peptides xxx (2006) xxx–xxx available at www.sciencedirect.com 7 8 9 10 OO 6 Marisa Rangel a,b,c,*, Marisa P. Prado b, Katsuhiro Konno c, Hideo Naoki d, José C. Freitas a,c, Glaucia M. Machado-Santelli b PR 5 a Department of Physiology, Biosciences Institute, University of Sao Paulo, Sao Paulo 05508-900, Brazil Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo 05508-900, Brazil c Center for Applied Toxinology, Butantan Institute, Sao Paulo 05503-900, Brazil d Okinawa Health Biotechnology Research Development Center, 12-75 Suzaki, Gushikawa, Okinawa 904-2234, Japan b article info abstract Article history: Crude extracts of the marine sponge Geodia corticostylifera from Brazilian Coast have previously Received 17 February 2006 shown antibacterial, antifungal, cytotoxic, hemolytic and neurotoxic activities. The present Received in revised form work describes the isolation of the cyclic peptides geodiamolides A, B, H and I from G. ED 4 Cytoskeleton alterations induced by Geodia corticostylifera depsipeptides in breast cancer cells 5 April 2006 corticostylifera (1–4) and their anti-proliferative effects against sea urchin eggs and human Accepted 6 April 2006 breast cancer cell lineages. Its structure–activity relationship is discussed as well. In an initial CT 1 2 3 F journal homepage: www.elsevier.com/locate/peptides series of experiments these peptides inhibited the first cleavage of sea urchin eggs (Lytechinus variegatus). Duplication of nuclei without complete egg cell division indicated the mechanism of action might be related to microfilament disruption. Further studies showed that the Cancer cell geodiamolides have anti-proliferative activity against human breast cancer cell lines (T47D Cytoskeleton and MCF7). Using fluorescence techniques and confocal microscopy, we found evidence that Sea urchin egg the geodiamolides A, B, I and H act by disorganizing actin filaments of T47D and MCF7 cancer Marine sponge cells, in a way similar to other depsipeptides (such as jaspamide 5 and dolastatins), keeping the Depsipeptide normal microtubule organization. Normal cells lines (primary culture human fibroblasts and Geodiamolide BRL3A rat liver epithelial cells) were not affected by the treatment as tumor cells were, thus RR E Keywords: 13 11 12 14 15 16 1. 17 18 19 20 21 22 23 Studies on marine life forms in the last few years have led to the discovery of a variety of organic compounds with known or novel pharmacological and toxic activities on mammalian species. Available evidence suggests that the sea offers a rich source of new organic molecules which, either structurally modified or not, may be used as medicines, or as biochemical, UN Introduction CO indicating the biomedical potential of these compounds. # 2006 Elsevier Inc. All rights reserved. physiological or pharmacological tools in biomedical research [17,29,40]. Chemical defense through synthesis or accumulation of large amounts of toxic or deterrent natural products is usually found in Porifera [5]. Many of the compounds isolated from marine sponges exhibit neurotoxic, bactericidal, ichthyotoxic, cytotoxic, haemolytic and other toxic properties [37]. * Corresponding author at: Laboratorio Especial de Toxinologia Aplicada, Instituto Butantan, Av. Vital Brasil, 1500, CEP 05503-900 Sao Paulo, SP, Brazil. Tel.: +55 11 37261024; fax: +55 11 37261024. E-mail address: [email protected] (M. Rangel). 0196-9781/$ – see front matter # 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.peptides.2006.04.021 PEP 66763 1–11 23 24 25 26 27 28 29 30 31 2 evidence for the biomedical potential of these compounds, the cytoskeleton proteins of two different normal cell lineages (human fibroblasts and rat liver cells) were also analyzed under confocal microscope after incubation with the sponge depsipeptides. 2.1. Extraction and isolation of compounds OO 97 PR Specimens of G. corticostylifera (2.7 kg) were collected by Dr. Marcio dos Reis Custodio on June 2001 off the coast of Sao Paulo State, Brazil, then homogenized in methanol (1:3, w/v) and filtered. The filtered material was evaporated and partitioned with water/methylene chloride (1:1, v/v). The non-polar fraction volume was reduced in a vacuum evaporator and partitioned in methanol–water (9:1, v/v)/ n-hexane (1:2, v/v). The methanol–water fraction was fractionated by a Sep Pak Vac C18 cartridge with step-wise elution of 20, 50 and 90% CH3CN in water. Successive purification of the 50–90% acetonitrile fractions by reversed-phase HPLC using CAPCELL PAK C18 (10 mm 250 mm) with isocratic elution of 45% CH3CN/H2O at a flow rate of 2.5 ml/min over 40 min, monitored by UV absorption at 215 nm yielded the anti-proliferative compounds geodiamolide A (8.8 mg), B (4.6 mg), H (12.2 mg) and I (5.9 mg). ED RR E CO F Materials and methods 2.2. 91 92 93 94 95 96 2. CT Bioassay guided fractionation is the most frequently used technique to isolate sponge peptides, which are usually cyclic and lipophilic [18]. The depsipeptides isolated from marine sponges or associated organisms are usually described as cytotoxic substances, such as jaspamide, or jasplakinolide [14,45,46], geodiamolides [10,12,13,15,43], hemiasterlins and criamides [12], halicilindramides [23]. Nevertheless, there are sponge depsipeptides which presented anti-inflammatory activity, as the halipeptins A and B [36], and anti-HIV activity, as papuamides A and microspinosamide [16,44]. From the genus Geodia, compounds other than depsipeptides have also presented interesting biological activities; for example, the brominated cyclopeptides from the marine sponge Geodia barretti, barretin and 8,9-dihydrobarettin, which showed potent antifouling effects inhibiting the settlement of cyprid larvae of the barnacle Balanus improvisus [41]. A Geodia species collected from southern Australian waters of the Great Australian Bight has yielded a potent new in vitro nematocidal agent identified as geodin A Mg salt (1), a new macrocyclic polyketide lactam tetramic acid magnesium salt [9]. Additionally, two proteins from Geodia mesotriaena, named geodiastatins 1 and 2, presented antineoplastic activity against the murine P388 lymphocytic leukemia [33], and geodiatoxins 1 and 2, related proteins from the same species, were very toxic to mice (i.p.)[34]. The present is focused on the anti-proliferative compounds from the marine sponge Geodia corticostylifera (Porifera, Demospongiae). This species can be found in Southeastern Brazilian coast and Venezuela [21]. Its orange color, the apparent lack of predators and other sponge species nearby, suggests the presence of chemical defense mechanisms. Previous studies indicated that the crude extracts of G. corticostylifera displayed bactericidal and fungicidal activities [30], and cytotoxicity to sea urchin eggs, neurotoxicity to crab sensory nerve, and haemolytic activity in mice erythrocytes [38]. Neurotoxic and haemolytic activities were recently related to a pore-forming substance in the extract, which incorporates channels on artificial lipid bilayers. These channels have small conductance levels, are rectifiers and cation selective [37]. The cyclic depsipeptides geodiamolides A (1), B (2), H (3) and I (4) were previously isolated and characterized from the Caribbean marine sponge Geodia sp. [10,43]. Geodiamolides A and B presented antifungal activity [10], while geodiamolide H was active against cancer cell lineages (lung, HOP 92; central nervous system, SF-268; ovarian, OV car-4; kidney, A498 and UO-31; breast, MDA-MB-231/ATCC and HS 578T), although geodiamolide I was considered inactive in the same screening [43]. Compounds 1–4 were also isolated from the marine sponge G. corticostylifera, collected on the Brazilian coast, during our studies on purifying the pore-forming substance, and the anti-proliferative effects of these peptides were investigated against sea urchin eggs (Lytechinus variegatus), and T47D and MCF7 human breast cancer cells lineages. Using fluorescence techniques and confocal microscopy the effects of the geodiamolides A, B, I and H on cancer cell cytoskeleton and nucleus were observed. In an attempt to give UN 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 peptides xxx (2006) xxx–xxx Spectroscopic analysis 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 The compounds purified by HPLC were detected by positive electrospray ionization (ESI) mass spectrometry. Typical conditions were a capillary voltage of 2.4 kV, a cone voltage of 32 V, and a desolvation gas temperature of 150 8C. The experiments were done with a Q-ToF mass spectrometer (Micromass, UK) in Qq-orthogonal time-of-flight configuration. The spectra were recorded by the use of MassLynx 4.0 software (Micromass). NMR spectra were recorded on a JEOL EX-400 (at 400 MHz) or a Bruker DMX-750 spectrometer (at 750 MHz) in CD3OD or CDCl3. 116 117 118 119 120 121 122 123 124 125 126 2.3. 127 Sea urchin eggs development experiments Antimitotic activity was monitored initially as the ability of the geodiamolides A, B, H and I to inhibit the first cleavage of L. variegatus sea urchin eggs. The animals were collected in Sao Sebastiao, off the north coast of Sao Paulo State, Brazil. Germ cell release was induced by KCl injections (0.5 M, up to 3 ml) into the perivisceral cavity of the sea urchins. The eggs were washed three times in filtered seawater to remove the jelly coat. The sperm was maintained under refrigeration and not diluted until it was used. The geodiamolides A, B, H and I were diluted in filtered sea water/MeOH (19:1; v/v) in different concentrations. The final volume was 200 ml of seawater/MeOH mixture, plus 200 ml of egg suspension (prepared with 50 ml of washed eggs + 50 ml of sperm and observation of formation of fertilization layer). Control tubes contained 200 ml of sea water/MeOH mixture. When PEP 66763 1–11 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 3 peptides xxx (2006) xxx–xxx 143 144 145 146 147 148 149 the first division in control tubes occurred, the material was fixed in formaldehyde 10%, observed under microscope and photographed [25]. The results were analyzed according to their logarithms of mean and respective standard errors (n = 3). Dose response curves were plotted, and the EC50 values were calculated by means of non-linear regression. 150 2.4. 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 The T47D and MCF7 human breast cancer cells were grown in Sigma culture medium (DMEM) with 10% fetal bovine serum (Cultilab) in cellular culture multiplates (24 wells) with an initial density of 5 104 cells/well and incubated during 24 h. After this period, the medium was changed to a new one with the geodiamolides A, B, H and I. After 48 h the medium was removed, each well was washed with PBS, and trypsin was added to release the cells from the bottom. Then the trypsin was neutralized with medium, and 20 ml of Trypan Blue were added. The living cells were counted in a Neubauer chamber. The results were analyzed according to their mean log and respective standard errors (n = 3). Dose–response curves were plotted and EC50 were calculated by means of non-linear regression. For the fluorescence techniques, cells were plated on coverslips within culture Petri dishes. After 48 h incubating with either geodiamolides or control, the medium was removed, and the cells were fixed with formaldehyde (3.7%). After washing with PBS, the cells were treated with RNAase, stained with phalloidin-FITC (actin), and propidium iodide (nuclei). Monoclonal anti-tubulin and secondary antimouse-CY were used in immunofluorescence reactions. The preparations were mounted on slides with anti-fading (VectaShield, Vector). The fluorescent images were obtained by confocal laser scanning microscope (Zeiss LSM 510) with lasers of argon (458, 488 and 514 nm), helium–neon 1 (543 nm) and helium–neon 2 (633 nm) connected to an inverted fluorescence microscope, Zeiss Axiovert 100 M [26]. The effect of geodiamolides A and H (50 ng/ml) on microfilaments of T47D cells was also observed at different incubation times (2, 4, 8, 12 and 48 h), using phalloidin-FITC stained cells and confocal laser scanning microscope, as described above. 184 2.5. 185 186 187 188 Primary culture human fibroblasts and BRL3A rat liver epithelial cells were tested against geodiamolides A and H, stained with phalloidin-FITC and observed under a confocal laser scanning microscope, as described above. F OO 195 196 197 198 3.2. 199 PR comparison with literature revealed that the structures of GC503/901-1, -2, -3, and -4 were identical to geodiamolides B (2), A (1), I (4), and H (3), respectively, cyclic depsipeptides previously described [10,43]. ED Biological activities In the experiments with L. variegatus sea urchin eggs, the geodiamolides A, B, H and I inhibited the first cleavage in a dose-dependent form (Fig. 1), and the geodiamolides A and B acted at much lower concentrations than H and I (Table 1). Light microscope images of sea urchin eggs revealed a particularity of the depsipeptides anti-mitotic effect: nuclei duplications without complete cytokinesis at lower concentrations, and cells deformations at high concentration treatment (Fig. 2). In the breast cancer cells experiments, the values of EC50 for the geodiamolides A, B, H and I were also obtained in nM range (Table 1). Geodiamolides A and H were more effective against T47D cells, while geodiamolides B and A had a stronger effect on MCF7 cells. T47D and MCF7 cells growth inhibition curves by the compounds are shown in Fig. 3. The observation of T47D cells stained for actin filaments and nuclei in confocal microscope showed that geodiamolides A, B, H and I act upon F-actin, disorganizing the filaments and gathering them in the cytoplasm, in a dosedependent manner (Fig. 4). At the concentration of 100 ng/ml (135–170 nM), nuclei were displaced from central position in the cytoplasm and their shape changed when compared to control cells (Fig. 4). Notwithstanding, microtubule organization remained unchanged, as observed in immunofluor- CT 3. Results 3.1. Extraction and isolation of compounds 190 191 192 193 194 Fig. 1 – Dose–response curves of sea urchin eggs first cleavage inhibition induced by the geodiamolides (n = 3). RR E CO Normal cells experiments UN 189 Breast cancer cells experiments Reversed-phase HPLC purification of Sep Pak Vac prepared extract fractions GC503 and GC901 yielded four anti-mitotic peaks (screened in sea urchin eggs), named 1–4. Further mass spectrometry and NMR analysis of these peaks as well as Table 1 – EC50 values of geodiamolides (in nM) tested in L. variegatus sea urchin eggs, and in T47D and MCF7 human breast cancer cell lineages Compound Geodiamolide Geodiamolide Geodiamolide Geodiamolide A B H I Sea urchin eggs T47D MCF7 101.2 98.8 595.6 620.0 18.82 113.90 38.36 115.30 17.83 9.82 89.96 65.70 PEP 66763 1–11 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 4 UN CO RR E CT ED PR OO F peptides xxx (2006) xxx–xxx Fig. 2 – Light microscope observation of sea urchin eggs first cleavage in the absence of (control (A)) and under treatment with the geodiamolides A ((B) and (C)) (0.1 and 15 mg/ml), B ((D) and (E)) (0.1 and 15 mg/ml), H ((F) and (G)) (0.5 and 50 mg/ml) and I ((H) and (I)) (0.5 and 15 mg/ml). PEP 66763 1–11 5 ED PR OO F peptides xxx (2006) xxx–xxx 240 241 242 243 244 245 246 247 248 249 250 251 RR E 4. Discussion CO 239 escence preparations under confocal microscope, showing that the geodiamolides do not disassemble microtubules (Fig. 5). Disorganization of microfilaments of T47D cells induced by the geodiamolides A and H was perceived within 2 h of the treatment (the first time interval chosen in our experiment), and progressed along the incubation time (Fig. 6). In our experiments using normal cells lines, only geodiamolide A induced a slight disorganization of the human fibroblasts microfilaments at 100 ng/ml concentration, while geodiamolide H did not cause cytoskeleton alterations (Fig. 7(A)–(C)). The same concentration of both peptides had no effect on rat liver epithelial cells (BRL-3A) Factin (Fig. 7(D)–(F)). UN 225 226 227 228 229 230 231 232 233 234 235 236 237 238 CT Fig. 3 – Dose–response curves showing the growth inhibition of T47D and MCF7 human breast cancer cells induced by the depsipeptides of the sponge G. corticostylifera (n = 3). Cyclic peptides and depsipeptides are metabolite classes that have not been reported in sponges until recent years. These metabolite classes were described in species of four different orders: Axinellida, Choristida (to which the genus Geodia belongs), Halichondrida and Lithistida. A number of peptides and depsipeptides are characterized by the presence of new amino acids [39]. Depsipeptides, besides presenting unusual amino acids in their molecules, are also characterized by ester bonds and carboxylic acid. According to Tinto et al. [43], who first described the structures of the geodiamolides H and I, only geodiamolide H presented cytotoxic activity, inhibiting the growth of tumor cells in vitro, while geodiamolide I was completely devoid of activity. Our results with sea urchin eggs (Figs. 1 and 2) showed that both peptides are active under similar concentrations. Additionally, geodiamolides A and B from G. corticostylifera inhibited first division at smaller concentrations than geodiamolides H and I (Fig. 1). Observation under light microscope of sea urchin eggs showed that at the higher concentrations, the geodiamolides induced cell deformation, and at lower concentrations multinucleated cells were present (Fig. 2). Inhibition of sea urchin egg cleavage may occur in several processes in different stages of the cell cycle; for example, DNA, RNA or protein synthesis. Nevertheless, initial cleavage inhibition in these cells is due to either DNA synthesis blockade or inhibition of cytoskeleton protein organization [19], since in the first period of development the embryo does not synthesize RNA once all the mRNA it needs comes from the oocyte [6]. When there is microtubule disorganization, spots corresponding to nuclei duplication are perceptible in the cytoplasm, without having fulfilled mitosis [19]. Moreover, actin filaments sustain the cytoplasm, and form contractile rings during cell division [1]. Thus the effects induced by the geodiamolides in the sea urchin eggs seem to be due to a disorganization of cytoskeleton, specially the microfilaments. Similar results were found when the macrolides, isolated from an unidentified nudibranch whose chemical structures are similar to swinholide A produced by the Red Sea sponge Theonella swinhoei, inhibited the development of starfish embryos, producing multinucleated cells and unusually shaped nuclei [19]. Also, the diterpenoids of the sponge PEP 66763 1–11 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 6 UN CO RR E CT ED PR OO F peptides xxx (2006) xxx–xxx Fig. 4 – Laser scanning confocal microscope images of T47D cells stained with phalloidin-FITC (actin, green) and propidium iodide (nuclei, red); controls, and after 48 h incubation with the geodiamolides A, B, H and I. 282 283 284 285 Strongylophora strongylata from Japan inhibited the maturation of starfish oocytes Asterina pectinifera, and this effect may be due to either cyclinB/cdc2 or microtubule assembly inhibition [24]. Though a series of cyclic depsipeptides geodiamolides have been found and described as in vitro cytotoxic substances [10,12,13,15,43], their mechanism of action was not investigated in as much detail as jaspamide (5) [7,8,11,28,31,35], PEP 66763 1–11 286 287 288 289 7 peptides xxx (2006) xxx–xxx 290 291 CO RR E CT ED 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 UN 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 PR OO F 333 334 365 Fig. 5 – Laser scanning confocal microscope images of T47D cells stained with phalloidin-FITC (actin) and Cy-5 (tubulin); controls, and treated with 100 ng/ml of the geodiamolides A, B, H and I. PEP 66763 1–11 366 367 368 369 370 371 372 373 8 UN CO RR E CT ED PR OO F peptides xxx (2006) xxx–xxx Fig. 6 – Laser scanning confocal microscope images of T47D cells stained with phalloidin-FITC (actin), treated with 50 ng/ml of geodiamolides A and H and observed at different time intervals: ((A) and (G)) control; geodiamolide A: (B) 2 h, (C) 4 h, (D) 8 h, (E) 12 h, (F) 24 h; geodiamolide H: (H) 2 h, (I) 4 h, (J) 8 h, (K) 12 h, (L) 24 h. PEP 66763 1–11 9 CT ED PR OO F peptides xxx (2006) xxx–xxx RR E Fig. 7 – Laser scanning confocal microscope images of normal cells stained with phalloidin-FITC (actin). Human fibroblasts from primary culture: (A) control; (B) geodiamolide A and (C) geodiamolide H (100 ng/ml) and rat liver epithelial cells BRL-3A: (D) control; (E) Geodiamolide A and (F) Geodiamolide H (100 ng/ml). whose chemical structure is very similar to the geodiamolides (1–4). 292 293 294 295 296 297 298 299 The cytoskeleton is one of the main targets of substances in the tests applied to search for new compounds with potential anti-tumor activity [2,27]. It is known that jaspamide, isolated from the sponge Jaspis johnstoni, exerts its cytotoxic activity through actin filament stabilization, competing with phalloidin binding to F-actin [7,8,32]. Other cytotoxic marine depsipeptides, isolated from the sea hare Dolabella auricularia, dolastatin 11 [4] and doliculide [3], also stabilize actin filaments, in a way similar to jaspamide. More recently, bistramide A, a cyclic polyether from the ascidia Lissoclinum bistratum was found to 300 301 induce actin depolymerization by direct binding to F-actin [42]. Laser scanning confocal microscope analysis of T47D cells stained for actin and nucleus showed that the geodiamolides A, B, H and I act by affecting the cell microfilaments, disorganizing them and forming aggregates in the cytoplasm, in a dose-dependent manner (Fig. 4). In addition, under high doses the nucleus shapes were altered, as well as their location in the cytoplasm. In the present work a- and b-tubulin 302 303 304 305 306 307 308 309 UN CO 290 291 PEP 66763 1–11 10 364 365 366 PR The authors wish to thank Dr. Marcio Reis Custodio, from the Department of Physiology of Biosciences Institute, University of Sao Paulo, for collecting and identifying the sponge. This work was supported by FAPESP. MR and MPP received FAPESP fellowships and GMMS a CNPq fellowship. Conclusions The geodiamolides A, B, H and I presented anti-proliferative activity against breast cancer cells. This effect is related to the 375 376 377 378 379 references 380 [1] Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. Molecular biology of the cell New York: Garland Science; 2002. [2] Altmann KH. Microtubule-stabilizing agents: a growing class of important anticancer drugs. Curr Opin Chem Biol 2001;5:424–31. [3] Bai R, Covell DG, Liu C, Ghosh AK, Hamel E. (_)-Doliculide, a new macrocyclic depsipeptide enhancer of actin assembly. J Biol Chem 2002;277:32165–71. [4] Bai R, Verdier-Pinard P, Gangwar S, Stessman CC, Mcclure KJ, Sausville EA, et al. Dolastatin 11, a marine depsipeptide, arrests cells at cytokinesis and induces hyperpolymerization of purified actin. Mol Pharmacol 2001;59:462–9. [5] Braekman JC, Daloze D. Chemical defense in sponges. Pure Appl Chem 1986;58:357–64. [6] Brandhorst BP. Informational content of the echinoderm egg.. In: Browder LW, editor. Developmental biology: a compreensive synthesis. Oogenesis. New York: Plenum Press; 1985. p. 525–76. [7] Bubb MR, Senderowicz AMJ, Sausville EA, Duncan KLK, Korn ED. Jasplakinolide, a cytotoxic natural product, induces actin polymerization and competitively inhibits the binding of phalloidin to F-actin. J Biol Chem 1994;269:14869–71. [8] Bubb MR, Spector I, Beyer BB, Fosen KM. Effects of jasplakinolide on the kinetics of actin polymerization. J Biol Chem 2000;275:5163–70. [9] Capon RJ, Skene C, Lacey E, Gill JH, Wadsworth D, Friedel T. Geodin A magnesium salt: a novel nematocide from a Southern Australian marine sponge, Geodia. J Nat Prod 1999;62:1256–9. [10] Chan WR, Tinto W, Manchand PS, Todaro LJ. Stereostructures of geodiamolides A and B, novel cyclodepsipeptides from the marine sponge Geodia sp.. J Org Chem 1987;52:3091–3. [11] Cioca DP, Kitano K. Induction of apoptosis and CD10/ neutral endopeptidase expression by jaspamide in HL-60 line cells. Cell Mol Life Sci 2002;59:1377–87. [12] Coleman JE, de Silva E, Kong F, Andersen RJ, Allen TM. Cytotoxic peptides from the marine sponge Cymbastela sp.. Tetrahedron 1995;51:10653–62. [13] Coleman JE, Soest RV, Andersen RJ. New geodiamolides from the Sponge Cymbastela sp. collected in Papua New Guinea. J Nat Prod 1999;62:1137–41. 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 ED RR E CO 5. 367 368 369 370 371 372 373 374 Acknowledgments OO 6. F actin depolymerization, as observed in the confocal microscopy analysis of stained cells. Normal cell lines, however, did not show cytoskeleton alterations after treatment with the peptides, which is beneficial to the biomedical potential of such compounds. Interestingly, differences in peptide potencies are associated with an amino acid substitution or with the presence of bromide or iodide. CT immunofluorescence was utilized to show that the geodiamolides do not affect microtubule assembly (Fig. 5). Usually, the microtubule and microfilament networks interact during a variety of cellular processes, including vesicle and organelles transport, cleavage orientation, cell migration control, mitotic spindle rotation, and nuclear migration. Thus, substances that specifically affect microtubules or microfilaments may impair the processes accomplished by the cooperation of both of them [20]. Since the geodiamolides A, B, H and I disorganize actin microfilaments, they must be impairing the mechanisms involved in mitosis, and may be inducing cell death by apoptosis, like jaspamide does [11,28,31,35]. The disassembly of microfilaments of T47D cells induced by the geodiamolides A and H (Fig. 6) was perceived within 2 h of the treatment, and progressed along the incubation time. Jaspamide effect was evident after 2 h incubation, and continued to grow until 24 h of treatment [8]. Other peptides can affect the microfilaments in shorter incubation time, such as doliculide [3] and dolastatin 11 [4], which start to disrupt actin filaments of kangaroo rat kidney epithelial cells (PtK1 and 2) within 30 min of the beginning of treatment. The results of our experiments using normal cells lines (Fig. 7) are encouraging, considering that at the same concentration of the geodiamolides 70% of the breast cancer cells died in culture, and at 25 ng/ml a microfilaments disorganization was very clear (Fig. 4), thus indicating the biomedical potential of these marine depsipeptides. It is noteworthy that the structure–activity relationships of these compounds seem to vary according to the cell type. For instance, in sea urchin eggs, geodiamolides A and B are much more potent than geodiamolides H and I, a difference which apparently resides in the structural difference of alanine versus b-tyrosine. In contrast, this structural difference does not matter with T47D cells, but the halogen substituent X in the phenol ring of N-methyltyrosine moiety is crucial in this case because geodiamolides A and H (X = I) are much more potent than geodiamolides B and I (X = Br). More interestingly, in case of MCF7 cells, the trend is similar to that of sea urchin eggs, but distinct from that of T47D cells, despite the fact that these are mammalian cancer cell lines similar to T47D cells. Thus, we found that small structural alteration in geodiamolides largely affects the rank order of potency in each cell line, indicating that there would be different cellular sensitivity depending on its phenotype. Disruption of cytoskeleton elements such as microtubule and microfilament has been shown to interfere with the invasiveness and adhesion of tumor cells during the initial phases of metastasis formation [22]. New drugs acting in specific manner of this process may contribute to establishing new therapeutic approaches focusing on different phases of tumor progression. UN 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 peptides xxx (2006) xxx–xxx PEP 66763 1–11 11 peptides xxx (2006) xxx–xxx [35] [36] [37] ED [38] F [34] OO [33] PR [32] like protease-dependent pathway. Clin Diagn Lab Immunol 2000;7:947–52. Ou GS, Chen ZL, Yuan M. Jasplakinolide reversibly disrupts actin filaments in suspension-cultured tobacco BY-2 cells. Protoplasma 2002;219:168–75. Pettit GR, Rideout JA, Hasler JA. Isolation of geodiastatins 1 and 2 from the marine sponge Geodia mesotriaena. J Nat Prod 1981;44:588–92. Pettit GR, Rideout JA, Hasler JA. Isolation of geodiatoxins 2– 4 from the marine sponge Geodia mesotriaena. Comp Biochem Physiol C 1990;96:305–6. Posey SC, Bierer BE. Actin stabilization by jasplakinolide enhances apoptosis induced by cytokine deprivation. J Biol Chem 1999;274:4259–65. Randazzo A, Bifulco D, Giannini C, Bucci MR, Debitus C, Cirino G, et al. Halipeptins A and B: rwo novel potent antiinflammatory cyclic depsipeptides from the Vanuatu marine sponge Haliclona species. J Am Chem Soc 2001;123:10870–6. Rangel M, Konno K, Brunaldi K, Procopio J, Freitas JC. Neurotoxic activity induced by a haemolytic substance in the extract of the marine sponge Geodia corticostylifera. Comp Biochem Physiol (Part C) 2005;141:207–15. Rangel M, Sanctis B, Freitas JC, Polatto JM, Granato AC, Berlinck RGS, et al. Cytotoxic and neurotoxic activities in extracts of marine sponges (Porifera) from southeastern Brazilian coast. J Exp Mar Biol Ecol 2001;262:31–40. Schmitz F. Cytotoxic compounds from sponges and associated microfauna. In: Van Soest RWM, Van Kempen TMG, Braekman JC, Balkema AA, editors. Sponges in time and space: biology, chemistry, paleontology. Proceedings of the fourth international Porifera congress. Rotterdam; 1994. p. 485–96. Sheuer PJ. Marine natural products: diversity in molecular structure and bioactivity. Adv Exp Med Biol 1996;391:1–8. Sjögren M, Göransson U, Johnson AL, Dahlström M, Andersson R, Bergman J, et al. Antifouling activity of brominated cyclopeptides from the marine sponge Geodia barretti. J Nat Prod 2004;67:368–72. Statsuk AV, Bai R, Baryza JR, Verma VA, Hamel E, Wender PA, et al. Actin is the primary cellular receptor of bistramide A. Nat Chem Biol 2005;1:383–8. Tinto WF, Lough AJ, Mclean S, Reynolds WF, Yu M, Chan WR. Geodiamolides H and I, further cyclodepsipeptides from the marine sponge Geodia sp.. Tetrahedron 1998;54:4451–8. Tziveleka LA, Vagias C, Roussis V. Natural products with anti-HIV activity from marine organisms. Curr Top Med Chem 2003;3:1512–35. Zabriskie TM, Klocke JA, Ireland CM, Marcus AH, Molinski TF, Faulkner DJ, et al. Jaspamide, a modified peptide from a Jaspis sponge, with insecticidal and antifungal activity. J Am Chem Soc 1986;108:3123–4. Zampella A, Giannini C, Debitus C, Roussakis C, D’auria MV. New jaspamide derivatives from the marine sponge Jaspis splendans collected in Vanuatu. J Nat Prod 1999;62:332–4. [39] CO RR E CT [14] Crews P, Manes LV, Boehler M. Jasplakinolide, a cyclodepsipeptide from the marine sponge, Jaspis sp.. Tetrahedron Lett 1986;27:2797–800. [15] de Silva E, Andersen RJ, Allen TM. Geodiamolides C to F, new cytotoxic cyclodepsopeptides from the marine sponge Pseudoaxinyssa sp.. Tetrahedron Lett 1990;31: 489–92. [16] Donia M, Hamann MT. Marine natural products and their potential applications as anti-infective agents. Lancet Infect Dis 2003;3:338–48. [17] Faulkner DJ. Marine natural products. Nat Prod Rep 2002;19:1–48. [18] Fusetani N, Matsunaga S. Bioactive sponge peptides. Chem Rev 1993;93:1793–806. [19] Fusetani N. Marine metabolites which inhibit development of echinoderm embryos.. In: Scheuer PJ, editor. Bioorganic marine chemistry. Berlin: Springer-Verlag; 1987. p. 61–92. [20] Goode BL, Drubin DG, Barnes G. Functional cooperation between the microtubule and actin cytoskeletons. Curr Opin Cell Biol 2000;12:63–71. [21] Hajdu E, Muricy G, Custodio M, Russo C, Peixinho S. Geodia corticostylifera (Demospongiae, Porifera) new astrophorid from the Brazilian Coast (Southwestern Atlantic). Bull Mar Sci 1992;51:204–17. [22] Korb T, Schlüter K, Enns A, Spiegel HU, Senninger N, Nicolson GL, et al. Integrity of actin fibers and microtubules influences metastatic tumor cell adhesion. Exp Cell Res 2004;299:236–47. [23] Li H, Matsunaga S, Fusetani N. Halicylindramides A–C, antifungal and cytotoxic depsipeptides from the marine sponge Halichondria cylindrata. J Med Chem 1995;38:338–43. [24] Liu H, Namikoshi M, Akano K, Kobayashi H, Nagai H, Yao X. Seven new meroditerpenoids, from the marine sponge Strongylophora strongylata, that inhibited the maturation of starfish oocytes. J Asian Nat Prod Res 2005;7:661–70. [25] Malpezzi ELA, Freitas JC. Antimitotic effect of an extract of the sea anemone Bunodosoma caissarum on sea urchin egg development. Braz J Med Biol Res 1990;23:811–4. [26] Manelli-Oliveira R, Machado-Santelli G. Cytoskeletal and nuclear alterations in human lung tumor cells: a confocal microscope study. Histochem Cell Biol 2001;115:403–11. [27] McConnel OJ, Longley RE, Kohen FE. In: Gullo VP, editor. The discovery of natural products with therapeutical potential. Butterworth-Heineman; 1994. p. 109–74. [28] Morley SC, Sun GP, Bierer BE. Inhibition of actin polymerization enhances commitment to and execution of apoptosis induced by withdrawal of trophic support. J Cell Biochem 2003;88:1066–76. [29] Munro MHG, Blunt JW, Dumdei EJ, Hickfort SJH, Lill RE, Li S, et al. The discovery and development of marine compounds with pharmaceutical potential. J Biotechnol 1999;70:15–25. [30] Muricy G, Hajdu E, Araujo FV, Hagler AN. Antimicrobial activity of southwestern atlantic shallow-water marine sponges (Porifera). Sci Mar 1993;57:427–32. [31] Odaka C, Sanders ML, Crews P. Jasplakinolide induces apoptosis in various transformed cell lines by a caspase-3- UN 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 [40] [41] [42] [43] [44] [45] [46] 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 PEP 66763 1–11