Planta Medica International Open 2016; 3(03): e55-e59
DOI: 10.1055/s-0042-112226
Letter
Georg Thieme Verlag KG Stuttgart · New York

Naturally Occurring Anti-TB Agents: Isolation, Chemical Transformations and In Vitro Antitubercular Activities of Secondary Metabolites of Rhizomes of Alpinia galanga

Tushar R. Valkute
1   Division of Organic Chemistry, CSIR-National Chemical Laboratory (CSIR-NCL), Pune, India
,
Manisha Arkile
2   Combi-Chem Bioresource Centre, CSIR-National Chemical Laboratory (CSIR-NCL), Pune, India
,
Dhiman Sarkar
2   Combi-Chem Bioresource Centre, CSIR-National Chemical Laboratory (CSIR-NCL), Pune, India
,
Asish K. Bhattacharya
1   Division of Organic Chemistry, CSIR-National Chemical Laboratory (CSIR-NCL), Pune, India
› Author Affiliations
Further Information

Correspondence

Dr. Asish K. Bhattacharya
Division of Organic Chemistry
CSIR-National Chemical Laboratory (CSIR-NCL)
Dr. Homi Bhabha Road
Pune 411 008
India
Phone: +91 20 25 90 23 09   
Fax: +91 20 25 90 22 69   

Publication History

received 27 April 2016
revised 17 June 2016

accepted 04 July 2016

Publication Date:
15 September 2016 (online)

 

Abstract

A bioactivity-guided chemical examination of the acetone extract of the rhizomes of Alpinia galanga led to the isolation of six secondary metabolites, eucalyptol derivative (1) and phenylpropanoids (26). The structures of all of the isolated compounds (1–6) were elucidated on the basis of their spectral data. The isolated compounds (1–6) were in vitro assayed against active and dormant phenotypes of Mycobacterium tuberculosis H37Ra, respectively. Interestingly, 1′S-1′-acetoxychavicol acetate (2) showed good antitubercular activities against both active and dormant phenotypes of M. tuberculosis with IC50 values of 1.04 µM and 2.69 µM, respectively. Tsuji-Trost and homodimerization reactions of the active compound (2) respectively resulted in the formation of two analogues, 7 and 8. Both of these synthesized analogues were also found to be active in vitro against active [IC50 s of 3.24 and 3.87 µM, respectively, for compounds 7 and 8] and dormant [IC50 s of 8.33 and 2.41 µM, respectively, for compounds 7 and 8] phenotypes of M. tuberculosis H37Ra, respectively.


#

Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (MTB), is a leading cause of death worldwide. The World Health Organization (WHO) reported [1] that approximately 9 million people were infected with TB globally in the year 2013 alone, which resulted in 1.5 million deaths, out of which an estimated 360 000 were infected with both human immunodeficiency virus (HIV) as well as tuberculosis. It is estimated that more than half of the TB-infected population is from Southeast Asia and Western Pacific Regions with China and India alone accounting for 11 % and 24 % of total cases, respectively. The treatment requires long spells due to which several patients discontinue the treatment in between, which results in the development of multidrug resistance (MDR) and extensively drug-resistant (XDR) TB. Both of these forms of TB are highly fatal, and the treatment is both expensive and complicated, thereby further complicating the prevention, control, and treatment of TB [2], [3], [4]. Although, at present, isoniazid, ethambutol, pyrazinamide, and rifampicin are available as effective anti-TB drugs, the threat posed by the development of multidrug resistance tuberculosis (MDR-TB) against the first-line as well as the second-line drugs is a serious issue [5], [6]. Hence, the need for the development of new naturally occurring molecules to effectively treat TB and also address MDR and XDR assumes significance.

The Zingiberaceae plant, Alpinia galanga (L.) Willd., is commonly known as galangal and is widely cultivated in China, India, and Southeast Asian countries such as Thailand, Indonesia, and the Philippines [7], [8]. The rhizomes of this plant are extensively used as a spice or ginger substitute for flavoring foods. The rhizome has found several uses in the traditional system of medicine such as stomachic in China, or for carminative, antiflatulent, antifungal, and anti-itching in Thailand. In India, it has been traditionally used as a nervine tonic and for a stimulant effect [9]. Also, the use of the extract of the rhizome as an aphrodisiac, anti-inflammatory, revulsive, antiproliferative activity, antioxidant, anticholinergic, immunostimulating activity, hypoglycemic, and antimicrobial has been reported [8], [9], [10], [11], [12], [13], [14], [15], [16], [17]. The chemical examination of A. galanga has resulted in the isolation of several bioactive molecules [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35]. The pungent principal compound 1′S-1′-acetoxychavicol acetate (2) of A. galanga has been reported to possess various biological activities, such as antioxidative [36], antitumor [37], [38], [39], [40], [41], anti-inflammatory [42], xanthine oxidase inhibitory activity [43], and antifungal [44].

In continuation of our work on naturally occurring bioactive secondary metabolites [45], [46], [47], [48], [49], we initiated a systematic chemical examination of A. galanga for its antitubercular secondary metabolites. The dried rhizomes of A. galanga were successively extracted with acetone and MeOH to furnish acetone and MeOH extracts, respectively, which were assayed against both active and dormant phenotypes of M. tuberculosis. The acetone extract showed antitubercular activity ([Table 1]) against both the active and dormant phenotypes of M. tuberculosis with MIC90 (IC50) values of 17.80 (10.44) and 18.27 (10.87) µM, respectively, however, the MeOH extract was found to be totally inactive. Bioactive crude acetone was taken up for the isolation of the bioactive secondary metabolites and was fractionated over SiO2 column (100–200 mesh) into nine fractions (A–I). Fraction B was flash chromatographed using a RediSep® column (SiO2, 12 g) to furnish a pale yellow viscous oil that was identified as 2-acetoxy-1,8-cineole (1) by comparison with its reported spectral data [20], [22]. Silica gel column chromatography of fraction C resulted in 12 subfractions (C1 to C12). These subfractions were further flash chromatographed and resulted in the isolation of four compounds that were identified on the basis of their spectral data as 1′S-1′-acetoxychavicol acetate (2) [24], [25], [29], [34], trans-p-coumaryl diacetate (3) [24], [29], 1′S-1′-acetoxyeugenol acetate (4) [24], [29], [34], [43], and trans-coniferyl diacetate (5) [43]. Further, fraction D on flash chromatography furnished a viscous liquid that was identified as 1′S-1′-hydroxychavicol acetate (6) by comparison with its reported spectral data [24], [29].

Table 1In vitro antitubercular activity of pure isolated compounds (16) and their synthetic analogues (7 and 8).

Extract/Compound

Antitubercular activity against M. tuberculosis H37Ra in µM with SD values

Active state

Dormant state

MIC90

IC50 (µM)

MIC90

IC50 (µM)

NA: not active; both IC50 and MIC90 are > 100 µg/mL; SD (±): standard deviation

Acetone ext.

17.80 ± 0.17

10.44 ± 0.37

18.27 ± 1.02

10.87 ± 0.57

MeOH ext.

NA

NA

1

NA

NA

2

3.27 ± 0.08

1.04 ± 0.04

4.73 ± 0.58

2.69 ± 0.14

3

40.95 ± 1.05

25.04 ± 1.36

60.11 ± 1.20

27.12 ± 1.10

4

9.18 ± 1.29

5.04 ± 0.23

8.53 ± 0.20

5.40 ± 0.40

5

23.16 ± 1.19

17.38 ± 0.65

15.07 ± 0.78

7.80 ± 1.44

6

21.98 ± 0.09

7.98 ± 1.13

21.03 ± 0.20

9.33 ± 0.20

7

14.53 ± 1.88

3.24 ± 0.71

11.98 ± 1.10

8.33 ± 0.20

8

5.01 ± 0.41

3.87 ± 0.61

4.04 ± 0.30

2.41 ± 0.30

Rifampicin

0.048

0.0018

0.043

0.0014

The isolated compounds 16 ([Fig. 1]) were assayed in vitro against active and dormant phenotypes of M. tuberculosis H37Ra, respectively, using an established XTT reduction menadione assay (XRMA) antitubercular screening protocol [50], [51], [52]. The first-line antitubercular drug rifampicin (Sigma) was used as a reference standard and data obtained are presented in [Table 1]. Interestingly, 1′S-1′-acetoxychavicol acetate (2) was found to be the most active amongst all of the isolated metabolites against both active and dormant phenotypes of M. tuberculosis, having IC50 values of 1.04 µM and 2.69 µM, respectively. However, out of all of the isolated secondary metabolites viz. trans-p-coumaryl diacetate (3), 1′S-1′-acetoxyeugenol acetate (4), trans-coniferyl diacetate (5), and 1′S-1′-hydroxychavicol acetate (6), only compound 4 showed moderate activities with IC50 values of 5.04 µM and 5.40 µM against active and dormant phenotypes of M. tuberculosis, respectively.

Zoom Image
Fig. 1 Isolated compounds (16) from the acetone extract of the rhizomes of A. galanga.

Since compound 2, 1′S-1′-acetoxychavicol acetate (yield 562 mg), showed antitubercular activities compared to other isolated secondary metabolites against both active and dormant phenotypes of M. tuberculosis, we thought of carrying out synthesis of the analogues of 1′S-1′-acetoxychavicol acetate (2) and evaluate their in vitro antitubercular activities in order to further improve activities. The presence of allylic acetateʼs functionality in compound 2 prompted us to attempt a palladium-catalyzed Tsuji-Trost reaction [53] to synthesize its analogue (7) via a C–C bond-forming reaction ([Fig. 2]). Reaction of 1′S-1′-acetoxychavicol acetate (2) with cyclohexanone in DMSO catalyzed by Pd(OAc)2 at room temperature furnished a reaction mixture that was flash chromatographed using RediSep® column (SiO2, 12 g) and eluted with petroleum ether : ethyl acetate (0 → 10 %) to furnish compound 7 as a viscous liquid (37 %). Homodimerization [54] of compound 2 was carried out using Grubbʼs Ist generation catalyst to furnish homodimer 8 (91 %) as a colorless solid [m. p. 83–85 °C; [α]D 25 − 36.6 (c 1, CHCl3)].

Zoom Image
Fig. 2 Scheme of the preparation of derivatives of 2. Reagents and conditions: (i) Pd(OAc)2, PPh3, DMSO, cyclohexanone, pyrrolidine, rt, 3 h, 37 %; (ii) Grubbʼs Ist generation catalyst, CH2Cl2, rt, 16 h, 91 %.

The synthesized analogues 7 and 8 were assayed in vitro against both active and dormant phenotypes of M. tuberculosis for their antitubercular activities ([Table 1]). Both compounds showed in vitro antitubercular activities. Compound 7 was found to possess IC50 values of 3.24 µM and 8.33 µM, whereas compound 8 had IC50 values of 3.87 µM and 2.41 µM against active and dormant phenotypes of M. tuberculosis, respectively.

Materials and Methods

Plant material

The rhizomes of A. galanga were collected and identified by Prof. Kornkanok Ingkainan, Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Thailand from Phitsanulok, Thailand in May 2008. A herbarium specimen (003 566) is being maintained at the Department of Biology, Faculty of Pharmaceutical Sciences, Naresuan University, Thailand.


#

Synthesis of compound 7

A mixture of 1′S-1′-acetoxychavicol acetate (2; 40 mg, 0.5 mmol), Pd(OAc)2 (10 mol%), and ligand PPh3 (25 mg) in DMSO (2 mL) was stirred at room temperature for 5 min. Next, cyclohexanone (1.5 mmol, 3 equiv.) and pyrrolidine (30 mol%) were added and the reaction mixture was further stirred at room temperature for 3 h. After completion of the reaction (TLC), the reaction mixture was quenched with H2O (5 mL) and was extracted with EtOAc (3 × 25 mL). The organic layers were pooled together and washed with brine solution (1 × 25 mL). The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The crude reaction mixture was flash chromatographed using a RediSep® column (SiO2, 12 g) and eluted with petroleum ether : ethyl acetate (0 → 10 %) to furnish pure compound 7 as a viscous liquid (17 mg, 37 %); R f 0.30 (EtOAc-petroleum ether, 1 : 4); [α]D 25 + 0.23 (c 1, CHCl3); 1 H NMR (200 MHz, CDCl3) δ H: 7.33 (d, J = 8.6 Hz, 2 H), 7.00 (d, J = 8.6 Hz, 2 H), 6.44–6.29 (m, 1 H), 6.24–6.05 (m, 1 H), 2.75–2.57 (m, 1 H), 2.52–2.32 (m, 3 H), 2.29 (s, 3 H), 2.24–2.01 (m, 4 H), 1.88 (dd, J = 3.4, 8.3 Hz, 1 H), 1.74–1.59 (m, 2 H); 13 C NMR (50 MHz, CDCl3) δ C: 212.5, 169.6, 149.6, 135.4, 130.7, 128.7, 126.9, 121.6, 77.7, 77.0, 76.4, 50.7, 42.2, 33.6, 33.0, 27.9, 25.1, 21.2; ESI-MS: m/z 295.1 [M + Na]+; HRMS (ESI): calcd. for C17H20O3Na [M + Na]+ 295.1305, found 295.1298.


#

Synthesis of compound 8

A stirred solution of 1′S-1′-acetoxychavicol acetate (2; 40 mg) was dissolved in dry CH2Cl2 (2 mL) and degassed for 15 min. Then Grubbʼs Ist generation catalyst (15 mol%) was added to the reaction mixture and stirring was continued for a further 16 h at room temperature under an argon atmosphere. After the completion of reaction (TLC), the solvent was removed under reduced pressure. The crude reaction mixture was flash chromatographed using RediSep® column (SiO2, 12 g) and eluted with petroleum ether : ethyl acetate (0 → 20 %) to furnish pure homodimer 8 as a colorless solid (68 mg, 91 %); R f 0.30 (EtOAc-petroleum ether, 3 : 7); m. p. 83–85 °C; [α]D 25 − 36.6 (c 1, CHCl3); 1 H NMR (400 MHz, CDCl3) δ H: 7.34 (d, J = 8.7 Hz, 4 H), 7.07 (d, J = 8.2 Hz, 4 H), 6.27–6.32 (m, 2 H), 5.90 (dd, J = 2.7, 1.4 Hz, 2 H), 2.29 (s, 6 H), 2.09 (s, 6 H); 13 C NMR (100 MHz, CDCl3) δ C: 169.9, 169.5, 150.6, 136.3, 130.6, 130.6, 128.6, 121.9, 74.4, 21.3, 21.2; ESI-MS: m/z 463.1 [M + Na]+; HRMS (ESI): calcd. for C24H24O8Na [M + Na]+ 463.1363, found 463.1351.


#

Antitubercular assay using the XTT reduction menadione assay protocol

Crude extracts and pure compounds 1 – 8 were evaluated for their in vitro effects against the active and dormant phase of M. tuberculosis H37Ra (MTB) using the XRMA protocol [51]. M. tuberculosis H37Ra (ATCC 25 177) was obtained from MTCC. MTB (ATCC No. 25 177) were grown to the logarithmic phase (O. D. 1.0) in a Mycobacterium phlei medium. The stock culture was maintained at − 70 °C and subcultured once in M. phlei medium before inoculation into the experimental culture. All experiments were performed in triplicate, and IC50 and MIC values were calculated from their dose-response curves.

%Inhibition = 100 – (A1 – blank)/(A2 – blank) × 100

where A1 is the culture absorbance at 470 nm in the presence of the compound after the addition of menadione, A2 is the culture absorbance at 470 nm (DMSO solvent control) after the addition of menadione, and blank is the culture absorbance at 470 nm of the respective data points before the addition of XTT/menadione [51].


#

XTT reduction menadione assay protocol

Activity against MTB was determined through the XRMA, reading absorbance at 470 nm, as per the protocol [51]. A compound solution (2.5 µL) was added in a total volume of 250 µL of M. pheli medium consisting of the MTB, sealed with plate sealers and allowed to incubate for 8 (active phase) and 12 (dormant phase) days at 37 °C. The XRMA was then carried out to estimate the viable cells present in different wells of the assay plate. To all wells, 200 µM of XTT were added and incubated at 37 °C for another 20 min. It was followed by the addition of 60 µM of menadione and incubated at 37 °C for 40 min. The optical density was measured using a microplate reader (SpectraMax Plus 384 plate reader, Molecular Devices, Inc.) at 470 nm filter against a blank prepared from a well free of cells. Absorbance obtained from the cells treated with 1 % DMSO alone was considered 100 % cell growth. The %inhibition in the presence of test material is calculated by using formula,

%Inhibition = (average of control – average of compound)/(average of control – average of blank) × 100

where control is culture medium with cells and DMSO and blank are culture medium without cells. For all samples, each compound concentration was tested in triplicate in a single experiment and the quantitative value is expressed as the mean ± standard deviation (S. D.).


#

Supporting information

The general experimental procedures, extractions of the plant material, isolation of compounds, and antitubercular assay protocol as well as copies of their 1H, 13C, DEPT, LCMS, and HRMS spectra are available as Supporting Information.


#
#

Acknowledgments

This work was supported by the Council of Scientific and Industrial Research (CSIR)-New Delhi sponsored network projects, NaPAHA (CSC0130) and NORMS (CSC0406). The authors are grateful to Prof. Kornkanok Ingkainan, Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Thailand for the collection and identification of A. galanga rhizomes.


#
#

Conflict of Interest

The authors declare no conflict of interest.

Supporting Information

  • References

  • 1 World Health Organization. Global tuberculosis report: WHO report 2014. Geneva, Switzerland: WHO Press; 2014
  • 2 Sankar MM, Singh J, Diana SCA, Singh S. Molecular characterization of Mycobacterium tuberculosis isolates from North Indian patients with extrapulmonary tuberculosis. Tuberculosis 2013; 93: 75-83
  • 3 Young DB, Perkins MD, Duncan K, Barry CE. Confronting the scientific obstacles to global control of tuberculosis. J Clin Invest 2008; 118: 1255-1265
  • 4 Lienhardt C, Vernon A, Raviglione MC. New drugs and new regimens for the treatment of tuberculosis: review of the drug development pipeline and implications for national programmes. Curr Opin Pulm Med 2010; 16: 186-193
  • 5 Shaikh MH, Subhedar DD, Nawale L, Sarkar D, Khan FA, Sangshetti JN, Shingate BB. 1,2,3-Triazole derivatives as antitubercular agents: synthesis, biological evaluation and molecular docking study. Medchemcomm 2015; 6: 1104-1116
  • 6 Singh MM. XDR-TB–danger ahead. Indian J Tuberc 2007; 54: 1-2
  • 7 Rao K, Chodisetti B, Gandi S, Mangamoori LN, Giri A. Direct and indirect organogenesis of Alpinia galanga and the phytochemical analysis. Appl Biochem Biotechnol 2011; 165: 1366-1378
  • 8 Gupta P, Bhatter P, Dʼsouza D, Tolani M, Daswani P, Tetali P, Birdi T. Evaluating the anti Mycobacterium tuberculosis activity of Alpinia galanga (L.) Willd. axenically under reducing oxygen conditions and in intracellular assays. BMC Complement Altern Med 2014; 14: 84
  • 9 Warrier PK, Nambiar VPK. Indian medicinal plants: a compendium of 500 species, Vol. 1. New Delhi: Orient Longman Pvt Ltd.; 1994: 104
  • 10 Samarghandian S, Hadjzadeh M, Afshari JT, Hosseini M. Antiproliferative activity and induction of apoptotic by ethanolic extract of Alpinia galanga rhizhome in human breast carcinoma cell line. BMC Complement Altern Med 2014; 14: 192
  • 11 Köse LP, Gülçin İ, Gören AC, Namiesnik J, Martinez-Ayala AL, Gorinstein S. LC–MS/MS analysis, antioxidant and anticholinergic properties of galanga (Alpinia officinarum Hance) rhizomes. Ind Crops Prod 2015; 74: 712-721
  • 12 Khattak S, Shah HU, Ahmad W, Ahmad M. Biological effects of indigenous medicinal plants Curcuma longa and Alpinia galanga . Fitoterapia 2005; 76: 254-257
  • 13 Bendjeddou D, Lalaoui K, Satta D. Immunostimulating activity of the hot water-soluble polysaccharide extracts of Anacyclus pyrethrum, Alpinia galanga and Citrullus colocynthis . J Ethnopharmacol 2003; 88: 155-160
  • 14 Akhtar MS, Khan MA, Malik MT. Hypoglycaemic activity of Alpinia galanga rhizome and its extracts in rabbits. Fitoterapia 2002; 73: 623-628
  • 15 Chan EW, Ng VP, Tan VV, Low YY. Antioxidant and antibacterial properties of Alpinia galanga, Curcuma longa, and Etlingera elatior (Zingiberaceae). Pharmacog J 2011; 3: 54-61
  • 16 Trakranrungsie N, Chatchawanchonteera A, Khunkitti W. Ethnoveterinary study for antidermatophytic activity of Piper betle, Alpinia galanga and Allium ascalonicum extracts in vitro . Res Vet Sci 2008; 84: 80-84
  • 17 Oonmetta-Aree J, Suzuki T, Gasaluck P, Eumkeb G. Antimicrobial properties and action of galangal (Alpinia galanga Linn.) on Staphylococcus aureus . LWT Food Sci Technol 2006; 39: 1214-1220
  • 18 Ling ZH, Lv-Yi CH, Liang JY. Two new phenylpropanoids isolated from the rhizomes of Alpinia galanga . Chinese J Nat Med 2012; 10: 370-373
  • 19 Xiao-Lu ZH, Ming-Hua YA, Jian-Guang LU, Huang XF, Ling-Yi KO. A new phenylpropanoid from Alpinia galanga . Chinese J Nat Med 2009; 7: 19-20
  • 20 Kubota K, Nakamura K, (Murayama) Kobayashi A, Amaike M. Acetoxy-1, 8-cineoles as aroma constituents of Alpinia galanga Willd. J Agric Food Chem 1998; 46: 5244-5247
  • 21 Barik BR, Kundu AB, Dey AK. Two phenolic constituents from Alpinia galanga rhizomes. Phytochemistry 1987; 26: 2126-2127
  • 22 De Pooter HL, Omar MN, Coolsaet BA, Schamp NM. The essential oil of greater galanga (Alpinia galanga) from Malaysia. Phytochemistry 1985; 24: 93-96
  • 23 Yang WQ, Gao Y, Li M, Miao DR, Wang F. New chalcones bearing a long-chain alkylphenol from the rhizomes of Alpinia galanga . J Asian Nat Prod Res 2015; 17: 783-787
  • 24 Roy SK, Pahwa S, Nandanwar H, Jachak SM. Phenylpropanoids of Alpinia galanga as efflux pump inhibitors in Mycobacterium smegmatis mc2 155. Fitoterapia 2012; 83: 1248-1255
  • 25 Singh JH, Alagarsamy V, Diwan PV, Kumar SS, Nisha JC, Reddy YN. Neuroprotective effect of Alpinia galanga (L.) fractions on Aβ (25–35) induced amnesia in mice. J Ethnopharmacol 2011; 138: 85-91
  • 26 Latha C, Shriram VD, Jahagirdar SS, Dhakephalkar PK, Rojatkar SR. Antiplasmid activity of 1′-acetoxychavicol acetate from Alpinia galanga against multi-drug resistant bacteria. J Ethnopharmacol 2009; 123: 522-525
  • 27 Phitak T, Choocheep K, Pothacharoen P, Pompimon W, Premanode B, Kongtawelert P. The effects of p-hydroxycinnamaldehyde from Alpinia galanga extracts on human chondrocytes. Phytochemistry 2009; 70: 237-243
  • 28 Yasuhara T, Manse Y, Morimoto T, Qilong W, Matsuda H, Yoshikawa M, Muraoka O. Acetoxybenzhydrols as highly active and stable analogues of 1′S-1′-acetoxychavicol, a potent antiallergic principal from Alpinia galanga . Bioorg Med Chem Lett 2009; 19: 2944-2946
  • 29 Matsuda H, Pongpiriyadacha Y, Morikawa T, Ochi M, Yoshikawa M. Gastroprotective effects of phenylpropanoids from the rhizomes of Alpinia galanga in rats: structural requirements and mode of action. Eur J Pharmacol 2003; 471: 59-67
  • 30 Tamura S, Shiomi A, Kimura T, Murakami N. Halogenated analogs of 1′-acetoxychavicol acetate, Rev-export inhibitor from Alpinia galanga, designed from mechanism of action. Bioorg Med Chem Lett 2010; 20: 2082-2085
  • 31 Tamura S, Shiomi A, Kaneko M, Ye Y, Yoshida M, Yoshikawa M, Kimura T, Kobayashi M, Murakami N. New Rev-export inhibitor from Alpinia galanga and structure-activity relationship. Bioorg Med Chem Lett 2009; 19: 2555-2557
  • 32 Hasima N, Aun LI, Azmi MN, Aziz AN, Thirthagiri E, Ibrahim H, Awang K. 1′S-1′-Acetoxyeugenol acetate: a new chemotherapeutic natural compound against MCF-7 human breast cancer cells. Phytomedicine 2010; 17: 935-939
  • 33 Matsuda H, Ando S, Morikawa T, Kataoka S, Yoshikawa M. Structure–activity relationships of 1′S-1′-acetoxychavicol acetate for inhibitory effect on NO production in lipopolysaccharide-activated mouse peritoneal macrophages. Bioorg Med Chem Lett 2005; 15: 1949-1953
  • 34 Zeng QH, Lu CL, Zhang XW, Jiang JG. Isolation and identification of ingredients inducing cancer cell death from the seeds of Alpinia galanga, a Chinese spice. Food Funct 2015; 6: 431-443
  • 35 Phanthong P, Lomarat P, Chomnawangb MT, Bunyapraphatsaraa N. Antibacterial activity of essential oils and their active components from Thai spices against foodborne pathogens. Sci Asia 2013; 39: 472-476
  • 36 Kubota K, Ueda Y, Yasuda M, Masuda A. Occurrence and antioxidative activity of 1′-acetoxychavicol acetate and its related compounds in the rhizomes of Alpinia galanga during cooking. In: Spanier AM, Shahidi F, Parliment TH, Mussinan C, Ho CT, Tratras Contis E, editors. Food flavors and chemistry: advances of the new millennium. Cambridge, UK: The Royal Society of Chemistry 2001; 274: 601-607
  • 37 Moffatt J, Hashimoto M, Kojima A, Kennedy DO, Murakami A, Koshimizu K, Ohigashi H, Matsui-Yuasa I. Apoptosis induced by 1′-acetoxychavicol acetate in Ehrlich ascites tumor cells is associated with modulation of polyamine metabolism and caspase-3 activation. Carcinogenesis 2000; 21: 2151-2157
  • 38 Itokawa H, Morita H, Sumitomo T, Totsuka N, Takeya K. Antitumour principles from Alpinia galanga . Planta Med 1987; 53: 32-33
  • 39 Kondo A, Ohigashi H, Murakami A, Suratwadee J, Koshimizu K. 1′-acetoxychavicol acetate as a potent inhibitor of tumor promoter-induced Epstein-Barr virus activation from Languas gaianga, a traditional Thai condiment. Biosci Biotechnol Biochem 1993; 57: 1344-1345
  • 40 Zheng Q, Hirose Y, Yoshimi N, Murakami A, Koshimizu K, Ohigashi H, Sakata K, Matsumoto Y, Sayama Y, Mori H. Further investigation of the modifying effect of various chemopreventive agents on apoptosis and cell proliferation in human colon cancer cells. J Cancer Res Clin Oncol 2002; 128: 539-546
  • 41 Murakami A, Toyota K, Ohura S, Koshimizu K, Ohigashi H. Structure-activity relationships of (1′S)-1′-acetoxychavicol acetate, a major constituent of a southeast Asian condiment plant Languas galanga, on the inhibition of tumor-promoter-induced Epstein-Barr virus activation. J Agric Food Chem 2000; 48: 1518-1523
  • 42 Nakamura Y, Murakami A, Ohto Y, Torikai K, Tanaka T, Ohigashi H. Suppression of tumor promoter-induced oxidative stress and inflammatory responses in mouse skin by a superoxide generation inhibitor 1′-acetoxychavicol acetate. Cancer Res 1998; 58: 4832-4839
  • 43 Noro T, Sekiya T, Katoh M, Oda Y, Miyase T, Kuroyanagi M, Ueno A, Fukushima S. Inhibitors of xanthine oxidase from Alpinia galanga . Chem Pharm Bull 1988; 36: 244-248
  • 44 Janssen AM, Scheffer JJ. Acetoxychavicol acetate, an antifungal component of Alpinia galangal . Planta Med 1985; 51: 507-511
  • 45 Bhattacharya AK, Chand HR, John J, Deshpande MV. Clerodane type diterpene as a novel antifungal agent from Polyalthia longifolia var. pendula . Eur J Med Chem 2015; 94: 1-7
  • 46 Bhattacharya AK, Rana KC. Antimycobacterial agent, (E)-phytol and lauric amide from the plant Lagascea mollis . Ind J Chem 2013; 52?B: 901-903
  • 47 Bhattacharya AK, Pathak AK, Sharma RP. Semi-synthesis of deoxyartemisinin. Mendeleev Commun 2007; 17: 27-28
  • 48 Bhattacharya AK, Pal M, Jain DC, Joshi BS, Roy R, Rychlewska U, Sharma RP. Stereoselective reduction of arteannuin B and its chemical transformations. Tetrahedron 2003; 59: 2871-2876
  • 49 Bhattacharya AK, Jain DC, Sharma RP, Roy R, McPhail AT. Boron trifluoride-acetic anhydride catalysed rearrangement of dihydroarteannuin B. Tetrahedron 1997; 53: 14975-14990
  • 50 Sarkar S, Sarkar D. Potential use of nitrate reductase as a biomarker for the identification of active and dormant inhibitors of Mycobacterium tuberculosis in a THP1 infection model. J Biomol Screening 2012; 17: 966-973
  • 51 Singh U, Akhtar S, Mishra A, Sarkar D. A novel screening method based on menadione mediated rapid reduction of tetrazolium salt for testing of anti-mycobacterial agents. J Microbiol Methods 2011; 84: 202-207
  • 52 Khan A, Sarkar S, Sarkar D. Bactericidal activity of 2-nitroimidazole against the active replicating stage of Mycobacterium bovis BCG and Mycobacterium tuberculosis with intracellular efficacy in THP-1 macrophages. Int J Antimicrob Agents 2008; 32: 40-45
  • 53 Ibrahem I, Córdova A. Direct catalytic intermolecular α‐allylic alkylation of aldehydes by combination of transition‐metal and organocatalysis. Angew Chem Int Ed 2006; 45: 1952-1956
  • 54 Rosebrugh LE, Herbert MB, Marx VM, Keitz BK, Grubbs RH. Highly active ruthenium metathesis catalysts exhibiting unprecedented activity and Z-selectivity. J Am Chem Soc 2013; 35: 1276-1279

Correspondence

Dr. Asish K. Bhattacharya
Division of Organic Chemistry
CSIR-National Chemical Laboratory (CSIR-NCL)
Dr. Homi Bhabha Road
Pune 411 008
India
Phone: +91 20 25 90 23 09   
Fax: +91 20 25 90 22 69   

  • References

  • 1 World Health Organization. Global tuberculosis report: WHO report 2014. Geneva, Switzerland: WHO Press; 2014
  • 2 Sankar MM, Singh J, Diana SCA, Singh S. Molecular characterization of Mycobacterium tuberculosis isolates from North Indian patients with extrapulmonary tuberculosis. Tuberculosis 2013; 93: 75-83
  • 3 Young DB, Perkins MD, Duncan K, Barry CE. Confronting the scientific obstacles to global control of tuberculosis. J Clin Invest 2008; 118: 1255-1265
  • 4 Lienhardt C, Vernon A, Raviglione MC. New drugs and new regimens for the treatment of tuberculosis: review of the drug development pipeline and implications for national programmes. Curr Opin Pulm Med 2010; 16: 186-193
  • 5 Shaikh MH, Subhedar DD, Nawale L, Sarkar D, Khan FA, Sangshetti JN, Shingate BB. 1,2,3-Triazole derivatives as antitubercular agents: synthesis, biological evaluation and molecular docking study. Medchemcomm 2015; 6: 1104-1116
  • 6 Singh MM. XDR-TB–danger ahead. Indian J Tuberc 2007; 54: 1-2
  • 7 Rao K, Chodisetti B, Gandi S, Mangamoori LN, Giri A. Direct and indirect organogenesis of Alpinia galanga and the phytochemical analysis. Appl Biochem Biotechnol 2011; 165: 1366-1378
  • 8 Gupta P, Bhatter P, Dʼsouza D, Tolani M, Daswani P, Tetali P, Birdi T. Evaluating the anti Mycobacterium tuberculosis activity of Alpinia galanga (L.) Willd. axenically under reducing oxygen conditions and in intracellular assays. BMC Complement Altern Med 2014; 14: 84
  • 9 Warrier PK, Nambiar VPK. Indian medicinal plants: a compendium of 500 species, Vol. 1. New Delhi: Orient Longman Pvt Ltd.; 1994: 104
  • 10 Samarghandian S, Hadjzadeh M, Afshari JT, Hosseini M. Antiproliferative activity and induction of apoptotic by ethanolic extract of Alpinia galanga rhizhome in human breast carcinoma cell line. BMC Complement Altern Med 2014; 14: 192
  • 11 Köse LP, Gülçin İ, Gören AC, Namiesnik J, Martinez-Ayala AL, Gorinstein S. LC–MS/MS analysis, antioxidant and anticholinergic properties of galanga (Alpinia officinarum Hance) rhizomes. Ind Crops Prod 2015; 74: 712-721
  • 12 Khattak S, Shah HU, Ahmad W, Ahmad M. Biological effects of indigenous medicinal plants Curcuma longa and Alpinia galanga . Fitoterapia 2005; 76: 254-257
  • 13 Bendjeddou D, Lalaoui K, Satta D. Immunostimulating activity of the hot water-soluble polysaccharide extracts of Anacyclus pyrethrum, Alpinia galanga and Citrullus colocynthis . J Ethnopharmacol 2003; 88: 155-160
  • 14 Akhtar MS, Khan MA, Malik MT. Hypoglycaemic activity of Alpinia galanga rhizome and its extracts in rabbits. Fitoterapia 2002; 73: 623-628
  • 15 Chan EW, Ng VP, Tan VV, Low YY. Antioxidant and antibacterial properties of Alpinia galanga, Curcuma longa, and Etlingera elatior (Zingiberaceae). Pharmacog J 2011; 3: 54-61
  • 16 Trakranrungsie N, Chatchawanchonteera A, Khunkitti W. Ethnoveterinary study for antidermatophytic activity of Piper betle, Alpinia galanga and Allium ascalonicum extracts in vitro . Res Vet Sci 2008; 84: 80-84
  • 17 Oonmetta-Aree J, Suzuki T, Gasaluck P, Eumkeb G. Antimicrobial properties and action of galangal (Alpinia galanga Linn.) on Staphylococcus aureus . LWT Food Sci Technol 2006; 39: 1214-1220
  • 18 Ling ZH, Lv-Yi CH, Liang JY. Two new phenylpropanoids isolated from the rhizomes of Alpinia galanga . Chinese J Nat Med 2012; 10: 370-373
  • 19 Xiao-Lu ZH, Ming-Hua YA, Jian-Guang LU, Huang XF, Ling-Yi KO. A new phenylpropanoid from Alpinia galanga . Chinese J Nat Med 2009; 7: 19-20
  • 20 Kubota K, Nakamura K, (Murayama) Kobayashi A, Amaike M. Acetoxy-1, 8-cineoles as aroma constituents of Alpinia galanga Willd. J Agric Food Chem 1998; 46: 5244-5247
  • 21 Barik BR, Kundu AB, Dey AK. Two phenolic constituents from Alpinia galanga rhizomes. Phytochemistry 1987; 26: 2126-2127
  • 22 De Pooter HL, Omar MN, Coolsaet BA, Schamp NM. The essential oil of greater galanga (Alpinia galanga) from Malaysia. Phytochemistry 1985; 24: 93-96
  • 23 Yang WQ, Gao Y, Li M, Miao DR, Wang F. New chalcones bearing a long-chain alkylphenol from the rhizomes of Alpinia galanga . J Asian Nat Prod Res 2015; 17: 783-787
  • 24 Roy SK, Pahwa S, Nandanwar H, Jachak SM. Phenylpropanoids of Alpinia galanga as efflux pump inhibitors in Mycobacterium smegmatis mc2 155. Fitoterapia 2012; 83: 1248-1255
  • 25 Singh JH, Alagarsamy V, Diwan PV, Kumar SS, Nisha JC, Reddy YN. Neuroprotective effect of Alpinia galanga (L.) fractions on Aβ (25–35) induced amnesia in mice. J Ethnopharmacol 2011; 138: 85-91
  • 26 Latha C, Shriram VD, Jahagirdar SS, Dhakephalkar PK, Rojatkar SR. Antiplasmid activity of 1′-acetoxychavicol acetate from Alpinia galanga against multi-drug resistant bacteria. J Ethnopharmacol 2009; 123: 522-525
  • 27 Phitak T, Choocheep K, Pothacharoen P, Pompimon W, Premanode B, Kongtawelert P. The effects of p-hydroxycinnamaldehyde from Alpinia galanga extracts on human chondrocytes. Phytochemistry 2009; 70: 237-243
  • 28 Yasuhara T, Manse Y, Morimoto T, Qilong W, Matsuda H, Yoshikawa M, Muraoka O. Acetoxybenzhydrols as highly active and stable analogues of 1′S-1′-acetoxychavicol, a potent antiallergic principal from Alpinia galanga . Bioorg Med Chem Lett 2009; 19: 2944-2946
  • 29 Matsuda H, Pongpiriyadacha Y, Morikawa T, Ochi M, Yoshikawa M. Gastroprotective effects of phenylpropanoids from the rhizomes of Alpinia galanga in rats: structural requirements and mode of action. Eur J Pharmacol 2003; 471: 59-67
  • 30 Tamura S, Shiomi A, Kimura T, Murakami N. Halogenated analogs of 1′-acetoxychavicol acetate, Rev-export inhibitor from Alpinia galanga, designed from mechanism of action. Bioorg Med Chem Lett 2010; 20: 2082-2085
  • 31 Tamura S, Shiomi A, Kaneko M, Ye Y, Yoshida M, Yoshikawa M, Kimura T, Kobayashi M, Murakami N. New Rev-export inhibitor from Alpinia galanga and structure-activity relationship. Bioorg Med Chem Lett 2009; 19: 2555-2557
  • 32 Hasima N, Aun LI, Azmi MN, Aziz AN, Thirthagiri E, Ibrahim H, Awang K. 1′S-1′-Acetoxyeugenol acetate: a new chemotherapeutic natural compound against MCF-7 human breast cancer cells. Phytomedicine 2010; 17: 935-939
  • 33 Matsuda H, Ando S, Morikawa T, Kataoka S, Yoshikawa M. Structure–activity relationships of 1′S-1′-acetoxychavicol acetate for inhibitory effect on NO production in lipopolysaccharide-activated mouse peritoneal macrophages. Bioorg Med Chem Lett 2005; 15: 1949-1953
  • 34 Zeng QH, Lu CL, Zhang XW, Jiang JG. Isolation and identification of ingredients inducing cancer cell death from the seeds of Alpinia galanga, a Chinese spice. Food Funct 2015; 6: 431-443
  • 35 Phanthong P, Lomarat P, Chomnawangb MT, Bunyapraphatsaraa N. Antibacterial activity of essential oils and their active components from Thai spices against foodborne pathogens. Sci Asia 2013; 39: 472-476
  • 36 Kubota K, Ueda Y, Yasuda M, Masuda A. Occurrence and antioxidative activity of 1′-acetoxychavicol acetate and its related compounds in the rhizomes of Alpinia galanga during cooking. In: Spanier AM, Shahidi F, Parliment TH, Mussinan C, Ho CT, Tratras Contis E, editors. Food flavors and chemistry: advances of the new millennium. Cambridge, UK: The Royal Society of Chemistry 2001; 274: 601-607
  • 37 Moffatt J, Hashimoto M, Kojima A, Kennedy DO, Murakami A, Koshimizu K, Ohigashi H, Matsui-Yuasa I. Apoptosis induced by 1′-acetoxychavicol acetate in Ehrlich ascites tumor cells is associated with modulation of polyamine metabolism and caspase-3 activation. Carcinogenesis 2000; 21: 2151-2157
  • 38 Itokawa H, Morita H, Sumitomo T, Totsuka N, Takeya K. Antitumour principles from Alpinia galanga . Planta Med 1987; 53: 32-33
  • 39 Kondo A, Ohigashi H, Murakami A, Suratwadee J, Koshimizu K. 1′-acetoxychavicol acetate as a potent inhibitor of tumor promoter-induced Epstein-Barr virus activation from Languas gaianga, a traditional Thai condiment. Biosci Biotechnol Biochem 1993; 57: 1344-1345
  • 40 Zheng Q, Hirose Y, Yoshimi N, Murakami A, Koshimizu K, Ohigashi H, Sakata K, Matsumoto Y, Sayama Y, Mori H. Further investigation of the modifying effect of various chemopreventive agents on apoptosis and cell proliferation in human colon cancer cells. J Cancer Res Clin Oncol 2002; 128: 539-546
  • 41 Murakami A, Toyota K, Ohura S, Koshimizu K, Ohigashi H. Structure-activity relationships of (1′S)-1′-acetoxychavicol acetate, a major constituent of a southeast Asian condiment plant Languas galanga, on the inhibition of tumor-promoter-induced Epstein-Barr virus activation. J Agric Food Chem 2000; 48: 1518-1523
  • 42 Nakamura Y, Murakami A, Ohto Y, Torikai K, Tanaka T, Ohigashi H. Suppression of tumor promoter-induced oxidative stress and inflammatory responses in mouse skin by a superoxide generation inhibitor 1′-acetoxychavicol acetate. Cancer Res 1998; 58: 4832-4839
  • 43 Noro T, Sekiya T, Katoh M, Oda Y, Miyase T, Kuroyanagi M, Ueno A, Fukushima S. Inhibitors of xanthine oxidase from Alpinia galanga . Chem Pharm Bull 1988; 36: 244-248
  • 44 Janssen AM, Scheffer JJ. Acetoxychavicol acetate, an antifungal component of Alpinia galangal . Planta Med 1985; 51: 507-511
  • 45 Bhattacharya AK, Chand HR, John J, Deshpande MV. Clerodane type diterpene as a novel antifungal agent from Polyalthia longifolia var. pendula . Eur J Med Chem 2015; 94: 1-7
  • 46 Bhattacharya AK, Rana KC. Antimycobacterial agent, (E)-phytol and lauric amide from the plant Lagascea mollis . Ind J Chem 2013; 52?B: 901-903
  • 47 Bhattacharya AK, Pathak AK, Sharma RP. Semi-synthesis of deoxyartemisinin. Mendeleev Commun 2007; 17: 27-28
  • 48 Bhattacharya AK, Pal M, Jain DC, Joshi BS, Roy R, Rychlewska U, Sharma RP. Stereoselective reduction of arteannuin B and its chemical transformations. Tetrahedron 2003; 59: 2871-2876
  • 49 Bhattacharya AK, Jain DC, Sharma RP, Roy R, McPhail AT. Boron trifluoride-acetic anhydride catalysed rearrangement of dihydroarteannuin B. Tetrahedron 1997; 53: 14975-14990
  • 50 Sarkar S, Sarkar D. Potential use of nitrate reductase as a biomarker for the identification of active and dormant inhibitors of Mycobacterium tuberculosis in a THP1 infection model. J Biomol Screening 2012; 17: 966-973
  • 51 Singh U, Akhtar S, Mishra A, Sarkar D. A novel screening method based on menadione mediated rapid reduction of tetrazolium salt for testing of anti-mycobacterial agents. J Microbiol Methods 2011; 84: 202-207
  • 52 Khan A, Sarkar S, Sarkar D. Bactericidal activity of 2-nitroimidazole against the active replicating stage of Mycobacterium bovis BCG and Mycobacterium tuberculosis with intracellular efficacy in THP-1 macrophages. Int J Antimicrob Agents 2008; 32: 40-45
  • 53 Ibrahem I, Córdova A. Direct catalytic intermolecular α‐allylic alkylation of aldehydes by combination of transition‐metal and organocatalysis. Angew Chem Int Ed 2006; 45: 1952-1956
  • 54 Rosebrugh LE, Herbert MB, Marx VM, Keitz BK, Grubbs RH. Highly active ruthenium metathesis catalysts exhibiting unprecedented activity and Z-selectivity. J Am Chem Soc 2013; 35: 1276-1279

Zoom Image
Fig. 1 Isolated compounds (16) from the acetone extract of the rhizomes of A. galanga.
Zoom Image
Fig. 2 Scheme of the preparation of derivatives of 2. Reagents and conditions: (i) Pd(OAc)2, PPh3, DMSO, cyclohexanone, pyrrolidine, rt, 3 h, 37 %; (ii) Grubbʼs Ist generation catalyst, CH2Cl2, rt, 16 h, 91 %.