Next Article in Journal
Antibacterial Activity of Sulfated Galactans from Eucheuma serra and Gracilari verrucosa against Diarrheagenic Escherichia coli via the Disruption of the Cell Membrane Structure
Previous Article in Journal
Preventive Effects of Three Polysaccharides on the Oxidative Stress Induced by Acrylamide in a Saccharomyces cerevisiae Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Unique Polyhalogenated Peptides from the Marine Sponge Ircinia sp.

1
Natural Products Department, PharmaMar S.A., Pol. Ind. La Mina Norte, Avda. de los Reyes 1, 28770 Colmenar Viejo (Madrid), Spain
2
Research Center for Biotechnology, Indonesian Institute of Sciences, Jl. Raya Jakarta-Bogor No. Km46, Cibinong, Bogor, Jawa Barat 16911, Indonesia
3
Research Center for Oceanography, Indonesian Institute of Sciences, Jl. Pasir Putih I, Ancol Timur, Jakarta 14430, Indonesia
*
Author to whom correspondence should be addressed.
Mar. Drugs 2020, 18(8), 396; https://doi.org/10.3390/md18080396
Submission received: 7 July 2020 / Revised: 21 July 2020 / Accepted: 27 July 2020 / Published: 28 July 2020

Abstract

:
Two new bromopyrrole peptides, haloirciniamide A (1) and seribunamide A (2), have been isolated from an Indonesian marine sponge of the genus Ircinia collected in the Thousand Islands (Indonesia). The planar structure of both compounds was assigned on the basis of extensive 1D and 2D NMR spectroscopy and mass spectrometry. The absolute configuration of the amino acid residues in 1 and 2 was determined by the application of Marfey’s method. Compound 1 is the first dibromopyrrole cyclopeptide having a chlorohistidine ring, while compound 2 is a rare peptide possessing a tribromopyrrole ring. Both compounds failed to show significant cytotoxicity against four human tumor cell lines, and neither compound was able to inhibit the enzyme topoisomerase I or impair the interaction between programmed cell death protein PD1 and its ligand, PDL1.

1. Introduction

Indonesia is located at the center of a biodiversity hotspot, and around 750 structures from Indonesian waters have been published in the last 50 years [1]. The structural diversity and bioactive properties of the compounds isolated from this region encouraged us to continue to investigate this area, which still remains largely unexplored. Thus, one of the recent PharmaMar expeditions was carried out in the Thousand Islands, in collaboration with Indonesian Institute of Sciences (LIPI). The Thousand Islands archipelago is located about 25 miles from the coast to the northeast of Jakarta; the collection site is an area full of gentle rock and coral slopes, and it is potentially a highly productive area, both in terms of the quantity and the nature of the biodiversity. In this paper, we describe the isolation of two new peptides isolated from an Ircinia specimen from this area.
Of all the marine organisms investigated, sponges (Porifera) are the most primitive multicellular animals with ample time to evolve into more complex living organisms. In fact, marine sponges are recognized as the richest sources of MNP, contributing to nearly 30% of all marine natural products discovered so far [2]. Previous reports revealed that Marine sponges of the genus Ircinia are known as a rich source of varied bioactive natural products, including fatty acids [3], steroids [4,5], terpenes [6,7], macrolides [8,9], and peptides [10], many of which have biological activities. This structural diversity could be due to the fact that sponges harbor diverse microorganisms and in numerous cases, bacteria isolated from sponges or symbiotic bacteria are the true producers of the compounds found in their extracts [11]. Specifically, an intriguing group of Ircinia-derived peptides are assumed to be of microbial origin due to the presence of both d-amino acids and unusual amino acids, as illustrated by the cyclic hexapeptide waiakeamide from Ircinia dendroides [12] and the cyclotheonamides E4 and E5, which are cyclic pentapeptides also from Ircinia species [13].
Undeniably, among the compounds isolated from marine sources, linear and cyclic peptides are recognized as an important class with great structural diversity and a wide range of bioactivities, and these include the antimalarial carmabin A [14], the antiproliferative jaspamides [15], and the cytotoxic patellamides [16]. Furthermore, two marine peptide-derived products have reached the market: ziconotide [17] for analgesic use and a synthetic derivative of dolastatin 10 [18] linked to an antibody for the treatment of Hodgkin’s lymphoma. PharmaMar has also developed a marine natural peptide Aplidin, which was originally found in the Ascidian Aplidium albicans and has recently been approved for commercialization in Australia for the treatment of multiple myeloma. Recent studies suggest that Aplidin may also have antiviral properties, and a clinical trial to treat patients with COVID-19 has been initiated.
In the course of our screening program to isolate novel compounds with antitumor properties from marine sources, the organic extract of an Ircinia specimen collected off the coast of the Thousand Islands showed hints of activity, and although the fractions did not confirm cytotoxicity, the chromatographic profiles along with the mass spectra showed peaks that were interesting enough for us to purify them. Thus, we have isolated two unique peptides haloirciniamide A (1) and seribunamide A (2). It is worth mentioning that the number of known peptides with a halogenated pyrrole ring is limited, with only cyclocinamides and corticiamide A [19] as well as gunungamide A [20] having been described as possessing chlorinated pyrrole rings. Although there are dozens of dibromopyrrolecarboxamide derivatives from porifera such as nagelamide [21] and carteramine [22], mainly from Agela and Stylissa species, compound 1 is the first example of a peptide containing a halogenated pyrrole ring in its structure. Indeed, haloirciniamide A represents a structurally unique cyclopeptide, since it also has an unprecedented chlorohistidine moiety. Furthermore, there are only two examples of tribromopyrrole rings derived from natural sources: 2,3,4-tribromopyrrole itself, which was isolated from the marine Poychaete Polyphysia crassa [23], and tribromopyrrol-2-methylphenol isolated from a coralline algal-associated Pseudoalteromone [24], with compound 2 being the first of its class.
Details of the isolation and structural elucidation of the new halogenated peptides 1 and 2 are provided. The results of cytotoxicity and other related antitumor programmed cell death protein (PD1) and TOPO-I screenings are also described.

2. Results and Discussion

Isolation and Structure Elucidation

The sponge Ircinia sp. was collected by hand while diving in the Thousand Islands (Indonesia). The specimen was repeatedly extracted using CH2Cl2:MeOH (1:1 v/v). The combined concentrated extracts, after vacuum liquid chromatography (VLC) and semipreparative reverse-phase HPLC separations, led to the isolation of the two pure compounds shown in Figure 1.
Compound 1 was isolated as an amorphous white solid. The isotopic distribution observed in the (+)-LRESIMS mass spectrum with four protonated ion [M+Na]+ peaks at m/z 830, 832, 834, and 836 in the ratio 3:6:3:1 respectively, showed the presence of two bromine atoms and a chlorine atom in the molecule. The presence of these halogens in the structure was confirmed by (+)-HRESI-TOFMS analysis, with the ion peak observed at m/z 830.0400 [M+Na]+ corresponding to the molecular formula C25H3279Br235ClN11O8Na (calcd. 830.0383). Interpretation of the mono NMR data (1H, 13C and 1D-TOCSY) compiled in Table 1 and two-dimensional NMR spectra (gHSQC, gCOSY, gHMBC, and 2D-TOCSY) in CD3OD led to identification of 6 spin systems. Taking into consideration the seven carbonyl carbon resonances (δC 161.4–175.8) and the number of α-amino acid proton signals (δH 4.06–4.84), the peptide nature of compound 1 was expected. This hypothesis was confirmed by the NH signals observed in the 1H NMR spectrum in CD3OH (δH 7.13–12.68) and DMSO-d6H 7.37–12.03), with the latter solvent being chosen for full structural elucidation. The COSY correlations observed between methines at δH 4.23/δC 50.5 and δH 4.49/δC 51.5 with the diastereotopic methylenes at δH 2.87, 3.13/δC 49.2 and δH 3.22, 4.04/δC 40.5 respectively, indicated the presence of two 2,3-diaminopropionic acid units (Figure 2). Both amino acids were directly connected based on the HMBC correlation of the NH signals at δH 9.11 for Dap1 and δH 7.72 for Dap2 with the same carbonyl carbon at δC 169.5. The next amino acid present in the peptide core was an isoserine with a methylene group at δH 2.75, 3.47/δC 42.8, a methine carbon at δH 4.13/δC 67.8, and an NH signal at δH 8.23. Isoserine was linked to the Dap2 by the correlation between NH at δH 8.23 and the carbonyl carbon of Dap2 at δC 170.6. In addition, an HMBC correlation of the methine carbon at δH 4.13 and the NH signal at δH 7.13, which belongs to a unit of isoasparagine (δH 4.58/δC 48.4, δH 2.80, 3.09/δC 35.0), with the carbonyl group at δC 170.6 allowed the sequence of amino acids to be continued. Finally, the peptide ring was closed by an NMe-histidine (δH 3.99/δC 65.6, δH 3.05/δC 25.0), whose N-methyl group showed an HMBC correlation with carbons belonging to the carbonyl group of iAsn at δC 173.5 and its own methine at δC 65.6, with an additional HMBC correlation between the α-aminoacid proton signal of NMe-histidine and the NH signal at δH 9.11 of Dap1 with the carbonyl carbon at δC 169.0 (Figure 2). NMe-histidine ring shifts at δC 110.5, δC 128.2, and δH 6.76/δC 109.5 revealed that the non-protonated carbon at δC 128.2 bore one of the three halogen atoms.
To complete the structure elucidation, the two remaining doublets with a small coupling constant value of 2.7 Hz were assigned to an sp2 methine at δH 6.30/δC 110.4 and a significant downfield NH signal at δH 12.68. An HMBC correlation of these two protons with three non-protonated sp2 carbons (δC 96.9, 118.1, and 123.2), demonstrated the existence of a trisubstituted pyrrol moiety, with two of these three positions bearing halogens. The placement of this heterocycle was established by the HMBC correlation of the methylene group of Dap2 with a carbonyl group at δC 158.6. To confirm the direct connection between the pyrrol moiety and this carbonyl group, a new gHMBC experiment with J = 3 Hz was conducted. The position of the sp2 methine in the pyrrol unit and the bond to the ring with the cyclopeptide was settled by the HMBC correlation between the methine proton at δH 6.30 and the carbonyl group at δC 158.6. Although the chlorine and two bromine atoms were undoubtedly located on the three free positions of the heterocycle ring and the carbon shifts (δC 96.9 and 123.2) suggested that both bromine atoms were on the pyrrol moiety, this evidence was insufficient to fully confirm this proposal. Fortunately, this could be resolved by detailed study of the peptide structure by (+)-HRESI-TOFMS and QTOFMS (Figure 3), which showed significant cluster ions at m/z 158.0487/160.0460 in a 3:1 ratio corresponding to the iAsn moiety. These m/z values, the mass error observed, and the isotopic distribution clearly confirmed the presence of a chlorine atom on the NMeHis amino acid (Figure S30).
The absolute stereochemistry of compound 1 was established on the basis of Marfey’s analysis with the 1-fluoro-2,4-di-nitrophenyl-5-l-alanine amide (l-FDAA) [25]. Compound 1 was hydrolyzed in strong acid conditions and derivatization of the free amino acids with l-FDAA allowed an exhaustive analysis by HPLC-MS. A comparison of the retention times of the derivatized amino acids present in 1 and the suitably derivatized pure amino acid standards unambiguously demonstrated the absolute configuration as l-Dap, l-iSer, and d-Asn. The absolute configuration of NMeClHis could not be determined due to the absence of the standard amino acid.
Compound 2 was isolated as an amorphous white solid. Its (+)-LRESI showed an m/z = 825 [M + H]+ with a characteristic cluster corresponding to the presence of three bromine atoms. The molecular formula C29H4379Br3N6O7 was established by (+)-HRESI-TOFMS analysis of the [M + H]+ at m/z 825.0806 (calcd. 825. 0816). The peptide nature of 2 was evident from its 1H and 13C NMR spectra (Table 2). 1H NMR in DMSO showed the characteristic α-proton resonances of four α-amino acids in the range δH 5.35 to 4.10 ppm, five interchangeable protons at δH 12.53, 8.56, 8.08, 7.19, and 6.77 ppm, and two NMe signals at δH 3.61 and 3.04 ppm. 13C NMR data displayed six carbonyl signals between δC 173.5 and 159.1 ppm, four adjacent methine carbons in the range δC 59.1–51.5 ppm, and two NMe signals at δC 35.7 and 30.5 ppm. Extensive 2D NMR analysis, including COSY, TOCSY, HSQC, and HMBC was used to determine the identity of the four amino acids and to assign the NMR signals. As a result of these studies, the amino acids were found to be one Ile, one NMe-Leu, one Pro, and one Asn unit. A long-range correlation between protons at δH 7.19/6.77 and 2.15/2.11 ppm with the carbonyl group at δC 173.5 ppm and the observation of ROESY cross-peaks between protons at δH 7.19/6.77 ppm and the CH2 of position 4 at δH 2.15/2.11 ppm established the presence of a Gln. A N-methyl-2,3,4-bromopyrrol unit was inferred by the presence of four aromatic non-protonated carbons at δC 128.5, 107.9, 100.7 and 93.4 ppm with chemical shifts similar to those described for bromopseudoceratines [23].
The sequencing for compound 2 was carried out using a combination of HMBC and ROESY data. Long-range correlations from α-protons, NH, and NMe to carbonyl carbons of adjacent amino acids plus ROESY correlations between α-protons, NH, and NMe protons of adjacent amino acids (see Figure 2) allowed us to establish the sequence as Br3Py-Ile-NMeLeu-Pro-Gln.
The absolute configurations of the amino acids were determined by comparing the hydrolysis products of 2 (6 N HCl, 110 °C, 18 h) after derivatization with Marfey’s reagent (N-(3-fluoro-4,6-dinitrophenyl)-l-alaninamide, l-FDAA), with appropriate amino acid standards using HPLC-MS chromatography. As a result, all the amino acids were determined to be L.

3. Materials and Methods

3.1. General Experimental Procedures

Optical rotations were determined using a Jasco P-1020 polarimeter. UV spectra were performed using an Agilent 8453 UV−vis spectrometer. IR spectra were obtained with a Perkin-Elmer Spectrum 100 FT-IR spectrometer with ATR sampling. NMR spectra were recorded on a Varian “Unity 500” spectrometer at 500/125 MHz (1H/13C). Chemical shifts were reported in ppm using residual CD3OH (δ 3.31 ppm for 1H and 49.0 ppm for 13C) and DMSO-d6 (δ 2.50 ppm for 1H and 39.5 ppm for 13C) as an internal reference. HRESI-TOFMS was performed on an Agilent 6230 TOF LC/MS chromatograph spectrometer. (+)-ESIMS were recorded using an Agilent 1100 Series LC/MSD spectrometer. HRESI-TOFMS was performed on an Agilent 6230 TOF LC/MS chromatograph spectrometer. ESI(+) and MSe were performed on an Waters UHPLC-QTOF Acquity I-Class + Xevo G2-XS.

3.2. Biological Material

The sponge Ircinia sp. (158 g) was collected by hand using a diving rebreather system in the Thousand Islands (Indonesia). The sponge was immediately frozen and kept under these conditions until extraction. The specimen was identified by María Jesús Uriz at CEAB, Blanes, Spain. A voucher specimen (ORMA155272) was deposited at PharmaMar facilities (Madrid, Spain).

3.3. Extraction and Isolation

The sponge Ircinia sp. (158 g) was triturated and exhaustively extracted with MeOH:DCM (1:1, 3 × 500 mL). The combined extracts were concentrated to yield a crude mass of 7.9 g. The crude product was subjected to VLC on a Lichroprep RP-18 with a stepped gradient from H2O to MeOH to CH2Cl2. The fractions eluting with H2O:MeOH (3:1, 606 mg) and H2O:MeOH (1:1, 89.6 mg) were subjected to semipreparative HPLC (Symmetry Prep C18 5 μm, 10 × 150 mm; 3 min isocratic H2O + 0.04% TFA: CH3CN + 0.04% TFA 95:5 and then gradient from 5% to 68% CH3CN + 0.04% TFA in 25 min, flow 3 mL/min, UV detection) to obtain 4.3 mg of compound 1. The fraction eluting with H2O:MeOH (1:3, 43.9 mg) was subjected to semipreparative HPLC (Symmetry Prep C18 5 μm, 10 × 150 mm; 3 min. isocratic H2O + 0.04% TFA: CH3CN + 0.04% TFA 90:10 and then gradient from 10% to 75% CH3CN + 0.04% TFA in 25 min, flow 3 mL/min, UV detection) to obtain 3.0 mg of compound 2.
Haloirciniamide A (1): amorphous white solid; [α]25D –62.7º (c 0.1, MeOH); IR υmax 3314, 2920, 2850, 1644, 1523, 1416, 1311, 1239, 1199, 1041 cm−1; UV (MeOH) λmax 198, 268 nm. 1H NMR (500 MHz) and 13C NMR (125 MHz) see Table 1; (+)-HREI-TOFMS m/z 830.0400 [M+Na]+ (calcd for C25H3279Br2N11O8Na m/z 830.0383).
Seribunamide A (2): amorphous white solid; [α]25D –38.4º (c 0.2, MeOH); IR υmax 3352, 2932, 2850, 1658, 1515, 1320, 1236, and 1035 cm−1; UV (MeOH) λmax 197, 266 nm. 1H NMR (500 MHz) and 13C NMR (125 MHz) see Table 2; (+)-HREI-TOFMS m/z 825.0806 [M + H]+ (calcd for C29H4479Br3N6O7 m/z 825.0816).

3.4. Absolute Configuration

Absolute Configuration of 1. First, 0.5 mg of haloirciniamide A was hydrolyzed in 0.5 mL of 6 N HCl at 110 °C for 15 h. The excess aqueous HCl was removed under a N2 stream, and a solution of 700 µg of l-FDAA (N-(3-fluoro-4,6-dinitrophenyl)-l-alanine-amide) in acetone (160 µL), H2O (100 µL), and NaHCO3 1N (50 µL) was added to the dry hydrolysate. The resulting mixture was heated at 40 ºC for 1 h, before being cooled to 23 ºC, quenched by addition of 2N HCl (20 µL), dried, and dissolved in H2O (800 μL). The resultant aqueous solution was subjected to reversed-phase LC/MS (column: Waters Symmetry 4.6 × 150 mm, 3.5 µm, flow rate 0.8 mL/min) in three different gradients.
Gradient 1 for iSer (mobile phase CH3CN + 0.04% formic acid /H2O + 0.04% formic acid, using a linear gradient from 5% to 20% CH3CN in 10 min and then from 20% to 35% CH3CN in 25 min): the retention time was 23.9 min for l-iSer.
Gradient 2 for Asp (mobile phase CH3CN + 0.04% formic acid/H2O + 0.04% formic acid, using a linear gradient from 5% to 30% CH3CN in 10 min and then from 30% to 50% CH3CN in 30 min): the retention time was 17.0 min for d-Asp.
Gradient 3 for Dap: mobile phase CH3CN + 0.04% formic acid/H2O + 0.04% formic acid, using a linear gradient from 5% to 10% CH3CN in 5 min and then from 10% to 35% CH3CN in 25 min): the retention times was 15.5 min for l-Dap.
Retention times for the derivatized amino acids standards were as follows: gradient 1 (23.2 min for d-iSer and 23.8 min for the l-iSer); gradient 2 (16.2 min for l-Asp and 17.0 min for d-Asp), and gradient 3 (15.5 min for l-Dap and 16.6 min for the d-Dap).
Absolute Configuration of 2. First, 0.3 mg of seribunamide A was hydrolyzed in 0.4 mL of 6 N HCl, 110 °C for 15 h. The excess aqueous HCl was removed under a N2 stream, and a solution of 400 µg of l-FDAA (N-(3-fluoro-4,6-dinitrophenyl)-l-alanine-amide) in acetone (160 µL), H2O (100 µL), and NaHCO3 1N (50 µL) was added. The vial was heated at 40 ºC for 1 h, and the contents were neutralized with 2N HCl (20 µL) after cooling to room temperature. The resulting solution was dried in vacuum and reconstituted in H2O (600 μL) before being analyzed by HPLC-MS using two different methods.
Ile was analyzed using Lux Cellulose-4, 5 µm, flow 1 mL/min, H2O/AcN + 0.04%TFA isocratic 65:35 in 60 min. The retention time of the l-FDAA amino acid in the hydrolysate of 2 was established as l-Ile 36.1 min. Retention times for the derivatized amino acids standards were as follows: l-allo-Ile 31.7 min, l-Ile 36.1 min, d-allo-Ile 38.1 min, and d-Ile 51.4 min.
Pro, NMeLeu, and Glu were analyzed using Symmetry 4.6 × 150 mm, 3.5 µm, flow 0.8 mL/min, H2O+ 0.04%TFA/CH3CN + 0.04%TFA from 20% to 50% in 30 min. The retention time of the l-FDAA amino acids in the hydrolysate of 2 were established as l-Glu 14.0 min, l-Pro 16.8 min, and NMe-l-Leu 28.5 min. Retention times for the derivatized amino acids standards were as follows: l-Glu 14.1 min, d-Glu 15.2 min, l-Pro 16.8 min, d-Pro 14.01 min, NMe-l-Leu 28.5 min, and NMe-d-Leu 30.1 min.

3.5. Biological Activity

The cytotoxic activity of 1 and 2 was tested against four human tumor cell lines, lung (A-549), colon (HT-29), breast (MDA-MB-231), and pancreas PSN-1, and both compounds displayed a GI50 > 1.2 × 10−5 M in all cell lines. Since these compounds did not show cytotoxicity, we further evaluated an anticancer response through other targets. Thus, compound 1 was further tested for the capacity to inhibit the enzyme topoisomerase I, but it showed no inhibition of the enzyme at 1.0 × 10−5 M, and therefore, it was not considered active in inhibiting this enzyme. Likewise, compounds 1 and 2 were unable to impair the interaction between the programmed cell death protein PD-1 and its natural ligand PDL1 as demonstrated by their lack of effect in a cell-based assay whose final readout was dependent on the interaction between the two proteins (Table 3).

4. Conclusions

In summary, two new peptides bearing unprecedented halogenated moieties, haloirciniamide A (1) and seribunamide A (2), were isolated from a marine sponge belonging to the Irnicia genus, which was selected for further studies. The sample was collected around the Thousand Islands (Indonesia) by the Pharmamar expedition team in collaboration with the Research Center for Oceanography, Indonesian Institute of Sciences (RCO-LIPI). The planar structures of the novel compounds were determined by a combination of extensive NMR and HPLC-MS experiments. The absolute configuration was achieved by Marfey’s analysis after acid hydrolysis. Cytotoxic activity in the four cancer cell lines tested was not observed for 1 and 2, as in the case of gunungamide A and cyclocinamide B. In addition, an anticancer immune response was also evaluated, and neither compound was able to impair PD1–PDL1 interaction. Moreover, compound 1 failed to inhibit the enzyme topoisomerase I. Unfortunately, the amount of the compounds isolated was not enough for further assays. This work is the first example of the isolation and structural elucidation of novel compounds with unique structural features from an Ircinia sponge, which highlights this gender and its microbiota as a distinctive source of novel structures.

Supplementary Materials

The following are available online at https://www.mdpi.com/1660-3397/18/8/396/s1, Figure S1: Picture of the fresh sponge; Figures S2–S16: NMR spectra of compound 1. Figures S17–S29: NMR spectra of compound 2. Figures S30: QTOFMS spectrum of compound 1 and fragments found. Figures S31–S38: Analysis by Marfey’s method.

Author Contributions

Sample collection: T.A.H., A.B. and S.B.; Conceptualization and supervision: C.C. and M.Y.P.; Methodology and writing: R.F.; Investigation: A.B.; Original draft preparation and writing: M.P.; Review and editing: C.C., M.P. and M.Y.P. All the authors read, reviewed, and agreed with the structure and content of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The present research was financed in part by Grants from Ministerio de Ciencia, Innovación y Universidades of Spain (AGL2015-63740-C2-2-R and RTC-2016 4611-1, Inmunotop project), cofunded by the FEDER Programme from the European Union.

Acknowledgments

We gratefully acknowledge the help of our PharmaMar colleagues C. Crespo for her excellent technical assistance, E. Millán for performing the ESI(+) and MSe experiments, J. M. Dominguez for the biological assays and S. Munt for revision of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanif, N.; Murni, A.; Tanaka, C.; Tanaka, J. Marine Natural Products from Indonesian Waters. Mar. Drugs 2019, 17, 364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mehbub, M.F.; Lei, J.; Franco, C.; Zhang, W. Marine Sponge Derived Natural Products between 2001 and 2010: Trends and Opportunities for Discovery of Bioactives. Mar. Drugs 2014, 12, 4539–4577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kawakami, A.; Miyamoto, T.; Higuchi, R.; Uchiumi, T.; Kuwano, M.; Van Soet, R.W.M. Structure of a novel multidrug resistance modulator, irciniasulfonic acid, isolated from a marine sponge Ircinia sp. Tetrahedron Lett. 2001, 42, 3335–3337. [Google Scholar] [CrossRef]
  4. Sica, D.; Piccialli, V.; Pronzato, R. Sterols from the sponges Ircinia pipetta and Dysidea avara identification of cholestatrienol. Comp. Biochem. Physiol. 1987, 88, 293–296. [Google Scholar]
  5. Venkateswarlu, Y.; Reddy, M.V.R.; Rao, M.N. A new epoxy sterol from the sponge Ircinia fasciculata. J. Nat. Prod. 1996, 59, 876–877. [Google Scholar] [CrossRef]
  6. Issa, H.H.; Tanaka, J.; Higa, T. New cytotoxic furanosesterterpenes from an Okinawan marine sponge Ircinia sp. J. Nat. Prod. 2003, 66, 252–254. [Google Scholar] [CrossRef]
  7. Lai, Y.Y.; Lu, M.C.; Wang, L.H.; Chen, J.J.; Fang, L.S.; Wu, Y.C.; Sung, P.J. New scalarane sesterterpenoids from the Formosan sponge Ircinia felix. Mar. Drugs 2015, 13, 4296–4309. [Google Scholar] [CrossRef] [Green Version]
  8. Rashid, M.A.; Gustafson, K.R.; Boyd, M.R. New chondropsin macrolide lactams from marine sponges in the genus Ircinia. Tetrahedron Lett. 2001, 42, 1623–1626. [Google Scholar] [CrossRef]
  9. Chevallier, C.; Bugni, T.S.; Feng, X.; Harper, M.K.; Orendt, A.M.; Ireland, C.M. Tedanolide C: A potent new 18-membered-ring cytotoxic macrolide isolated from the Papua New Guinea marine sponge Ircinia sp. J. Org. Chem. 2006, 71, 2510–2513. [Google Scholar] [CrossRef]
  10. Feng, Y.; Caroll, A.R.; Pass, D.M.; Archbold, J.K.; Avery, V.M.; Quin, R.J. Polydiscamides B−D from a marine sponge Ircinia sp. as potent human sensory neuron-specific G protein coupled receptor agonists. J. Nat. Prod. 2008, 71, 8–11. [Google Scholar] [CrossRef]
  11. Mohamed, A.M.; Rao, V.; Hamann, M.T.; Kelly, M.; Hill, R.T. Monitoring Bacterial Diversity of the Marine Sponge Ircinia strobilina upon Transfer into Aquaculture. Appl. Environ. Microbiol. 2008, 74, 4133–4143. [Google Scholar] [CrossRef] [Green Version]
  12. Mau, C.; Nakao, Y.; Yoshida, W.Y.; Scheuer, P.J.; Kelly-Borges, M. Waiakeamide, a Cyclic Hexapeptide from the Sponge Ircinia dendroides. J. Org. Chem. 1996, 61, 6302–6304. [Google Scholar] [CrossRef] [PubMed]
  13. Murakami, Y.; Takei, M.; Shindo, K.; Kitazume, C.; Tanaka, J.; Higa, T.; Fukamachi, H. Cyclotheonamide E4 and E5, New Potent Tryptase Inhibitors from an Ircinia Species of Sponge. J. Nat. Prod. 2002, 65, 259–261. [Google Scholar] [CrossRef]
  14. Hooper, G.J.; Orjala, J.; Schatzman, R.C.; Gerwick, W.H. Carmabins A and B, New Lipopeptides from the Caribbean Cyanobacterium Lyngbya majuscula. J. Nat. Prod. 1998, 614, 529–533. [Google Scholar] [CrossRef] [PubMed]
  15. Ebada, S.S.; Wray, V.; De Voogd, N.J.; Deng, Z.; Lin, W.; Proksch, P. Two New Jaspamide Derivatives from the Marine Sponge Jaspis splendens. Mar. Drugs 2009, 7, 435–444. [Google Scholar] [CrossRef] [PubMed]
  16. Fu, X.; Do, T.; Schmitz, F.J.; Andrusevich, V.; Engel, M.H. New Cyclic Peptides from the Ascidian Lissoclinum patella. J. Nat. Prod. 1998, 61, 1547–1551. [Google Scholar] [CrossRef] [PubMed]
  17. McIntosh, M.; Cruz, L.J.; Hunkapiller, M.W.; Gray, W.R.; Olivera, B.M. Isolation and structure of a peptide toxin from the marine snail Conus magus. Arch. Biochem. Biophys. 1982, 218, 329–334. [Google Scholar] [CrossRef]
  18. Pettit, G.R.; Kamano, Y.; Herald, C.L.; Tuinman, A.A.; Boettner, F.E.; Kizu, H.; Schmidt, J.M.; Baczynskyj, L.; Tomer, K.B.; Bontems, R.J. The isolation and structure of a remarkable marine animal antineoplastic constituent: Dolastatin 10. J. Am. Chem. Soc. 1987, 109, 6883–6885. [Google Scholar] [CrossRef]
  19. Laird, D.W.; LaBarbera, D.V.; Feng, X.D.; Bugni, T.S.; Harper, M.K.; Ireland, C.M. Halogenanted Cyclic Peptides Isolated from the Sponge Corticium sp. J. Nat. Prod. 2007, 70, 741–746. [Google Scholar] [CrossRef] [Green Version]
  20. Tarazona, G.; Fernández, R.; Cruz, P.G.; Pérez, M.; Rodríguez, J.; Jiménez, C.; Cuevas, C. Combining JBCA and Marfey’s methodology to determine the absolute configuration of threonines: The case of gunungamide A, a new cyclic depsideptide containing chloropyrrole from the sponge Discodermia sp. Org. Chem. Front. 2019, 6, 15–21. [Google Scholar] [CrossRef]
  21. Yasuda, T.; Araki, A.; Kubota, T.; Ito, J.; Mikami, Y.; Fromont, J.; Kobayashi, J. Bromopyrrole Alkaloids from Marine Sponges of the Genus Agelas. J. Nat. Prod. 2009, 72, 488–491. [Google Scholar] [CrossRef] [PubMed]
  22. Kobayashi, H.; Kitamura, K.; Nagai, K.; Nakao, Y.; Fusetani, N.; van Soest, R.W.M.; Matsunaga, S. Carteramine A, an inhibitor of neutrophil chemotaxis, from the marine sponge Stylissa carteri. Tetrahedron Lett. 2007, 48, 2127–2129. [Google Scholar] [CrossRef]
  23. Emrich, R.; Weyland, H.; Weber, K. 2,3,4-Tribromopyrrole from the marine Polychaete Polyphysia crassa. J. Nat. Prod. 1990, 53, 703–705. [Google Scholar] [CrossRef]
  24. Tebben, J.; Motti, C.; Tapiolas, D.; Thomas-Hall, P.; Harder, T. A Coralline Algal-Associated Bacterium, Pseudoalteromonas Strain J010, Yields Five New Korormicins and a Bromopyrrole. Mar. Drugs 2014, 12, 2802–2815. [Google Scholar] [CrossRef] [Green Version]
  25. Marfey, P. Determination of D-amino acids. II. Use of a bifunctional reagent, 1,5-difluoro-2,4-dinitro-benzene. Carlsberg Res. Commun. 1984, 49, 591–596. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Chemical structures of the compounds 1 and 2 isolated from Ircinia sp.
Figure 1. Chemical structures of the compounds 1 and 2 isolated from Ircinia sp.
Marinedrugs 18 00396 g001
Figure 2. Selected key COSY (bold), HMBC (red), and ROESY (blue) correlations for 1 and 2.
Figure 2. Selected key COSY (bold), HMBC (red), and ROESY (blue) correlations for 1 and 2.
Marinedrugs 18 00396 g002
Figure 3. Fragment found for 1 by QTOF.
Figure 3. Fragment found for 1 by QTOF.
Marinedrugs 18 00396 g003
Table 1. NMR spectroscopy data for 1 (1H NMR MHz, 13C NMR 125 MHz).
Table 1. NMR spectroscopy data for 1 (1H NMR MHz, 13C NMR 125 MHz).
PosδC, Mult aδH, Mult (J in Hz) aδC, Mult bδH, Mult (J in Hz) b
Dap11169.5, C 172.3, C
250.3, CH4.23 ddd (6.2, 6.2, 2.8)52.2, CH4.54 dd (5.6, 2.9)
349.2, CH22.87 d (13.9, 6.2)50.5, CH23.17 dd (14.6, 5.6)
3.13 d (13.9, 2.8) 3.54 d (14.2)
NH 9.11 d (6.2) 8.97 d (6.6)*
NH2
NMeClHis1169.0, CO 170.9, CO
265.6, CH3.99 dd (10.6, 3.9)67.0, CH4.06 dd (10.1, 4.5)
324.9, CH23.05 m25.9, CH23.19 dd (15.2, 10.1)
3.30 dd (15.2, 4.5)
4110.5, C 135.6, C
5109.5, CH6.76 s120.8, CH6.96 s
6128.2, C 131.0, C
NMe39.6, CH32.84 s40.4, CH33.02 s
iAsn1173.5, C 175.8, C
235.0, CH22.80 dd (16.6, 2.7)36.2, CH23.02 m
3.09 dd (16.6, 5.8) 3.27 m
348.4, CH4.58 ddd (8.0, 5.8, 2.7)50.3, CH4.84 m
4172.2, CO 173.8, CO
NH 7.13 d (8.0) 7.61 d (6.3) *
NH2
iSer1171.9, C 173.3, C
267.8, CH4.13 dd (9.0, 4.2)69.5, CH4.40 dd (9.5, 4.0)
342.8, CH22.75 ddd (9.0, 9.4, 5.4)44.2, CH23.00 m
3.47 m 3.75 dd (13.2, 4.0)
NH 8.23 t (5.4) 8.25 s *
Dap21170.5, C 172.8, C
251.5, CH4.49 ddd (9.2, 9.2, 6.3)53.4, CH4.84 m
340.1, CH23.22 m41.6, CH23.57 dd (13.8, 8.6)
4.04 ddd (12.9, 6.3, 6.3) 4.22 dd (13.8, 5.4)
NH-1 7.72 d (9.2) 7.98 d (9.5) *
NH-2 7.20 t (6.3, 6.3) 7.37 t (6.0) *
Br2Py1158.6, CO 161.4, CO
2118.0, C 120.2, C
3110.4, CH6.30 d (2.7)112.0, CH6.15 s
496.9, C 99.4, C
5123.2, C 124.2, C
NH 12.68 d (2.7) 12.03 s
a In DMSO-d6. b In CD3OD (* CD3OH).
Table 2. NMR spectroscopy data for 2 (1H NMR 500 MHz, 13C NMR 125 MHz).
Table 2. NMR spectroscopy data for 2 (1H NMR 500 MHz, 13C NMR 125 MHz).
PosδH, Mult (J in Hz) aδC, Mult aδH, Mult (J in Hz) bδC, Mult b
Br3Py1-161.8, CO-159.1, CO
2-128.6, C-128.5, C
3-102.7, C-100.7, C
4-101.3, C-93.4, C
5-110.8, C-107.9, C
NMe3.76, s36.7, CH33.61, s35.7, CH3
Ile1-174.3, CO-171.4, CO
24.84, m55.7, CH4.65, dd, 8.35, 8.553.7, CH
31.96, m38.0, CH1.87, m35.8, CH
41.73, m, 1.22, m25.9, CH21.57, m; 1.21, m24.2, CH2
50.95, t, 7.411.2, CH30.83, t, 7.410.7, CH3
60.99, d, 6.815.7, CH30.85, d, 6.915.1, CH3
NH8.24, d, 8.1-8.56, d, 8.2-
NMeLeu1-171.8, CO-168.8, CO
25.51, dd, 10.3, 4.754.3, CH5.35, dd, 10.1, 4.351.7, CH
31.77, m; 1.61, m38.0, CH21.59, m; 1.42, m36.7, CH2
41.56, m25.7, CH1.43, m23.9, CH
50.97, d, 6.223.6, CH30.87, d, 6.223.1, CH3
60.93, d, 6.122.3, CH30.83, d, 6.221.8, CH3
NMe3.21, s31.8, CH33.04, s30.5, CH3
Pro1-174.5, CO-171.4, CO
24.41, m61.6, CH4.31, dd, 8.3, 4.159.1, CH
32.23, m; 2.00, m30.5, CH22.02, m; 1.80, m28.9, CH2
42.08, m; 1.92, m26.0, CH21.90, m; 1.77, m24.4, CH2
53.75, m; 3.69, m48.8, CH23.53, m; 3.50; m46.7, CH2
Gln1-174.7, CO2H12.53, brs173.3, CO2H
24.41, m52.8, CH4.10, ddd, 8.6, 8.5, 5.251.5, CH
32.27, m; 1.92, m30.5, CH21.93, m; 1.74, m27.0, CH2
42.41, m; 2.32, m32.6, CH22.15, m; 2.11, m31.3, CH2
5-177.9, CONH27.19, s; 6.77, s173.5, CONH2
NH--8.08, brs-
a In CD3OD. b In DMSO-d6.
Table 3. % PD-1 and TOPO-I inhibition for compounds 1 and 2. PD-1: programmed cell death protein.
Table 3. % PD-1 and TOPO-I inhibition for compounds 1 and 2. PD-1: programmed cell death protein.
CompoundTarget%Inhibition at 1 × 10−5 M
1Top-I3
1PD-10.3
2PD-1−3.5

Share and Cite

MDPI and ACS Style

Fernández, R.; Bayu, A.; Aryono Hadi, T.; Bueno, S.; Pérez, M.; Cuevas, C.; Yunovilsa Putra, M. Unique Polyhalogenated Peptides from the Marine Sponge Ircinia sp. Mar. Drugs 2020, 18, 396. https://doi.org/10.3390/md18080396

AMA Style

Fernández R, Bayu A, Aryono Hadi T, Bueno S, Pérez M, Cuevas C, Yunovilsa Putra M. Unique Polyhalogenated Peptides from the Marine Sponge Ircinia sp. Marine Drugs. 2020; 18(8):396. https://doi.org/10.3390/md18080396

Chicago/Turabian Style

Fernández, Rogelio, Asep Bayu, Tri Aryono Hadi, Santiago Bueno, Marta Pérez, Carmen Cuevas, and Masteria Yunovilsa Putra. 2020. "Unique Polyhalogenated Peptides from the Marine Sponge Ircinia sp." Marine Drugs 18, no. 8: 396. https://doi.org/10.3390/md18080396

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