complexes exhibited higher hMAO-inhibitory activity than the related silver analogues. Notably, all the gold-based complexes displayed a higher inhibition potency against hMAO-A. Specifically, complexes 4bP and 2bP emerged as the most active hMAO-A inhibitors. Complex 4bP, bearing a benzene ring fused to the carbene, demonstrated, approximately, a 10-fold-higher hMAO-A-inhibitory activity than complex 2bP, which bore two chlorine atoms at the carbene ring (the IC50 values for complexes 4bP and 2bP for hMAO-A were (7.13 ± 0.97) × 10−2 and 0.72 ± 0.02 µM, respectively). The activity of both complexes against hMAO-B was similar, with IC50 values of 2.10 ± 0.20 µM and 1.94 ± 0.09 µM for complexes 4bP and 2bP, respectively. The remaining gold complexes, 1bP and 3bP, showed lower and similar activity against both enzymes. Thus, the presence of either two hydrogen atoms (complex 1bP) or two phenyl groups (complex 3bP) at the carbene ring reduced the inhibitory activity against hMAOs. Regarding the silver complexes, the presence of a benzene scaffold fused with the carbene core in complex 4aP resulted in the highest inhibitory activity against hMAO-A (IC50 = 3.32 ± 0.15 µM). Conversely, complex 2aP, with two chlorine atoms in the carbene core, exhibited greater selectivity towards hMAO-B (IC50 = 9.82 ± 0.84 µM). Finally, consistent with the trend observed for gold complexes, the presence of two hydrogen atoms or two phenyl groups at the carbene core made complexes 1aP and 3aP completely inactive against both enzymes at concentrations of up to 25 µM. These findings suggest that the nature of the substituents in the carbene core plays a critical role in modulating the inhibitory activity of metal complexes against hMAO-A and hMAO-B. Moreover, the inhibitory activity against hMAO-A and -B mirrored that observed against ChEs. The recorded trend highlights the potential of NHC-Au and Ag complexes as promising starts for the development of MAO inhibitors to be further exploited for therapeutic applications, and, most importantly, the latter outcomes represent the first report that fills the gaps in the scientific literature data. Indeed, to date, many compounds bearing different chemical scaffolds have been reported to inhibit MAO isoforms, but they are not structurally related to the present series [7,28,29].

2.4. Anti-Inflammatory Activity

A growing amount of research highlights the role of multiple inflammatory pathways in the development and progression of different NDDs, suggesting that chronic neuroinflammation could significantly amplify disease progression. Thus, regulating the neuroinflammatory response could decrease the production of neurotoxic substances and potentially lead to improved clinical outcomes [30].
For this reason, the newly synthesized gold and silver complexes were tested for their potential anti-inflammatory activity, particularly their inducible nitric oxide synthase (iNOS)-inhibitory activity, by means of a Griess-based assay in lipopolysaccharide (LPS)-stimulated murine macrophages, RAW 264.7. As a positive control, the nonsteroidal anti-inflammatory drug (NSAID) Indomethacin (Ind) was used [31]. The RAW 264.7 cell viability after exposure to our complexes and the IC50 values for iNOS were calculated and are reported in Table 3. First, as can be observed in Figure 2, amongst the tested complexes, only complex 3bP produced a great and significant inhibition of NO production of about 52% in LPS-stimulated RAW 264.7 cells at a concentration of 5 µM. For the 3bP complex, the observed decrease in NO production with respect to that achieved with the LPS-only treatment (%) was statistically significant (p < 0.0001). At the same concentration, the silver-based analogue 3aP inhibited the NO production by approximately 8%. The obtained data demonstrate that both gold and the two phenyl groups at the carbene ring are important structural features required to obtain compounds able to inhibit iNOS. In fact, as can be seen from Table 3, the IC50 values related to the inhibition of NO production were 4.88 µM and 30.88 µM for complexes 3bP and 3aP, respectively. Furthermore, both complexes showed no toxicity effects on RAW 264.7 cells, as is evident from the cell viability IC50 values calculated for the RAW 264.7 cells (20.27 ± 1.26 µM and >50 µM for complexes 3bP and 3aP, respectively).
It should be noticed that Ind produced an NO inhibition of about 20% at 25 µM, making it 2.6 times less potent at a concentration five-fold higher compared to complex 3bP. Anti-inflammatory activity comparable to that of complex 3aP was also observed for complex 2bP, which showed a similar IC50 value for NO production of 32.31 ± 1.82 µM, without exhibiting toxicity towards murine macrophages up to a concentration of 50 µM (see Table 3). Moreover, complex 4bP possessed a good IC50 value for NO inhibition of 13.05 ± 0.92 µM, but it was proved to be toxic against RAW 264.7 cells (IC50 for cell viability = 12.12 ± 1.03 µM). The toxicity of complex 4bP effectively invalidated the NO inhibition data, as the observed reduction in NO was simply a consequence of cellular death. All the other complexes did not exhibit any anti-inflammatory activity at concentrations up to 50 µM under the employed experimental conditions. The data strongly suggest that the presence of gold and the two phenyl groups in the carbene core are key determinants of iNOS inhibition activity in this series of complexes. Contrarily, the concurrent presence of gold and a benzene ring fused with the carbene core strongly influenced the toxicity of the complexes for the murine macrophage RAW 264.7.

2.5. Antioxidant Activity

Considering that the exacerbation of intracellular oxidative stress has been implicated in several diseases, including NDDs, in this context, we evaluated the potential ROS scavenging activity of Ag (1aP4aP) and Au (1bP4bP) complexes in 3T3-L1 murine fibroblasts using a dihydro-2′,7′-dichlorofluorescein diacetate (H2DCF-DA) fluorescent probe. Menadione (Men, 25 µM, 15 min) was used to induce ROS production within the cells (Figure 3, brown histograms). N-acetyl-cysteine (NAC), a powerful antioxidant, was used as a positive control. The lack of cytotoxicity for all the compounds used in the antioxidant assay at 1 µM (compound 3bP) or 10 µM (compounds 1aP4aP, 1bP, 2bP, and 4bP) was ascertained using an MTT test performed under the same experimental conditions (Figure 3, salmon histograms). Only the concentrations that did not significantly affect the cell viability (around 95–99%) were chosen. The best antioxidant complexes were 4bP and 3bP, which reduced the ROS production to approximately 60 and 70% at 10 µM and 1 µM, respectively, compared to the Men treatment alone. These activities proved to be highly interesting, considering that NAC reduced the ROS formation to approximately 70%, but only at a concentration of 20 mM. The other complexes decreased the ROS generation by approximately 10–20%. These results clearly demonstrate that all the gold-based complexes showed better in vitro antioxidant capacities compared to the silver analogues. Among the gold-based complexes, the presence of the two phenyl groups as substituents in the carbene core made complex 3bP the best antioxidant, even at a concentration of 1 µM. It should be noted that complex 3bP also exhibited the best anti-inflammatory activity amongst all the considered complexes. Furthermore, the benzene scaffold fused with the carbene ring allowed complex 4bP to inhibit ROS production, although to a lesser extent. Conversely, the presence of two chlorine atoms or the absence of substituents produced a marked reduction in the antioxidant activity, as was evident for the gold complexes 2bP and 1bP.

2.6. Docking Studies

Molecular docking simulations were performed to investigate the molecular recognition profiles of a series of gold and silver complexes for hChEs, hMAOs, and iNOS. The theoretical binding energies were calculated as described in the Experimental Section. The predicted binding energies did not show a direct correlation with the experimentally determined IC50 values. This divergence may be attributed to several factors, including the limitations inherent to docking methodologies, the differential activity of enantiomers, the intrinsic limitations of the molecular mechanics energy evaluation of metal complexes, and the influence of solvent effects that were not fully captured in the simulations. Based on the experimental data, attention was focused on the complexes most active against the specified targets mentioned (2aP, 2bP, 4aP, and 4bP for hChEs and hMAOs and 3aP and 3bP for iNOS; Tables S1, S2, and S3).
In the case of hAChE, the docking results indicated that both 4aP and 4bP oriented the benzimidazole moiety toward the catalytic active site (CAS). Compound 4aP exhibited π–π stacking interactions with the key residues Trp82, Tyr337, and Tyr341. Similarly, 4bP formed a hydrogen bond with Tyr124 and π–π stacking with Trp82, His447, and Tyr124. For 2aP, the imidazole ring was directed toward the CAS, establishing π–π stacking and hydrogen bonds with Tyr341, Tyr337, and Tyr124. Conversely, 2bP adopted a flipped orientation toward the peripheral anionic site (PAS), where it interacted with Tyr341, Tyr124, and Trp286. The evaluated complexes also displayed favourable binding conformations within the hBChE active site. (R)4aP formed hydrogen bonds and π–π stacking interactions with Thr120 and Trp82, while (S)4aP engaged Trp231, Phe329, and His438 through π–π interactions. Compound 4bP showed enantiomer-dependent binding modes: (S)4bP primarily interacted with His438, whereas (R)4bP engaged Trp321 and Phe329. Regarding 2aP, the (R)-enantiomer established hydrogen bonds and π–π stacking with Phe329 and Leu286, while the (S)-enantiomer interacted with Trp231, Phe329, and Pro285. (S)2bP engaged in π–π stacking with Phe329, whereas (R)2bP adopted a flipped pose, establishing hydrogen and halogen bonds with His438 (Figure 4).
Docking simulations for hMAO-A revealed that the benzimidazole moiety of both 4aP and 4bP derivatives was oriented toward the flavin adenine dinucleotide (FAD) cofactor. Specifically, 4aP engaged in π–π stacking interactions with Tyr407 and Tyr444. The (S)-enantiomer further established additional π–π stacking and hydrogen bonds with Phe208 and Tyr407. In the case of 4bP, the (S)-enantiomer mainly established van der Waals interactions, whereas the (R)-enantiomer formed π–π stacking interactions with Phe352 and Tyr407, along with a hydrogen bond involving Asn181. For 2aP and 2bP, the imidazole ring was directed toward the FAD cofactor. An exception was observed with (R)2bP, which adopted a flipped orientation. The primary interactions were van der Waals interactions or, in some cases, π–π stacking with Phe352 for (R)2bP and with Tyr407 for the (S)-enantiomer. In hMAO-B, the docking results indicated that the benzimidazole group of 4aP and 4bP derivatives pointed toward the entrance of the catalytic cavity. 4aP showed enantiomer-specific interactions: the (R)-enantiomer exhibited π–π stacking with Trp189, and the (S)-enantiomer formed hydrogen bonds with Pro108. The imidazole moiety of 2aP and 2bP was oriented toward the entrance of the cavity. Notably, only the (S)-enantiomer of 2bP displayed a well-defined interaction pattern, including π–π stacking with Phe168, one halogen bond with Leu64, and one hydrogen bond with Pro102. The remaining derivatives were predominantly stabilized by non-specific van der Waals interactions (Figure 5).
Docking analyses targeting iNOS revealed that both 3aP and 3bP oriented the imidazole moiety toward the interior of the catalytic pocket, establishing key interactions with essential active site residues. In particular, the hydroxyphenylethyl side chain of both compounds engaged in a cation–π interaction with Arg375, while one of the biphenyl–imidazole units formed π–π stacking interactions with Tyr367 (Figure 6).
An in-depth analysis of the metals’ contribution to the activity of our compounds was carried out (Table S6), showing substantial equivalence between Ag and Au. In general, the desolvation energies, as computed by Autodock, revealed that the higher electronegativity of Au leads to a stronger penalty for Au-containing complexes with respect to that for the corresponding Ag analogues. On the other hand, the same property resulted in better electrostatic interaction between the Au-based compounds and the investigated targets.
In summary, the molecular docking studies provided valuable insights into the binding modes and interaction profiles of the most promising gold and silver complexes. The identified interactions with residues known to stabilize enzyme inhibitors support the experimentally observed activities. These findings offer a structural basis for the further optimization and design of more potent derivatives targeting these enzymes.

3. Materials and Methods

All the reagents were purchased from Merck Italy (Milan, Italy) and TCI Chemicals (Zwijndrecht, Belgium) and used without any purification. All the solvents were bought from Carlo Erba Reagents srl (Milano, Italy) and were distilled over appropriate drying agents under nitrogen before use. The synthesis of silver(I) and gold(I) complexes was carried out under a nitrogen atmosphere by using Schlenk techniques in the dark. The glassware used was dried in an oven at 120 °C overnight. Deuterated solvents were dried on molecular sieves. 1H and 13C nuclear magnetic resonance spectra (NMR) were acquired on a Bruker Avance 300 spectrometer (300 MHz for 1H; 75 MHz for 13C), a Bruker AVANCE 400 spectrometer (400 MHz for 1H; 100 MHz for 13C), and a Bruker AVANCE 600 spectrometer (600.13 MHz for 1H, 150.90 MHz for 13C) operating at 298 K. NMR samples were prepared by dissolving about 15 mg of the compound in 0.5 mL of deuterated solvent (Eurisotop Cambridge Isotope Laboratories, Cambridge, UK). The chemical shifts in the 1H-NMR and 13C-NMR spectra were referenced using the residual proton impurities of the deuterated solvents. The 1H-NMR spectra were reported relative to DMSO-d6 1H δH = 2.50 ppm, while the 13C-NMR spectra were reported relative to 13C δC = 39.52 ppm. The spectrum multiplicities were indicated as follows: singlet (s), doublet (d), triplet (t), multiplet (m), and broad (br). MALDI-MS mass spectra were obtained using a Bruker SolariX XR Fourier transform ion cyclotron resonance mass spectrometer (Bruker Daltonik GmbH, Bremen, Germany) with a 7 T refrigerated actively shielded superconducting magnet (Bruker Biospin, Wissembourg, France). A MALDI ion source (Bruker Daltonik GmbH, Bremen, Germany) was used when examining the samples in the positive ion mode. The mass range was set to m/z 200–3000. The laser power was 28%, and 22 laser shots were utilized for each scan. The mass spectra were calibrated externally using a mix of peptide clusters in the MALDI ionization positive ion mode.

3.1. Chemistry

The precursor halide complexes 14a/b were synthesized according to the literature procedures reported by us [20,32,33].

3.1.1. Synthesis of Boc-L-Proline Ag(I) Salt

Boc-protected L-proline silver (I) salt was synthesized according to the literature procedures [21]. Boc-L-proline (1.00 g, 4.64 mmol) was dissolved in 0.1 M NaOH (40 mL); to the resulting solution, a silver nitrate (1.7 M in H2O) solution was added under stirring. After 15 min, the mixture was partially reduced in volume, and cold Et2O was added to obtain an off-white precipitate, which was filtered and dried.
Synthesis of Co-Assembled Boc-Proline Silver(I) Complexes 1–4aP [23]
The Boc amino acid silver(I) NHC complexes 14aP were obtained through counterion exchange using silver NHC complexes and Boc-L-Pro silver salt. The silver(I) iodide complexes 1–4a (0.20 mmol) and Boc-L-Pro silver salt (0.22 mmol) were dissolved in 30 mL of CH2Cl2/CH3OH (6:4) and stirred for 3 h at room temperature, with the exclusion of light. The mixture was filtered to remove the AgI byproduct. The silver NHC-Ag(I)-Boc-L-Pro complex was obtained by removing the solvent at a reduced pressure. The 1H-, 13C-NMR spectra of complexes 1-4aP are reported in Figures S1–S8.
Ijms 26 06116 i001
[N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-imidazole-2-ylidine silver(I)] Boc-L-Pro (1aP, 0.0387 g, 36%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.65 (m, 2H, CH2NCHCHN), 7.40–7.29 (m, 5H, C6H5CH(OH)), 4.93 (m, 1H, C6H5CH(OH)CH2N), 4.41 (m, 1H, Jgem = 14.8 Hz, C6H5CH(OH)CH2N), 4.24 (m, 1H, Jgem = 14.8 Hz, C6H5CH(OH)CH2N), 3.85–3.78 (overlapping signals, 4H, NCH3 and NCHCH2 (L-Pro)), 3.30–3.12 (overlapping signals, 2H, NCH2 (L-Pro)), 1.89–1.61 (overlapping signals, 4H, NCHCH2CH2 (L-Pro)), 1.34 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 179.0, 175.0, 163.8, 137.6, 128.7, 126.5, 77.8, 71.0, 61.3, 56.2, 46.4, 36.1, 31.3, 28.6, 24.2, 23.5.
(MALDI-ICR FTMS) m/z: HRMS calculated for C24H28AgN4O2+: 512.3822, found: 512.3825.
Ijms 26 06116 i002
4,5-dichloro [N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-imidazole-2-ylidine silver(I)] Boc-L-Pro (2aP, 0.0756 g, 64%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.48–7.10 (m, 5H, C6H5CH(OH)), 5.29 (br d, 1H, C6H5CH(OH)CH2N), 3.86 (m, 1H, C6H5CH(OH)CH2N), 3.78 (m, 1H, J = 13.5 Hz, 7.5 Hz, C6H5CH(OH)CH2N), 3.64 (m, 1H, J = 13.5 Hz, 7.5 Hz, C6H5CH(OH)CH2N), 3.40–3.19 (overlapping signals, 4H, NCH3 and NCHCH2 (L-Pro)), 3.30–3.12 (overlapping signals, 2H, NCH2 (L-Pro)), 1.96–1.61 (overlapping signals, 4H, NCHCH2CH2 (L-Pro)), 1.34 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 180.8, 175.0, 158.1, 154.5, 153.7, 149.0, 135.7, 129.4, 128.7, 128.2, 126.4, 120.3, 77.5, 65.9, 61.1, 55.7, 46.7, 33.4, 31.4, 28.6, 24.2, 23.9.
(MALDI-ICR FTMS) m/z: HRMS calculated for C24H24AgCl4N4O2+: 650.1502, found: 650.1500.
Ijms 26 06116 i003
4,5-diphenyl [N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-imidazole-2-ylidine silver(I)] Boc-L-Pro (3aP, 0.0230 g, 17%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.47–7.39 (m, overlapping signals, 9H, Ar-H), 7.28–7.26 (m, overlapping signals, 4H, Ar-H), 7.09 (dd, 2H, J = 7.0, 1.4 Hz, Ar-H), 4.73 (br d, 1H, C6H5CH(OH)CH2N), 4.22 (br dd, 1H, C6H5CH(OH)CH2N), 4.15 (dd, 1H, J = 8.5 Hz, 1.4 Hz, C6H5CH(OH)CH2N), 3.86 (br dd, 1H, C6H5CH(OH)CH2N), 3.80 (s, 3H, NCH3), 3.32–3.12 (overlapping signals, 3H, NCHCH2 (L-Pro) and NCH2 (L-Pro)), 1.93–1.62 (overlapping signals, 4H, NCHCH2CH2 (L-Pro), 1.34 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 175.6, 174.6, 154.7, 153.8, 152.1, 141.9, 138.0, 131.8, 131.3, 130.6, 130.5, 129.4, 128.8, 128.3, 126.1, 125.6, 125.4, 77.8, 70.6, 61.4, 61.1, 54.4, 46.6, 46.4, 34.9, 31.2, 30.9, 28.7, 28.5, 24.2, 23.5.
(MALDI-ICR FTMS) m/z: HRMS calculated for C48H44AgN4O2+: 816.7742, found: 816.7737.
Ijms 26 06116 i004
[N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-benzoimidazole-2-ylidine silver(I)] Boc-L-Pro (4aP, 0.0183 g,16%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.98 (d, 2H, J = 8.7 Hz, Ar-H), 7.62 (2H, m, Ar-H), 7.48–7.27 (m, overlapping signals, 5H, Ar-H), 5.07 (br d, 1H, C6H5CH(OH)CH2N), 4.75 (dd, J = 13.8, 2.7 Hz, 1H, C6H5CH(OH)CH2N), 4.58 (m, 1H, C6H5CH(OH)CH2N), 4.10 (s, 3H, NCH3), 3.78 (br dd, 1H, C6H5CH(OH)CH2N), 3.19–3.12 (overlapping signals, 3H, NCHCH2 (L-Pro) and NCH2 (L-Pro)), 1.89–1.58 (overlapping signals, 4H, NCHCH2CH2 (L-Pro)), 1.31 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 176.6, 175.4, 163.9, 154.6, 153.7, 152.2, 144.2, 141.7, 139.7, 132.2, 131.8, 128.7, 128.4, 128.2, 126.7, 125.3, 124.2, 114.3, 113.8, 77.6, 70.4, 62.0, 61.6, 53.9, 46.5, 46.3, 33.7, 31.3, 30.9, 28.8, 28.7, 24.2, 23.4.
(MALDI-ICR FTMS) m/z: HRMS calculated for C32H32AgN4O2+: 612.5022, found: 612.5019.
Synthesis of Co-Assembled Boc-Proline Gold(I) Complexes 1–4bP [23]
The Boc amino acid gold(I) NHC complexes 14bP were obtained through counterion exchange using gold NHC complexes and Boc-L-Pro silver salt. The gold(I) chloride complexes 14b (0.20 mmol) and Boc-L-Pro silver salt (0.22 mmol) were dissolved in 30 mL of CH2Cl2/CH3OH (6:4) and stirred for 3 h at room temperature, with the exclusion of light. The mixture was filtered to remove the AgCl byproduct. The gold NHC-Au(I)-Boc-L-Pro complex was obtained by removing the solvent at a reduced pressure. The 1H-, 13C-NMR spectra of complexes 1-4bP are reported in Figures S9–S16.
Ijms 26 06116 i005
[N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-imidazole-2-ylidine gold(I)] Boc-L-Pro (1bP, 0.0957 g, 78%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.45 (m, 2H, CH2NCHCHN), 7.32–7.21 (m, 5H, C6H5CH(OH)), 4.99 (m, 1H, C6H5CH(OH)CH2N), 4.31 (m, 1H, C6H5CH(OH)CH2N), 4.04–3.40 (m, overlapping signals, 2H, C6H5CH(OH)CH2N), 3.79 (s, 3H, NCH3), 3.24 (m, 1H, NCHCH2 (L-Pro)), 2.10–2.02 (overlapping signals, 2H, NCH2 (L-Pro)), 1.82–1.70 (overlapping signals, 4H, NCHCH2CH2 (L-Pro)), 1.34 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 175.6, 171.1, 154.1, 128.9, 126.3, 124.6, 123.6, 79.6, 78.4, 60.2, 46.6, 38.0, 30.9, 28.7, 24.0, 23.3.
(MALDI-ICR FTMS) m/z: HRMS calculated for C24H28AuN4O2+: 601.4806, found: 601.4809.
Ijms 26 06116 i006
4,5-dichloro [N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-imidazole-2-ylidine gold(I)] Boc-L-Pro (2bP, 0.0749 g, 55%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.43–7.27 (m, 5H, C6H5CH(OH)), 5.95 (br d, 1H, C6H5CH(OH)CH2N), 5.00 (m, 1H, C6H5CH(OH)CH2N), 4.36 (m, 1H, C6H5CH(OH)CH2N), 4.01 (m, 1H, C6H5CH(OH)CH2N), 3.88 (m, 3H, NCH3), 3.79 (m, 1H, NCHCH2 (L-Pro)), 3.25–3.20 (overlapping signals, 2H, NCH2 (L-Pro)), 2.07–1.71 (overlapping signals, 4H, NCHCH2CH2 (L-Pro)), 1.34 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 183.4, 176.2, 154.1, 141.8, 129.2, 128.5, 126.7, 118.6, 118.0, 80.0, 79.5, 78.4, 60.4, 57.3, 46.4, 28.6, 24.5, 23.4.
(MALDI-ICR FTMS) m/z: HRMS calculated for C24H24AuCl4N4O2+: 739.2486, found: 739.2487.
Ijms 26 06116 i007
4,5-diphenyl [N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-imidazole-2-ylidine gold(I)] Boc-L-Pro (3bP, 0.0325 g, 23%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.47–7.39 (m, overlapping signals, 9H, Ar-H), 7.28–7.26 (m, overlapping signals, 4H, Ar-H), 7.09 (dd, 2H, J = 7.0, 1.4 Hz, Ar-H), 4.73 (br d, 1H, C6H5CH(OH)CH2N), 4.22 (br dd, 1H, C6H5CH(OH)CH2N), 4.15 (dd, 1H, J = 8.5 Hz, 1.4 Hz, C6H5CH(OH)CH2N), 3.86 (br dd, 1H, C6H5CH(OH)CH2N), 3.80 (s, 3H, NCH3), 3.32–3.12 (overlapping signals, 3H, NCHCH2 (L-Pro) and NCH2 (L-Pro)), 1.93–1.62 (overlapping signals, 4H, NCHCH2CH2 (L-Pro)), 1.34 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 175.6, 174.6, 154.7, 153.8, 152.1, 141.9, 138.0, 131.8, 131.3, 130.6, 130.5, 129.4, 128.8, 128.3, 126.1, 125.6, 125.4, 77.8, 70.6, 61.4, 61.1, 54.4, 46.6, 46.4, 34.9, 31.2, 30.9, 28.7, 28.5, 24.2, 23.5.
(MALDI-ICR FTMS) m/z: HRMS calculated for C48H44AuN4O2+: 905.8726, found: 905.8729.
Ijms 26 06116 i008
[N-methyl, N′-(2-hydroxy-2-phenyl) ethyl)-benzoimidazole-2-ylidine gold(I)] Boc-L-Pro (4bP, 0.0942 g, 71%).
1H-NMR (400 MHz, DMSO-d6, ppm) δ: 7.95 (d, 1H, J = 6.5 Hz, Ar-H), 7.87 (1H, d, J = 6.5 Hz, Ar-H), 7.57–7.42 (m, overlapping signals, 7H, Ar-H), 5.92 (br s, 1H, C6H5CH(OH)CH2N), 5.21 (dd, J = 14.0, 6.0 Hz, 1H, C6H5CH(OH)CH2N), 4.76 (m, 1H, C6H5CH(OH)CH2N), 4.18 (d, J = 6.9 Hz, 3H, NCH3), 4.05 (br dd, 1H, C6H5CH(OH)CH2N), 3.30–3.24 (overlapping signals, 1H, NCHCH2 (L-Pro)), 2.10–2.03 (overlapping signals, 2H, NCH2 (L-Pro)), 1.83–1.74 (overlapping signals, 4H, NCHCH2CH2 (L-Pro)), 1.38 (s, 9H, C(CH3)3).
13C-NMR (100 MHz, DMSO-d6, ppm) δ: 191.1, 176.6, 154.2, 153.9, 142.5, 134.3, 134.1, 128.8, 128.7, 128.2, 126.7, 124.9, 113.5, 113.4, 112.6, 112.5, 112.2, 78.3, 72.4, 61.0, 60.8, 55.9, 46.6, 46.4, 35.4, 31.3, 30.5, 28.8, 28.7, 24.3, 23.6.
(MALDI-ICR FTMS) m/z: HRMS calculated for C32H32AuN4O2+: 701.6006, found: 701.60.

3.2. Evaluation of Cholinesterase-Inhibitory Activity

The enzyme-inhibiting properties of the Au and Ag complexes against electric eel acetylcholinesterase (eeAChE) and equine butyrylcholinesterase (eqBChE) were assessed via a modified Ellman assay [34]. Briefly, the enzyme solutions (eeAChE: 0.4 U/mL; eqBChE: 0.6 U/mL), DTNB (2.14 mM), and the substrates (1.5 mM ATCI and 5.12 mM BTCI) were dissolved and incubated in a 96-well format with the test complexes/standards (concentration range of 0.01–50 µM). The absorbance at 412 nm was monitored over five minutes. A buffer-only control was included. The inhibition potency (IC50) was calculated from triplicate dose–response data, presented as the mean ± SD.

3.3. Evaluation of Human Monoamine Oxidase (hMAO)-Inhibitory Activity

The inhibitory activity of the gold and silver complexes against hMAO-A and hMAO-B was evaluated using a previously published protocol [35]. To ensure comparable enzymatic activity, the concentrations of hMAO-A (3 ng/mL) and hMAO-B (12 ng/mL) were adjusted to achieve a maximum velocity (Vmax) of 50 pmol/min for both isoforms. Data analysis was performed using GraphPad PRISM 9 (GraphPad Software®, San Diego, CA, USA). IC50 values were determined from the dose–response curves and were presented as the mean ± SD of three independent experiments, each conducted in triplicate.

3.4. Anti-Inflammatory Activity

To evaluate the anti-inflammatory capacity of the Au and Ag complexes, the nitric oxide (NO) generation in RAW 264.7 murine macrophage cells was quantified using the Griess method [36]. Specifically, the cells were seeded in 48-well plates, treated with varying concentrations of the complexes (range: 1–50 µM; dissolved in DMSO) or Indomethacin (used as a positive control) for a 24 h period, and subsequently stimulated with lipopolysaccharide (LPS, 1 µg/mL). Then, the cell culture medium was combined with an equal volume of Griess reagent at 40 mg/mL (ratio 1:1), and after a 30 min incubation with agitation, the optical density was measured at 540 nm. The levels of NO vs. LPS were calculated from the absorbance measurements using the following formula:
I N O   v s   L P S   ( % ) = A B S L P S A B S S a m p l e A B S L P S 100
where ABSLPS represents the average absorbance of cells treated with LPS alone, and ABSsample represents the average absorbance of cells treated with both LPS and the tested complexes or Indomethacin.
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) was added to the cells in order to measure the cell viability at 24 h, as previously reported [36]. IC50 values were determined from the dose–response curves for both NO inhibition (%) and the cell viability (%) using GraphPad Prism 9 software (GraphPad Software, La Jolla, CA, USA) and were presented as the mean ± SD of three independent experiments, each conducted in triplicate.

3.5. Antioxidant Activity

The antioxidant capacity of the Au and Ag complexes was evaluated in 3T3-L1 murine fibroblasts by assessing the intracellular reactive oxygen species (ROS) levels using a DCFH2-DA assay, following a previously established method [36]. The cells were seeded in 96-well plates and incubated with the tested complexes at concentrations of between 0.1 and 20 µM for a 24 h period. Subsequently, ROS generation was triggered by the addition of menadione (25 µM) for 15 min. N-acetylcysteine (NAC), a recognized antioxidant, served as a positive control at 20 mM. Then, the cells were loaded with 25 µM DCFH2-DA for 40 min under controlled conditions (dark, 37 °C, 5% CO2). At the end of the assay, the fluorescence intensity was measured using a plate reader (λex of 485 nm, λem of 535 nm). The % of ROS production inhibition (IROS) vs. that of the menadione-treated cells was calculated using the following formula:
I R O S   v s   M e n   ( % ) = F M e n F S a m p l e F M e n 100
where FMen represents the average fluorescence of the cells treated with menadione alone, and Fsample represents the average fluorescence of the cells treated with both menadione and the tested complexes or NAC.
A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was conducted in the same experimental conditions in order to measure the cell viability, as previously reported [36]. IC50 values were determined from the cell viability (%) using GraphPad Prism 9 software (GraphPad Software, La Jolla, CA, USA) and were presented as the mean ± SD of three independent experiments, each conducted in triplicate.

3.6. Docking Studies

Gold and silver proline complexes were designed using the Maestro graphical user interface [37]. Both the R and S configurations of the carbon 17 stereocenter, derived from the racemic phenyl epoxide synthon (Scheme 1A), were considered. A library of 18 hits was finally obtained. In order to obtain a reasonable starting conformation and a suitable electrostatic partial charge distribution of the ligands (Table S4), the compounds were optimized by means of a Density Functional Theory (DFT) calculation at the B3LYP level, considering a LACV3P** basis set as implemented by Jaguar [38]. All the simulations were carried out in a vacuum environment. A Mulliken electrostatic distribution analysis was used for the next docking simulation.
The Protein Data Bank (PDB) [39] was searched for crystallographic models for hAChE, hBChE, hMAO-A, hMAO-B, and NOS2. The PDB entries corresponding to the codes 4EYZ [40], 5NN0 [41], 2Z5X [42], 6FW0 [43], and 3E7M [44] were selected for each mentioned target, respectively. Prior to the docking calculations, the PDB target models underwent suitable optimization, including the removal of water molecules and other co-crystallized chemical entities. Gaistager charges were assigned to the target models by means of Autodock tools ver. 1.5.6 (ADT) [45], and a Mulliken DFT-derived electrostatic distribution was adopted for the ligands. For each target, a 40 × 40 × 40 Å grid centred on the co-crystallized ligand was considered to mimic the binding sites. AutoDock version 4.2.6 [45] was employed with the default parameters. Ten runs of the Lamarckian Genetic Algorithm were applied to explore the ligand–target configurations. Silver and gold atom parameters were obtained from the Autodock Forum [46,47] and implemented in the AutoDock force field (Table S5). The most energy-stable complexes were visually inspected.

3.7. Statistical Analysis

The data were analyzed for statistical significance (p < 0.05) using a one-way ANOVA followed by Dunnett’s multiple comparison test, performed using GraphPad Prism 9 and in triplicate as indicated in the description of each experiment (n = 3). The standard deviations (SD) are shown.

4. Conclusions

Despite extensive scientific progress, NDDs continue to impose a significant global burden of disability and morbidity, particularly on the ageing population. The current NDD therapies are often limited by the multifactorial nature of these disorders, and the interconnected presence of neuroinflammation and oxidative stress exacerbates the progression of these diseases. In this context, the exploration of Au- and Ag-NHC metal complexes as potential therapeutic agents for NDDs represents a new and hopeful area. Notably, no information concerning the inhibition of MAOs by Au- and Ag-NHC metal complexes has been reported to date. Our study aimed to contribute to this field by investigating the multi-faceted potential of novel Au- and Ag-NHC complexes bearing a Boc-N-protected proline as an anionic ligand, specifically evaluating their antagonism against both ChEs and MAOs, as well as their antioxidant and anti-inflammatory properties. Our findings reveal that Au-based complexes generally exhibit superior inhibitory activity against both eeAChE and eqBChE and hMAO-A and hMAO-B compared to their silver analogues. Notably, the Au complex 4bP, bearing a benzene ring fused to the carbene, demonstrated potent eqBChE inhibition and moderate hMAO-A inhibition, while complex 3bP, bearing two phenyl groups on the carbene, acted as a selective eeAChE inhibitor and exhibited remarkable anti-inflammatory and antioxidant properties. These results underscore the critical role of the metal and the substituents in the NHC core in modulating the biological activity of these metal complexes. Importantly, our study provides the first evidence of NHC-metal complexes’ inhibition of MAOs and of AChE/BChE inhibition by Au-NHC complexes, filling a significant gap in the current literature. While further in-depth investigations are warranted to fully elucidate their therapeutic potential and safety, the observed multi-target profiles offer a strong rationale for pursuing the development of Au-based NHC-proline complexes as promising novel lead compounds in the fight against NDDs.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms26136116/s1.

Author Contributions

Conceptualization, F.O., F.B. and P.L.; methodology, M.S.S., P.L., F.O. and F.B.; software, F.P. and F.O.; validation, D.I., A.M. and D.C.; formal analysis, D.I. and A.D.; investigation, J.C., A.M., A.D. and D.C.; resources, A.M., J.C. and D.I.; data curation, A.D., J.C. and F.P.; writing—original draft preparation, J.C., A.M., A.D., D.I. and F.P.; writing—review and editing, M.S.S., P.L., F.O. and F.B.; visualization, D.I., A.M. and F.O.; supervision, P.L., F.B., F.O. and M.S.S.; project administration, P.L., M.S.S., A.M. and A.D.; funding acquisition, P.L., M.S.S., A.M. and A.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by PRIN 2022 PNRR, Code P20222BLAZ—Enhanced pharmacological activity of noble metal carbene-N-heterocyclic complexes by oligopeptide counterion (CUP MASTER: D53D23016900001)—and PRIN 2022, Code 2022HARH5W—HyMTA (Hybrid Multi-Target Agents) Synthesis and biological evaluation of chimeric hybrid molecules containing NHC-metal complexes and carbazole moieties, as innovative multi-target anticancer and antiviral agents (CUP MASTER: C53D23004490001). This work was also funded by FEDER funds provided using national funds by the FCT—Foundation for Science and Technology—under the research grants UIDB/00081/2020 (https://doi.org/10.54499/UIDB/00081/2020, accessed on 22 June 2025. CIQUP), UIDP/00081/2020 (https://doi.org/10.54499/UIDP/00081/2020, accessed on 22 June 2025. CIQUP), LA/P/0056/2020 (https://doi.org/10.54499/LA/P/0056/2020, accessed on 22 June 2025. IMS), and UID/BIM/04308/2019 (MedinUP). D.C.’s contract (CEECINSTLA/00022/2022) is also supported by the FCT and FEDER/COMPETE.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ateleanu, D.M.; Niculescu, A.-G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef]
  2. Olufunmilayo, E.O.; Gerke-Duncan, M.B.; Holsinger, R.M.D. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants 2023, 12, 517. [Google Scholar] [CrossRef] [PubMed]
  3. Teleanu, R.I.; Niculescu, A.-G.; Roza, E.; Vladâcenco, O.; Grumezescu, A.M.; Teleanu, D.M. Neurotransmitters—Key Factors in Neurological and Neurodegenerative Disorders of the Central Nervous System. Int. J. Mol. Sci. 2022, 23, 5954. [Google Scholar] [CrossRef]
  4. Talesa, V.N. Acetylcholinesterase in Alzheimer’s disease. Mech. Ageing Dev. 2001, 122, 1961–1969. [Google Scholar] [CrossRef] [PubMed]
  5. Zhou, S.; Huang, G. The biological activities of butyrylcholinesterase inhibitors. Biomed. Pharmacother. 2022, 146, 112556. [Google Scholar] [CrossRef]
  6. Finberg, J.P.M.; Rabey, J.M. Inhibitors of MAO-A and MAO-B in Psychiatry and Neurology. Front. Pharmacol. 2016, 7, 340. [Google Scholar] [CrossRef]
  7. Shaban, N.Z.; Masoud, M.S.; Awad, D.; Mawlawi, M.A.; Sadek, O.M. Effect of Cd, Zn and Hg complexes of barbituric acid and thiouracil on rat brain monoamine oxidase-B (in vitro). Chem.-Biol. Interact. 2014, 208, 37–46. [Google Scholar] [CrossRef]
  8. Nitin, V.; Anand, T.; Singh, C.K.; Sodhi, K.K. Role of organometallic complexes in targeted therapies of different diseases: Infectious diseases, cancer and neurodegenerative Diseases. J. Organomet. Chem. 2024, 1022, 123389. [Google Scholar] [CrossRef]
  9. Sales, T.A.; Prandi, I.G.; de Castro, A.A.; Leal, D.H.S.; da Cunha, E.F.F.; Kuca, K.; Ramalho, T.C. Recent Developments in Metal-Based Drugs and Chelating Agents for Neurodegenerative Diseases Treatments. Int. J. Mol. Sci. 2019, 20, 1829. [Google Scholar] [CrossRef]
  10. Navale, G.R.; Ahmed, I.; Lim, M.H.; Ghosh, K. Transition Metal Complexes as Therapeutics: A New Frontier in Combatting Neurodegenerative Disorders through Protein Aggregation Modulation. Adv. Healthc. Mater. 2024, 13, 2401991. [Google Scholar] [CrossRef]
  11. Iacopetta, D.; Costabile, C.; La Chimia, M.; Mariconda, A.; Ceramella, J.; Scumaci, D.; Catalano, A.; Rosano, C.; Cuda, G.; Sinicropi, M.S.; et al. NHC-Ag(I) and NHC-Au(I) Complexes with N-Boc-Protected α-Amino Acidate Counterions Powerfully Affect the Growth of MDA-MB-231 Cells. ACS Med. Chem. Lett. 2023, 14, 1567–1575. [Google Scholar] [CrossRef] [PubMed]
  12. Johnson, N.A.; Southerland, M.R.; Youngs, W.J. Recent Developments in the Medicinal Applications of Silver-NHC Complexes and Imidazolium Salts. Molecules 2017, 22, 1263. [Google Scholar] [CrossRef] [PubMed]
  13. Mariconda, A.; Iacopetta, D.; Sirignano, M.; Ceramella, J.; Costabile, C.; Pellegrino, M.; Rosano, C.; Catalano, A.; Saturnino, C.; El-Kashef, H.; et al. N-Heterocyclic Carbene (NHC) Silver Complexes as Versatile Chemotherapeutic Agents Targeting Human Topoisomerases and Actin. ChemMedChem 2022, 17, e202200345. [Google Scholar] [CrossRef]
  14. Mora, M.; Gimeno, M.C.; Visbal, R. Recent advances in gold–NHC complexes with biological properties. Chem. Soc. Rev. 2019, 48, 447–462. [Google Scholar] [CrossRef]
  15. Bal, S.; Demirci, Ö.; Şen, B.; Taşkın Tok, T.; Taslimi, P.; Aktaş, A.; Gök, Y.; Aygün, M.; Gülçin, İ. Silver N-heterocyclic carbene complexes bearing fluorinated benzyl group: Synthesis, characterization, crystal structure, computational studies, and inhibitory properties against some metabolic enzymes. Appl. Organomet. Chem. 2021, 35, e6312. [Google Scholar] [CrossRef]
  16. Kazancı, A.; Gök, Y.; Kaya, R.; Aktaş, A.; Taslimi, P.; Gülçin, İ. Synthesis, characterization and bioactivities of dative donor ligand N-heterocyclic carbene (NHC) precursors and their Ag(I)NHC coordination compounds. Polyhedron 2021, 193, 114866. [Google Scholar] [CrossRef]
  17. Lasmari, S.; Ikhlef, S.; Boulcina, R.; Mokrani, E.H.; Bensouici, C.; Gürbüz, N.; Dündar, M.; Karcı, H.; Özdemir, İ.; Koç, A.; et al. New silver Nheterocyclic carbenes complexes: Synthesis, molecular docking study and biological activities evaluation as cholinesterase inhibitors and antimicrobials. J. Mol. Struct. 2021, 1238, 130399. [Google Scholar] [CrossRef]
  18. Gladkevich, A.; Bosker, F.; Korf, J.; Yenkoyan, K.; Vahradyan, H.; Aghajanov, M. Proline-rich polypeptides in Alzheimer’s disease and neurodegenerative disorders—Therapeutic potential or a mirage? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2007, 31, 1347–1355. [Google Scholar] [CrossRef]
  19. Yenkoyan, K.; Safaryan, K.; Chavushyan, V.; Meliksetyan, I.; Navasardyan, G.; Sarkissian, J.; Galoyan, A.; Aghajanov, M. Neuroprotective action of proline-rich polypeptide-1 in β-amyloid induced neurodegeneration in rats. Brain Res. Bull. 2011, 86, 262–271. [Google Scholar] [CrossRef]
  20. Sirignano, M.; D’Amato, A.; Costabile, C.; Mariconda, A.; Crispini, A.; Scarpelli, F.; Longo, P. Hydroamination of alkynes catalyzed by NHC-Gold(I) complexes: The non-monotonic effect of substituted arylamines on the catalyst activity. Front. Chem. 2023, 11, 1260726. [Google Scholar] [CrossRef]
  21. Ivry, E.; Ben-Asuly, A.; Goldberg, I.; Lemcoff, N.G. Amino acids as chiral anionic ligands for ruthenium based asymmetric olefin metathesis. Chem. Commun. 2015, 51, 3870–3873. [Google Scholar] [CrossRef] [PubMed]
  22. Legault, C.Y.; Kendall, C.; Charette, A.B. Structure and reactivity of a new anionic N-heterocyclic carbene silver (i) complex. Chem. Commun. 2005, 30, 3826–3828. [Google Scholar] [CrossRef] [PubMed]
  23. Ciardulli, M.C.; Mariconda, A.; Sirignano, M.; Lamparelli, E.P.; Longo, R.; Scala, P.; D’Auria, R.; Santoro, A.; Guadagno, L.; Della Porta, G.; et al. Activity and Selectivity of Novel Chemical Metallic Complexes with Potential Anticancer Effects on Melanoma Cells. Molecules 2023, 28, 4851. [Google Scholar] [CrossRef]
  24. Ellman, G.L.; Courtney, K.D.; Andres, V.; Featherstone, R.M. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  25. Sandeli, A.E.-K.; Khiri-Meribout, N.; Benzerka, S.; Gürbüz, N.; Dündar, M.; Karcı, H.; Bensouici, C.; Mokrani, E.H.; Özdemir, İ.; Koç, A.; et al. Silver (I)-N-heterocyclic carbene complexes: Synthesis and characterization, biological evaluation of Anti-Cholinesterase, anti-alpha-amylase, anti-lipase, and antibacterial activities, and molecular docking study. Inorg. Chim. Acta 2021, 525, 120486. [Google Scholar] [CrossRef]
  26. Behçet, A.; Aktaş, A.; Gök, Y.; Kaya, R.; Taslimi, P.; Gülçin, İ. Novel silver(I)N-heterocyclic carbene complexes bearing 2-(4-hydroxyphenyl)ethyl group: Synthesis, characterization, and enzyme inhibition properties. J. Heterocycl. Chem. 2020, 58, 603–611. [Google Scholar] [CrossRef]
  27. Behl, T.; Kaur, D.; Sehgal, A.; Singh, S.; Sharma, N.; Zengin, G.; Andronie-Cioara, F.L.; Toma, M.M.; Bungau, S.; Bumbu, A.G. Role of Monoamine Oxidase Activity in Alzheimer’s Disease: An Insight into the Therapeutic Potential of Inhibitors. Molecules 2021, 26, 3724. [Google Scholar] [CrossRef]
  28. Mateev, E.; Georgieva, M.; Mateeva, A.; Zlatkov, A.; Ahmad, S.; Raza, K.; Azevedo, V.; Barh, D. Structure-Based Design of Novel MAO-B Inhibitors: A Review. Molecules 2023, 28, 4814. [Google Scholar] [CrossRef]
  29. Shaban, N.Z.; Ali, A.E.; Masoud, M.S. Effect of cadmium and zinc ethanolamine complexes on rat brain monoamine oxidase-B activity in vitro. J. Inorg. Biochem. 2003, 95, 141–148. [Google Scholar] [CrossRef]
  30. Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms Underlying Inflammation in Neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef]
  31. Nalamachu, S.; Wortmann, R. Role of Indomethacin in Acute Pain and Inflammation Management: A Review of the Literature. Postgrad. Med. 2015, 126, 92–97. [Google Scholar] [CrossRef] [PubMed]
  32. Mariconda, A.; Sirignano, M.; Costabile, C.; Longo, P. New NHC- silver and gold complexes active in A3-coupling (aldehyde-alkyne-amine) reaction. Mol. Catal. 2020, 480, 110570. [Google Scholar] [CrossRef]
  33. Saturnino, C.; Barone, I.; Iacopetta, D.; Mariconda, A.; Sinicropi, M.S.; Rosano, C.; Campana, A.; Catalano, S.; Longo, P.; Andò, S.N. Heterocyclic Carbene Complexes of Silver and Gold as Novel Tools Against Breast Cancer Progression. Future Med. Chem. 2016, 8, 2213–2229. [Google Scholar] [CrossRef] [PubMed]
  34. Oliveira, C.; Cagide, F.; Teixeira, J.; Amorim, R.; Sequeira, L.; Mesiti, F.; Silva, T.; Garrido, J.; Remião, F.; Vilar, S.; et al. Hydroxybenzoic Acid Derivatives as Dual-Target Ligands: Mitochondriotropic Antioxidants and Cholinesterase Inhibitors. Front. Chem. 2018, 6, 126. [Google Scholar] [CrossRef]
  35. Chavarria, D.; Cagide, F.; Pinto, M.; Gomes, L.R.; Low, J.N.; Borges, F. Development of piperic acid-based monoamine oxidase inhibitors: Synthesis, structural characterization and biological evaluation. J. Mol. Struct. 2019, 1182, 298–307. [Google Scholar] [CrossRef]
  36. Mariconda, A.; Iacopetta, D.; Sirignano, M.; Ceramella, J.; D’Amato, A.; Marra, M.; Pellegrino, M.; Sinicropi, M.S.; Aquaro, S.; Longo, P. Silver and Gold Complexes with NHC-Ligands Derived from Caffeine: Catalytic and Pharmacological Activity. Int. J. Mol. Sci. 2024, 25, 2599. [Google Scholar] [CrossRef]
  37. Schrödinger Release 2024-1: Maestro, Schrödinger LLC. 2024. Available online: www.schrodinger.com (accessed on 2 December 2024).
  38. Bochevarov, A.D.; Harder, E.; Hughes, T.F.; Greenwood, J.R.; Braden, D.A.; Philipp, D.M.; Rinaldo, D.; Halls, M.D.; Zhang, J.; Friesner, R.A. Jaguar: A high-performance quantum chemistry software program with strengths in life and materials sciences. Int. J. Quantum Chem. 2013, 113, 2110–2142. [Google Scholar] [CrossRef]
  39. Berman, H.M. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef]
  40. Kobe, B.; Levy-Assaraf, M.; Voronov-Goldman, M.; Rozman Grinberg, I.; Weiserman, G.; Shimon, L.J.W.; Jindou, S.; Borovok, I.; White, B.A.; Bayer, E.A.; et al. Crystal Structure of an Uncommon Cellulosome-Related Protein Module from Ruminococcus flavefaciens That Resembles Papain-Like Cysteine Peptidases. PLoS ONE 2013, 8, e56138. [Google Scholar] [CrossRef]
  41. Košak, U.; Brus, B.; Knez, D.; Žakelj, S.; Trontelj, J.; Pišlar, A.; Šink, R.; Jukič, M.; Živin, M.; Podkowa, A.; et al. The Magic of Crystal Structure-Based Inhibitor Optimization: Development of a Butyrylcholinesterase Inhibitor with Picomolar Affinity and In Vivo Activity. J. Med. Chem. 2017, 61, 119–139. [Google Scholar] [CrossRef]
  42. Son, S.-Y.; Ma, J.; Kondou, Y.; Yoshimura, M.; Yamashita, E.; Tsukihara, T. Structure of human monoamine oxidase A at 2.2-Å resolution: The control of opening the entry for substrates/inhibitors. Proc. Natl. Acad. Sci. USA 2008, 105, 5739–5744. [Google Scholar] [CrossRef] [PubMed]
  43. Reis, J.; Manzella, N.; Cagide, F.; Mialet-Perez, J.; Uriarte, E.; Parini, A.; Borges, F.; Binda, C. Tight-Binding Inhibition of Human Monoamine Oxidase B by Chromone Analogs: A Kinetic, Crystallographic, and Biological Analysis. J. Med. Chem. 2018, 61, 4203–4212. [Google Scholar] [CrossRef] [PubMed]
  44. Garcin, E.D.; Arvai, A.S.; Rosenfeld, R.J.; Kroeger, M.D.; Crane, B.R.; Andersson, G.; Andrews, G.; Hamley, P.J.; Mallinder, P.R.; Nicholls, D.J.; et al. Anchored plasticity opens doors for selective inhibitor design in nitric oxide synthase. Nat. Chem. Biol. 2008, 4, 700–707. [Google Scholar] [CrossRef] [PubMed]
  45. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef]
  46. Rodríguez Arce, E.; Mosquillo, M.F.; Pérez-Díaz, L.; Echeverría, G.A.; Piro, O.E.; Merlino, A.; Coitiño, E.L.; Maríngolo Ribeiro, C.; Leite, C.Q.F.; Pavan, F.R.; et al. Aromatic amine N-oxide organometallic compounds: Searching for prospective agents against infectious diseases. Dalton Trans. 2015, 44, 14453–14464. [Google Scholar] [CrossRef]
  47. Forum, A. ADL, Parameters for Docking with Metal Ions in Receptor. Available online: https://www.researchgate.net/profile/Dr-Rajesh-Patil/post/Autodock-docking-with-gold-nanoparticles/attachment/59d6477f79197b80779a2567/AS%3A462677517574144%401487322324251/download/ADL_+Parameters+for+docking+with+metal+ions+in+receptor.pdf (accessed on 2 December 2024).
Figure 1. Ag (1aP4aP) and Au (1bP4bP) complexes stabilized by NHC ligands and bearing carboxylate salt of tert-butyloxycarbonyl (Boc)-N-protected proline as anionic ligand.
Figure 1. Ag (1aP4aP) and Au (1bP4bP) complexes stabilized by NHC ligands and bearing carboxylate salt of tert-butyloxycarbonyl (Boc)-N-protected proline as anionic ligand.
Ijms 26 06116 g001
Scheme 1. (A) Synthesis of precursor metallic complexes 14a/b; (B) synthesis of co-assembled NHC silver(I) and gold(I) L-proline complexes 14a/bP.
Scheme 1. (A) Synthesis of precursor metallic complexes 14a/b; (B) synthesis of co-assembled NHC silver(I) and gold(I) L-proline complexes 14a/bP.
Ijms 26 06116 sch001
Figure 2. Brown histograms: Anti-inflammatory activity in terms of NO production inhibition (%), measured in vitro using RAW 264.7 murine macrophages. NO production was induced by LPS (1 µg/mL). Data were plotted as % NO inhibition with respect to inhibition with LPS-only treatment. Salmon histograms: MTT assays performed in same experimental conditions in order to verify effect on RAW 264.7 cells’ viability of Indomethacin and tested complexes. Data were plotted as % of cell viability with respect to viability with LPS-only treatment (1 µg/mL). Indomethacin (Ind) was used at concentration of 25 µM, while all other complexes were used at 5 µM, for 24 h. Bars ± SDs are shown (n = 3). ** p < 0.01 and **** p < 0.0001 for complex 3bP and Ind vs. LPS only; not significant (n.s.) where not indicated.
Figure 2. Brown histograms: Anti-inflammatory activity in terms of NO production inhibition (%), measured in vitro using RAW 264.7 murine macrophages. NO production was induced by LPS (1 µg/mL). Data were plotted as % NO inhibition with respect to inhibition with LPS-only treatment. Salmon histograms: MTT assays performed in same experimental conditions in order to verify effect on RAW 264.7 cells’ viability of Indomethacin and tested complexes. Data were plotted as % of cell viability with respect to viability with LPS-only treatment (1 µg/mL). Indomethacin (Ind) was used at concentration of 25 µM, while all other complexes were used at 5 µM, for 24 h. Bars ± SDs are shown (n = 3). ** p < 0.01 and **** p < 0.0001 for complex 3bP and Ind vs. LPS only; not significant (n.s.) where not indicated.
Ijms 26 06116 g002
Figure 3. Brown histograms: Antioxidant activity in terms of inhibition of intracellular ROS production (%), measured in vitro using 3T3-L1 murine fibroblasts. ROS production was induced by menadione (Men, 25 µM, 15 min). Data were plotted as % ROS inhibition with respect to inhibition with Men-only treatment. Salmon histograms: MTT assays were performed in order to verify effect on 3T3-L1 cells’ viability of N-acetyl cysteine (NAC) and tested complexes. Data were plotted as % of cell viability with respect to viability with CTRL-only treatment (vehicle only). NAC was used at concentration of 20 mM, while all other complexes were used at 10 µM, except for 3bP, used at 1 µM, for 24 h. Bars ± SDs are shown (n = 3). ** p < 0.01, *** p < 0.001, and **** p < 0.0001 for complexes 4bP and 3bP and NAC vs. only Men; other complexes were not significant (n.s.).
Figure 3. Brown histograms: Antioxidant activity in terms of inhibition of intracellular ROS production (%), measured in vitro using 3T3-L1 murine fibroblasts. ROS production was induced by menadione (Men, 25 µM, 15 min). Data were plotted as % ROS inhibition with respect to inhibition with Men-only treatment. Salmon histograms: MTT assays were performed in order to verify effect on 3T3-L1 cells’ viability of N-acetyl cysteine (NAC) and tested complexes. Data were plotted as % of cell viability with respect to viability with CTRL-only treatment (vehicle only). NAC was used at concentration of 20 mM, while all other complexes were used at 10 µM, except for 3bP, used at 1 µM, for 24 h. Bars ± SDs are shown (n = 3). ** p < 0.01, *** p < 0.001, and **** p < 0.0001 for complexes 4bP and 3bP and NAC vs. only Men; other complexes were not significant (n.s.).
Ijms 26 06116 g003
Figure 4. Docking poses of the best-scoring enantiomers of compounds 2aP, 2bP, 4aP, and 4bP in hAChE (AD) and hBChE (EH). The ligands are represented as pink or green sticks and the residues involved in the interaction as violet or blue sticks. (A) (S)2aP; (B) (R)2bP; (C) (S)4aP; (D) (S)4bP; (E) (S)2aP; (F) (R)2bP; (G) (R)4aP; (H) (S)4bP.
Figure 4. Docking poses of the best-scoring enantiomers of compounds 2aP, 2bP, 4aP, and 4bP in hAChE (AD) and hBChE (EH). The ligands are represented as pink or green sticks and the residues involved in the interaction as violet or blue sticks. (A) (S)2aP; (B) (R)2bP; (C) (S)4aP; (D) (S)4bP; (E) (S)2aP; (F) (R)2bP; (G) (R)4aP; (H) (S)4bP.
Ijms 26 06116 g004
Figure 5. Docking poses of the best-scoring enantiomers of compounds 2aP, 2bP, 4aP, and 4bP in hMAO-A (AD) and hMAO-B (EH). The ligands are represented as pink or orange sticks and the residues involved in the interaction as blue or green sticks. (A) (S)2aP; (B) (R)2bP; (C) (S)4aP; (D) (S)4bP; (E) (S)2aP; (F) (S)2bP; (G) (S)4aP; (H) (R)4bP.
Figure 5. Docking poses of the best-scoring enantiomers of compounds 2aP, 2bP, 4aP, and 4bP in hMAO-A (AD) and hMAO-B (EH). The ligands are represented as pink or orange sticks and the residues involved in the interaction as blue or green sticks. (A) (S)2aP; (B) (R)2bP; (C) (S)4aP; (D) (S)4bP; (E) (S)2aP; (F) (S)2bP; (G) (S)4aP; (H) (R)4bP.
Ijms 26 06116 g005
Figure 6. Docking poses of the best-scoring enantiomers of compounds 3aP and 3bP in iNOS. The ligands are represented as green sticks and the residues involved in the interaction as blue sticks. (A) (R)3aP; (B) (R)3bP; (C) (S)3aP; (D) (S)3bP.
Figure 6. Docking poses of the best-scoring enantiomers of compounds 3aP and 3bP in iNOS. The ligands are represented as green sticks and the residues involved in the interaction as blue sticks. (A) (R)3aP; (B) (R)3bP; (C) (S)3aP; (D) (S)3bP.
Ijms 26 06116 g006
Table 1. Evaluation of eeAChE/eqBChE-inhibitory activities of Ag (1aP4aP) and Au (1bP4bP) complexes and reference inhibitor Donepezil. Values, expressed as µM, are means ± S.D.s of three different experiments performed in triplicate.
Table 1. Evaluation of eeAChE/eqBChE-inhibitory activities of Ag (1aP4aP) and Au (1bP4bP) complexes and reference inhibitor Donepezil. Values, expressed as µM, are means ± S.D.s of three different experiments performed in triplicate.
IC50 (µM)SI a
CompoundseeAChEeqBChE
Donepezil(2.30 ± 0.53) × 10−22.13 ± 0.120.01
1aP>25>25-
2aP>2518.24 ± 2.4>1.37
3aP>2517.38 ± 0.98>1.44
4aP13.95 ± 0.725.19 ± 0.952.68
1bP18.37 ± 1.436.19 ± 0.912.97
2bP4.68 ± 0.473.14 ± 0.331.49
3bP5.89 ± 0.56>25>0.24
4bP3.72 ± 0.440.45 ± 0.068.27
a eqBChE selectivity index = IC50(eeAChE)/IC50(eqBChE).
Table 2. Evaluation of hMAO-inhibitory activities of Ag (1aP4aP) and Au (1bP4bP) complexes and reference MAOIs (Clorgyline for hMAO-A and (R)-(–)-Deprenyl for hMAO-B). Values, expressed as µM, are means ± S.D.s of three different experiments performed in triplicate.
Table 2. Evaluation of hMAO-inhibitory activities of Ag (1aP4aP) and Au (1bP4bP) complexes and reference MAOIs (Clorgyline for hMAO-A and (R)-(–)-Deprenyl for hMAO-B). Values, expressed as µM, are means ± S.D.s of three different experiments performed in triplicate.
IC50 (µM)SI a
CompoundshMAO-AhMAO-B
Clorgyline(2.00 ± 0.17) × 10−32.44 ± 0.178.19 × 10−4
(R)-(–)-Deprenyl17.49 ± 2.67(4.92 ± 0.87) × 10−2356.94
1aP>25>25-
2aP15.04 ± 1.029.82 ± 0.841.53
3aP24.97 ± 2.95>250.99
4aP3.32 ± 0.1510.40 ± 1.950.32
1bP1.32 ± 0.023.78 ± 0.160.35
2bP0.72 ± 0.021.94 ± 0.090.37
3bP1.84 ± 0.184.75 ± 0.750.39
4bP(7.13 ± 0.97) × 10−22.10 ± 0.200.03
a hMAO-B selectivity index = IC50(hMAO-A)/IC50(hMAO-B).
Table 3. IC50 values for NO inhibition and the cell viability (%), measured in vitro using RAW 264.7 murine macrophages for the Ag (1aP4aP) and Au (1bP4bP) complexes and Indomethacin (used as a reference molecule). The values, expressed as µM, are the means ± SDs of three different experiments, performed in triplicate.
Table 3. IC50 values for NO inhibition and the cell viability (%), measured in vitro using RAW 264.7 murine macrophages for the Ag (1aP4aP) and Au (1bP4bP) complexes and Indomethacin (used as a reference molecule). The values, expressed as µM, are the means ± SDs of three different experiments, performed in triplicate.
IC50 (µM)
CompoundsNO InhibitionCell Viability
Indomethacin>50>50
1aP>50>50
2aP>50>50
3aP30.88 ± 1.74>50
4aP>50>50
1bP>50>50
2bP32.31 ± 1.82>50
3bP4.88 ± 0.5820.27 ± 1.26
4bP13.05 ± 0.9212.12 ± 1.03
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ceramella, J.; D’Amato, A.; Procopio, F.; Mariconda, A.; Chavarria, D.; Iacopetta, D.; Ortuso, F.; Longo, P.; Borges, F.; Sinicropi, M.S. Novel Au(I)- and Ag(I)-NHC Complexes with N-Boc-Protected Proline as Potential Candidates for Neurodegenerative Disorders. Int. J. Mol. Sci. 2025, 26, 6116. https://doi.org/10.3390/ijms26136116

AMA Style

Ceramella J, D’Amato A, Procopio F, Mariconda A, Chavarria D, Iacopetta D, Ortuso F, Longo P, Borges F, Sinicropi MS. Novel Au(I)- and Ag(I)-NHC Complexes with N-Boc-Protected Proline as Potential Candidates for Neurodegenerative Disorders. International Journal of Molecular Sciences. 2025; 26(13):6116. https://doi.org/10.3390/ijms26136116

Chicago/Turabian Style

Ceramella, Jessica, Assunta D’Amato, Francesca Procopio, Annaluisa Mariconda, Daniel Chavarria, Domenico Iacopetta, Francesco Ortuso, Pasquale Longo, Fernanda Borges, and Maria Stefania Sinicropi. 2025. "Novel Au(I)- and Ag(I)-NHC Complexes with N-Boc-Protected Proline as Potential Candidates for Neurodegenerative Disorders" International Journal of Molecular Sciences 26, no. 13: 6116. https://doi.org/10.3390/ijms26136116

APA Style

Ceramella, J., D’Amato, A., Procopio, F., Mariconda, A., Chavarria, D., Iacopetta, D., Ortuso, F., Longo, P., Borges, F., & Sinicropi, M. S. (2025). Novel Au(I)- and Ag(I)-NHC Complexes with N-Boc-Protected Proline as Potential Candidates for Neurodegenerative Disorders. International Journal of Molecular Sciences, 26(13), 6116. https://doi.org/10.3390/ijms26136116

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop