Malyngamide 3 and cocosamides A and B from the marine cyanobacterium Lyngbya majuscula from Cocos Lagoon, Guam.
Journal: 2011/July - Journal of Natural Products
ISSN: 1520-6025
Abstract:
Malyngamide 3 (1) and cocosamides A (2) and B (3) were isolated from the lipophilic extract of a collection of Lyngbya majuscula from Cocos Lagoon, Guam. The planar structures of compounds 1-3 were determined by spectroscopic methods. The absolute configuration of 1 was determined by modified Mosher's method, NOESY data, and comparison with lyngbic acid (4). The absolute configurations of 2 and 3 were assigned by enantioselective HPLC analysis and comparison with the closely related compound pitipeptolide A (5). Compounds 1-3 showed weak cytotoxicity against MCF7 breast cancer and HT-29 colon cancer cells.
Relations:
Content
Citations
(5)
References
(6)
Chemicals
(3)
Organisms
(2)
Processes
(1)
Anatomy
(1)
Similar articles
Articles by the same authors
Discussion board
J Nat Prod 74(4): 871-876

Malyngamide 3 and Cocosamides A and B from the Marine Cyanobacterium <em>Lyngbya majuscula</em> from Cocos Lagoon, Guam

Experimental Section

General Experimental Procedures

The optical rotations were recorded on a Perkin Elmer model 343 polarimeter. UV spectrophotometric data were acquired on a Hitachi U-3010 spectrophotometer. IR spectroscopic data were obtained on a Bruker Vector 22 FT-IR spectrometer. NMR data were collected on a JEOL ECA-600 spectrometer operating at 600.17 MHz for H and 150.9 MHz for C. The edited-HSQC experiment was optimized for JCH = 140 Hz and the HMBC spectrum was optimized for JCH = 8 Hz. H NMR chemical shifts (referenced to residual CHCl3 observed at δ 7.25) were assigned using a combination of data from 2D DQF COSY and multiplicity-edited HSQC experiments. Similarly, C NMR chemical shifts (referenced to CDCl3 observed at δ 77.0) were assigned on the basis of multiplicity-edited HSQC experiments. The HRMS data were obtained using an Agilent 6210 LC-TOF mass spectrometer equipped with an APCI/ESI multimode ion source detector at the Mass Spectrometer Facility at the University of California, Riverside, California. Silica gel 60 (EMD Chemicals, Inc. 230–400 mesh) was used for column chromatography. All solvents used were of HPLC grade (Fisher Scientific).

Collection, Extraction and Isolation

The sample of cyanobacterial assemblage of Lyngbya majuscula for this study was collected in February 2001 from a patch reef near Cocos Island, Guam. This was a collection of ECO 27, first collected in March 1999. This chemotype of L. majuscula grew during winter months on Guam (January – March) and consistently produced malyngamides A and B, and majusculamides A and B. The samples were identified by one of us (VJP) based on morphological characteristics of the genus, and a voucher specimen (VP-ECO 27) is maintained at the Smithsonian Marine Station, Fort Pierce, FL. The freeze dried material (328 g) was extracted with EtOAc–MeOH (1:1). This lipophilic extract was partitioned between EtOAc and H2O and the aqueous portion subsequently partitioned between n-BuOH and H2O. Concentration of these extracts furnished 9.56 g (2.9%) of EtOAc-soluble fraction and 2.24 g (0.6%) of BuOH-soluble material. The EtOAc-soluble fraction (9.56 g) was chromatographed on a column of SiO2 (100 g) using a hexanes–EtOAc step gradient system followed by an EtOAc–MeOH step gradient to give thirteen sub-fractions. The combined sub-fractions 7 to 9 (2.0 g), eluting with hexanes–75% EtOAc was further chromatographed on a Si-column (100 g) using a hexanes–EtOAc step gradient system to give eight sub-fractions. Sub-fraction 6 (36 mg), eluting with hexanes–25% EtOAc was further purified by reversed-phase HPLC (semi-prep 250 × 10 mm, 5 μm, RP-18, flow 3.0 mL/min) using 15% H2O–MeOH to give 3 mg of impure cocosamide B, 20 mg of majusculamides A and B, 4.0 mg of malyngamide 3 (1, yield, 0.001% dry wt), and 1.4 mg of cocosamide A (2, yield, 0.0004% dry wt). The impure cocosamide B fraction was further separated by reversed-phase HPLC using 30% H2O–MeOH to give 2.0 mg of cocosamide B (3, yield 0.0006% dry wt).

Malyngamide 3 (1)

colorless, amorphous powder; [α]D −10.1 (c 0.36, MeOH); UV (MeOH) λmax (log ε) 205 (4.19), 274 (3.40) nm; IR (film) νmax 3320, 2932, 1725, 1636 cm; H and C NMR data, see Table 1, assignments were made by interpretation of 2D DQF COSY, edited-HSQC, HMBC and NOESY data; HRESI/TOFMS m/z 559.3146 [M + H] (calcd for C28H48ClN2O7, 559.3145).

Cocosamide A (2)

white solid; [α]D −77.7 (c 0.12, MeOH); UV (MeOH) λmax (log ε) 208 (4.36), 260 (3.26) nm; IR (film) νmax 3330, 2920, 1665, 1634, 1527, 1197 cm; H and C NMR data, see Table 2, assignments were made by interpretation of 2D DQF COSY, edited-HSQC, HMBC and NOESY data; HRESI/TOFMS m/z 744.4330 [M + H] (calcd for C42H58N5O7, 744.4331).

Cocosamide B (3)

white solid; [α]D −103 (c 0.18, MeOH); UV (MeOH) λmax (log ε) 208 (4.54), 260 (3.35) nm; IR (film) νmax 3416, 2950, 1665, 1634, 1541, 1032 cm; H and C NMR data, see Table 2, assignments were made by interpretation of 2D DQF COSY, edited-HSQC, HMBC and NOESY data; HRESI/TOFMS m/z 742.4186 [M + H] (calcd for C42H56N5O7, 772.4174).

Preparation of (R)-MTPA and (S)-MTPA Esters of 1

Compound 1 (1.0 mg) was dissolved in CHCl3 (50 μL) and added pyridine (50 μL) and a catalytic amount of 4-DMAP. The solution was treated with S(+)-MTPA chloride (1.0 μL) and stirred at room temperature for 12 h. The reaction was terminated with the addition of MeOH (200 μL) and the solvent was evaporated to give the (R)-MTPA ester of 1. Similarly, the (S)-MTPA ester of 1 was prepared with R(−)-MTPA chloride using the same procedure. Both esters were subjected to HPLC (semi-prep 250 × 10 mm, 5 μm, SiO2, flow 3.0 mL/min) using EtOAc– 3% MeOH to yield the pure (R)-MTPA ester of 1 (0.4 mg) and (S)-MTPA esters of 1 (0.3 mg).

R-MTPA ester of 1

H NMR δ (only key resonances are listed) 5.921 (1H, t, J = 5.4 Hz, NH), 5.292 (1H, m, H-9), 3.584 (3H, s, OMe-12), 3.536 (2H, s, H2-6), 2.624 (2H, d, J = 6.9 Hz, H2-10); ESIMS m/z 777.4 [M + H]; HRESI/TOFMS m/z 777.3404 [M + H] (calcd for C38H57ClF3N2O9, 777.3422).

S-MTPA ester of 1

H NMR δ (only key resonances are listed) 5.732 (1H, t, J = 5.4 Hz, NH), 5.292 (1H, m, H-9), 3.656 (3H, s, OMe-12), 3.535 (2H, s, H2-6), 2.652 (2H, d, J = 6.9 Hz, H2-10); ESIMS m/z 777.4 [M + H]; HRESI/TOFMS m/z 777.3443 [M + H] (calcd for C38H57ClF3N2O9, 777.3422).

Base Hydrolysis of Malyngamide 3

Compound 1 (1.9 mg) was dissolved in a 0.5 mL solution of 10% KOH in 80% aqueous EtOH and refluxed for 12 h. The hydrolysate was concentrated in vacuo and partitioned between H2O and CH2Cl2. The H2O layer was separated, acidified, and extracted with CH2Cl2 to yield lyngbic acid (5, 0.4 mg): colorless oil; [α]D −12 (c 0.04, CHCl3) [lit. −12.6 (c 0.8, MeOH)16]; H NMR (600 MHz, CDCl3) δ 5.48 (2H, m), 3.31 (3H, s, OMe), 3.15 (1H, quin, J = 5.5 Hz), 2.42 (2H, t, J = 7.5 Hz), 2.34 (2H, m), 2.18 (2H, m), 1.43 (2H, m), 1.27 (10H, m), 0.87 (3H, t, J = 7.0 Hz); HRESI/TOFMS m/z 257.2113 [M + H] (calcd for C15H29O3, 257.2114).

Acid Hydrolysis and Enantioselective HPLC analysis

Compounds 2 and 3 (0.1 mg each) were suspended in 6 N HCl (0.3 mL) and heated at 115 °C for 18 h in two sealed tubes. The hydrolysates were concentrated to dryness. The residues were reconstituted in 0.3 mL of H2O and analyzed by enantioselective HPLC, comparing the retention times with those of authentic standards [Phenomenex Chirex (d) Penicillamine, 4.6 × 250 mm, 5 μm]; solvent mixtures of 2.0 mM CuSO4– CH3CN (85:15 or 90:10); detection at 254 nm. Using 2.0 mM CuSO4 – CH3CN (90:10) with a flow rate of 0.8 mL/min, the retention times (tR min) for authentic standards were l-Pro (10.0) and d-Pro (19.6) and with a flow rate of 1.0 mL/min the retention times (tR min) for authentic standards were l-Val (17.0) and d-Val (22.9). Using 2.0 mM CuSO4–CH3CN (85:15) with a flow rate of 1.0 mL/min, the retention times (tR min) for authentic standards were N-Me-l-Phe (34.2) and N-Me-d-Phe (36.6). The retention times in min (and respective HPLC conditions) of the amino acids in the hydrolysates of 2 and 3 were 10.0 (90:10, 0.8 mL/min), 17.0 (90:10, 1.0 mL/min), and 34.2 (85:15, 1.0 ml/min), indicating the presence of l-Pro, l-Val and N-Me-l-Phe.

Cell Viability Assays

Cells were propagated and maintained in DMEM (Invitrogen) supplemented with 10% FBS (Hyclone) at 37 °C humidified air and 5% CO2. Cells were seeded in 96-well plates (MCF7 10,500 cells/well; HT-29 13,000 cells/well). After 24 h, cells were treated with various concentrations of the test compound, or solvent control (1% EtOH). After 48 h of incubation, cell viability was measured using MTT according to the manufacturer’s instructions (Promega). Paclitaxel was used as a positive control; IC50 values were 7 nM and 6 nM in HT-29 and MCF7 cell lines, respectively. Experiments were done in duplicate. IC50 values were determined using non-linear regression in GraphPad Prism.

General Experimental Procedures

The optical rotations were recorded on a Perkin Elmer model 343 polarimeter. UV spectrophotometric data were acquired on a Hitachi U-3010 spectrophotometer. IR spectroscopic data were obtained on a Bruker Vector 22 FT-IR spectrometer. NMR data were collected on a JEOL ECA-600 spectrometer operating at 600.17 MHz for H and 150.9 MHz for C. The edited-HSQC experiment was optimized for JCH = 140 Hz and the HMBC spectrum was optimized for JCH = 8 Hz. H NMR chemical shifts (referenced to residual CHCl3 observed at δ 7.25) were assigned using a combination of data from 2D DQF COSY and multiplicity-edited HSQC experiments. Similarly, C NMR chemical shifts (referenced to CDCl3 observed at δ 77.0) were assigned on the basis of multiplicity-edited HSQC experiments. The HRMS data were obtained using an Agilent 6210 LC-TOF mass spectrometer equipped with an APCI/ESI multimode ion source detector at the Mass Spectrometer Facility at the University of California, Riverside, California. Silica gel 60 (EMD Chemicals, Inc. 230–400 mesh) was used for column chromatography. All solvents used were of HPLC grade (Fisher Scientific).

Collection, Extraction and Isolation

The sample of cyanobacterial assemblage of Lyngbya majuscula for this study was collected in February 2001 from a patch reef near Cocos Island, Guam. This was a collection of ECO 27, first collected in March 1999. This chemotype of L. majuscula grew during winter months on Guam (January – March) and consistently produced malyngamides A and B, and majusculamides A and B. The samples were identified by one of us (VJP) based on morphological characteristics of the genus, and a voucher specimen (VP-ECO 27) is maintained at the Smithsonian Marine Station, Fort Pierce, FL. The freeze dried material (328 g) was extracted with EtOAc–MeOH (1:1). This lipophilic extract was partitioned between EtOAc and H2O and the aqueous portion subsequently partitioned between n-BuOH and H2O. Concentration of these extracts furnished 9.56 g (2.9%) of EtOAc-soluble fraction and 2.24 g (0.6%) of BuOH-soluble material. The EtOAc-soluble fraction (9.56 g) was chromatographed on a column of SiO2 (100 g) using a hexanes–EtOAc step gradient system followed by an EtOAc–MeOH step gradient to give thirteen sub-fractions. The combined sub-fractions 7 to 9 (2.0 g), eluting with hexanes–75% EtOAc was further chromatographed on a Si-column (100 g) using a hexanes–EtOAc step gradient system to give eight sub-fractions. Sub-fraction 6 (36 mg), eluting with hexanes–25% EtOAc was further purified by reversed-phase HPLC (semi-prep 250 × 10 mm, 5 μm, RP-18, flow 3.0 mL/min) using 15% H2O–MeOH to give 3 mg of impure cocosamide B, 20 mg of majusculamides A and B, 4.0 mg of malyngamide 3 (1, yield, 0.001% dry wt), and 1.4 mg of cocosamide A (2, yield, 0.0004% dry wt). The impure cocosamide B fraction was further separated by reversed-phase HPLC using 30% H2O–MeOH to give 2.0 mg of cocosamide B (3, yield 0.0006% dry wt).

Malyngamide 3 (1)

colorless, amorphous powder; [α]D −10.1 (c 0.36, MeOH); UV (MeOH) λmax (log ε) 205 (4.19), 274 (3.40) nm; IR (film) νmax 3320, 2932, 1725, 1636 cm; H and C NMR data, see Table 1, assignments were made by interpretation of 2D DQF COSY, edited-HSQC, HMBC and NOESY data; HRESI/TOFMS m/z 559.3146 [M + H] (calcd for C28H48ClN2O7, 559.3145).

Cocosamide A (2)

white solid; [α]D −77.7 (c 0.12, MeOH); UV (MeOH) λmax (log ε) 208 (4.36), 260 (3.26) nm; IR (film) νmax 3330, 2920, 1665, 1634, 1527, 1197 cm; H and C NMR data, see Table 2, assignments were made by interpretation of 2D DQF COSY, edited-HSQC, HMBC and NOESY data; HRESI/TOFMS m/z 744.4330 [M + H] (calcd for C42H58N5O7, 744.4331).

Cocosamide B (3)

white solid; [α]D −103 (c 0.18, MeOH); UV (MeOH) λmax (log ε) 208 (4.54), 260 (3.35) nm; IR (film) νmax 3416, 2950, 1665, 1634, 1541, 1032 cm; H and C NMR data, see Table 2, assignments were made by interpretation of 2D DQF COSY, edited-HSQC, HMBC and NOESY data; HRESI/TOFMS m/z 742.4186 [M + H] (calcd for C42H56N5O7, 772.4174).

Preparation of (R)-MTPA and (S)-MTPA Esters of 1

Compound 1 (1.0 mg) was dissolved in CHCl3 (50 μL) and added pyridine (50 μL) and a catalytic amount of 4-DMAP. The solution was treated with S(+)-MTPA chloride (1.0 μL) and stirred at room temperature for 12 h. The reaction was terminated with the addition of MeOH (200 μL) and the solvent was evaporated to give the (R)-MTPA ester of 1. Similarly, the (S)-MTPA ester of 1 was prepared with R(−)-MTPA chloride using the same procedure. Both esters were subjected to HPLC (semi-prep 250 × 10 mm, 5 μm, SiO2, flow 3.0 mL/min) using EtOAc– 3% MeOH to yield the pure (R)-MTPA ester of 1 (0.4 mg) and (S)-MTPA esters of 1 (0.3 mg).

R-MTPA ester of 1

H NMR δ (only key resonances are listed) 5.921 (1H, t, J = 5.4 Hz, NH), 5.292 (1H, m, H-9), 3.584 (3H, s, OMe-12), 3.536 (2H, s, H2-6), 2.624 (2H, d, J = 6.9 Hz, H2-10); ESIMS m/z 777.4 [M + H]; HRESI/TOFMS m/z 777.3404 [M + H] (calcd for C38H57ClF3N2O9, 777.3422).

S-MTPA ester of 1

H NMR δ (only key resonances are listed) 5.732 (1H, t, J = 5.4 Hz, NH), 5.292 (1H, m, H-9), 3.656 (3H, s, OMe-12), 3.535 (2H, s, H2-6), 2.652 (2H, d, J = 6.9 Hz, H2-10); ESIMS m/z 777.4 [M + H]; HRESI/TOFMS m/z 777.3443 [M + H] (calcd for C38H57ClF3N2O9, 777.3422).

Base Hydrolysis of Malyngamide 3

Compound 1 (1.9 mg) was dissolved in a 0.5 mL solution of 10% KOH in 80% aqueous EtOH and refluxed for 12 h. The hydrolysate was concentrated in vacuo and partitioned between H2O and CH2Cl2. The H2O layer was separated, acidified, and extracted with CH2Cl2 to yield lyngbic acid (5, 0.4 mg): colorless oil; [α]D −12 (c 0.04, CHCl3) [lit. −12.6 (c 0.8, MeOH)16]; H NMR (600 MHz, CDCl3) δ 5.48 (2H, m), 3.31 (3H, s, OMe), 3.15 (1H, quin, J = 5.5 Hz), 2.42 (2H, t, J = 7.5 Hz), 2.34 (2H, m), 2.18 (2H, m), 1.43 (2H, m), 1.27 (10H, m), 0.87 (3H, t, J = 7.0 Hz); HRESI/TOFMS m/z 257.2113 [M + H] (calcd for C15H29O3, 257.2114).

Acid Hydrolysis and Enantioselective HPLC analysis

Compounds 2 and 3 (0.1 mg each) were suspended in 6 N HCl (0.3 mL) and heated at 115 °C for 18 h in two sealed tubes. The hydrolysates were concentrated to dryness. The residues were reconstituted in 0.3 mL of H2O and analyzed by enantioselective HPLC, comparing the retention times with those of authentic standards [Phenomenex Chirex (d) Penicillamine, 4.6 × 250 mm, 5 μm]; solvent mixtures of 2.0 mM CuSO4– CH3CN (85:15 or 90:10); detection at 254 nm. Using 2.0 mM CuSO4 – CH3CN (90:10) with a flow rate of 0.8 mL/min, the retention times (tR min) for authentic standards were l-Pro (10.0) and d-Pro (19.6) and with a flow rate of 1.0 mL/min the retention times (tR min) for authentic standards were l-Val (17.0) and d-Val (22.9). Using 2.0 mM CuSO4–CH3CN (85:15) with a flow rate of 1.0 mL/min, the retention times (tR min) for authentic standards were N-Me-l-Phe (34.2) and N-Me-d-Phe (36.6). The retention times in min (and respective HPLC conditions) of the amino acids in the hydrolysates of 2 and 3 were 10.0 (90:10, 0.8 mL/min), 17.0 (90:10, 1.0 mL/min), and 34.2 (85:15, 1.0 ml/min), indicating the presence of l-Pro, l-Val and N-Me-l-Phe.

Cell Viability Assays

Cells were propagated and maintained in DMEM (Invitrogen) supplemented with 10% FBS (Hyclone) at 37 °C humidified air and 5% CO2. Cells were seeded in 96-well plates (MCF7 10,500 cells/well; HT-29 13,000 cells/well). After 24 h, cells were treated with various concentrations of the test compound, or solvent control (1% EtOH). After 48 h of incubation, cell viability was measured using MTT according to the manufacturer’s instructions (Promega). Paclitaxel was used as a positive control; IC50 values were 7 nM and 6 nM in HT-29 and MCF7 cell lines, respectively. Experiments were done in duplicate. IC50 values were determined using non-linear regression in GraphPad Prism.

Supplementary Material

1_si_001

1_si_001

Click here to view.(2.0M, pdf)

Acknowledgment

This research was supported by the NIH, NIGMS Grant P41GM806210. We thank E. Cruz-Rivera for collecting the sample. We also thank the Harbor Branch Oceanographic Institute at Florida Atlantic University spectroscopy facility for 600 MHz NMR spectrometer time and UV measurements and the Florida Atlantic University, Jupiter Campus, for the use of their polarimeter and infrared spectrometer. The high resolution mass spectrometric analysis was performed by the UCR mass spectrometer facility, Department of Chemistry, University of California at Riverside. This is contribution number 840 from the Smithsonian Marine Station at Fort Pierce.

Smithsonian Marine Station at Ft. Pierce, 701 Seaway Drive, Ft. Pierce, FL 34949, and Department of Medicinal Chemistry, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32610
To whom correspondence should be addressed. Tel: (772) 462-0982. Fax: (772) 461-8154. ude.is@luap
Smithsonian Marine Station
Department of Medicinal Chemistry, University of Florida

Abstract

Malyngamide 3 (1) and cocosamides A (2) and B (3) were isolated from the lipophilic extract of a collection of Lyngbya majuscula from Cocos Lagoon, Guam. The planar structures of compounds 13 were determined by spectroscopic methods. The absolute configuration of 1 was determined by modified Mosher’s method, NOESY data and comparison with lyngbic acid (4). The absolute configurations of 2 and 3 were assigned by enantioselective HPLC analysis and comparison with the closely related compound pitipeptolide A (5). Compounds 13 showed weak cytotoxicity against MCF7 breast cancer and HT-29 colon cancer cells.

Abstract

Marine cyanobacteria of the genus Lyngbya are a prolific source of chemically diverse bioactive secondary metabolites.1 Malyngamides are small amides first discovered in the late 1970s and early 1980s from L. majuscula by Richard E. Moore’s research group.26 There are now over 30 known examples of malyngamides and the majority are reported from cyanobacteria.7 Recently, William Gerwick’s group reported the newest addition malyngamide 2 isolated from L. sordida collected from Papua New Guinea.8 Malyngamides are characterized by a fatty acid side chain, which is most commonly 7S-methoxytetradec-4(E)-enoic acid (lyngbic acid). The other part of the malyngamides usually encloses a cyclic unit. In one notable example (malyngamide J) the cyclic ketone has a pendant 2,4-dimethoxyxylose.9 Malyngamides O and P are the only examples10 of acyclic molecules in this series. Malyngamide 3 (1) described here is the next example of an acyclic malyngamide.

Cyclic depsipeptides containing a unique 2,2-dimethyl-3-hydroxy-7-octynoic acid (Dhoya), 2,2-dimethyl-3-hydroxy-7-octenoic acid (Dhoea) or 2,2-dimethyl-3-hydroxyoctanoic acid (Dhoaa) were first reported by Scheuer’s group from a marine mollusk.1112 Subsequently, Richard Moore’s13 and William Gerwick’s14 groups have isolated several of these unique cyclic depsipeptides from L. majuscula. Here, we report the isolation, structure determination and biological activity determination of two new cyclic depsipeptides that possess these distinctive moieties (Dhoea/Dhoya), namely cocosamides A (2) and B (3), from L. majuscula collected from Cocos Lagoon, Guam. Interestingly, this is the first report of cyclic depsipeptides in this series with one ester linkage, while other related compounds reported thus far with these unique acids have two or more ester linkages.1114

An external file that holds a picture, illustration, etc.
Object name is nihms-275445-f0001.jpg

The sample of the marine cyanobacterium L. majuscula was collected from a patch reef near Cocos Island, Guam in February 2001. The freeze-dried material was extracted with a mixture of EtOAc–MeOH (1:1) to afford a lipophilic extract, which was subsequently partitioned between EtOAc and H2O. The EtOAc-soluble portion was repeatedly fractionated by SiO2 chromatography followed by reversed-phase C18 HPLC to give three new compounds malyngamide 3 (1), and cocosamides A (2) and B (3) in addition to the known compounds malyngamide A,4 malyngamide B,3 and an unresolved mixture of majusculamides A and B.15

Malyngamide 3 (1) was obtained as a colorless, amorphous powder. The molecular formula C28H47ClN2O7 was determined from HRESIMS data. Its infrared spectrum contained absorption due to amide proton at 3320 cm, ester carbonyl at 1725 cm, and an amide carbonyl at 1636 cm. The H and C NMR spectra (Table 1) showed signature signals for the presence of the characteristic 7-methoxy-tetradec-4(E)-enoic acid moiety suggesting compound 1 to be an analogue of the malyngamides.

Table 1

NMR Spectroscopic Data for Malyngamide 3 (1) in CDCl3 (H 600 MHz, C 150 MHz)

positionδC mult.δH (J in Hz)COSYaHMBCNOESYb
1a45.9, CH24.24, d (−15.1)2, 3, 4, 1′13
1b4.17, d (−15.1)32, 3, 4, 1′13
2131.4, C
3120.7, CH6.08, s1, 41, 2, 44b
4a46.8, CH23.20, d (17.1)31, 2, 3, 53, 6
4b3.15, d (17.1)31, 2, 3, 51b, 3, 6
5202.2, C
649.7, CH23.39, s5, 74, NH
7166.6, C
8a45.2, CH23.52, ddd (14.2, 7.0, 5.4)NH, 97, 9, 109, NH
8b3.21, ddd (14.2, 9.0, 5.4)NH, 97, 9, 109, NH
967.2, CH4.17, m8, 108, 10, 118a, 8b, 10
1038.9, CH22.50, d (6.9)98, 9, 118a, 8b, 9
11172.4, C
1251.9, CH33.68, s11
1335.5, CH32.93, s1, 1′1a, 1b, 2′
N–H7.26, t (5.4)87, 86, 8a, 8b
1′174.0, C
2′33.1, CH22.35, m3′1′, 3′, 4′13, 4′
3′28.1, CH22.28, m2′, 4′1′, 2′, 4′, 55′
4′130.8, CH5.43, dt (15.7, 3.5)3′, 5′2′, 6′
5′127.6, CH5.49, dt (15.7, 3.5)3′, 4′, 6′3′, 7′, 15′
6′36.4, CH22.17, m5, 74′, 5′, 7′, 8′4′, 7′
7′80.8, CH3.14, m6, 85′, 9′, 15′5′, 6′, 15′
8′33.4, CH21.41, m7, 96′, 7′, 9′, 10′
9′25.4, CH21.32, m
1.26, m
8, 108′, 10′
10′29.4, CH21.27, m
11′29.9, CH21.27, m
12′31.9, CH21.27, m
13′22.7, CH21.27, m
14′14.2, CH30.86, t (6.9)1312′, 13′
15′56.5, CH33.30, s7′5′, 7′
1H-H COSY
NOESY correlations are from proton(s) stated to the indicated proton(s).

Following the interpretation of DQF COSY and edited HSQC experiments, the H and C NMR signals were assignable to three partial structures C-1 to C-4, C-8 to C-10, C-2′ to C-14′ and an isolated C-6 methylene group. In addition, the H, C and edited HSQC spectra indicated the presence of signals for two O-Me groups (C-12, δH 3.68, δC 51.9 and C-15′, δH 3.30, δC 56.5), one N-Me (C-13, δH 2.93, δC 35.5) and four carbonyl groups (C-5, δC 202.2; C-7, δC 166.6; C-11, δC 172.4 and C-1′, δC 174.0). The chemical shift values for C-2 (δC 131.4, C) and C-3 (δH 6.08, s; δC 120.7, CH) indicated the presence of a chloromethylene moiety in the C-1 to C-4 partial structure as in other malyngamides4516 and accounted for the chlorine atom in the molecular formula. HMBC correlations (Table 1) from H-3 (δH 6.08) to C-1 (δC 45.9), C-2 (δC 131.4) and C-4 (δC 46.8) confirmed the position of the chloromethylene moiety. Similarly, HMBC correlations from H-4a and H-4b (δH 3.20, 3.15) to C-5 (δC 202.2) and H2-6 (δH 3.39) to the C-5 and C-7 (δC 166.6) carbonyl groups extended the carbon chain to the amide carbonyl group. The HMBC correlations N-H (δH 7.26) to C-7 and C-8 (δC 45.2), H-9 (δH 4.17) to C-8, C-10 (δC 38.9) and C-11 (δC 172.4), and H3-12 (δH 3.68) to the C-11 carbonyl group established the planar structure for the right hand end of the molecule. The HMBC correlations from H3-13 (δH 2.93) to the C-1′ carbonyl (δC 174.0) and to C-1 connected the fatty acid chain to the right hand end of the molecule via an amide linkage, resembling other malyngamides. An E configuration was assigned for the C-4′/C-5′ olefin on the basis of the coupling constant (15.7 Hz).17 The NOESY spectrum of 1 did not show any cross-peaks between H-3 and H2-1, nor between H-3 and H3-13. However, the presence of a strong cross-peak between H-3 (δH 6.08) and H-4b (δH 3.15) established a Z configuration for the chloromethylene moiety in 1 as in isomalyngamides A and B.17 In order to determine the configuration at C-7′, compound 1 was hydrolyzed under basic conditions to give lyngbic acid (4). The observed specific rotation of 4 ([α]D −12) was comparable to the reported value for 7(S)-methoxytetradec-4(E)-enoic acid ([α]D −12.6),16 and thus, established a 7(S) configuration at the C-7′ position in 1. The absolute configuration at C-9 of 1 was determined by the modified Mosher’s method.18 Compound 1 was converted to (S)- and (R)-MTPA esters. The δ (= δS − δR) values of (S)- and (R)-MTPA esters (−0.078 for Ha-8; −0.031 for Hb-8; +0.028 for H2-10; +0.072 for CH3-12) revealed the R configuration at C-9. These data confirmed the structure 1 for malyngamide 3.

Cocosamides A (2) and B (3) were obtained as white solids. The molecular weights of 2 and 3 differ by two mass units on the basis of HRESI/TOFMS analysis. The H and C NMR spectra were indicative of depsipeptides (Table 2).

Table 2

NMR Spectroscopic Data for Cocosamides A (2) and B (3) in CDCl3 (H 600 MHz, C 150 MHz)

Cocosamide A (2)
Cocosamide B (3)
unitpositionδC mult.δH (J in Hz)HMBCaNOESYbbδC mult.δH (J in Hz)
Dhoea/Dhoyad1176.5, C176.5, C
248.9, C46.3, C
377.8, CH5.19, dd (11.0, 2.1)1, 9, 414a, 4b, 1077.3, CH5.20, br. d (11.0)
4a27.9, CH21.54, m53, 1027.6, CH21.75, m
4b1.48, m53, 91.58, m
525.0, CH21.34, m73, 624.6, CH21.51, m
633.3, CH22.06, m5, 718.1, CH22.21, m
7138.0, CH5.76, ddt (17.0, 11.6, 6.9)66, 8a83.6, C
8a115.2, CH24.96, dd (11.6, 3.4)6769.2, CH1.96, t (2.7)
8b5.04, dd (17.0, 3.4)6
917.5, CH31.25, s1, 3, 104a, 4b, NH (Val)17.6, CH31.28, s
1023.5, CH31.17, s1, 3, 93, 4a, 923.5, CH31.20, s
Val11172.2, C172.3, C
1255.5, CH4.33, dd (7.7, 7.5)1, 11, 14, 1513, NH (Val), 2555.6, CH4.32, dd (7.7, 7.5)
1330.5, CH1.91, m1212, 14, 1530.6, CH1.93, m
1419.1, CH30.97, d (6.9)12, 13, 1512, 13, 1519.2, CH30.97, d (6.9)
1518.7, CH30.88, d (6.9)12, 13, 1412, 13, 1418.8, CH30.90, d (6.9)
NH5.82 d (7.5)1, 129, 12, 13, 145.83 d (7.6)
N-Me-Phe-116168.9, C169.0, C
1754.2, CH5.08, dd (12.4, 3.9)18a, 18b, 25, 2754.3, CH5.03, br.d (12.4)
18a37.7, CH23.18, dd (12.4, 12.3)16, 19, 20/2417, 20/24, 2537.8, CH23.18, dd (12.4, 12.3)
18b3.02, dd (12.4, 3.9)16, 19, 20/2417, 20/24, 253.02, dd (12.4, 3.9)
19137.6, C137.6, C
20/24129.8, CH7.41, d (7.6, 2.7)1817, 18, 21/23129.9, CH7.40, d (7.6, 2.7)
21/23128.5, CH7.24, m1920/24, 22128.6, CH7.24, m
22127.0, CH7.18, m20/2421/23127.0, CH7.18, m
2532.3, CH33.58, s11, 1712, 17, 18a32.3, CH33.58, s
Pro26171.5, C171.6, C
2756.0, CH3.08, dd (8.2, 1.8)28, 29, 3017, 28a, 3256.1, CH3.06, dd (8.2, 1.8)
28a29.8, CH20.46, m2627, 28b, 29b29.9, CH20.47, m
28b−0.18, m28a, 32−0.19, m
29a21.9, CH21.31, m28b, 29b, 30a22.0, CH21.31, m
29b1.21, m28a, 29a, 30b1.21, m
30a46.2, CH23.38, m1629a, 30b46.0, CH23.37, m
30b3.22, m1629b, 30a3.21, m
N-Me-Phe-231169.4, C169.5, C
3263.5, CH3.93, dd (9.7, 3.5)31, 33, 4027, 33, NH (Gly)63.6, CH3.94, dd (9.7, 3.5)
33a34.9, CH23.68, dd (12.0, 3.5)32, 34, 35/3932, 33b, 35/3935.0, CH23.68, dd (12.0, 3.5)
33b2.75, dd (12.0, 9.7)32, 34, 35/3932, 33a, 35/392.75, dd (12.0, 9.7)
34138.1, C138.2, C
35/39129.4, CH7.05, d (7.6)33, 3733, 36/38129.5, CH7.05, d (7.6)
36/38128.8, CH7.23, m3435/39, 37128.9, CH7.23, m
37127.0, CH7.22, m35, 3936/38127.1, CH7.22, m
4031.0, CH32.82, s31.0, CH32.82, s
Gly41168.3, C168.4, C
42a41.9, CH24.80, dd (16.8, 8.9)4142b, NH (Gly)41.9, CH24.78, dd (16.8, 8.9)
42b3.66, dd (16.8, 1.0)4142a, NH (Gly)3.68, dd (16.8, 1.0)
NH8.91, dd (8.9, 1.0)3132, 40, 42b8.91, dd (8.3, 1.0)
HMBC correlations, optimized for JCH = 8 Hz, are from proton(s) stated to the indicated carbon.
NOESY correlations are from proton(s) stated to the indicated proton(s).
Dhoea moiety in cocosamide A.
Dhoya moiety in cocosamide B.

Following the interpretation of DQF COSY, edited HSQC and HMBC experiments, the H and C NMR signals of 2 and 3 were assignable to six partial structures, which accounted for all atoms in both molecules. These partial structures consisted of the amino acids valine, proline, glycine, two N-Me-phenylalanines, besides 2,2-dimethyl-3-hydroxy-7-octenoic acid (Dhoea) in 2 and 2,2-dimethyl-3-hydroxy-7-octynoic acid (Dhoya) in 3. The H NMR spectra showed the presence of three olefinic protons (δH 5.76, H-7; δH 4.96, 5.04, H-8a, b) in the spectrum of 2, while no olefinic protons were seen in the spectrum of 3, which instead revealed a characteristic acetylenic proton (δH 1.96, t, J = 2.7 Hz, H-8). The C spectrum of 2 indicated olefinic signals (δC 138.0, CH, C-7; δC 115.2, CH2, C-8), while the C spectrum of 3 indicated acetylenic signals (δC 83.6, C, C-7; δC 69.2, CH, C-8). These data together with other data presented in Table 2 confirmed the presence of a 2,2-dimethyl-3-hydroxy-7-octenoic acid in 2 and 2,2-dimethyl-3-hydroxy-7-octynoic acid in 3, respectively. The residue sequences for 2 and 3 were determined from HMBC data which showed linkages: Val-NH to C-1, Me-25 to C-11, CH2-30 to C-16, Me-40 to C-26, Gly-NH to C-31 and H-3 to C-41, and these connections were confirmed by NOESY correlations. These data established the residue sequences as 1,6-anhydro[Dhoea-Val-N-Me-Phe(1)-Pro-N-Me-Phe(2)-Gly] for 2 and 1,6-anhydro[Dhoya-Val-N-Me-Phe(1)-Pro-N-Me-Phe(2)-Gly] for 3. The absolute configurations of the amino acids were determined by enantioselective HPLC analysis of the acid hydrolysates of 2 and 3. The analysis revealed l-configurations for valine, proline and both N-Me-phenylalanines in compounds 2 and 3. Because we have isolated only small quantities of 2 and 3, the configuration at C-3 of the hydroxy acids (Dhoea and Dhoya) was investigated by comparison of NOE data with pitipeptolide A (5).13 Pitipeptolide A (5) is a cyclic depsipeptide that has a (S)-Dhoya moiety, which is connected to l-valine and glycine forming the amide and ester linkages similar to compounds 2 and 3. The NOE data for 5 were not previously reported,13 therefore, we used 5 that we isolated from another Lyngbya sample for NOE comparison studies. The H NMR spectrum, HRMS and specific rotation data for this sample matched that reported in the literature.1319 The NOESY spectrum of 5 showed a strong correlation between H-3 (δH 4.94) and H3-10 (δH 1.15) and another correlation between H-4a (δH 1.80) and H3-10. Similarly, strong correlations were observed between H-4b (δH 1.58) and H3-9, δH 1.29), and methyl (H3-9 and l-Val-NH (δH 6.08). There was no correlation seen between H-3 and H3-9. These data clearly indicated that in 5 the methine (H-3) is closer to methyl H3-10 and away from methyl H3-9. Cocosamides A (2) and B (3) showed the same patterns of NOE correlations for H-3, H3-9 and H3-10. These data suggested a 3S configuration at C-3 in compounds 2 and 3.

Compounds 13 were tested for antiproliferative activity against MCF7 breast cancer and HT-29 colon cancer cells and found to be weakly active. Malyngamide 3 (1) showed cytotoxic activity against MCF7 and HT-29 cells with IC50 values of 29 and 48 μM, respectively. This is an about 10-fold weaker activity than reported for the closely related analogue malyngamide O.10 Cocosamides A (2) and B (3) showed cytotoxic activity against HT-29 cells with IC50 values of 24 μM and 11 μM, respectively. MCF7 cells were slightly less susceptible to both compounds with IC50 values of 30 μM for 2 and 39 μM for 3. The closely related pitipeptolides A and B exert similar activity against cancer cells.13

Footnotes

Supporting Information Available:H, C, and 2D NOESY NMR spectra in CDCl3 for malyngamide 3 (1) and cocosamide A (2). H, C, COSY, HMBC and 2D NOESY NMR spectra in CDCl3 for cocosamide B (3). H and 2D NOESY NMR spectra in CDCl3 for pitipeptolide A (5). This material is available free of charge via the Internet at http://pubs.acs.org.

Footnotes

References and Notes

References and Notes

References

  • 1. Tidgewell K, Clark BT, Gerwick WH In: Comprehensive Natural Products Chemistry. 2nd ed. Moore B, Crews P, editors. Elsevier Limited; Oxford: 2010. in press. [PubMed][Google Scholar]
  • 2. Ainslie RD, Barchi JJ, Kuniyoshi M, Moore RE, Mynderse JS. J. Org. Chem. 1985;50:2859–2862.[PubMed]
  • 3. Cardellina JH, Dalietos D, Marner F-J, Mynderse JS, Moore RE. Phytochem. 1978;17:2091–2095.[PubMed]
  • 4. Cardellina JH, Marner F-J, Moore RE. J. Am. Chem. Soc. 1979;101:240–242.[PubMed]
  • 5. Moore RE In: Marine Natural Products: Chemical and Biological Perspectives. Scheuer PJ, editor. IV. Academic Press; New York: 1981. pp. 1–52. [PubMed][Google Scholar]
  • 6. Mynderse JS, Moore RE. J. Org. Chem. 1978;43:4359–4363.[PubMed]
  • 7. Blunt JW, Copp BR, Munro MHG, Northcote PT, Prinsep MR. Nat. Prod. Rep. 2010;27:165–237. and references therein.
    [PubMed]
  • 8. Malloy KL, Villa FA, Engene N, Matainaho T, Gerwick L, Gerwick HW. J. Nat. Prod. 2010 December 14; ASAP article. [PubMed]
  • 9. Wu M, Milligan KE, Gerwick WH. Tetrahedron. 1997;53:15983–15990.[PubMed]
  • 10. Gallimore WA, Scheuer PJ. J. Nat. Prod. 2000;63:1422–1424.[PubMed]
  • 11. Reese MT, Gulavita NK, Nakao Y, Hamann MT, Yoshida WY, Covel SJ, Scheuer PJ. J. Am. Chem. Soc. 1996;118:11081–11084.[PubMed]
  • 12. Nakao Y, Yoshida WY, Szabo CM, Baker BJ, Scheuer PJ. J. Org. Chem. 1998;63:3272–3280.[PubMed]
  • 13. Luesch H, Pangilinan R, Yoshida WY, Moore RE, Paul VJ. J. Nat. Prod. 2001;64:304–307.[PubMed]
  • 14. Sitachitta N, Williamson RT, Gerwick WH. J. Nat. Prod. 2000;63:197–200.[PubMed]
  • 15. Marner F-J, Moore RE, Hirotsu K, Clardy J. J. Org. Chem. 1977;42:2815–2819.[PubMed]
  • 16. Kwan JC, Teplitski M, Gunasekera SP, Paul VJ, Luesch H. J. Nat. Prod. 2010;73:463–466.
  • 17. Kan Y, Sakamoto B, Fujita T, Nagai H. J. Nat. Prod. 2000;63:1599–1602.[PubMed]
  • 18. Ohtani I, Kusumi T, Kashman Y, Kakisawa H. J. Am Chem. Soc. 1991;113:4092–4096.[PubMed]
  • 19. Pitipeptolide A: HRESI/TOFMS m/z 808.4875 [M + H] (calcd for C44H66N5O9, 808.4855); [α]D –110 (c 1.0, MeOH).
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.