Skip to main content
Erschienen in:

Open Access 25.11.2024 | Original Paper

Sulfoquinovosyl diacylglycerol, a component of Holy Basil Ocimum tenuiflorum, inhibits the activity of the SARS-CoV-2 main protease and viral replication in vitro

verfasst von: Hinako Koze, Masayuki Sudoh, Satoaki Onitsuka, Hiroaki Okamura, Takeshi Ishikawa, Fumito Tani, Yukako Miyata-Yabuki, Mikako Shirouzu, Masanori Baba, Mika Okamoto, Toshiyuki Hamada

Erschienen in: Journal of Natural Medicines | Ausgabe 1/2025

Abstract

The persistence of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the emergence of new mutant strains continue to present a substantial threat with potential for future pandemics. Safe, effective, and readily available COVID-19 therapeutics are urgently needed to prepare for future coronavirus pandemics. To help identify new antiviral agents, the present study focused on natural products in the extracts of Holy Basil, Ocimum tenuiflorum L., which show potential inhibitory effects against the SARS-CoV-2 main protease (Mpro). Bioassay-guided isolation of the MeOH extracts of O. tenuiflorum led to the identification of a sulfur-containing glyceroglycolipid, sulfoquinovosyl diacylglycerol (SQDG: 1), as a potent Mpro inhibitor that effectively inhibited Mpro activity (IC50: 0.42 µM). SQDG (1) also markedly suppressed SARS-CoV-2 replication (EC50, 51.2 µM) in vitro while displaying no cytotoxicity (CC50 > 100 µM). Further inhibition kinetic studies and docking simulations clearly demonstrated that SQDG strongly inhibited SARS-CoV-2 Mpro in a competitive and mixed-inhibition manner. These findings highlight SQDG as a promising lead compound for COVID-19 therapy and emphasize the need to explore new drugs from natural sources.

Graphical Abstract

Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s11418-024-01855-6.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The coronavirus disease 2019 (COVID-19) pandemic, caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which emerged in December 2019, has significantly impacted global health [1]. World Health Organization (WHO) declared COVID-19 a public health emergency in January 2020 and later, in May 2023, declared the end of the emergency phase. However, owing to the persistence of the virus and the emergence of new mutant strains [24], COVID-19 continues to pose a significant threat with potential for future pandemics. Therefore, the development of safer and more effective therapies for COVID-19 remains an urgent priority.
The development of new anti-coronaviral drugs focuses on targeting two key enzymes: RNA-dependent RNA polymerase and the main protease (Mpro or 3-chymotrypsin-like protease; 3CLpro) [5, 6]. Mpro plays a crucial role in viral replication by cleaving polypeptides translated from the RNA of SARS-CoV-2 [7]. Mpro shows glutamine-specific cleavage activity not present in human proteases [8, 9] and is highly conserved across coronaviruses, such as SARS and Middle East respiratory syndrome [10], making it an ideal target for drug discovery [11]. Mpro-specific inhibitors are predicted to have negligible off-target activity and thus have limited side effects [1214]. These features make Mpro an attractive therapeutic target for COVID-19 treatment. Two Mpro inhibitors, nirmatrelvir [11] and ensitrelvir [15], have recently been developed. However, nirmatrelvir requires the concurrent use of ritonavir as a metabolic modifier, whereas ensitrelvir still has side effects. Therefore, there is an urgent need to develop more effective and specific anti-SARS-CoV-2 drugs.
Although synthetic Mpro inhibitors represent significant advances in COVID-19 therapy, the exploration of natural compounds provides a complementary approach. Leveraging biodiversity to discover novel antiviral agents is crucial for the development of effective treatments. Natural compounds from medicinal plants remain one of the major sources for the discovery of lead compounds for drug development. Recently, some natural compounds originating from medicinal plants have been found to exhibit Mpro inhibitory activity [16, 17], encouraging us to identify more potent SARS-CoV-2 Mpro inhibitors from herbal medicines. In this context, Holy Basil (Ocimum tenuiflorum L., syn. O. sanctum) or Tulsi, known for its extensive medicinal properties, emerges as a promising candidate. Holy Basil, cultivated in Southeast Asia for religious and medicinal purposes [18], has various properties, including antifertility, anticancer, antidiabetic, antifungal, antimicrobial, cardioprotective, analgesic, antispasmodic, and adaptogenic effects [19]. Its extracts are used to treat bronchitis, malaria, diarrhea, dysentery, skin conditions, arthritis, eye diseases, and insect bites [20]. A recent study also demonstrated the therapeutic potential of Holy Basil against SARS-CoV-2 infection. Specifically, eugenol, a phenolic compound found in this basil, inhibited the interaction between SARS-CoV-2 spike S1 and ACE2 to induce therapeutic responses [21].
In this study, we aimed to isolate and purify the active components of the methanol extract of the dried aerial parts of O. tenuiflorum using a bioassay-guided method and test its efficacy against SARS-CoV-2 Mpro. We identified a sulfur-containing glyceroglycolipid, sulfoquinovosyl diacylglycerol (SQDG: 1) (Fig. 1), which inhibits SARS-CoV-2 Mpro activity. SQDG (1) also inhibited the replication of SARS-CoV-2 in vitro. We also performed kinetic analysis and docking simulation to examine the inhibitory mechanisms of SQDG (1) against SARS-CoV-2 Mpro, with the goal of aiding the development of new SARS-CoV-2 inhibitors.

Results and discussion

Bioassay-guided purification from O. tenuiflorum

Upon screening the natural products for inhibitory effects against SARS-CoV-2 main protease Mpro, the crude methanol extract of the aerial parts of O. tenuiflorum showed 46% inhibition at a sample concentration of 200 µg/mL (Fig. 2). The methanol extract was partitioned between H2O and EtOAc and the H2O-soluble portion was further partitioned between n-BuOH and H2O. The n-BuOH extract, showing 63% inhibition at 200 μg/mL, was subjected to ODS vacuum column chromatography, further yielding 16 fractions. Among these, the 14th fraction exhibited ~91% inhibition at 200 µg/mL. Further fractionation and purification of the 14th fraction using a SiO2 column and reverse-phase HPLC yielded compound 1 (fr. 14-9-8, 9.5 mg) and 2 (fr. 14-6, 23.3 mg). Although the chemical structures of the two isolated compounds (1 and 2) belonged to the glycolipid family and appeared to be very similar, their Mpro inhibitory activities showed extremely different outcomes; the IC50 values were 0.42 and >200 µM, respectively. To understand these findings, we solved the chemical structures of 1 and 2 and compared the Mpro inhibitory activities of these compounds and related analogs.

Structures of 1 and 2

Compound 1 was isolated as a colorless, amorphous solid. High-resolution fast atom bombardment mass spectrometry (HR-FAB-MS) analysis of compound 1 in positive-ion mode displayed an [M + H]+ ion peak at m/z 839.4983, along with an [M+ Na]+ ion peak at m/z 861.6 (Online Resource Fig. S1), indicating a molecular formula of C45H74O12S. 1H-NMR and attached proton test (APT) spectra of 1 (Online Resource Figs. S2 and S3) showed signals corresponding to glycerol, carbohydrate, and unsaturated fatty acid components. Extensive NMR analysis (Fig. 3) was conducted using COSY (Online Resource Fig. S6), HMQC (Online Resource Fig. S4), and HMBC (Online Resource Fig. S5). Based on 2D NMR spectra, C-1 (δC 64.8 / δH 4.17 and 4.49), C-2 (δC 72.2 / δH 5.28), and C-3 (δC 67.7/δH 3.56 and 4.06) were assigned to the glycerol moiety. The other signals at δH 2.9–4.8 and δC 54–101 were assigned to one glycosyl residue. The 1H-1H COSY correlations revealed the presence of one consecutive spin system, H-1‴ to H-6‴, with a quinovosyl unit. The α-configuration of anomeric carbon (δC 100.6; C-1‴) was determined from the three bond coupling constant values (3.8 Hz) between H-1‴ (δH 4.75) and H-2‴ (δH 3.40). The other configurations of the quinovosyl unit were determined from the coupling constant of each proton. HSQC (Online Resource Fig. S4) confirmed the presence of the CH2-S group in the sulfodeoxyhexosyl unit, with CH2-6″ resonances at an unusually high frequency at δC 54.8 / δH 2.92 and 3.35. The HMBC correlations (Online Resource, Fig. S5) of H-1‴ with C-3 and H2-3 with C-1‴ confirmed that C-3 was O-glycosylated by sulfoquinovosyl groups. Furthermore, the HMBC correlations of H2-1 with C-1′ and H-2 with C-1″ indicated that CH2-1 and CH-2 were esterified by the fatty acids. The presence of lipids was confirmed by 13C signals at δC 15.2 (CH3-18′) and from δC 22.0 to 35.5 (CH2) with several double bond signals at δC 128–133/δH 5.27–5.39. Based on this structural information, compound 1 was identified as a SQDG. The presence of α-linolenic acid (C18:3) was confirmed using GC-MS. The stereochemistry of C-2S of glycerol and D-sulfoquinovose was determined by comparing the optical rotation value ([α]D 48.2°) and the chemical shifts of glycerol H-1 (δH 4.13 and 4.34) in DMSO-d6 (Online Resource Fig. S8) with those reported previously [22]. This comparison confirmed that the chemical structure of compound 1 was 1,2S-di-(9Z,12Z,15Z-octadecatrienoyl)-3-(6′-sulfo-α-d-quinovosyl)-sn-glycerol.
Compound 2 was obtained as a colorless amorphous solid. FAB-MS analysis in the positive-ion mode of compound 2 displayed [M + H]+ and [M + Na]+ ion peaks at m/z 937.6 and 959.6, respectively (Online Resource Fig. S9), which corresponds to the molecular formula C51H84O15. The 1H-NMR and APT signals for compound 2 (Online Resource Figs. S10 and S11) were similar to those of compound 1, except for the carbohydrate region. The HSQC (Online Resource Fig. S12) revealed two anomeric signals at δC 105.8/δH 4.24 (J = 7.2 Hz; H-1‴) and δC 101.2/δH 4.86 (J = 3.7 Hz; H-1‴′), along with two CH2 signals at δC 68.3/δH 3.66 and 3.91 and at δC 63.4/δH 3.69, corresponding to CH2-6‴ and CH2-6‴′, respectively (Fig. 3). These findings suggested the presence of an α-galactosyl-β-(1→6)-galactosyl unit. These results confirmed that compound 2 was a digalactosyl diacylglycerol (DGDG). GC-MS analysis of compound 2 after hydrolysis-methyl esterification revealed the structure of the fatty acids to be α-linolenic acid (C18:3). DGDG and monogalactosyl diacylglycerol (MGDG; 3) along with SQDG (1) are significant metabolites commonly found in the chloroplast thylakoid membranes of photosynthetic plants (approximately 90% of the total lipid molecules in the thylakoid membrane) [23]. Assuming that the absolute stereochemistry of the sugar and glycerol parts in 2 were the same, the chemical structure of compound 2 was determined to be 1,2-di-(9Z,12Z,15Z-octadecatrienoyl)-3-O-(α-d-galactosyl-1-6-β-d-galactosyl)-sn-glycerol.

Inhibitory activity on Mpro

We evaluated the inhibitory activities of SQDG (1) and DGDG (2) against SARS-CoV-2 Mpro, along with those of the commercially available compounds MGDG (3), sulfoquinovose (4), and methyl linolenate (5) (Fig. 4A). Enzymatic assays were performed at concentrations of 20 and 200 µM for all compounds. Among compounds 13, which belong to the glyceroglycolipid family, only compound 1 demonstrated significant potency, with an Mpro inhibition percentage of 88.5% and an IC50 value of 0.42 ± 0.15  µM (Fig. 4B). DGDG (2) and MGDG (3) had virtually no effect, even at 200 µM, indicating that the sulfoquinovose moiety of SQDG is essential for Mpro inhibition. Sulfoquinovose (4) and methyl linolenate (5) displayed very weak Mpro inhibitory activity, although the IC50 value of 5 was approximately 100 µM. These results indicated that the combined structure of the sulfate group and fatty acids in SQDG plays a crucial role in its potent Mpro inhibitory activity.

Kinetic analysis of SQDG on Mpro

SQDG exhibited strong Mpro inhibitory activity; however, its mode of inhibition differed from that of common Mpro inhibitors, such as nirmatrelvir and ensitrelvir. This discrepancy may be attributed to the structural differences between these inhibitors and SQDG. To further investigate the inhibitory mechanisms of SQDG (1), an inhibition kinetics study was conducted. As shown in Fig. 5, the Lineweaver–Burk plot of 1 against SARS-CoV-2 Mpro demonstrated that compound 1 strongly inhibited Mpro in a mixed-inhibition manner. The Dixon plot indicates a ki value of 1.9 μM with a competitive mode of inhibition. These results suggest that SQDG (1) acts as a mixed-type inhibitor of SARS-CoV-2 Mpro, implying that 1 may bind to multiple locations on this key target enzyme. These results were also supported by docking simulations of compound 1 on a dimer crystal structure of SARS-CoV-2 Mpro [24], i.e., four pockets (two catalytic sites of both chains and two non-catalytic sites located at the dimer interface) were identified as possible ligand-binding pockets of this key enzyme (Fig. 6 and Online Resource Fig. S16). These results indicate that the mode of inhibition, as shown in the Lineweaver plot, is a mixed-type.

Inhibitory effect on viral replication in vitro

To further explore the therapeutic potential of SQDG against SARS-CoV-2 infection, we assessed the inhibitory effects of compounds 1–5 on viral replication (Fig. 7). When VeroE6/TMPRSS2 cells were infected with SARS-CoV-2 and left untreated for 3 days, the virus-induced cytopathic effect caused near-complete destruction of the cells. SQDG (1) showed dose-dependent protection of the infected cells from virus-induced cell destruction, with an EC50 value of 51 µM (Fig. 7A), while this compound did not show apparent cytotoxicity at concentrations up to 100 µM. These results indicated that SQDG selectively inhibits SARS-CoV-2 replication. It is important to note that amphiphilic compounds such as SQDG may not be fully internalized into cells, potentially leading to lower antiviral activity compared to their inhibitory activity against the Mpro enzyme. Compounds 2 and 5 also inhibited cell death caused by SARS-CoV-2 in a concentration-dependent manner (Fig. 7B, E); however, their inhibitory activity against Mpro was weak. This suggests that other mechanisms, such as viral adsorption or inhibition of intracellular entry, may be involved in suppressing viral replication.
This study found that SQDG isolated from Holy Basil, O. tenuiflorum, displayed strong inhibition of SARS-CoV-2 Mpro and demonstrated concentration-dependent inhibition of SARS-CoV-2-induced cell death. Previous studies have shown that SQDGs exhibit strong antiviral against human immunodeficiency virus (HIV), antitumor, and anti-inflammatory activities [25, 26]. For example, SQDG identified in cyanobacteria (blue-green algae) exhibits antiviral activity [27], whereas those obtained from the marine red alga Gigartina tenella and pteridophyte Athyrium niponicum have been shown to inhibit the activities of DNA polymerase and HIV reverse transcriptase [28, 29]. Similarly, SQDG from spinach (Spinacia oleracea L.) inhibited DNA polymerase and suppressed the proliferation of human gastric cancer cells (NUGC-3) [30]. These findings suggest that SQDG may interact with other enzymes, such as DNA- or RNA-polymerase, which could contribute to its ability to inhibit SARS-CoV-2 replication.
In conclusion, the present study demonstrates that SQDG inhibits SARS-CoV-2 Mpro with an IC50 value of 0.42 µM and effectively suppresses viral replication. While the development of a novel therapeutic agent from SQDG against COVID-19 is complex, discovering a compound that selectively inhibits SARS-CoV-2 replication is highly significant. Further research into the structure-activity relationship and mechanism of action of this sulfosaccharide-containing glyceroglycolipid could pave the way for new therapeutic agents, not only for COVID-19 but also for other infectious diseases.

Experimental section

General procedures

Optical rotation was measured at 25°C using a JASCO P-1030 Polarimeter (Jasco International Co. Ltd., Tokyo, Japan). NMR spectra were acquired at 300 K using a JEOL JNM-ECA 600 (JEOL Ltd., Tokyo, Japan) and Avance III 600 Cryo-probe Spectrometers (Bruker BioSpin Group, Faellanden, Switzerland). UV–VIS and IR spectra were recorded on Shimadzu UV-2700 (Shimadzu Co., Kyoto, Japan) and JASCO IRT-3000 Spectrometer equipped with an ATR-30-Z accessory (Jasco International Co. Ltd., Tokyo, Japan), respectively. FAB mass spectra were obtained using a JEOL JMS-700 MStation system (JEOL Ltd.). Column chromatography was performed using silica gel 60 (Merck, 70–230 mesh) and Cosmosil 75C18-OPN (Nacalai Tesque, Kyoto, Japan), and thin-layer chromatography was performed using silica gel 60 F254 plates (Merck, 0.25 mm thick). HPLC was performed using a Shimadzu LC-10AT instrument equipped with an SPD-20A detector. The HPLC separation was performed using a C18 column, Cosmosil 5C18-MS-II (Nacalai Tesque; 10 mmI.D. × 250 mm) (Fig. 1).

Materials

Holy basil (Ocimum tenuiflorum L.) was collected from farms managed by Botanical Factory Co. Ltd. (www.​botanical.​co.​jp, Japan), Minami-Osumi-cho, Kagoshima Prefecture, Japan, in the early summer of 2023. Fresh samples were dried naturally and frozen at −30°C until extraction. Compound 4, Sulfoquinovose (CAS No.: MC9551), was purchased from MCAT GmbH (Donaueschingen, Germany). Compound 3 (MGDG; CAS No.: 1932659-76-1) and Compound 5 (Methyl linolenate; CAS No.: 301-00-8) were purchased from Sigma-Aldrich (Tokyo, Japan). Supelco 37 Component FAME Mix (CAS No.: CRM47885) was used as the methylated fatty acid standard.

Extraction and isolation

The dried plants (480 g) were homogenized with MeOH (3 L × 3), and the extract was concentrated under reduced pressure at 40–45°C. The residue (57.5 g) was partitioned between EtOAc (3 L) and H2O (1 L), and the H2O layer (26.2 g) was further partitioned between n-BuOH (3 L) and H2O. The n-BuOH layer (8.20 g), which showed Mpro inhibitory activity, was subjected to vacuum column chromatography (ODS, mobile phase: stepwise elution with 50% MeOH/H2O –80% MeOH/H2O –100% MeOH–10% CH2Cl2/MeOH–100% CH2Cl2) to obtain 16 fractions. Among these 16 fractions, Fraction 14 (fr. 14; 380 mg) exhibited the most potent inhibitory activity. Therefore, further fractionation of fr. 14 was performed using flash column chromatography (SiO2, mobile phase: 20% EtOAc /n-hexane –100% EtOAc) followed by reverse-phase HPLC, resulting in the isolation of compound 2 (fr. 14-6; 23.3 mg) and compound 1 (fr. 14-9-8; 9.5 mg).

Measurement of fatty acids using GC-MS

Methyl esterification of the fatty acids in 1 and 2 (each 1.0 mg) and purification of the FAMEs were conducted using the Nacalai Tesque fatty acid methylation kit (p/N: 06482-04) and a methylated fatty acid purification kit (P/N: 06483-94) following the manufacturers’ instructions. The resultant solutions were allowed to evaporate under a stream of N2 gas and measured using GC-MS after dissolution in 1 mL of n-hexane. GC-MS analysis of the FAMEs was performed on a 7890A/5975C GC/MSD system (Agilent Technologies, CA, USA) equipped with a DB-WAX capillary column (J&W Scientific Inc., CA, USA; 30 m × 0.25 mm I.D., 0.25 µm film thickness). The injection port was then heated to 250°C. The column was heated to 50°C for 5 min, and the temperature was increased at 10°C/min to a final temperature of 240°C, which was maintained for 10 min. The MS conditions were as follows: ionization voltage, 70 eV; emission current, 40 mA; mass range, 50–800 amu; scan rate, 1.0 scan/s; split ratio, 5:1. Helium was used as the carrier gas at a flow rate of 1 mL/min. FAMEs were identified by comparing the retention times of FAMEs with those of a standard mixture and by matching the mass spectra with the NIST Mass Spectral Library.

1,2-di-(9Z,12Z,15Z-octadecatrienoyl)-3-(6′-sulfo-α-d-quinovosyl)-sn-glycerol (SQDG) (1)

Colorless amorphous solid; \({[\alpha ]}_{D}^{20}\) +48.2 (c 1.67); IR (film): 3432, 1733, 1645, 1374, 1171, 1034 cm-1; 1H and 13C NMR (CD3OD) see Table 1; HR-FAB-MS m/z 839.4983 [M+H]+ (calculated for C45H75O12S, 839.4987).
Table 1
NMR spectral data for compounds 1 and 2 in CD3OD at 300 K
No.
1
2
δC (mult.)
δH (mult., J =Hz)
δC (mult.)
δH (mult., J =Hz)
1
64.8 (t)
4.49 (dd, 12.0, 2.9)
64.5 (t)
4.43 (dd, 12.1, 2.7)
  
4.17 (dd, 12.0, 6.8)
 
4.22 (dd, 12.1, 6.6)
2
72.2 (d)
5.28a
72.27 (d)
5.24 (m)
3
67.7 (t)
4.08 (brdd, 10.7, 5.3)
69.3 (t)
3.93 (dd, 10.9, 5.4)
  
3.56 (dd, 10.7, 6.2)
 
3.72a
1' or 1"
175.4 or 175.6 (s)
175.5 or 175.6 (s)
2' or 2"
35.5 (t)
2.34 (t, 7.6) or 2.32 (t, 7.6)
35.5 (t)
2.32 (t, 7.6) or 2.31 (t, 7.6)
3', 3"
26.6 (t)
1.60 (m)
26.6 (t)
1.60 (m)
4', 4"
30.8 (t)
1.32a
30.8 (t)
1.32a
5', 5"
30.9–31.0a (t)
1.28–1.30a
30.9–31.0a (t)
1.28–1.30a
6', 6"
31.2 (t)
1.28–1.30a
31.2 (t)
1.28–1.30a
7', 7"
31.3 (t)
1.34a
31.3 (t)
1.34a
8', 8"
28.7 (t)
2.08a
28.7 (t)
2.06a
9', 9"
129.4–131.6a (d)
5.27–5.35a
128.8–131.6a (d)
5.26–5.36a
10', 10"
129.4–131.6a (d)
5.27–5.35a
128.8–131.6a (d)
5.26–5.36a
11', 11"
27.1 (t)
2.80 (t, 6.0)
27.1 (t)
2.80 (t, 6.0)
12', 12"
129.4–131.6a (d)
5.27–5.35a
128.8–131.6a (d)
5.26–5.36a
13', 13"
129.4–131.6a (d)
5.27–5.35a
128.8–131.6a (d)
5.26–5.36a
14', 14"
27.0 (t)
2.80 (t, 6.0)
27.0 (t)
2.80 (t, 6.0)
15', 15"
128.8 (d)
5.31a
128.8–131.6a (d)
5.26–5.36a
16', 16"
133.3 (d)
5.34a
133.3 (d)
5.26–5.36a
17', 17"
22.0 (t)
2.08a
22.0 (t)
2.06a
18', 18"
15.2 (q)
0.98 (t, 7.5)
15.2 (q)
0.96 (t, 7.5)
1"'
100.6 (d)
4.75 (d, 3.8)
105.8 (d)
4.24 (d, 7.2)
2"'
74.0 (d)
3.40 (dd, 9.5, 3.8)
72.9 (d)
3.50 (dd, 9.7, 7.2)
3"'
75.4 (d)
3.62 (dd, 9.5, 9.3)
75.2 (d)
3.47 (dd, 9.7, 3.2)
4"'
75.5 (d)
3.09 (dd, 9.5, 9.3)
70.6 (d)
3.87a
5"'
70.3 (d)
4.06a
75.1 (d)
3.74a
6"'
54.8 (t)
3.35a
68.3 (t)
3.91a
  
2.92 (dd, 14.3, 9.1)
 
3.66 (dd, 10.0, 6.0)
1""
  
101.2 (d)
4.86 (d, 3.7)
2""
  
70.8 (d)
3.78 (dd, 10.1, 3.7)
3""
  
72.0(d)
3.72a
4""
  
71.7 (d)
3.87a
5""
  
73.0 (d)
3.84 (brt, 6.6)
6""
  
63.4 (t)
3.69a
aSuperimposed by other signals

1,2-di-(9Z,12Z,15Z-octadecatrienoyl)-3-O-(α-d-galactosyl-1-6-β-d-galactosyl)-sn-glycerol (DGDG) (2)

Colorless amorphous solid; \({[\alpha ]}_{D}^{20}\) +25.2 (c 0.15); IR (film): 3393, 1733, 1150, 1070 cm-1; 1H and 13C NMR (CD3OD) see Table 1; FAB-MS m/z 937.6 [M+H]+ and 959.6 [M + Na]+.

In vitro Mpro assays

SARS-CoV-2 Mpro was purchased from Sigma-Aldrich (Cat# SAE0172). The fluorogenic substrate (Dabcyl-KTSAVLQ ↓SGFRKME-Edans-NH2) for Mpro was purchased from the Peptide Institute, Inc., Japan. The inhibition assay relies on fluorescence resonance energy transfer (FRET), employing a fluorescent protein-based substrate established in a previous study [31]. Briefly, a 20 nM portion of purified SARS-CoV-2 Mpro was preincubated in a 96-well black plate (Cat#237105; Thermo Scientific, USA) with compounds in 20 mM 4-(2-hydroxyethyl) piperazine-1-ethanesulfonic acid (HEPES) (pH 6.5), 120 mM NaCl, 0.4 mM EDTA, and 4 mM DTT for approximately 5 min before the reaction was initiated by the addition of 10 µM substrate. Protease activity was monitored at 30°C by FRET with excitation and emission wavelengths of 340 and 485 nm, respectively, using a multiplate reader (TECAN Infinite 200, USA). The reduction in fluorescence at 485 nm was analyzed using single exponential decay to obtain the observed rate constant (kobs). The relative activity of Mpro was calculated as the ratio of kobs to inhibitors compared with that without inhibitors. The relative IC50 values of the compounds were determined by fitting the relative activities of different inhibitor concentrations to a four-parameter logistic equation.

Docking calculations

The crystal structure of Mpro (PDB ID: 6XHU [32]) used in this study was obtained from the RCSB PDB. The 2D structure of SQDG (1) was downloaded from PubChem (PubChem ID: 15719992, https://​pubchem.​ncbi.​nlm.​nih.​gov/​) in the SDF format, and Open Babel [33] was used to generate the 3D structure. This structure was further optimized by quantum chemical calculations at the B3LYP/6-31G level using the Gaussian 16 software [34]. AutoDock Vina [24] was used as docking software. To search for binding structures of SQDG (1) across the entire Mpro dimer, five independent docking calculations were performed with different cubic spaces (Fig. S16) as the target region. Each docking calculation was configured to output 100 predicted binding structures (NUM_MODES=100), resulting in a total of 500 predicted binding structures. The top 20 structures obtained from each docking calculation (i.e., a total of 100 structures) are shown in Fig. 6. Four sites within the Mpro dimer (two catalytic sites on chains A and B and two non-catalytic sites located at the dimer interface) were identified as potential SQDG (1) binding sites.

Antiviral assays

VeroE6 cell line expressing transmembrane protease serine 2 (VeroE6/TMPRSS2), which is highly susceptible to SARS-CoV-2 infection [35], was used for anti-SARS-CoV-2 assays. Briefly, VeroE6/TMPRSS2 cells (2 × 104 cells/well) were cultured in 96-well microtiter plates and incubated at 37 °C. After 24 h, the cells were mock-infected or infected with SARS-CoV-2 (WK-521 strain, GISAID database ID EPI_ISL_408667) at a multiplicity of infection of 0.002 and cultured in the absence or presence of various concentrations of test compounds. After three days, cell viability was assessed using the tetrazolium dye method [36]. All experiments involving SARS-CoV-2 were conducted at the biosafety level 3 (BSL3) facilities at Kagoshima University.

Acknowledgements

We wish to thank Yasuyuki Kuroki of the Botanical Factory Co., Ltd. for supplying the Holy Basil materials. We thank Mariko Ikeda (RIKEN) for helping with SARS-CoV-2 Main protease production for the SPR study and Dr. Mutsuko Kukimoto-Niino (RIKEN) for the helpful discussion. We thank the National Institutes of Biomedical Innovation, Health and Nutrition, and the National Institute of Infectious Diseases for kindly providing VeroE6/TMPRSS2 cells and SARS-CoV-2 (WK-521 strain), respectively. We would like to thank Editage (www.​editage.​com) for the English language editing.

Declarations

Conflict of interest

The authors declare no competing financial interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
4.
Zurück zum Zitat Stevens LJ, Pruijssers AJ, Lee HW, Gordon CJ, Tchesnokov EP, Gribble J, George AS, Hughes TM, Lu X, Li J, Perry JK, Porter DP, Cihlar T, Sheahan TP, Baric RS, Gotte M, Denison MR (2022) Mutations in the SARS-CoV-2 RNA-dependent RNA polymerase confer resistance to remdesivir by distinct mechanisms. Sci Transl Med 14:eabo0718. https://doi.org/10.1126/scitranslmed.abo0718CrossRefPubMed Stevens LJ, Pruijssers AJ, Lee HW, Gordon CJ, Tchesnokov EP, Gribble J, George AS, Hughes TM, Lu X, Li J, Perry JK, Porter DP, Cihlar T, Sheahan TP, Baric RS, Gotte M, Denison MR (2022) Mutations in the SARS-CoV-2 RNA-dependent RNA polymerase confer resistance to remdesivir by distinct mechanisms. Sci Transl Med 14:eabo0718. https://​doi.​org/​10.​1126/​scitranslmed.​abo0718CrossRefPubMed
7.
Zurück zum Zitat Rota PA, Oberste MS, Monroe SS, Nix WA, Campagnoli R, Icenogle JP, Penaranda S, Bankamp B, Maher K, Chen MH, Tong S, Tamin A, Lowe L, Frace M, Derisi JL, Chen Q, Wang D, Erdman DD, Peret TC, Burns C, Ksiazek TG, Rollin PE, Sanchez A, Liffick S, Holloway B, Limor J, Mccaustland K, Olsen-Rasmussen M, Fouchier R, Gunther S, Osterhaus AD, Drosten C, Pallansch MA, Anderson LJ, Bellini WJ (2003) Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science 300:1394–1399. https://doi.org/10.1126/science.1085952CrossRefPubMed Rota PA, Oberste MS, Monroe SS, Nix WA, Campagnoli R, Icenogle JP, Penaranda S, Bankamp B, Maher K, Chen MH, Tong S, Tamin A, Lowe L, Frace M, Derisi JL, Chen Q, Wang D, Erdman DD, Peret TC, Burns C, Ksiazek TG, Rollin PE, Sanchez A, Liffick S, Holloway B, Limor J, Mccaustland K, Olsen-Rasmussen M, Fouchier R, Gunther S, Osterhaus AD, Drosten C, Pallansch MA, Anderson LJ, Bellini WJ (2003) Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science 300:1394–1399. https://​doi.​org/​10.​1126/​science.​1085952CrossRefPubMed
8.
Zurück zum Zitat Goetz DH, Choe Y, Hansell E, Chen YT, McDowell M, Jonsson CB, Roush WR, Mckerrow J, Craik CS (2007) Substrate specificity profiling and identification of a new class of inhibitor for the major protease of the SARS coronavirus. Biochemistry 46:8744–8752. https://doi.org/10.1021/bi0621415CrossRefPubMed Goetz DH, Choe Y, Hansell E, Chen YT, McDowell M, Jonsson CB, Roush WR, Mckerrow J, Craik CS (2007) Substrate specificity profiling and identification of a new class of inhibitor for the major protease of the SARS coronavirus. Biochemistry 46:8744–8752. https://​doi.​org/​10.​1021/​bi0621415CrossRefPubMed
9.
Zurück zum Zitat Zhang L, Lin D, Kusov Y, Nian Y, Ma Q, Wang J, Von Brunn A, Leyssen P, Lanko K, Neyts J, De Wilde A, Snijder EJ, Liu H, Hilgenfeld R (2020) Alpha-Ketoamides as broad-spectrum inhibitors of coronavirus and enterovirus replication: Structure-based design, synthesis, and activity assessment. J Med Chem 63:4562–4578. https://doi.org/10.1021/acs.jmedchem.9b01828CrossRefPubMed Zhang L, Lin D, Kusov Y, Nian Y, Ma Q, Wang J, Von Brunn A, Leyssen P, Lanko K, Neyts J, De Wilde A, Snijder EJ, Liu H, Hilgenfeld R (2020) Alpha-Ketoamides as broad-spectrum inhibitors of coronavirus and enterovirus replication: Structure-based design, synthesis, and activity assessment. J Med Chem 63:4562–4578. https://​doi.​org/​10.​1021/​acs.​jmedchem.​9b01828CrossRefPubMed
15.
Zurück zum Zitat Unoh Y, Uehara S, Nakahara K, Nobori H, Yamatsu Y, Yamamoto S, Maruyama Y, Taoda Y, Kasamatsu K, Suto T, Kouki K, Nakahashi A, Kawashima S, Sanaki T, Toba S, Uemura K, Mizutare T, Ando S, Sasaki M, Orba Y, Sawa H, Sato A, Sato T, Kato T, Tachibana Y (2022) Discovery of S-217622, a noncovalent oral SARS-CoV-2 3CL protease inhibitor clinical candidate for treating COVID-19. J Med Chem 65:6499–6512. https://doi.org/10.1021/acs.jmedchem.2c00117CrossRefPubMedPubMedCentral Unoh Y, Uehara S, Nakahara K, Nobori H, Yamatsu Y, Yamamoto S, Maruyama Y, Taoda Y, Kasamatsu K, Suto T, Kouki K, Nakahashi A, Kawashima S, Sanaki T, Toba S, Uemura K, Mizutare T, Ando S, Sasaki M, Orba Y, Sawa H, Sato A, Sato T, Kato T, Tachibana Y (2022) Discovery of S-217622, a noncovalent oral SARS-CoV-2 3CL protease inhibitor clinical candidate for treating COVID-19. J Med Chem 65:6499–6512. https://​doi.​org/​10.​1021/​acs.​jmedchem.​2c00117CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Zhang YN, Zhu GH, Liu W, Chen XX, Xie YY, Xu JR, Jiang MF, Zhuang XY, Zhang WD, Chen HZ, Ge GB (2023) Discovery of the covalent SARS-CoV-2 Mpro inhibitors from antiviral herbs via integrating target-based high-throughput screening and chemoproteomic approaches. J Med Virol 95:29208. https://doi.org/10.1002/jmv.29208CrossRef Zhang YN, Zhu GH, Liu W, Chen XX, Xie YY, Xu JR, Jiang MF, Zhuang XY, Zhang WD, Chen HZ, Ge GB (2023) Discovery of the covalent SARS-CoV-2 Mpro inhibitors from antiviral herbs via integrating target-based high-throughput screening and chemoproteomic approaches. J Med Virol 95:29208. https://​doi.​org/​10.​1002/​jmv.​29208CrossRef
20.
Zurück zum Zitat Sinoriya SK, Singh K (2024) Review on therapeutic uses of tulsi with its phytochemical constituents in different kind of extracts. J Plant Sci Res 11(2):263 Sinoriya SK, Singh K (2024) Review on therapeutic uses of tulsi with its phytochemical constituents in different kind of extracts. J Plant Sci Res 11(2):263
27.
Zurück zum Zitat Loya S, Reshef V, Mizrachi E, Silberstein C, Rachamim Y, Carmeli S, Hizi A (1998) The inhibition of the reverse transcriptase of HIV-1 by the natural sulfoglycolipids from cyanobacteria: contribution of different moieties to their high potency. J Nat Prod 61:891–895. https://doi.org/10.1021/np970585jCrossRefPubMed Loya S, Reshef V, Mizrachi E, Silberstein C, Rachamim Y, Carmeli S, Hizi A (1998) The inhibition of the reverse transcriptase of HIV-1 by the natural sulfoglycolipids from cyanobacteria: contribution of different moieties to their high potency. J Nat Prod 61:891–895. https://​doi.​org/​10.​1021/​np970585jCrossRefPubMed
31.
34.
Zurück zum Zitat Frisch MJ, Trucks GW, Schlegel HB, Scuseria GE, Robb M, Cheeseman JR, Scalmani G, Barone VPGA, Petersson GA, Nakatsuji HJRA et al (2016) Gaussian16, revision A.03, vol 3. Gaussian Inc, Wallingford CT Frisch MJ, Trucks GW, Schlegel HB, Scuseria GE, Robb M, Cheeseman JR, Scalmani G, Barone VPGA, Petersson GA, Nakatsuji HJRA et al (2016) Gaussian16, revision A.03, vol 3. Gaussian Inc, Wallingford CT
Metadaten
Titel
Sulfoquinovosyl diacylglycerol, a component of Holy Basil Ocimum tenuiflorum, inhibits the activity of the SARS-CoV-2 main protease and viral replication in vitro
verfasst von
Hinako Koze
Masayuki Sudoh
Satoaki Onitsuka
Hiroaki Okamura
Takeshi Ishikawa
Fumito Tani
Yukako Miyata-Yabuki
Mikako Shirouzu
Masanori Baba
Mika Okamoto
Toshiyuki Hamada
Publikationsdatum
25.11.2024
Verlag
Springer Nature Singapore
Erschienen in
Journal of Natural Medicines / Ausgabe 1/2025
Print ISSN: 1340-3443
Elektronische ISSN: 1861-0293
DOI
https://doi.org/10.1007/s11418-024-01855-6