1. Introduction
Alzheimer’s disease (AD) remains an incurable condition despite significant global research efforts and investments aimed at developing effective treatments. In the United States, approximately 10% of the elderly population is affected by AD, and this number is projected to double to approximately 15 million by 2050 [
1]. The public health impact of AD can be significantly mitigated through interventions aimed at preventing, halting, or slowing the disease; however, current medications have limited efficacies in reducing disease severity or slowing disease progression [
2]. Such challenges in developing effective treatments for AD may stem from the narrow focus on disease treatment. In addition, the failure to develop treatments for dementia as a broader category presents another significant challenge [
3]. Despite substantial investment to date, only seven treatments for AD have been approved by the Food and Drug Administration (FDA) [
4,
5]. These medications include cholinesterase inhibitors, NMDA receptor antagonists, and anti-amyloid beta (Aβ) monoclonal antibodies. Among these, five drugs are used to address the symptoms of AD, namely donepezil, rivastigmine, galantamine, memantine, and aducanumab (Aduhelm). Recently, the FDA accelerated the approval of lecanemab (Lequembi) for the treatment of early-stage AD owing to its validated efficacy [
6]. Additionally, the beta-saccharide 1 (BACE1) inhibitor donanemab has been shown to slow the progression of cognitive decline by approximately 25% in early-stage patients [
7]. However, these antibody-based treatments have not been effective in patients with advanced AD, and face numerous challenges, such as high costs and undesirable side effects [
8,
9].
Previous research has identified the potential of Ramalin, derived from the Antarctic lichen, as an anti-Alzheimer’s treatment owing to its strong antioxidant and anti-inflammatory properties [
10]. Initial experiments showed promise; however, Ramalin was ultimately ruled out as a viable treatment because of its instability and toxicity, including cytotoxicity, splenomegaly, and genotoxicity. To address these issues, various Ramalin derivatives have been prepared to reduce its toxicity and enhance its physical characteristics [
11]. For example, it was proposed that its stability could be improved by introducing a methyl group to hydrazine- or benzamido-type derivatives, creating a more stable structure by preventing oxidation (
Figure 1). The structural instability of Ramalin is presumed to be caused by oxidation through its reaction with oxygen. This is evidenced by the increased stability when dissolved in deoxygenated water [
12] compared to water containing dissolved oxygen. After dissolving Ramalin in water and deoxygenated water at a concentration of 10 mM and leaving it for 48 h at 25 °C, it was found that only about 53% of Ramalin remained in regular water, whereas 92% remained in deoxygenated water. Based on the mass product ion scan results (
Supplementary Materials, Figure S33), as shown in
Figure 2, it is expected that Ramalin decomposes into the RA-Azo form. Although attempts were made to isolate and purify RA-Azo, it was found to decompose too rapidly, making isolation and purification impossible. In an effort to prevent the oxidation of Ramalin into the RA-Azo form, we designed derivatives to improve stability. We attempted modifications by introducing a methyl group into phenyl hydrazine to prevent conversion into the azo form or by linking the phenyl group to a benzophenone structure to block electron flow and mitigate oxidation, thereby enhancing stability.
In addition, it has been demonstrated that the structural instability resulting from the resonance between the hydrazine and phenol groups can vary depending on the phenyl functional group. Indeed, this was verified through the preparation of RA-PF derivatives, where the stability was enhanced by replacing the protons at positions 2 and 6 of the phenyl ring with fluoride [
10]. However, although the stability was found to increase based on the type and position of the functional group [
13], the obtained derivatives could not be unequivocally labeled as stable substances, because their stabilities were merely enhanced.
Antioxidant effects play a crucial role in the treatment of AD because oxidative stress is a key contributor to its pathogenesis [
14,
15,
16]. Antioxidants mitigate neuronal damage and slow disease progression by neutralizing free radicals. In addition, anti-inflammatory effects are critically important in the context of AD, since chronic inflammation is known to exacerbate AD pathology by promoting the accumulation of amyloid plaques and tau tangles [
17,
18]. It is therefore desirable to prepare derivatives that exhibit desirable anti-inflammatory properties to both prevent and treat AD by reducing neuroinflammation.
The inhibition of BACE-1 is another significant therapeutic strategy for AD treatment, as this enzyme is responsible for the production of Aβ peptides that aggregate to form amyloid plaques, a hallmark of AD. Previous studies have demonstrated that BACE-1 inhibitors can reduce Aβ levels, thereby slowing or halting the progression of the disease [
19]. Extensive research and development efforts in the area of BACE-1 inhibitors have led to the preparation of several nanomolar-range inhibitory compounds [
20].
The inhibition of acetylcholinesterase (AChE) is also a well-established mechanism in AD treatment, as it increases the levels of acetylcholine in the brain, a neurotransmitter that is typically reduced in patients with AD [
21]. Enhanced acetylcholine levels can improve cognitive function and alleviate the symptoms of dementia, thereby indicating the potential of AChE inhibitors to act as effective AD therapies [
22,
23].
Moreover, the tau inhibitory activity is also critical due to the ability of hyperphosphorylated tau proteins to form neurofibrillary tangles, another key pathological feature of AD [
24]. By inhibiting tau phosphorylation, tangle formation could be inhibited, thereby offering an alternative approach for the treatment and prevention of AD [
25,
26,
27].
Thus, in the current study, a range of derivatives are synthesized and tested for their ability to treat AD based on their antioxidant and anti-inflammatory effects, BACE-1 inhibitory activity, AChE inhibitory activity, and tau inhibitory activity. The primary objective of synthesizing these Ramalin derivatives is to evaluate whether the more stable derivatives exhibit comparable anti-AD activities to the parent compound, Ramalin. In addition, this study aims to address the stability and toxicity concerns associated with Ramalin, and the ultimate goal is to synthesize Ramalin derivatives that could potentially be developed into effective anti-AD drugs.
3. Discussion
This study focused on the synthesis and evaluation of various Ramalin derivatives for the treatment of AD. Ramalin, which is derived from the Antarctic lichen, is known to demonstrate strong antioxidant and anti-inflammatory properties, thereby rendering it a promising candidate for AD treatment. However, its inherent instability and toxicity significantly limit its potential as a viable therapeutic agent. Addressing these limitations by developing more stable and less toxic derivatives was therefore the primary motivation for this study.
Thus, four novel Ramalin derivatives were prepared, namely RA-Hyd-Me, RA-Hyd-Me-Tol, RA-Sali, and RA-Benzo, and their chemical stabilities were extensively evaluated under various conditions. The results revealed that these derivatives maintained their structural integrity for extended periods, even at elevated temperatures. In contrast, the parent compound, Ramalin, exhibited significant degradation, undergoing rapid decomposition within a few hours at 60 °C. The incorporation of hydrazino methyl and benzamido functional groups therefore appeared to play a crucial role in enhancing its stability by preventing oxidative degradation and stabilizing the electron flow within the structure. These findings are particularly significant as they address one of the major challenges associated with AD drug development, namely the stability of the potential therapeutic agent. Notably, stable compounds are essential for the development of reliable and effective drugs because instability can lead to inconsistent therapeutic outcomes, reduced efficacies, and increased side effects. By improving the stability of Ramalin, this study has taken a crucial step towards developing more reliable AD treatments.
The mutagenic potentials of the prepared derivatives were assessed using a mini Ames test with two bacterial strains, namely Salmonella typhimurium TA98 and TA100. The results indicated that unlike the parent compound Ramalin, which demonstrated significant mutagenic activity, particularly in the presence of the S9 metabolic activation system, the synthesized derivatives did not exhibit mutagenicity under the tested conditions. This lack of mutagenicity suggests that the structural modifications introduced into the derivatives effectively mitigated the mutagenic potential observed for Ramalin.
However, it should be noted that the above improvements come at the cost of reduced antioxidant activities, with the synthesized derivatives exhibiting lower antioxidant activities than Ramalin itself. These reductions were attributed to the structural modifications intended to inhibit oxidation by disrupting the electron flow between the hydrazine and phenyl groups. More specifically, RA-Benzo and RA-Sali exhibited diminished antioxidant effects, with their activities reduced to the mM level, while RA-Hyd-Me and RA-Hyd-Me-Tol maintained their antioxidant effect at concentrations of 12–14 µM. This tradeoff between stability and antioxidant activity underscores the complexity of drug development, wherein multiple factors must be balanced to achieve an optimal therapeutic profile.
Despite their reduced antioxidant activities, the synthesized derivatives exhibited other promising therapeutic properties, such as improved BACE-1 inhibitory activities and anti-tau activities, which are critical in the context of AD. The abilities of RA-Hyd-Me and RA-Sali to inhibit BACE-1, an enzyme involved in the production of Aβ, suggest that these compounds could reduce the formation of amyloid plaques, a hallmark of AD pathology. In particular, RA-Sali demonstrated the most effective BACE-1 inhibitory activity, with an IC50 value lower than that of the positive control, LY2811376. The effects of the various derivatives on tau phosphorylation were also evaluated using the ELISA approach. The cellular levels of p-tau were measured after treatment with the Ramalin derivatives (20 µM), and it was found that among the derivatives, RA-Sali exhibited the most significant inhibitory activity with an inhibition level of ~40%, further confirming the potential of these derivatives for use as multi-target therapeutic agents in AD treatment. The remaining three derivatives (RA-Hyd-Me, RA-Hyd-Me-Tol, and RA-Benzo) showed inhibitory activities of approximately 10–15%. Despite these promising findings, neither Ramalin nor any of its four derivatives exhibited inhibitory activities against AChE. This suggests that although these compounds may have potential as multi-target therapeutic agents for AD, they may not be effective in targeting AChE, a common target in existing AD treatments.
This study also examined the potential of the derivatives to cross the BBB. Given that the BBB is a significant obstacle to drug delivery into the central nervous system, the low molecular weights and favorable physicochemical properties of these derivatives suggest that they may effectively traverse the BBB and directly exert their therapeutic effects in the brain. This hypothesis is supported by the calculated HBD and HBA values, as well as the derivative molecular weights, which are within the ranges typically associated with BBB permeability [
39]. However, the passage of these derivatives through the BBB may still present challenges, particularly in the case of the RA-Sali derivative, which has a higher HBA value of 6, an HBD value of 5, and a polar surface area (PSA) of 141.75 Å
2 [
32]. Nevertheless, their low molecular weights provide a potential advantage for overcoming this limitation, especially when considering the possibility of enhancing the BBB permeability or performing further modifications to improve the physicochemical properties of Ramalin derivatives.
Thus, although the synthesized Ramalin derivatives show promise as potential therapeutic agents for AD, further optimization is necessary. Indeed, the current anti-AD effects are considered insufficient, and additional structural refinements are required to enhance their antioxidant, anti-inflammatory, anti-BACE-1, and anti-tau activities. Future studies should consider improvements in BBB permeability to ensure that these compounds can effectively reach their target in the brain. However, despite these challenges, this study represents a significant advancement in the development of stable and effective AD treatments and lays the groundwork for future therapeutic strategies.
4. Materials and Methods
4.1. General Experimental Information
All solvents and reagents were obtained from Merck (Darmstadt, Germany) or TCI (Tokyo, Japan) and used without further purification. All glassware was thoroughly washed and dried in a drying oven (60 °C) or flamed and cooled under a stream of dry argon prior to use. Filters were obtained from GE healthcare (GF/F, 0.7 µm, Whatman, UK). All reactions were performed under an inert argon atmosphere. Solvents and liquid reagents were transferred to a syringe prior to use. Organic extracts were dried over Na2SO4 and concentrated under reduced pressure in a rotary evaporator (Eyela, Tokyo, Japan). Residual solvent from the extracts was removed under a high vacuum (Vacuubrand RZ 2.5, Wertheim, Germany, 1 × 10−2 mbar). Purification was performed using a Yamazen Smart Flash EPCLC AI-580S (Yamazen, Osaka, Japan) medium-pressure liquid chromatography (MPLC) system. Accurate mass spectra were obtained using an AB Sciex Triple TOF 4600 (Framingham, MA, USA) instrument, with the interface in the direct injection mode. Infrared (IR) spectra were collected using a Bruker Vertex80V FT-IR spectrometer (Bruker, Billerica, MA, USA), equipped with a vacuum system. Nuclear magnetic resonance (NMR) spectra were obtained on a Jeol JNM ECP-400 spectrometer (Jeol Ltd., Tokyo, Japan) using a mixture of D2O (with 0.01 mg/mL sodium trimethylsilylpropanesulfonate (DSS))/acetone-d6 (6:1 v/v) or dimethyl sulfoxide (DMSO)-d6 as solvents. The internal reference or residual solvent signals were utilized for referencing (D2O (with DSS)/acetone-d6: dH 0.00/dC 29.8; DMSO-d6: dH 2.50/dC 39.5). The peak-splitting patterns were abbreviated as m, s, d, t, dd, ddd, and br for multiplets, singlets, doublets, triplets, doublets of doublets, doublets of doublets of doublets, and broad, respectively. Microplate (Thermo Scientific Inc., San Diego, CA, USA) and multimode plate readers (MultistkanTM GO, Thermo Scientific, Waltham, MA, USA) were used for absorbance analyses.
4.2. Synthesis and Characterization
4.2.1. General Method for the Synthesis of p-Glu-Hyd-Me
(S)-5-(benzyloxy)-4-(((benzyloxy)carbonyl)amino)-5-oxopentanoic acid (2.0 g, 5.39 mmol) was dissolved in 50 mL of DCM, and the reaction mixture was cooled to 0 °C. TEA (1.2 eq, 6.47 mmol, 902 µL) was gradually added to the mixture, and after 10 min, ECF (1.2 eq, 6.47 mmol, 615 µL) was added dropwise to the mixture over the course of 1 h. The mixture was then stirred at 0 °C for 4 h. Separately, phenyl hydrazine (1.2 eq, 6.47 mmol) and TEA (1.2 eq, 6.47 mmol, 902 µL) were added to a 100 mL pear-shaped flask. This mixture was then added slowly to the main reaction flask over 1 h while maintaining the temperature at 0 °C. Once the addition of hydrazine was complete, the reaction mixture was allowed to warm to RT and stirred for an additional 16 h. Upon completion of the reaction, the organic layer was sequentially washed with distilled water, 1 N HCl, 0.5 N NaHCO3, and again with distilled water, before being separated and collected. The organic phase was dried over Na2SO4, followed by concentration. The desired product was purified via recrystallization using an AcOEt/n-hexane mixture (1:5).
4.2.2. General Method for the Synthesis of RA-Hyd-Me Derivatives
The p-Glu-Hyd-Me analog (4.5 mmol) and palladium on carbon (10 wt%) were dissolved in MeOH (200 mL) and stirred under a hydrogen atmosphere (1 atm, using a hydrogen balloon) for 16 h. Once the reaction was complete, the mixture was passed through a 0.4 µm glass microfiber filter. The filtrate was then concentrated, followed by purification through recrystallization from a 1:5 mixture of methanol and AcOEt.
N5-(methyl(phenyl)amino)-L-glutamine (RA-Hyd-Me). From (S)-5-(benzyloxy)-4-(((benzyloxy)carbonyl)amino)-5-oxopentanoic acid; 1.0 g, 81%, white solid; IR (ν cm−1, KBr): 3195, 3030, 2932, 2856, 1656, 1582, 1517, 1451, 1403, 1309, 1250; 1H NMR (400 MHz, D2O/acetone-d6 6/1): δ 7.33 (m, 2H, PhH), 6.99 (m, 1H, PhH), 6.94 (m, 2H, PhH), 3.86 (t, J = 6.0 Hz, 1H, H-2), 3.09 (s, 3H, 1′-N-CH3), 2.61 (m, 2H, H-3), 2.26 (m, 2H, H-4); 13C NMR (100 MHz D2O/acetone-d6 6/1): δ 174.1, 174.0, 149.3, 129.6, 120.9, 114.0, 54.3, 40.6, 29.7, 26.3; HRESIMS m/z 252.1354 [M + H]+ (calcd for C12H18N3O3, 252.1348).
N5-(methyl(m-tolyl)amino)-L-glutamine (RA-Hyd-Me-Tol). From (S)-5-(benzyloxy)-4-(((benzyloxy)carbonyl)amino)-5-oxopentanoic acid; 1.24 g, 87%, white solid; IR (ν cm−1, KBr): 3225, 3030, 2918, 2613, 1655, 1582, 1515, 1450, 1403, 1309, 1251; 1H NMR (400 MHz, D2O/acetone-d6 6/2): δ 7.26 (t, J = 8.0 Hz, 1H, PhH), 6.85 (t, J = 7.2 Hz, 1H, PhH), 6.81 (s, 1H, PhH), 6.79 (m, 1H, PhH), 3.86 (t, J = 6.0 Hz, 1H, H-2), 3.11 (s, 3H, 1′-N-CH3), 2.61 (m, 2H, H-3), 2.33 (s, 3H, 3′-CH3), 2.26 (m, 2H, H-4); 13C NMR (100 MHz D2O/acetone-d6 6/2): δ173.9, 174.0, 149.5, 140.0, 129.6, 121.6, 114.5, 111.2, 54.4, 40.7, 29.8, 26.2, 20.8; HRESIMS m/z 266.1513 [M + H]+ (calcd for C13H20N3O3, 266.1505).
4.2.3. General Method for the Synthesis of Glu-Boc-RA
The starting material, (S)-5-(tert-butoxy)-4-((tert-butoxycarbonyl)amino)-5-oxopentanoic acid (2.0 g, 6.59 mmol), was dissolved in DCM (100 mL), and the temperature was reduced to 0 °C. Once the temperature was stable, TEA (1.2 eq, 7.91 mmol) was slowly introduced into the reaction and stirred for about 10 min. Then, ECF (1.2 eq, 7.91 mmol) was added dropwise over the course of an hour while keeping the temperature at 0 °C, followed by continuous stirring for 4 h. Separately, hydrazine HCl salt was dissolved in DCM (20 mL), followed by the addition of TEA (1.5 eq, 9.89 mmol). This solution was then slowly added to the main reaction mixture and stirred for approximately 10 min. After hydrazine addition, the temperature was allowed to rise to room temperature (approximately 24 °C), and the reaction was stirred for 16 h. Upon completion, the organic phase was washed with distilled water, 1 N HCl, 0.5 M NaHCO3, and distilled water again to separate the layers. The organic phase was then dried over MgSO4 and concentrated. Purification was performed using MPLC (C18 resin) with water and MeOH.
4.2.4. General Method for the Synthesis of RA-BA Derivatives
An appropriate Boc-glu-Hyd analog (7.0 mmol) was dissolved in 1 M HCl in AcOEt (100 mL, 100 mmol). The reaction was allowed to proceed at room temperature (24 °C) for approximately 18 h. The resulting white solid was filtered and washed with AcOEt and n-hexane. The filtered white solid was dried under a vacuum to obtain the RA-BA derivative.
N5-benzamido-L-glutamine hydrochloride (RA-Benzo). From (S)-5-(tert-butoxy)-4-((tert-butoxycarbonyl)amino)-5-oxopentanoic acid; 1.69 g, 85%, white solid; IR (ν cm−1, KBr): 3183, 2928, 1729, 1638, 1576, 1508, 1484, 1237; 1H NMR (400 MHz, DMSO-d6): δ 10.38 (s, 1H, 1′-NH), 10.09 (s, 1H, 5-NH), 8.49 (br s, 2H, 2-NH2) 7.86 (d, J = 7.3 Hz, 2H, PhH), 7.57 (t, J = 7.3 Hz, 1H, PhH), 7.50 (t, J = 7.3 Hz, 1H, PhH), 3.98 (t, J = 6.4 Hz, 1H, H-2), 2.52 (m, 2H, H-4), 2.08 (m, 3H, H-3); 13C NMR (100 MHz, DMSO-d6): δ170.7, 170.4, 165.5, 131.9, 128.5, 127.5, 132.4, 51.4, 28.9, 26.0; HRESIMS m/z 266.1139 [M + H]+ (calcd for C12H16N3O4, 266.1140).
N5-(2-hydroxybenzamido)-L-glutamine hydrochloride (RA-Sali). From (S)-5-(tert-butoxy)-4-((tert-butoxycarbonyl)amino)-5-oxopentanoic acid; 1.88 g, 90%, white solid; IR (ν cm−1, KBr): 2931, 1713, 1604, 1484, 1210; 1H NMR (400 MHz, DMSO-d6): δ 7.84 (dd, J = 1.7, 8.0 Hz, 1H, PhH), 7.51 (ddd, J = 1.7, 7.3, 8.7 Hz, 1H, PhH), 7.05 (m, 2H, PhH), 3.86 (t, J = 6.1 Hz, 1H, H-2), 2.61 (m, 2H, H-4), 2.26 (m, 3H, H-3); 13C NMR (100 MHz, DMSO-d6): δ173.8, 173.7, 168.6, 157.6, 135.3, 129.5, 120.7, 117.5, 115.2, 54.4, 29.7, 26.3; HRESIMS m/z 282.1087 [M + H]+ (calcd for C13H20N3O3, 282.1090).
4.3. DPPH Assay (In Vitro)
Following the method described by Blois et al. [
40], the DPPH radical scavenging activity of Ramalin and its derivatives was evaluated. In summary, 150 µL of Ramalin, its derivatives, and butylated hydroxyanisole (BHA) at concentrations of 10, 5, 2.5, and 1 µM in methanol was combined with 50 µL of 0.1 mM DPPH in methanol. The mixture was then kept in the dark at RT for 30 min. Afterward, the absorbance was measured at 540 nm.
4.4. BACE-1 Inhibition Assay
A BACE-1 inhibition assay was performed using a β-Secretase FRET kit (BACE-1, Thermo Fisher Scientific, San Diego, CA, USA) following the manufacturer’s instructions. The procedure followed the previously described protocol. A stock solution of Ramalin and its derivatives was prepared in deionized distilled water (DDW) at a concentration of 20 mM. This stock was further diluted in assay buffer to achieve final concentrations of 50, 25, 12.5, 6.25, 3.12, 1.56, 0.78, 0.39, 0.2, and 0.1 µM in each well. In black 96-well microplates, 10 µL of the BACE-1 substrate was added to the diluted samples. The reaction was initiated by adding 10 µL of 3× BACE-1 enzyme to each well. The plates were incubated for 60 min at RT in the dark. After incubation, the reaction was stopped by adding 10 µL of 2.5 mM sodium acetate to each well. A multimode plate reader (Multiskan™ GO, Thermo Scientific, Waltham, MA, USA) was used to measure fluorescence with an excitation wavelength of 545 nm and an emission wavelength of 585 nm. The IC50 value was determined by plotting the relative fluorescence units per hour (RFU/h) against the logarithmic inhibitor concentrations. All experiments were conducted in triplicate.
4.5. Tau Inhibition Activity Assay
4.5.1. Tissue Culture of Adherent Cells
SH-SY5Y cells were grown in a complete growth medium comprising Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin. For treatment, adherent SH-SY5Y cells were plated in 96-well plates at a density of 2 × 106 cells per well. Levosimendan (Sigma, St. Louis, MO, USA) was used as an inhibitor to verify the effect on tau inhibition. Upon treatment with 20 μM of each substance, the cells were cultured for 24 h. The supernatant was then collected and used in subsequent ELISA assays.
4.5.2. Tau ELISA
The tau levels were assessed using a Human Tau ELISA kit (Abcam, Cambridge, UK). After treatment, the supernatant was diluted five times. The diluted supernatant, capture antibody, and detection antibody were mixed in a 2:1:1 ratio and incubated for 1 h. The wells were then washed three times with a wash buffer. After removing the buffer, 100 µL of 3,3′,5,5′-tetramethylbenzidine developer was added and incubated for 10 min, followed by the addition of 100 µL stop solution. The optical density was measured at 450 nm. Data analysis was performed by calculating the mean and standard deviation using GraphPad Prism 8 (GraphPad, San Diego, CA, USA).
4.6. Cytotoxicity Assay
4.6.1. Cell Culture
The murine macrophage cell line RAW 264.7 (KCLB number 40071; Korean Cell Line Bank, Seoul, Republic of Korea) was cultured in DMEM (Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% heat-inactivated FBS (Invitrogen, Burlington, ON, Canada) and 1% (w/v) antibiotic–antimycotic solution (Invitrogen, Grand Island, NY, USA) under 5% CO2 at 37 °C.
4.6.2. MTT Assay
The cytotoxicity of the cells was evaluated using an MTT colorimetric assay (3-(4,5)-dimethylthiazol-2-yl-2,5-diphenyltetrazolium bromide, Amresco, Solon, OH, USA). RAW 264.7 cells were seeded at a concentration of 2 × 105 cells/mL in 96-well plates and treated with various concentrations of Ramalin and its derivatives for 24 h. Following treatment, 5 µL of MTT solution (5 mg/mL in PBS) was added to each well, and the cells were incubated for 4 h at 37 °C. After incubation, 100 µL of fresh DMSO was added to dissolve the formazan crystals, and the cells were incubated for 10 min. Absorbance at 570 nm was then measured using a microplate reader (Thermo Scientific Inc., San Diego, CA, USA). Relative cell viability was calculated by comparing the absorbance values to those of the untreated control group. All assays were performed in triplicate.
4.7. NO Assay and NLRP3 ELISA
4.7.1. Determination of Nitric Oxide Production
Nitrite accumulation served as an indicator of NO production in the medium, and the nitrite concentration was assessed by analyzing the culture supernatants using the Griess reagent (1% sulfanilamide, 0.1% N-(1-naphthyl)-ethylenediamine dihydrochloride, and 5% phosphoric acid). To quantify the nitrite levels, 1 × 106 cells/mL were seeded in 96-well plates, followed by treatment with the specified concentrations of Ramalin and its derivatives at 37 °C for 1 h. Afterward, the cells were stimulated with 0.5 μg/mL of lipopolysaccharide (LPS, 0.5 μg/mL, Sigma-Aldrich, St. Louis, MO, USA) for 24 h in a final volume of 200 μL. Subsequently, 100 μL of the cell culture supernatants were combined with 100 μL of Griess reagent in a 96-well plate. A standard curve was prepared using sodium nitrite, and the nitrite concentration was determined by measuring the absorbance at 540 nm using a microplate reader. All experiments were carried out in triplicate.
4.7.2. NLRP3 ELISA
RAW 264.7 macrophages were seeded at a density of 5 × 105 cells/well in 96-well plates and treated with 20 µM of RA-Hyd-Me, RA-Hyd-Me-tol, RA-Benzo, and RA-Sali for 1 h and then stimulated with 0.5 μg/mL LPS for 24 h. Culture supernatant levels of NOD-like receptor pyrine domain containing-3 (NLRP3) were determined using a commercially available ELISA kit (Lsbio, Seattle, WA, USA) and following the instructions of the manufacturer. The absorbance of the plate was read at a wavelength of 450 nm. NLRP3 was determined from a standard curve. The concentrations were expressed as ng/mL. To obtain statistical significance, all experiments were carried out in duplicate.
4.8. Mini Ames Test
The mutagenic potential of the test substances was evaluated using a microplate Ames assay (Xenometrics MPFTM Ames Assay, AG, Allschwil, Switzerland), which employs the bacterial strains Salmonella typhimurium TA98 and TA100. The assay was conducted according to the xenometric calculation method, a validated approach for determining the mutagenicity of chemical compounds. The AMES MPF Penta 2 mutagenicity assay kit was utilized for this study. The bacterial strains were cultured in a growth medium supplemented with 1% (v/v) penicillin/streptomycin at 37 °C, with shaking at 250 rpm for 12 to 16 h. The test substances were prepared by serially diluting them into six different concentration levels. Frozen bacterial strains were pre-cultured under the conditions described above, and the bacterial suspensions were exposed to the test substances, negative control (DMSO), and positive controls in the presence or absence of the metabolic activation system (S9) for 90 min at 37 °C. For the positive controls, the TA98 strain was exposed to 2-nitrofluorene (2-NF) without S9 and 2-aminoanthracene (2-AA) with S9, while the TA100 strain was exposed to 4-nitroquinoline 1-oxide (4-NQO) without S9 and 2-AA with S9. After the exposure period, the mixtures were combined with the indicator medium and distributed into 384-well plates, which were incubated at 37 °C for 24 to 48 h. The mutagenic potential was assessed by counting the number of revertant colonies in each well. Positive results were defined as a two-fold or greater increase in the number of revertant colonies compared to the negative control or as a dose-dependent increase in revertant colonies.
4.9. Acetylcholinesterase Activity Assay
AChE Assay
The AChE inhibitory activity was assayed following an adaptation of the spectrophotometric method reported by Ellman et al. [
41]. The Acetylcholinesterase Inhibitor Screening Kit (catalog number MAK324) and purified AChE (catalog number C3389) were purchased from Sigma-Aldrich. Enzyme solutions were prepared by dissolving lyophilized powder in double-distilled water. The AChE inhibitory activity was measured using a clear 96-well flat-bottom plate. The absorbance was read on a fluorescence spectrometer (Thermo Scientific Inc., San Diego, CA, USA) in duplicate experiments with two control wells: a standard (no enzyme) well and one well containing the AChE reference enzyme (no-inhibitor control). The experimental procedures for AChE activity assays were performed according to the technical bulletins of the acetylcholinesterase activity assay kit (MAK324; Sigma-Aldrich). Purified AChE was prepared to a concentration of 500 units/mL. A reaction mix for each well was prepared by mixing the following into a clean tube: 154 μL of assay buffer (catalog number MAK324A), 1 μL of substrate (100 mM, catalog number MAK324B), and 0.5 μL of 5,5′-dithiobis(nitrobenzoic acid) (DNTB, catalog number MAK324C). The reaction was initiated by the addition of 45 μL of the assay buffer, 5 μL of the enzyme, and the investigated compounds (5 μL) to the wells to obtain the final concentrations of 6.25, 12.5, 25, and 50 μM. A positive control of donepezil was used in the same range of concentrations. The plate was incubated for 15 min. The reaction mix (150 μL) was then added to each sample, the control (no enzyme), and the no-inhibitor control wells. The plate was tapped to mix. Absorbance was measured at 412 nm at 0 min and then at 10 min. The AChE inhibitory activity was calculated as the % of inhibition. All samples were assayed in triplicate.
4.10. Statistical Analysis
Graphs and statistical analyses were created using GraphPad Prism 8. Data were analyzed using a one-way analysis of variance (ANOVA), followed by Dunnett’s multiple comparison test. The results are expressed as the mean ± standard deviation of three independent experiments, with statistical significance levels set at * p < 0.05, ** p < 0.01, and *** p < 0.005.
4.11. Use of AI-Assisted Tools
In the preparation of this manuscript, ChatGPT-4, an AI language model developed by OpenAI, was used to assist in various tasks including translation, proofreading for typographical errors, and contextual review to ensure clarity and accuracy of the content. The AI tool was employed as a supplementary aid to improve the quality of the writing, and all outputs generated by the AI were carefully reviewed and edited by the authors to ensure the originality, validity, and integrity of the manuscript. The use of ChatGPT-4 does not meet the criteria for authorship, in accordance with MDPI’s guidelines, and was therefore not included as an author.
5. Conclusions
In this study, four novel Ramalin derivatives (RA-Hyd-Me, RA-Hyd-Me-Tol, RA-Sali, and RA-Benzo) were prepared and their stabilities and therapeutic potentials against Alzheimer’s disease (AD) were evaluated. The synthesized derivatives showed significantly improved stabilities compared to that of the parent compound, Ramalin, especially at elevated temperatures, thereby overcoming a major challenge in the development of reliable AD treatments. The incorporation of functional groups, such as methyl and benzamido groups, was found to play a crucial role in preventing oxidative degradation, thereby stabilizing the derivatized compounds. Despite a reduction in their antioxidant activities, particularly in the cases of RA-Benzo and RA-Sali, the prepared derivatives exhibited promising multi-target therapeutic properties. More specifically, RA-Sali demonstrated the most potent BACE-1 inhibitory activity, whilst significantly inhibiting tau phosphorylation, rendering it a promising candidate for further development. However, the obtained derivatives did not exhibit inhibitory activity against acetylcholinesterase, indicating that although these compounds have potential as multi-target therapeutic agents, they may not address all the pathways involved in AD. Additionally, the low molecular weights and favorable physicochemical properties of these derivatives indicate their potential ability to cross the blood–brain barrier (BBB) and exert therapeutic effects directly in the brain. However, further optimization is required to ensure effective permeability of the BBB, and to enhance their efficacies, particularly in terms of their antioxidant and anti-inflammatory activities. Thus, although the synthesized Ramalin derivatives (particularly RA-Sali) show promise as potential therapeutic agents for AD, further structural refinements and studies are required to fully understand their potential. This study represents a significant advancement in the development of stable and effective AD treatments and provides a foundation for future therapeutic strategies.