Next Article in Journal
Progranulin in Musculoskeletal Inflammatory and Degenerative Disorders, Focus on Rheumatoid Arthritis, Lupus and Intervertebral Disc Disease: A Systematic Review
Next Article in Special Issue
Marine Sponge Aaptos suberitoides Extract Improves Antiproliferation and Apoptosis of Breast Cancer Cells without Cytotoxicity to Normal Cells In Vitro
Previous Article in Journal
A Mechanism of Isoorientin-Induced Apoptosis and Migration Inhibition in Gastric Cancer AGS Cells
Previous Article in Special Issue
Cucurbitacin-B Exerts Anticancer Effects through Instigation of Apoptosis and Cell Cycle Arrest within Human Prostate Cancer PC3 Cells via Downregulating JAK/STAT Signaling Cascade
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Components with Anti-Diabetic Activity Isolated from the Leaves and Twigs of Glycosmis pentaphylla Collected in Vietnam

1
Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
2
Division of Pharmacy, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813204, Taiwan
3
National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
4
Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2022, 15(12), 1543; https://doi.org/10.3390/ph15121543
Submission received: 28 November 2022 / Revised: 9 December 2022 / Accepted: 9 December 2022 / Published: 12 December 2022

Abstract

:
A phytochemical investigation of the leaves and twigs of Glycosmis pentaphylla (Rutaceae), collected in Vietnam, yielded three new compounds named glyfuran (1), glyphyllamide (2), and glyphyllazole (3), along with twenty-five known compounds (428). The structures of isolates were determined by IR, MS, NMR, and UV data analyses. In the anti-diabetic activity screening, (+)-isoaltholacton (4), glycoborinine (17), 2′,4′-dihydroxy-4,6′-dimethoxychalcone (24), and flavokawain A (25) simultaneously exhibited inhibition of dipeptidyl peptidase-4 (DPP4) and stimulation of the glucagon-like peptide-1 (GLP-1) secretion on the murine intestinal secretin tumor cell line (STC-1).

1. Introduction

Diabetes mellitus is a metabolic disease characterized by excessive amounts of blood sugar, leading to a family of diseases that includes hypertension, osteoporosis, retinopathy, and urethritis. Along with the increasing popularity of a sedentary lifestyle, diabetes has gradually become a global epidemic. According to the World Health Organization, approximately 422 million people worldwide suffer from this disease, with up to 1.5 million dying yearly [1]. Owing to the eating habits of East Asians (rice is the staple food), the prevalence of diabetes is much higher than in other areas of the world. It is estimated that 8.5% of adults in Taiwan are afflicted with this disease, and the number of patients is predicted to increase in the future [2]. Anyway, the treatment of many diabetes complications is very costly and has become an economic burden for governments everywhere. Moreover, people with type 2 diabetes are twice as likely to develop liver or pancreatic cancer [3,4].
The leading causes of diabetes are the pancreas being unable to produce enough insulin (juvenile diabetes) or when the body becomes resistant to insulin (adult-onset diabetes). There is no cure for diabetes, but it can be treated and controlled, with treatment being divided into two kinds: subcutaneous insulin injection and oral hypoglycemic drugs. Oral hypoglycemic drugs induce effects that include the promotion of insulin secretion (Diamicron) with an increase in insulin receptor sensitivity (Metformin), α-glucosidase inhibitors (Acarbose), and dipeptidase-4 (DDP-4) inhibitors (Galvus). These medications often cause unpleasant side effects such as flatulence, diarrhea, hypoglycemia, weight gain, and even liver toxicity [1]. As a result, many people are turning to herbal medicines to treat diabetes [5]. So far, many Traditional Chinese Medicines or folk herbs have shown significant effects on lowering blood sugar or reducing the side effects of western medicines. For example, Gastrodia elata water extract can increase the quality of islet β cells and reduce cell apoptosis [6]; polyacetylene from Bidens pilosa var. radiata can decrease blood sugar and increase insulin release [7], while the polysaccharides in the flower buds of Lonicera japonica can lower blood sugar [8]. In terms of pure compounds, iminosugars and sugar derivatives were regarded as important antidiabetic agents [9,10,11,12,13].
Glucagon-like peptide-1 (GLP-1) is an incretin that can decrease blood sugar levels in a glucose-dependent manner by enhancing insulin secretion; moreover, the action of GLP-1 is preserved in patients with type 2 diabetes, and substantial pharmaceutical research has therefore been directed towards developing GLP-1-based treatment. However, endogenous GLP-1 is rapidly degraded primarily by dipeptidyl peptidase-4 (DPP-4) [14]. In our pre-screenings for anti-diabetes, the extract of Glycosmis pentaphylla (Retz.) DC. was able to simultaneously inhibit DPP-4 and stimulate the secretion of GLP-1 (Figure S1) in a dose-dependent manner (Figure S2), so G. pentaphylla has been proven to have the potential for development of an anti-diabetes drug and was accordingly selected for the following studies.
G. pentaphylla, a species of plant belonging to the family Rutaceae, is a shrub or small tree, 1.5–5.0 m high, and is widely distributed over India, Malaysia, Southern China, and the Philippine Islands to Vietnam. This plant is a Traditional Chinese Medicine (TCM) that can strengthen the stomach and relieve pain. In Vietnam, the roots, leaves, and branches of G. pentaphylla are collected year-round to relieve pain, to treat rheumatism, body aches, boils, impetigo, and snakebite. It is also used for postpartum women to cure uterine bleeding, eating indigestion, and abdominal distention [15,16]. Herein, the details of extraction, isolation, structural elucidation, anti-diabetes, and cytotoxicity of isolated compounds are described.

2. Results

The air-dried twigs and leaves of G. pentaphylla were extracted with 95% ethanol. After partition and column chromatography, three new compounds named glyfuran (1), glyphyllamide (2), and glyphyllazole (3), along with 25 known compounds, (+)-isoaltholactone (4) [17], (+)-altholactone (5) [18], 6R-goniothalamin (6) [19], (6R,7R,8S)-8-chlorogoniodiol (7) [20], (6R,7S,8R)-8-chlorogoniodiol (8) [21], 7-epi-(+)-gonidiol (9) [22], 8-epi-(+)-goniodiol (10) [23], (6S,7S,8S)-goniodiol (11) [24], (+)-9-deoxygoniopypyrone (12) [25], (−)-8-epi-9-deoxygoniopyrone (13) [25], leiocarpin C (14) [26], dictamine (15) [27], 2-hydroxy-6,8-dimethoxy-3-methyl-9H-carbazole (16) [28], glycoborinine (17) [29], N-(4-hydroxyphenethyl)cinnamamide (18) [30], uvariadiamide (19) [31], alpinetin (20) [32], tsugafolin (21) [33], naringenin trimethyl ether (22) [33], 4′,6′-dihyroxy-2′,4-dimethoxydihydrochalcone (23) [34], 2′,4′-dihyroxy-4,6′-dimethoxychalcone (24) [35], flavokawain A (25) [36], glycothiomin-A (26) [37], penangin (27) [38], and ellipeiopsol B (28) [39], were obtained (Figure 1).
Compound 1 was observed as a white amorphous powder with [ α ] D 24 + 54 (c 0.05, MeOH). The molecular formula C17H22O5 (seven indices of hydrogen deficiency) of 1 was deduced from a sodium adduct peak at m/z 329.13605 in the HRESIMS. The IR spectrum indicated the presence of hydroxy (3380 cm−1) and ester carbonyl (1684 cm−1) groups. The 1H NMR data (Table 1) of 1 clearly demonstrated the presence of one mono-substituted benzene ring [δH 7.29 (m, H-4), 7.36 (2H, m, H-3/H-5), 7.39 (2H, m, H-2/H-6)], two olefinic protons [δH 6.00 (dd, J = 11.8, 1.8 Hz, H-12), δH 6.46 (dd, J = 11.8, 6.5 Hz, H-11)], four oxymethines [δH 4.16 (t, J = 5.0 Hz, H-8), δH 4.60 (t, J = 5.0 Hz, H-9), 5.00 (d, J = 5.6 Hz, H-7), and 5.65 (ddd, J = 6.5, 5.6, 1.8 Hz, H-10)], and one methyl group [δH 0.95 (t, J = 7.5 Hz, H-4′)]. The 13C NMR and DEPT spectra (Table 1) exhibited seventeen carbons, containing one ester group (δC 167.2), one olefinic quaternary carbon (δC 140.2), seven olefinic methines (δC 120.9, 125.6, 125.6, 127.8, 128.5, 128.5, and 148.2), four oxymethines (δC 73.7, 78.7, 79.0, and 84.0), one oxymethylene (δC 64.8), two methylenes (δC 19.1 and 30.6), and one methyl group (δC 13.6).
In the COSY spectrum (Figure 2) of 1, three proton sequences of H-2/H-3/H-4/H-5/H-6, H-7/H-8/H-9/H-10/H-11/H-12, and H2-1′ (δH 4.15)/H2-2′ (δH 1.66)/H2-3′ (δH 1.40)/H3-4′ (δH 0.95) were observed. The proton spin-spin coupling systems of H-2/H-3/H-4/H-5/H-6 and the HMBC correlations (Figure 2) of H-7 to C-1 (δC 140.2) and H-2/H-6 to C-7 (δC 84.0) revealed the presence of a monosubstituted benzene ring (ring A), while the sequences of H-7/H-8/H-9/H-10 and the HMBC correlation from H-10 to C-7 (δC 84.0) suggested 1 contained a tetrahydrofuran ring (ring B). The n-butyl ester attached at C-12 was constructed by the COSY consequences H2-1′/H2-2′/H2-3′/H3-4′ and the HMBC correlation from H-11, H-12, and H-1′ to C-13 (δC 167.2). On the basis of these definitive 2D NMR analyses, the planar structure of 1 was established.
Moreover, in the NOESY spectrum of 1, the presence of NOESY correlations (Figure 2) between H-11 and H-12 validated the cis conformation of Δ11. The NOESY cross-peaks of H-8/H-9/H-10 suggested they were β-orientated; on the other hand, the NOESY correlations of H-1/H-7 revealed these protons were α-orientated.
Additionally, compound 4 is the major component of G. Pentaphylla (0.006%), suggesting compound 1 might be derived from 4 (Figure 3). After hydrolysis, 4 might become intermediate A, and A was esterificated to form 1. Through comparison between compounds 1 and 4, the absolute configuration of 1 was established and assigned the trivial name glyfuran.
Compound 2 was isolated as a white amorphous powder with   [ α ] D 24 − 10 (c 0.05, MeOH). The high-resolution ESIMS data showed a protonated molecule peak at [M + H]+ at m/z 336.18067, which indicated the molecular formula of C18H25NO5 and seven degrees of unsaturation. The IR spectrum of 2 showed absorption bands at 1736 and 1671 cm−1, revealing the presence of ester and amide functionality. The 1H NMR data (Table 2) of 2 possessed the signals of one monosubstituted benzene ring [δH 7.29 (2H, m, H-3/H-5), 7.31 (m, H-4), and 7.37 (2H, m, H-2/H-6)], two methylenes [δH 4.02 (q, J = 7.1 Hz, H-1‴) and δH 4.19 (q, J = 7.1 Hz, H-1″)], and two methyl groups [δH 0.93 (t, J = 7.4 Hz, H-4‴) and δH 1.24 (t, J = 7.1 Hz, H-2″)]. The 13C NMR and DEPT spectra exhibited eighteen carbons, including two ester carbonyls (δC 170.5 and 170.8), one amide carbonyl (δC 170.7), one olefinic quaternary carbon (δC 134.4), five olefinic methines (δC 127.4, 128.9, 128.9, 129.3, and 129.3), two oxymethylenes (δC 61.8 and 64.9), one N-bearing methine (δC 48.6), four methylenes (δC 19.0, 36.2, 30.4, and 43.6), and two methyl groups (δC 13.6 and 14.0). The planar structure of 2 was elucidated by dividing it into three partial structures, X, Y, and Z (Figure 2). A proton spin system of H-2 (δH 7.37)/H-3 (δH 7.29)/H-4 (δH 7.31)/H-5 (δH 7.29)/H-6 (δH 7.37) was observed from the COSY spectrum. In the HMBC spectrum, H2-7 (δH 3.62) showed correlations to C-1 (δC 134.4) and C-8 (δC 170.7), and the correlation from H-2/H-6 to C-1 revealed the presence of partial structure X. For the partial structure Y, a butyl group attached to an ester group was identified by the COSY correlations of H-1‴ (δH 4.02)/H-2‴ (δH 1.54)/H-3‴ (δH 1.33)/H-4‴ (δH 0.93) and the HMBC correlations from H2-3′ (δH 2.82 and 3.00) and H-1‴ to C-4′ (δC 170.8). Additionally, the HMBC correlations from H-1″ (δH 4.19) to C-1′ (δC 170.5), along with the proton spin system between H-1″ (δH 4.19) and H-2″ (δH 1.24) established the partial structure Z. These three parts were connected by the HMBC correlations from H-2′ (δH 4.83) to C-8, C-1′, and C-4′. According to the above 2D NMR analyses, the planar structure of 2 was established (Figure 2). Structurally, 2 showed similar 1H and 13C NMR data to a synthetic compound N-PhAc-D-Asp(OEt)OEt [40]. In addition, the negative optical rotation values of 2 (−10) and N-PhAc-D-Asp(OEt)OEt (−39) suggested the R configuration of C-2′.
Compound 3 was isolated as a brown-yellow solid and had a molecular formula of C15H15NO3 based on HRESIMS data (m/z 280.09466 [M + Na]+), accounting for nine degrees of unsaturation. The IR spectrum showed the absorption bands due to hydroxy (3412 cm−1), amide (1625 cm−1), and phenyl (1512 and 1443 cm−1) functional groups. The UV spectrum displayed absorption maxima at 307 and 259 nm; therefore, 3 was defined to have a carbazole skeleton [41]. The 1H NMR spectrum (Table 3) of 3 showed the presence of three singlet protons, one for a phenolic hydroxy group at δH 7.94 (brs, 2-OH) and the other two for aromatic methines at δH 6.83 (s, H-1) and 7.67 (s, H-4), as well as a couple of aromatic protons [δH 6.84 (d, J = 8.5 Hz, H-6) and 7.56 (d, J = 8.5 Hz, H-5)]. The 1H NMR spectrum also displayed a methyl at δH 2.39 (s, 3-Me), along with two methoxy groups at δH 3.96 (s, 7-OMe) and δH 4.00 (s, 8-OMe). The 13C NMR exhibited the signals of fifteen carbons, being twelve aromatic carbons (δC 96.8, 106.2, 114.3, 116.2, 117.8, 119.5, 121.4, 133.7, 134.0, 139.6, 149.4, and 152.5), one methyl carbon (δC 16.1), and two methoxy carbon groups (δC 56.9 and 60.9). The COSY correlation showed one fragment of H-5/H-6. In the HMBC experiment, 3J correlations from δH 3.96 to δC 149.4 (C-7) and from δH 4.00 to δC 133.7 (C-8) indicated the positions of two methoxy groups. In addition, the correlations from 3-Me to C-2 (δC 152.5), C-3 (δC 116.2), and C-4 (δC 121.4) revealed the position of a methyl group. The HMBC correlations from H-4 to C-5a (δC 119.5)/C-1a (δC 139.6) and from H-5 to C-4a (δC 117.8) were used to construct the pyrrole moiety. Therefore, compound 3 was determined to be 7,8-dimethoxy-3-methyl-9H-carbazol-2-ol, and the trivial name glyphyllazole was given.
The anti-diabetic activity of isolated secondary metabolites was evaluated by stimulating the secretion of GLP-1 and inhibiting DPP-4. As shown in Figure 4A, compounds 1, 4, 6, 7, 17, 24, and 25 exhibited stimulatory effects on GLP-1 secretion from murine intestinal secretin tumor cell line (STC-1) at a concentration of 100 μM. In the acute toxicity evaluation (Figure 4B), except for compound 7, all tested compounds retained the cell viability of STC-1 cells at the concentration of 100 μM. On the other hand, all tested compounds provided inhibitory effects on DPP-4 enzyme activities (Figure 4C); significantly, compounds 24, 9, 10, 1618, 20, 24, and 25 demonstrated inhibition rates over 50%.

3. Materials and Methods

3.1. General Experimental Procedures

The optical rotations, UV, and IR spectra were recorded on a Jasco P-2000 digital polarimeter, a Jasco V-530 UV/VIS spectrophotometer, and a Jasco FT-IR 4600 spectrometer, respectively. NMR spectra data were corrected in CDCl3 (δH 7.26 and δC 77.0) using solvent peaks as the internal standard on Bruker AVIII HD 700X NMR and Varian VNMRS 600 MHz spectrometers. HRESIMS data were obtained by using a Bruker 7T solariX spectrometer. Column chromatography was performed using Merck silica gel 60 (0.040–0.063 mm), C18 silica gel (0.040–0.075 mm), and Sephadex LH-20 material. NP-HPLC was composed of a Hitachi L-6000 pump and a Hitachi L-4000 UV detector with Phenomenex Luna silica (5 μm, 250 × 10 mm) and CN (5 μm, 250 × 10 mm) columns. RP-HPLC was carried out on a Shimadzu chromatography system consisting of a LC-20AT pump, a SPD-M20A PDA detector, and a CBM-20A system controller with Phenomenex Luna C18 (5 μm, 250 × 10 mm), phenyl-hexyl (5 μm, 250 × 10 mm), and Kinetex biphenyl (5 μm, 250 × 10 mm) columns.

3.2. Plant Material

The leaves and twigs of G. pentaphylla were purchased in July 2018 from Cu Lao Cham Island (Hoi An City, Quang Nam province, Vietnam) by Associate Professor Quang Vinh Nguyen (Institute of Biotechnology and Environment, Tay Nguyen University, Vietnam) and Associate Professor Chia-Hung Yen (Graduate Institute of Natural Products, Kaohsiung Medical University, Taiwan). A voucher specimen (code No. KMU-VN016) was deposited at Kaohsiung Medical University.

3.3. Extraction and Isolation

The dried leaves and twigs of G. pentaphylla (0.7 kg) were extracted by 95% EtOH (5 L × 3, each for 3 days) at room temperature to give an ethanolic extract, which was then partitioned between EtOAc and H2O (1:1) to provide an EtOAc layer. The EtOAc layer was further partitioned by hexanes/MeOH/H2O (4:3:1) to obtain a 75% MeOH(aq) layer (13.3 g). This methanol extract was subjected to a silica gel flash column (hexanes/CH2Cl2/MeOH, 60/10/1 to 0/0/1) to afford subfractions GP-1 to GP-6. GP-3 (5.2 g) was separated by a silica gel open column stepwise-eluted with hexanes/EtOAc (10/1 to 0/1) and EtOAc/MeOH (5/1 to 0/1) to yield 18 subfractions (GP-3-1 to GP-3-18). GP-3-7 (80.5 mg) was subjected to a Sephadex LH-20 open column eluting with CH2Cl2/MeOH (1/1) to obtain four fractions (GP-3-7-1 to GP-3-7-4). Compound 25 (2.1 mg) was purified from GP-3-7-4 (17.8 mg) by semi-preparative RP-HPLC (phenyl-hexyl column, flow = 2.0 mL/min, 65% MeCN(aq), isocratic, 360 nm). GP-3-10 (342.6 mg) was applied to chromatography on a silica gel open column (hexanes/CH2Cl2/MeOH, 1/1/0 to 0/0/1) to give 6 (32.5 mg) and fractions GP-3-10-2 to GP-3-10-4. GP-3-10-3 (220.9 mg) was further separated on a Sephadex LH-20 column (CH2Cl2/MeOH, 1/1) to afford seven fractions (GP-3-10-3-1 to GP-3-10-3-7). GP-3-10-3-5 (12.4 mg) was isolated by semi-preparative NP-HPLC (silica column, flow = 2.0 mL/min, hexanes/CH2Cl2/MeOH = 60/10/1, isocratic, 280 nm) to obtain 2 (3.1 mg). Compounds 1 (2.5 mg), 15 (1.1 mg), and 23 (38.1 mg) were purified from GP-3-10-3-6 (108.4 mg) by repeated semi-preparative NP-HPLC (silica and CN columns, flow = 2.0 mL/min, hexanes/CH2Cl2/MeOH = 70/10/1, isocratic, 280 nm). GP-3-11 (1.8 g) was fractionated by a Sephadex LH-20 column eluted with CH2Cl2/MeOH (1/1) to give five fractions (GP-3-11-1 to GP-3-11-5). GP-3-11-3 (167.3 mg) was subjected to semi-preparative RP-HPLC (C18 column, flow = 2.0 mL/min, 30% MeCN(aq), isocratic, 280 nm) to afford 4 (38.6 mg), 5 (4.0 mg), 7 (12.9 mg), 8 (1.5 mg), and 18 (1.4 mg). GP-3-11-4 (237.2 mg) was isolated by semi-preparative RP-HPLC (phenyl-hexyl column, flow = 2.0 mL/min, 50% MeCN(aq), isocratic, 280 nm) to yield 21 (1.4 mg) and 24 (43.2 mg). GP-3-11-5 (23.1 mg) was purified by semi-preparative RP-HPLC (C18 column, flow = 2.0 mL/min, 50% MeCN(aq), isocratic, 280 nm) to obtain 3 (3.5 mg), 16 (1.9 mg), and 17 (2.9 mg). GP-3-12 (315.4 mg) was subjected to Sephadex LH-20 column eluted with CH2Cl2/MeOH (1/1) to obtain five fractions (GP-3-12-1 to GP-3-12-5). Compound 22 (6.2 mg) was purified from GP-3-12-3 (55.8 mg) by semi-preparative RP-HPLC (C18 column, flow = 2.0 mL/min, 60% MeCN(aq), isocratic, 280 nm). Compound 28 (1.7 mg) was isolated from GP-3-12-4 (84.1 mg) by semi-preparative RP-HPLC (phenyl-hexyl column, flow = 2.0 mL/min, 30% MeCN(aq), isocratic, 210 and 280 nm). GP-3-13 (218.5 mg) was processed using a Sephadex LH-20 open column eluted with CH2Cl2/MeOH (1/1). GP-3-13-2 (153.3 mg) was fractionated over C18 open column (H2O/MeOH, 100/0 to 0/100) to give GP-3-13-2-1 to GP-3-13-2-5. Compounds 11 (15.3 mg) and 13 (4.0 mg) were obtained from GP-3-13-2-1 (24.1 mg) by semi-preparative RP-HPLC (phenyl-hexyl column, flow = 2.0 mL/min, 20% MeCN(aq), isocratic, 254 nm). Compounds 10 (12.7 mg) and 14 (13.5 mg) were isolated from GP-3-13-2-2 (65.2 mg) by repeated semi-preparative RP-HPLC (phenyl-hexyl and biphenyl columns, flow = 2.0 mL/min, 25% MeCN(aq), isocratic, 254 nm). GP-3-13-2-3 (17.5 mg) was purified by semi-preparative RP-HPLC (phenyl-hexyl column, flow = 2.0 mL/min, 40% MeOH(aq), isocratic, 254 nm) to yield 9 (4.7 mg). GP-3-13-2-4 (13.7 mg) was isolated by semi-preparative RP-HPLC (phenyl-hexyl column, flow = 2.5 mL/min, 60% MeOH(aq), isocratic, 280 nm) to afford 20 (3.2 mg). GP-3-14 (282.1 mg) was subjected to Sephadex LH-20 (CH2Cl2/MeOH, 1/1) and ODS (H2O/MeOH, 100/0 to 0/100) open columns, respectively. Compound 12 (2.9 mg) was purified from GP-3-14-2-2 (18.8 mg) by semi-preparative RP-HPLC (C18 column, flow = 2.0 mL/min, 30% MeCN(aq), isocratic, 254 nm). GP-3-15 (364.1 mg) was chromatographed by a Sephadex LH-20 open column eluted with CH2Cl2/MeOH (1/1) to obtain three fractions (GP-3-15-1 to GP-3-15-3). GP-3-15-3 (163.5 mg) was applied to a C18 gel open column (H2O/MeOH, 100/0 to 0/100) to obtain fractions GP-3-15-3-1 to GP-3-15-3-5. Compounds 26 (18.8 mg) and 27 (44.8 mg) were purified by semi-preparative RP-HPLC (biphenyl column, flow = 2.0 mL/min, 10% MeCN(aq), isocratic, 280 nm). GP-3-15-3-3 (18.6 mg) was isolated by semi-preparative RP-HPLC (C18 column, flow = 2.0 mL/min, 30% MeCN(aq), isocratic, 280 nm) to give 19 (8.8 mg).
Glyfuran (1): White amorphous powder; [ α ] D 24 + 54 (c 0.05, MeOH); UV (MeOH) λmax (log ε) 276 (3.23), 226 (3.20) nm; IR (KBr) vmax 3380, 2923, 2853, 1684, 1456, 1259, 1192, 1026 cm−1; 1H NMR and 13C NMR data, see Table 1; HRESIMS m/z 329.13605 [M + Na]+ (calcd for C17H22NaO5, 329.13594).
Glyphyllamide (2): White amorphous powder; [ α ] D 24 − 10 (c 0.05, MeOH); UV (MeOH) λmax (log ε) 285 (2.83), 213 (3.53) nm; IR (KBr) vmax 2925, 2854, 1736, 1671, 1457, 1179 cm−1; 1H NMR and 13C NMR data, see Table 2; HRESIMS m/z 336.18067 [M + H]+ (calcd for C18H26NO5, 336.18055).
Glyphyllazole (3): Brown-yellow solid; UV (MeOH) λmax (log ε) 307 (3.80), 259 (3.96), 237 (2.23), 212 (4.0) nm; IR (KBr) vmax 3142 (OH), 2925, 1626, 1513, 1443, 1296, 1229, 1140, 1068 cm−1; 1H NMR and 13C NMR data, see Table 3; HRESIMS m/z 280.09468 [M + Na]+ (calcd for C15H15NNaO3, 280.09441).

3.4. Anti-Diabetic Assays

3.4.1. Cell Culture

Murine enteroendocrine cell line (STC-1) purchased from American Type Culture Collection (ATCC) was maintained in Dulbecco’s modified Eagle’s medium (DMEM) containing 15% (v/v) horse serum (HS) and 5% (v/v) fetal bovine serum (FBS).

3.4.2. GLP-1 Secretion

Cells were seeded into a 24-well plate at 1.5 × 105 cells/well. After 72 h, cells were washed with glucose-free DMEM at 0.1% BSA three times and then replaced with DMEM (5.5 mM glucose) with/without VN016 or testing compound preparations for 1 h. At the end of treatment, the supernatant was collected and measured for GLP-1 using an active GLP-1 assay kit (Cisbio). The viability of cells after extract or compound treatment was measured by neutral red assay according to the previous description [42].

3.4.3. DPP-IV Activity Assay

According to the manufacturer’s instructions, the measurement of the activity and potential inhibition of DPP-IV, a type II membrane glycoprotein, was performed using the DPP-IV GloTM Protease Assay (Promega).

4. Conclusions

As part of an ongoing program to search for bioactive compounds from Vietnam’s medicinal plants, a chemical study on the leaves and twigs of G. pentaphylla has been carried out, resulting in the purification of three new (13) and twenty-five known (428) compounds. This research led to the isolation of eleven styryl-lactones (414) and five alkaloids (1519), which proved the genus Glycosmis is a rich source of both alkaloid and styryl-lactone. In anti-diabetic evaluation, compounds 4, 17, 24, and 25 displayed dual activities by stimulating GLP-1 secretion while inhibiting DPP-4 (which can rapidly degrade GLP-1) without cytotoxicity in STC-1 cells. Consequently, our pharmacological data supports G. pentaphylla as being a folk medicine with a hypoglycemic effect.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph15121543/s1, Figure S1: Preliminary screening of dual biological activities related to modulation of GLP-1 levels; Figure S2: dose-dependent effects of VN016-MeOH; Figures S3–S27: HR-ESI-MS, IR, NMR (1H, 13C, COSY, HSQC, HMBC, and NOESY), and UV spectra of compounds 13.

Author Contributions

Conception and design of the work: I.-C.H. and Y.-B.C.; purification, analysis and interpretation of the NMR data, structure elucidation, and manuscript preparation: M.T.T.N., Y.-C.L., S.-R.C. and F.-R.C.; bioactive assay: H.-K.L.; critical revision of the manuscript: Y.-B.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the Ministry of Science and Technology of Taiwan (MOST 110-2628-B-110-005), Zuoying Branch of Kaohsiung Armed Forces General Hospital (KAFGH-ZY_A_111021), and the National Research Institute of Chinese Medicine (PG11001-0344).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article and in the Supplementary Materials.

Acknowledgments

The authors wish to thank Quang Vinh Nguyen (Institute of Biotechnology and Environment, Tay Nguyen University, Buon Ma Thuot, Vietnam) for purchasing the plant material from Vietnam.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. World Health Organization. Diabetes. Available online: https://www.who.int/health-topics/diabetes#tab=tab_1 (accessed on 25 August 2020).
  2. Health Promotion Administration, Ministry of Health and Welfare. The Information about the Diabetes Patients in Taiwan. Available online: https://www.hpa.gov.tw/Pages/List.aspx?nodeid=359 (accessed on 25 August 2020).
  3. Coughlin, S.S.; Calle, E.E.; Teras, L.R.; Petrelli, J.; Thun, M.J. Diabetes mellitus as a predictor of cancer mortality in a large cohort of US adults. Am. J. Epidemiol. 2004, 159, 1160–1167. [Google Scholar] [CrossRef] [PubMed]
  4. Li, D.; Yeung, S.C.; Hassan, M.M.; Konopleva, M.; Abbruzzese, J.L. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology 2009, 137, 482–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Covington, M.B. Traditional Chinese Medicine in the treatment of diabetes. Diabetes Spectr. 2001, 14, 154–159. [Google Scholar] [CrossRef] [Green Version]
  6. Yang, H.J.; Kim, M.J.; Kwon, D.Y.; Kim, D.S.; Lee, Y.H.; Kim, J.E.; Park, S. Anti-diabetic activities of Gastrodia elata Blume water extracts are mediated mainly by potentiating glucose-stimulated insulin secretion and increasing β-cell mass in non-obese type 2 diabetic animals. Nutrients 2016, 8, 161. [Google Scholar] [CrossRef] [Green Version]
  7. Chien, S.C.; Young, P.H.; Hsu, Y.J.; Chen, C.H.; Tien, Y.J.; Shiu, S.Y.; Li, T.H.; Yang, C.W.; Marimuthu, P.; Tsai, L.F.; et al. Anti-diabetic properties of three common Bidens pilosa variants in Taiwan. Phytochemistry 2009, 70, 1246–1254. [Google Scholar] [CrossRef]
  8. Wang, D.; Zhao, X.; Liu, Y. Hypoglycemic and hypolipidemic effects of a polysaccharide from flower buds of Lonicera japonica in streptozotocin-induced diabetic rats. Int. J. Biol. Macromol. 2017, 102, 396–404. [Google Scholar] [CrossRef]
  9. Nash, R.J.; Kato, A.; Yu, C.-Y.; Fleet, G.W.J. Iminosugars as therapeutic agents: Recent advances and promising trends. Future Med. Chem. 2011, 3, 1513–1521. [Google Scholar] [CrossRef]
  10. Yang, L.-F.; Shimadate, Y.; Kato, A.; Li, Y.-X.; Jia, Y.-M.; Fleet, G.W.J.; Yu, C.-Y. Synthesis and glycosidase inhibition of N-substituted derivatives of DIM. Org. Biomol. Chem. 2020, 18, 999–1011. [Google Scholar] [CrossRef]
  11. Chennaiah, A.; Dahiya, A.; Dubbu, S.; Vankar, Y.D. A stereoselective synthesis of an imino glycal: Application in the synthesis of (−)-1-epi-adenophorine and a homoiminosugar. Eur. J. Org. Chem. 2018, 2018, 6574–6581. [Google Scholar] [CrossRef]
  12. Chennaiah, A.; Bhowmick, S.; Vankar, Y.D. Conversion of glycals into vicinal-1,2-diazides and 1,2-(or 2,1)-azidoacetates using hypervalent iodine reagents and Me3SiN3. Application in the synthesis of N-glycopeptides, pseudo-trisaccharides and an iminosugar. RSC Adv. 2017, 7, 41755–41762. [Google Scholar] [CrossRef]
  13. Rajasekaran, P.; Ande, C.; Vankar, Y.D. Synthesis of (5,6 & 6,6)-oxa-oxa annulated sugars as glycosidase inhibitors from 2-formyl galactal using iodocyclization as a key step. ARKIVOC 2022, 2022, 5–23. [Google Scholar]
  14. Green, B.D.; Liu, H.K.; McCluskey, J.T.; Duffy, N.A.; O’Harte, F.P.; McClenaghan, N.H.; Flatt, P.R. Function of a long-term, GLP-1-treated, insulin-secreting cell line is improved by preventing DPP IV-mediated degradation of GLP-1. Diabetes Obes. Metab. 2005, 7, 563–569. [Google Scholar] [CrossRef]
  15. Ahmed, I.; Islam, R.; Sikder, M.A.A.; Haque, M.R.; Al Mansur, M.A.; Rashid, M.A. Alkaloid, sterol and triterpenoids from Glycosmis pentaphylla (Retz.) DC. Dhaka Univ. J. Pharm. Sci. 2014, 13, 115–118. [Google Scholar] [CrossRef] [Green Version]
  16. Do, H.B.; Dang, Q.C.; Bui, X.C.; Nguyen, T.D.; Do, T.D.; Pham, V.H.; Vu, N.L.; Pham, D.M.; Pham, K.M.; Doan, T.N.; et al. Medicinal Plants and Medicinal Animals in Vietnam Episode 1 (Cây thuốc và động vật làm thuốc ở Việt Nam tập 1); Science and Technology Press: Hanoi, Vietnam, 2006; p. 541. [Google Scholar]
  17. Colegate, S.M.; Din, L.B.; Latiff, A.; Salleh, K.M.; Samsudin, M.W.; Skelton, B.W.; Tadano, K.; White, A.H.; Zakaria, Z. (+)-Isoaltholactone: A furanopyrone isolated from Goniothalamus species. Phytochemistry 1990, 29, 1701–1704. [Google Scholar] [CrossRef]
  18. Prasad, K.R.; Gholap, S.L. Stereoselective total synthesis of bioactive styryllactones (+)-goniofufurone, (+)7-epi-goniofufurone, (+)-goniopypyrone, (+)-goniotriol, (+)-altholactone, and (−)-etharvensin. J. Org. Chem. 2008, 73, 2–11. [Google Scholar] [CrossRef]
  19. Cavalheiro, A.J.; Yoshida, M. 6-[ω-Arylalkenyl]-5,6-dihydro-α-pyrones from Cryptocarya moschata (Lauraceae). Phytochemistry 2000, 53, 811–819. [Google Scholar] [CrossRef]
  20. Sharada, A.; Rao, K.L.S.; Yadav, J.S.; Rao, T.P.; Nagaiah, K. First stereoselective synthesis of (6R,7R,8S)-8-chlorogoniodiol. Synthesis 2017, 49, 2483–2487. [Google Scholar] [CrossRef]
  21. Lan, Y.H.; Chang, F.R.; Yu, J.H.; Yang, Y.L.; Chang, Y.L.; Lee, S.J.; Wu, Y.C. Cytotoxic styrylpyrones from Goniothalamus amuyon. J. Nat. Prod. 2003, 66, 487–490. [Google Scholar] [CrossRef]
  22. Yoshida, T.; Yamauchi, S.; Tago, R.; Maruyama, M.; Akiyama, K.; Sugahara, T.; Kishida, T.; Koba, Y. Syntheses of all stereoisomers of goniodiol from yeast-reduction products and their antimicrobiological activity. Biocis. Biotechnol. Biochem. 2008, 29, 2342–2352. [Google Scholar] [CrossRef] [Green Version]
  23. Ramachandran, P.V.; Chandra, J.S.; Ram Reddy, M.V. Stereoselective syntheses of (+)-goniodiol, (−)-8-epigoniodiol, and (+)-9-deoxygoniopypyrone via alkoxyallylboration and ring-closing metathesis. J. Org. Chem. 2002, 67, 7547–7550. [Google Scholar] [CrossRef]
  24. Ramesh, P.; Rao, T.P. Biosynthesis-inspired total synthesis of bioactive styryllactones (+)-goniodiol, (6S,7S,8S)-goniodiol, (−)-parvistone D, and (+)-parvistone E. J. Nat. Prod. 2016, 79, 2060–2065. [Google Scholar] [CrossRef] [PubMed]
  25. Tai, B.H.; Huyen, V.T.; Huong, T.T.; Nhiem, N.X.; Choi, E.M.; Kim, J.A.; Long, P.Q.; Cuong, N.M.; Kim, Y.H. New pyrano-pyrone from Goniothalamus tamirensis enhances the proliferation and differentiation of osteoblastic MC3T3-E1 cells. Chem. Pharm. Bull. 2010, 58, 521–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Chandra Rao, D.; Shekhar, V.; Kumar Reddy, D.; Chinnababu, B.; Venkateswarlu, Y. Concise stereoselective total synthesis of leiocarpin C. Helv. Chim. Acta 2013, 96, 2179–2184. [Google Scholar] [CrossRef]
  27. Tanaka, H.; Woong Ahn, J.; Katayama, M.; Wada, K.; Marumo, S.; Osaka, Y. Isolation of two ovicidal substances against two-spotted spider mite, Tetranychus urticae Koch, from Skimmia repens Nakai. Agric. Biol. Chem. 1985, 49, 2189–2190. [Google Scholar] [CrossRef]
  28. Schuster, C.; Roennefahrt, M.; Julich-Gruner, K.K.; Jaeger, A.; Schmidt, A.W.; Knoelker, H.J. Synthesis of the pyrano[3,2-a]carbazole alkaloids koenine, koenimbine, koenigine, koenigicine, and structural reassignment of mukonicine. Synthesis 2016, 48, 150–160. [Google Scholar]
  29. Chakravarty, A.K.; Sarkar, T.; Masuda, K.; Shiojima, K. Carbazole alkaloids from roots of Glycosmis arborea. Phytochemistry 1999, 50, 1263–1266. [Google Scholar] [CrossRef]
  30. Mahindroo, N.; Connelly, M.C.; Punchihewa, C.; Kimura, H.; Smeltzer, M.P.; Wu, S.; Fujii, N. Structure-activity relationships and cancer-cell selective toxicity of novel inhibitors of glioma-associated oncogene homologue 1 (Gli1) mediated transcription. J. Med. Chem. 2009, 52, 4277–4287. [Google Scholar] [CrossRef] [Green Version]
  31. Zarecki, A.P.; Kolanowski, J.L.; Markiewicz, W.T. Microwave-assisted catalytic method for a green synthesis of amides directly from amines and carboxylic acids. Molecules 2020, 25, 1761. [Google Scholar] [CrossRef]
  32. Itokawa, H.; Morita, M.; Mihashi, S. Phenolic compounds from the rhizomes of Alpinia speciosa. Phytochemistry 1981, 20, 2503–2506. [Google Scholar] [CrossRef]
  33. Seidel, V.; Bailleul, F.; Waterman, P.G. (Rel)-1β,2α-di-(2,4-dihydroxy-6-methoxybenzoyl)-3β,4α-di-(4-methoxyphenyl)-cyclobutane and other flavonoids from the aerial parts of Goniothalamus gardneri and Goniothalamus thwaitesii. Phytochemistry 2000, 55, 439–446. [Google Scholar] [CrossRef]
  34. Somsrisa, J.; Meepowpan, P.; Krachodnok, S.; Thaisuchat, H.; Punyanitya, S.; Nantasaen, N.; Pompimon, W. Dihydrochalcones with antiinflammatory activity from leaves and twigs of Cyathostemma argenteum. Molecules 2013, 18, 6898–6907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Thuy, T.T.; Porzel, A.; Ripperger, H.; Van Sung, T.; Adam, G. Chalcones and ecdysteroids from Vitex leptobotrys. Phytochemistry 1998, 49, 2603–2605. [Google Scholar] [CrossRef]
  36. Zenger, K.; Agnolet, S.; Schneider, B.; Kraus, B. Biotransformation of flavokawains A, B, and C, chalcones from Kava (Piper methysticum), by human liver microsomes. J. Agric. Food Chem. 2015, 63, 6376–6385. [Google Scholar] [CrossRef]
  37. Wu, T.S.; Chang, F.C.; Wu, P.L. Flavonoids, amidosulfoxides and an alkaloid from the leaves of Glycosmis citrifolia. Phytochemistry 1995, 39, 1453–1457. [Google Scholar] [CrossRef]
  38. Hinterberger, S.; Hofer, O.; Greger, H. Synthesis and corrected structures of sulphur-containing amides from Glycosmis species: Sinharines, penimides, and illukumbins. Tetrahedron 1994, 50, 6279–6286. [Google Scholar] [CrossRef]
  39. Kijjoa, A.; Bessa, J.; Pinto, M.M.; Anatachoke, C.; Silva, A.M.; Eaton, G.; Herz, W. Polyoxygenated cyclohexene derivatives from Ellipeiopsis cherrevensis. Phytochemistry 2002, 59, 543–549. [Google Scholar] [CrossRef]
  40. Conde, S.; López-Serrano, P. Regioselective lipase-catalysed amidation of N-blocked L-and D-aspartic acid diesters. Eur. J. Org. Chem. 2002, 2002, 922–929. [Google Scholar] [CrossRef]
  41. Pudlo, M.; Csányi, D.; Moreau, F.; Hajós, G.; Riedl, Z.; Sapi, J. First Suzuki–Miyaura type cross-coupling of ortho-azidobromobenzene with arylboronic acids and its application to the synthesis of fused aromatic indole-heterocycles. Tetrahedron 2007, 63, 10320–10329. [Google Scholar] [CrossRef]
  42. Sathyanarayana, A.R.; Lu, C.-K.; Liaw, C.-C.; Chang, C.-C.; Han, H.-Y.; Green, B.D.; Huang, W.-J.; Huang, C.; He, W.-D.; Lee, L.-C.; et al. 1,2,3,4,6-Penta-O-galloyl-d-glucose interrupts the early adipocyte lifecycle and attenuates adiposity and hepatic steatosis in mice with diet-induced obesity. Int. J. Mol. Sci. 2022, 23, 4052. [Google Scholar] [CrossRef]
Figure 1. Structures of compounds 128.
Figure 1. Structures of compounds 128.
Pharmaceuticals 15 01543 g001
Figure 2. Key COSY (bold), HMBC (arrow), and NOESY (double arrow) correlations of 13.
Figure 2. Key COSY (bold), HMBC (arrow), and NOESY (double arrow) correlations of 13.
Pharmaceuticals 15 01543 g002
Figure 3. The plausible biosynthetic pathway of 1 from 4.
Figure 3. The plausible biosynthetic pathway of 1 from 4.
Pharmaceuticals 15 01543 g003
Figure 4. The anti-diabetes effect of isolated compounds in STC-1 cells. (A) GLP-1 secretory effects of the methanol layer of G. pentaphylla (VN16) and isolated compounds. (B) Acute toxicity evaluation of the methanol layer of G. pentaphylla (VN16) and isolated compounds in STC-1 cells. (C) The effects of the methanol layer of G. pentaphylla (VN16) and isolated compounds on DPP4. Data represent mean ± SEM (n = 3–4). Ordinary one-way ANOVA with Dunnett’s multiple comparisons test. * p < 0.05, *** p < 0.001 when compared to None.
Figure 4. The anti-diabetes effect of isolated compounds in STC-1 cells. (A) GLP-1 secretory effects of the methanol layer of G. pentaphylla (VN16) and isolated compounds. (B) Acute toxicity evaluation of the methanol layer of G. pentaphylla (VN16) and isolated compounds in STC-1 cells. (C) The effects of the methanol layer of G. pentaphylla (VN16) and isolated compounds on DPP4. Data represent mean ± SEM (n = 3–4). Ordinary one-way ANOVA with Dunnett’s multiple comparisons test. * p < 0.05, *** p < 0.001 when compared to None.
Pharmaceuticals 15 01543 g004
Table 1. 1H NMR (600 MHz) and 13C NMR (150 MHz) spectroscopic data of 1 in CDCl3.
Table 1. 1H NMR (600 MHz) and 13C NMR (150 MHz) spectroscopic data of 1 in CDCl3.
No.1
δH (Mult, J in Hz)δC, Type
1140.2, C
27.39, m125.6, CH
37.36, m128.5, CH
47.29, m127.8, CH
57.36, m128.5, CH
67.39, m125.6, CH
75.00, d (5.6)84.0, CH
84.16, t (5.0)78.7, CH
94.60, t (5.0)73.7, CH
105.65, ddd (6.5, 5.6, 1.8)79.0, CH
116.46, dd (11.8, 6.5)148.2, CH
126.00, dd (11.8, 1.8)120.9, CH
13167.2, C
1′4.15, t (5.0)64.8, CH2
2′1.66, m30.6, CH2
3′1.40, m19.1, CH2
4′0.95, t (7.5)13.6, CH3
Table 2. 1H NMR (700 MHz) and 13C NMR (175 MHz) spectroscopic data of 2 in CDCl3.
Table 2. 1H NMR (700 MHz) and 13C NMR (175 MHz) spectroscopic data of 2 in CDCl3.
No.2
δH (Mult, J in Hz)δC, Type
1134.4, C
27.37, m128.9, CH
37.29, m129.3, CH
47.31, m127.4, CH
57.29, m129.3, CH
67.37, m128.9, CH
73.62, s43.6, CH2
8170.7, C
1′170.5, C
2′4.83, m48.6, CH
3′2.82, dd (17.0, 4.6)36.2, CH2
3.00, dd (17.0, 4.3)
4′170.8, C
1″4.19, q (7.1)61.8, CH2
2″1.24, t (7.1)14.0, CH3
1‴4.02, t (7.1)64.9, CH2
2‴1.54, m30.4, CH2
3‴1.33, m19.0, CH2
4‴0.93, t (7.4)13.6, CH3
NH6.46, d (7.6)
Table 3. 1H NMR (600 MHz) and 13C NMR (150 MHz) spectroscopic data of 3 in CDCl3.
Table 3. 1H NMR (600 MHz) and 13C NMR (150 MHz) spectroscopic data of 3 in CDCl3.
No.3
δH (Mult, J in Hz)δC, Type
16.83, s96.8, CH
1a139.6, C
2152.5, C
3116.2, C
47.67, s121.4, CH
4a117.8, C
57.56, d (8.5)114.3, CH
5a119.5, C
66.84, d (8.5)106.2, CH
7149.4, C
8133.7, C
8a134.0, C
2-OH7.94, br s
3-Me2.39, s16.1, CH3
7-OMe3.96, s56.9, CH3
8-OMe4.00, s60.9, CH3
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nguyen, M.T.T.; Hsu, I.-C.; Liu, H.-K.; Lin, Y.-C.; Chen, S.-R.; Chang, F.-R.; Cheng, Y.-B. Components with Anti-Diabetic Activity Isolated from the Leaves and Twigs of Glycosmis pentaphylla Collected in Vietnam. Pharmaceuticals 2022, 15, 1543. https://doi.org/10.3390/ph15121543

AMA Style

Nguyen MTT, Hsu I-C, Liu H-K, Lin Y-C, Chen S-R, Chang F-R, Cheng Y-B. Components with Anti-Diabetic Activity Isolated from the Leaves and Twigs of Glycosmis pentaphylla Collected in Vietnam. Pharmaceuticals. 2022; 15(12):1543. https://doi.org/10.3390/ph15121543

Chicago/Turabian Style

Nguyen, Minh Tuyet Thi, I-Chi Hsu, Hui-Kang Liu, Yu-Chi Lin, Shu-Rong Chen, Fang-Rong Chang, and Yuan-Bin Cheng. 2022. "Components with Anti-Diabetic Activity Isolated from the Leaves and Twigs of Glycosmis pentaphylla Collected in Vietnam" Pharmaceuticals 15, no. 12: 1543. https://doi.org/10.3390/ph15121543

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