Next Article in Journal
On the Role of Peripheral Sensory and Gut Mu Opioid Receptors: Peripheral Analgesia and Tolerance
Previous Article in Journal
The Polychromatic Woodburytype—Colour Tracking in Translucent, Patterned Gelatin/Pigment Films
Previous Article in Special Issue
1H NMR Metabolic Profile of Scyphomedusa Rhizostoma pulmo (Scyphozoa, Cnidaria) in Female Gonads and Somatic Tissues: Preliminary Results
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Collective Locomotion of Human Cells, Wound Healing and Their Control by Extracts and Isolated Compounds from Marine Invertebrates

Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
*
Author to whom correspondence should be addressed.
Molecules 2020, 25(11), 2471; https://doi.org/10.3390/molecules25112471
Submission received: 10 May 2020 / Revised: 22 May 2020 / Accepted: 25 May 2020 / Published: 26 May 2020
(This article belongs to the Special Issue Marine Bioinorganic and Bioorganic Chemistry)

Abstract

:
The collective migration of cells is a complex integrated process that represents a common theme joining morphogenesis, tissue regeneration, and tumor biology. It is known that a remarkable amount of secondary metabolites produced by aquatic invertebrates displays active pharmacological properties against a variety of diseases. The aim of this review is to pick up selected studies that report the extraction and identification of crude extracts or isolated compounds that exert a modulatory effect on collective cell locomotion and/or skin tissue reconstitution and recapitulate the molecular, biochemical, and/or physiological aspects, where available, which are associated to the substances under examination, grouping the producing species according to their taxonomic hierarchy. Taken all of the collected data into account, marine invertebrates emerge as a still poorly-exploited valuable resource of natural products that may significantly improve the process of skin regeneration and restrain tumor cell migration, as documented by in vitro and in vivo studies. Therefore, the identification of the most promising invertebrate-derived extracts/molecules for the utilization as new targets for biomedical translation merits further and more detailed investigations.

1. Introduction

The collective migration of cells is a complex integrated process at the basis of several different physiological events, such as embryo development, tissue shaping, and wound healing, whereas, in the course of diseases, like carcinogenesis, it is strongly implied in the step of tumor cell metastatization. In the recent years, a great deal of literature papers has focused on the molecular, biophysical and cytological aspects of cell locomotory patterns in the awareness of the clinical relevance shown by the advancement of knowledge on the mechanism and dynamism of collective cell migration. These studies shed light upon the mechanobiological bases of movement generation and speed/orientation setting and unveiled some of the signal transduction properties that are linked to the biochemical features of the migratory process. As a result of this research, the roles played in epithelial-mesenchymal transition (EMT) by integrin, junction protein and cytoskeletal dynamics, growth factor/cytokine and metalloprotease secretion, as well as cell interactions with components of the extracellular matrix (ECM) have been increasingly elucidated. A comprehensive overview of these topics can be found in several previously-published reviews [1,2,3].
Among the events of tissue repair involving collective cell motility, skin wound healing represents a multi-step coordinated process, which, following the initial inflammatory and hemostatic stages, associates re-epithelization, i.e., epidermal cell locomotion and growth on top of the granulation tissue, with tissue recovery implying the restoration of vascularization via angiogenesis and fibroplasia, which is the recruitment of activated fibroblasts and newly-differentiated myofibroblasts for ECM reconstitution and wound contraction [4,5,6,7,8]. Once the wound damage is repaired, a final tissue remodeling is undertaken to restore skin integrity and structure [9,10]. During wound re-epithelization, human keratinocytes move in a spatially- and directionally-organized way, whose molecular basis can be found in specific cell-ECM interactions, like the binding of type XVII collagen by both intermediate filament and actin cytoskeleton via hemidesmosomal actinin 4, and the creation of gradients of epidermal growth factor (EGF) immobilized within the ECM and presented to cell receptors [11,12]. Indeed, EGF is one of the most potent regulators of keratinocyte locomotory behavior along with transforming growth factor (TGF)-β, whose prominent regulatory effect on epidermal cell motility during wound repair via ERK signaling pathways is widely acknowledged [13]. Scientific interest has also focused upon the transcriptional factors that are up-regulated and activated in migrating keratinocytes during the repair process, leading to the identification, among others, of Smad7, Stat3, and c-Jun, which are functionally involved in the promotion of cutaneous wound healing, as extensively reviewed by Boudra and Ramsey [14].
Collective cell motility can be studied both in culture and in vivo. In the first case, the migratory behaviour of cell monolayers can be evaluated via the “in vitro wound healing” or “scratch” assay, in which a gap is created by scratching a pipette tip through a confluent sheet of cells in culture (Figure 1A), and the eventually-occurring bidimensional cell migration leading to the closure of the scratch being recorded by either phase-contrast microscopic images captured at regular intervals or time-lapse videomicroscopy [15].
When the model system studied is a tumor cell line, the scratch assay is commonly complemented by either the “Boyden chamber” or the “transwell migration/invasion” assays, as originally developed by Albini and coworkers [16], which measure the ability of a population of neoplastic cells to migrate individually through a physical barrier, i.e., an untreated or basement membrane (Matrigel)-coated porous polyester membrane, in response to a chemoattractant or a motility-promoting compound (Figure 1B) [17,18].
Dealing with in vivo experimental systems, the group locomotion of tumor cells might be examined through the chick embryo chorioallantoic membrane (CAM) assay, which implies the grafting of neoplastic cells on the CAM, their subsequent interaction with the blood vessels, and the invasion of the adjacent mesenchymal tissue, which can be studied by microscopical, immunohistochemical, and molecular techniques [19]. CAM might be used also to study the angiogenetic process that is characterized by migration of mesodermal blood vessels into the basement membrane and the ECM leading to their compartmentalization into new vessels in the ectoderm [20]. The incision of the rodent dorsum has been commonly used as an in vivo model to study the effect of molecules and extracts on the whole process of skin repair. Using this technique, the evolution of wound closure can be macroscopically assessed at different time points and, once the animals are sacrificed, histological observations, immunohistochemical analyses, biophysical evaluations, such as the quantitation of tensile strength, and biochemical/molecular assays can be performed on the biopsies. This enables collecting additional multi-perspective information on the fine tuning of tissue repair and also to evaluating the ability of supplemented substance(s) to promote a quick, robust, and histologically-shaped healing aimed to obtain the most correct skin replacement [21]. In vivo burn injury models may be created by pressing for few seconds a shaved and stretched area of the rodent dorsum skin with a metal block pre-heated at 100 °C or more, thus enabling studying the effect of novel compounds for topical therapeutics that are designed for the treatment of burn lesions [22]. Progresses in the knowledge of the molecular bases of wound repair and in the design of novel more efficacious medicaments are of basic importance to develop effective treatments for compromised healing occurring in pathologies, such as diabetes, arteriosclerosis, malnourishment, and malignant tumors. However, it is worth-mentioning that, although small animal models are commonly used for wound healing research, they display some biological limitations due to the differences in the histological architecture of rodent vs. human skin, and in the primary physiological act of healing, i.e., myofibroblast contraction for rodents vs. re-epithelization for humans. Other experimental models, such as the porcine one, have been proven to be more linkable with the human healing process, but the costs of housing and the difficulties in handling have limited their usability for first-line exploratory research [23,24]. Another aspect to consider is that “pure” scarring-free regeneration, which only sees new cells completely replacing the lost ones, is very uncommon in humans. In fact, regeneration is a well-known phenomenon in which animals restore lost parts of their body through four overlapping steps: (a) wound healing and histolysis, (b) blastema formation, (c) recognition of positional information, and (d) repatterning of the missing portion. As discussed by Krafts [25], the repair process in the context of the healing of human tissues consists of both regeneration and replacement, with the latter seeing the intervention of inflammatory cells and the sequential formation of granulation and fibrous tissues, which make the biological system more multifaceted.
It is widely recognized that seas and oceans, which account for about 97% of the whole water in the world, have a still little exploited asset in terms of biodiversity and, consequently, marine species-derived biological products. Much research interest has been focused on marine invertebrates, which represent more than 50% of the species inhabiting the aquatic environment in Europe. A remarkable amount of secondary metabolites is produced by aquatic invertebrates to aid in both the inter-individual signalization and the defense from predators, competitors, infective agents and UV radiation damages due to the adaptation mechanisms to the specific life conditions in the considerably differing marine ecosystems. An ever increasing number of these substances displays active pharmacological properties with an extensive spectrum of indications, making them prospective drugs of marine origin to be developed for therapeutic applications [26]. Within this context, the aim of this review is to pick up selected studies that report the extraction and identification of mixed fractions or isolated chemicals which exert a modulatory effect on collective cell migration and/or skin tissue reconstitution and recapitulate the molecular, biochemical, and/or physiological aspects, where available, which are associated to the metabolites or extracts under examination. Such compounds will be listed respecting the taxonomic hierarchy of the producing invertebrate species in dedicated paragraphs, in which the structural aspects of each organism and their contribution to “blue biotech” applications, if any, will also be briefly outlined.

2. Porifera

Porifera is the oldest metazoan group that is still widespread on the planet. They are very simple animals, which are devoid of real tissues or organs, and only consist of a few types of specialized cells that are surrounded by a collagen-rich extracellular matrix [27].
Chondrosia reniformis (Nardo, 1847; Demospongiae, Chondrosiida: Chondrosiidae) is a sessile species that lives in the shallow coasts of the Mediterranean Sea and the South-West coast of the Atlantic Ocean, which can reach a considerable body thickness (Figure 2).
This species shows remarkable regenerative properties and, for this reason, has been chosen as a model system for studies on the molecular pathways of tissue restoration in lower metazoan, highlighting the involvement of TGF superfamily proteins [28]. Unlike other sponges, this invertebrate lacks the silica spicules that commonly strengthen the other species’ body, instead obtaining its stiffness through a greater amount of collagen fibers that are tightly packed in a complex fibrillar network. Collagen extracts from C. reniformis have shown considerable potentiality in carrying out the role of nanoparticle vector and drug-coating material [29,30] in the design of biocompatible scaffolds for regenerative medicine [31]. Moreover, sticky hydrogels with self-healing properties have been obtained from collagen extracted from the ectosome and choanosome of this demosponge showing remarkable rheological properties that make them suitable as injectable matrices for biomedical applications [32]. Particular interest has been given to the cosmetic and wound healing effect of trypsin-digested C. reniformis collagen hydrolysates. In their paper, Pozzolini et al. [33] isolated different peptide fractions by HPLC and demonstrated that those ones particularly enriched in hydroxyproline-containing collagenous peptides promoted the migration of HaCaT keratinocytes and L929 fibroblasts in “scratch” assays, also accelerating cell proliferation at the site of the wound with respect to the untreated controls at the same time points. In addition, these preparations appeared able to trigger type I collagen synthesis and extracellular release by fibroblasts and protect keratinocytes from UV damage by enhancing cell viability after exposure and down-regulating the UV-induced expression of KRT1 and KRT10 keratin genes responsible of skin photoaging via epidermal thickening and decrement of elasticity. When also considering the radical scavenging activity shown by these collagen hydrolysates and their lack of toxicity towards different cultured cytotypes, the peptide fractions from C. reniformis collagen show considerable biotechnological potential in the development of formulations that are designed to mitigate the damages in UV-exposed or wounded skin.
Jaspis stellifera [Carter, 1879; Demospongiae, Tetractinellida: Ancorinidae; Figure 3A] is another sessile species typical of tropical and Indo-West Pacific environments. A number of anti-cancer bioactive molecules have been isolated from this marine invertebrate, such as jaspiferins, jaspiferals, stelliferins, and stellettin [34,35,36,37,38,39,40,41]. In their publication, Cheng et al. [42] produced additional data regarding the mobility control exercised by the triterpenoid stellettin B (3Z,3aS,5aR,9aR,9bS)-3a,6,6,9a-tetramethyl-3-[(3E,5E)-6-(5-methyl-6-oxopyran-2-yl)hepta-3,5-dien-2-ylidene]-4,5,5a,8,9,9b-hexahydro-1Hcyclopenta [a]naphthalene-2,7-dione; Figure 3B on human U87MG and GBM8401 glioblastoma cells. In particular, the migration and invasion of tumor cells was inhibited in both scratch wound healing and transwell chamber assays when exposed to an increasing concentration of the triterpene in the range between 1 and 10 μM. This appeared to be based, at least in part, upon the p-Akt downsignalling-mediated inhibition of the interaction between F-actin and the actin-binding protein P-girdin, responsible for pseudopoda extension at the cell leading edge, and the expression of matrix metalloprotease 2, phosphorylated integrin β1 and phosphorylated focal adhesion kinase by this cancer cytotype [43]. In consideration that glioblastoma cell survival and the angiogenetic processes in models in vitro and in vivo are also remarkably reduced in the presence of stellettin B [42], this molecule is a strong candidate as anti-cancer agent against malignant brain tumors.
Negombata magnifica (Keller, 1889; Demospongiae, Poecilosclerida: Podospongiidae), formerly named Latrunculia magnifica, is also known as toxic finger-sponge and it typically has a brilliant orange-red coloration and a digitate or leafy morphology with crooked branches (Figure 4A). It is distributed in the Indian Ocean and the Red Sea, where it intensively populates the coral reefs. When these sponges form colonies, they protect themselves via chemical defences, in particular releasing a reddish fluid containing latrunculins, toxic substances that are known to bind G-actin in a reversible manner and 1:1 stoichiometric ratio [44]. Latrunculin B (4R)-4-[(1R,4Z,8Z,10S,13R,15R)-15-hydroxy-5,10-dimethyl-3-oxo-2,14-dioxabicyclo[1 1.3.1] heptadeca-4,8-dien-15-yl]-1,3-thiazolidin-2-one; Figure 4B is a member of this family that is a promising agent for the development of anti-glaucoma and anti-arrythmia therapies [45,46]. This macrolide and its semi-synthetic derivatives 15-O-methyl-latrunculin B, N-hydroxymethyl- latrunculin B and N-acetyl-latrunculin B were found to restrain the migration of the murine brain-metastatic melanoma cell line B16B15b in both transwell chamber and scratch wound healing assays [47]. Latrunculin A ((4R)-4-[(1R,4Z,8E,10Z,12S,15R,17R)-17-hydroxy- 5,12-dimethyl-3-oxo-2,16dioxabicyclo[13.3.1] nonadeca-4,8,10-trien-17-yl]-1,3-thiazolidin-2-one; Figure 4C and its carbamate derivatives 17-O-[N-(phenyl)carbamoyl]-latrunculin A, 17-O-[N-(3-chloropropyl)carbamoyl] latrunculin A and 17-O-[N-(benzyl)carbamoyl]-latrunculin A appeared to be able to impair the invasive behavior of prostate cancer cells in Boyden chambers in the presence of Matrigel, as well as the disaggregation of spheroids in vitro [48]. In this paper, a putative inhibitory effect of these molecules on the activation and signalization of hypoxia-inducible factor (HIF)-1, a transcription factor that is involved in the metastatic attitude of prostate cancer cells [49], was presumed. Afterwards, the in vitro invasive phenotype of MDA-MB231 breast cancer cell was also found to be impaired in Boyden chamber assays by derivatives of latrunculin A, the most potent being 17-O-phenylethyl-latrunculin A and N-p-methoxyphenylacetyl-15-O-methyl-latrunculin A. Interestingly, the latter one is likely to recognize different and yet unknown target(s) than actin microfilaments. Of note, 15-O-methyl-latrunculin B also exhibited a powerful anti-angiogenic activity, whereas 17-O-phenylethyl-latrunculin A was significantly cytotoxic against MCF-7 breast cancer cells [50], with both properties being appropriate to potential anti-breast cancer drugs.
Pseudoceratina arabica (Keller, 1889; Demospongiae, Verongiida: Pseudoceratinidae; Figure 5A is distributed in the shallow sandy lagoons and seagrass beds of the southern Red Sea. It appears as a sprawling and branching sponge and his body might show varying characteristics, appearing dense, rubbery, flexible, or waxy. A number of literature data indicate that these sponges produce several compounds with varied biological activities, e.g., the antimicrobial and antifungal parasympatholytic moloka’iamine and moloka’iakitamide, and the skeletal α-chitin of particular interest for different biotechnological applications, such as electrochemistry and regenerative medicine [51,52]. The bromotyrosine derivative (1’R,5’S,6’S)-2-(3’,5’-dibromo-1’,6’ -dihydroxy-4’-oxocyclohex-2’-enyl) acetonitrile obtained from P. arabica was found to impair the TGFβ-stimulated epithelial-mesenchymal transition of A549 lung cancer cells and the related cell migration in scratch wound closure assays through the inhibition of both the activity of type I TGF-β receptor kinase ALK5 and the consequent motility-addressed actin microfilament reorganization thereby being a promising anti-tumoral pharmacological tool [53]. Noteworthy, the bidimensional locomotion of the highly-malignant MDA-MB231 breast cancer cells in wound closure assay was consistently restrained by the already-mentioned moloka’iamine (3-[4-(2-aminoethyl)-2,6- dibromophenoxy]propan-1-amine; Figure 5B and its P. arabica-obtained derivative ceratinin A, whereas the in vitro invasive activity of the same cell line in transwell assays was decreased by exposure to two other P. arabica-obtained moloka’iamine derivatives, i.e., ceratinin D and hydroxymoloka’iamine (2-amino-1- [4-(3-aminopropoxy)-3,5- dibromophenyl] ethanol; Figure 5C. In the same paper, evidence was shown that also the alkaloid subereamolline A produced by another verongid sponge distributed in the Southern Red and Western Arabian Sea, i.e., Suberea mollis (Row, 1911; Demospongiae, Verongiida: Aplysinellidae; Figure 5D) was able to inhibit the scratch wound healing in vitro by MDA-MB231 breast cancer cells [54]. Of note, S. mollis is a good producer of a number of compounds with overt anti-microbial and anti-oxidant hepatoprotective functions, such as aeroplysinins, the brominated arginine-derived alkaloids subereamines, and the brominated phenolic molecules subereaphenols, among others [55,56,57].
Siphonocalina (Callyspongia) siphonella (Lévi, 1965; Demospongiae, Haplosclerida: Callyspongiidae; Figure 6A), also named colonial tube-sponge for its appearance as a bunch of pale lavender long tubes, is a species typical of the Red Sea. This organism is a source of secondary metabolites endowed with anti-infective and anti-inflammatory properties, and cytotoxicity against cancer cells [58,59,60]. Sipholenol A ((3R,5aR,6R, 7S,9aR)-6- [2-[(1R,3aR,5S,8aR)-1-hydroxy-1,4,4,6-tetramethyl-2,3,3a,5,8,8a-hexahydroazulen-5-yl]ethyl]-2,2,5a,7-tetramethyl-4,5,6,8,9,9a-hexahydro-3H-benzo[b]oxepine-3,7-diol; Figure 6B is a triterpenoid that is produced by this demosponge displaying an anti-cancer activity that is based upon its ability to counteract the multidrug resistance of neoplastic cells via direct inhibition of P-glycoprotein [61]. Aliphatic polar and aromatic ester derivatives of this molecules exhibited a strong anti-migratory and anti-invasive effect towards MDA-MB231 malignant breast cells in scratch wound healing and Boyden chamber assay in the presence of basement membrane extract [62]. The down- regulation of cell motility appeared to be due, at least in part, to the inhibition of phosphorylation of protein tyrosine kinase-6 (PTK6), an anti-apoptotic and pro-metastatic factor for this cell line [63]. Akl et al. [64] demonstrated that the sipholenol A-4-O-3′,4′-dichlorobenzoate derivative was also active as potential anti-breast cancer compound. In particular, they found that this molecule inhibited the proliferation of a panel of breast tumor cell lines with different neoplastic phenotypes, impairing cell cycle progression, and it restrained both cell motility in transwell chamber assays in vitro and tumor mass expansion in MDA-MB231 xenograft nude mice model in vivo. Such effects appeared to be based, at least in part, upon the interference with the metastasis-associated breast cancer kinase (BRK) and focal adhesion kinase (FAK) signal cascades, in turn inhibiting the activation of some downstream signalling factors, such as AKT, mitogen-activated protein kinase (MAPK), and paxillin. Thus, sipholenol A appears to be a promising scaffold for the development of novel anti-malignant breast cancer drugs.
Xestospongia sp. (Demospongiae, Haplosclerida: Petrosiidae; Figure 7 is widely distributed in the Indian and South Pacific Oceans. These animals represent a rich source of secondary metabolites with very different biological properties that have been shown effective as enzymatic inhibitors, anti-malarial, anti-bacterial, vasodilatory, and anti-cancer agents [65,66,67,68,69,70]. Among them, ranieramycins are a group of biologically-active tetrahydroisoquinoline compounds. Halim et al. [71] reported a significant inhibitory effect exerted by a member of this family, ranieramycin M, on the motile and invasive behavior of H460 lung cancer cells in scratch wound healing and matrigel-containing transwell assays at non-toxic or growth-impairing concentrations.

3. Cnidaria

Cladiella australis (Macfadyen, 1936; Anthozoa, Alcyonacea: Alcyoniidae; Figure 8A), also known as the finger blanching soft coral, is a species distributed in the Western Pacific ocean, displaying brown or white color, depending on whether the polyps are extended or retracted. It is know that this organism produces several secondary metabolites with promising biological activities as hypoglicemic, anti-cancer and anti-melanogenic compounds, the latter having potential applications in medical cosmetology [72,73,74]. Among the molecules that were obtained from C. australis, research interest has been focused on the neuroprotective austrasulfone (4-ethenylsulfonylbutan-2-one; Figure 8B [75]. Its synthetic precursor, dihydroaustrasulfone alcohol, exhibited the ability to restrain the migratory behaviour of A549 lung tumor cells in both wound healing and transwell chamber assays in vitro. This inhibitory activity appeared to be the result of the inactivation of extracellular signal-regulated kinase (ERK) 1/2 intracellular signalling, which, in turn, down-regulated the levels of activated focal adhesion kinase (FAK), PI3K, and p-AKT. Such derangement of transduction mechanisms also determined the decrease of the production of matrix metalloproteinase (MMP)-2 and -9 enzymes, responsible for the observed reduced tumor size in a A549 xenograft nude mice model [76]. Interestingly, dihydroaustrasulfone alcohol was also proven to restrain the platelet-derived growth factor (PDGF)-BB promoted the proliferation and migration of vascular smooth muscle cells, as documented by wound healing, Boyden chamber, and transwell assays, thereby playing a vasoprotective role and being a good candidate for the treatment of vascular occlusive diseases [77,78].
Cladiella pachyclados (Klunzinger, 1877; Anthozoa, Alcyonacea: Alcyoniidae; Figure 9A) is a soft coral typical of reef-associated and subtropical, in particular Western Pacific, environments. It is known that this marine invertebrate produces eunicellin (i.e., [(1R,2R,3R,5S,6S,7S,8R,11S,14R)-5,6,14-triacetyloxy-6, 14-dimethyl-10-methylidene-3-propan-2- yl-15-oxatricyclo[6.6.1.02,7]pentadecan-11-yl]acetate)-based diterpenes with anti-inflammatory and anti-cancer properties. In particular, Hassan et al. [79] showed the powerful anti-migratory and anti-invasive role played in vitro on PC3 prostate cancer cells by some of these compounds, with the most effective in both experimental tests being pachycladin A ([(1R,2S,3R,6R,7R,8R,9R,12S,13S)-9-acetyloxy-12,13-dihydroxy-3,9,13- trimethyl-6-propan-2-yl- 15 oxatri cyclo[6.6.1.02,7]pentadecan-3-yl] butanoate; Figure 9B, sclerophytin F methyl ether ((1R,2R,6R,7R,8R,9S,12S,13S)-12-methoxy-9,13-dimethyl-3- methylidene-6-propan-2-yl-15 oxatricyclo[6.6.1.02,7]pentadecane-9,13-diol; Figure 9C), polyanthelin A ([(1R,2R,6S,8R,9S,10R,13R,14R)-2,6,10-trimethyl-13-propan-2-yl- 15,16- dioxatetracyclo [6.6.1.12,6.09,14]hexadecan-10-yl] acetate; Figure 9D, and sclerophytin A ((1R,2R,6R,7R,8R,9R,12S,13S)-9,13-dimethyl-3-methylidene-6-propan-2-yl-15oxatricyclo[6.6.1.02,7] pentadecane-9,12,13-triol; Figure 9E [79].
Sarcophyton crassocaule (Moser, 1919, Anthozoa, Alcyonacea: Alcyoniidae; Figure 10A is a subtropical species of sessile soft corals distributed in the Western Pacific area, New Caledonia, Taiwan and Ryukyu Island. A number of terpenoids with anti-tumoral properties have been extracted by this marine invertebrate [80]. Studies in vitro on the anti-migratory effects of compounds that are produced by S. crassocaule have been focussed on bladder and gastric tumor cell models. In the first case, the cembrenolide diterpene 13-acetoxysarcocrassolide ([(1S,3S,5S,8E,12E,14S,15S)-5,9,13- trimethyl-18-methylidene-17-oxo-4,16-dioxatricyclo[13.3.0.03, 5]octadeca-8,12-dien-14-yl] acetate; Figure 10B) was proven to impair locomotion in scratch wound healing assays, apart from inducing apoptotic cell death after 24 h of exposure. Western blot and proteomic analyses demonstrated the up- or down-regulation of a number of proteins, including members of the group of heat shock proteins, key enzymes of the Krebs cycle, and proteins that are associated to apoptosis and DNA repair, although no correlation between the biochemical data and the anti-migratory effect was shown [81]. Similar results on 13- acetoxysarcocrassolide-induced motility restraining were obtained with gastric carcinoma cells, in which the up-regulation of p38- and c-Jun N-terminal kinase (JNK)-transduction pathways and down-regulation of phosphoinositide 3-kinase (PI3K)/AKT signalization were also observed [82].
Sinularia nanolobata (Verseveldt, 1977; Anthozoa, Alcyonacea: Alcyoniidae) populates the coral reefs or rocks in shallows from East Africa to the Western Pacific area. The bioactive compound 11-dehydrosinulariolide (1R,3S,5S,8E,13R)-5,9,13-trimethyl-16-methylene-4,14-dioxa tricyclo[11.3.2.03,5] octadec-8-ene-12,15-dione11-dehydrosinulariolide; Figure 11A that is produced by this soft coral has been tested for the control of the migratory behavior of CAL-27 oral squamous cancer cells in scratch wound healing assays. The obtained results demonstrate that the wound repair function is down-regulated by the molecule, as well as cell growth and survival, whereas apoptotic death is promoted. Proteomic analyses showed that exposure to 11-dehydrosinulariolide determined the modification of the levels of proteins that are associated with cell growth, apoptosis, translation and protein folding, and energy metabolism [83]. Subsequent investigations on another bioactive molecule extracted from S. nanolobata, i.e., sinularin ((9E)-13-hydroxy-4,9,13-trimethyl-17-methylidene-5,15-dioxatricyclo[12.3.1.04,6] octadec-9-en-16-one; Figure 11B), confirmed this compound as an additional potential anti-cancer molecule. In fact, sinularin promoted the inhibition of growth and motility, the block at G2/M cell cycle phase, and the onset of apoptosis in A2058 melanoma cells and proteomic analyses found modifications in the levels of oncosuppressor and apoptosis-associated proteins consistent with the observed phenotypic changes [84].
Clavularia koellikeri (Dean, 1927; Anthozoa, Alcyonacea, Clavulariidae; Figure 12) is a widespread species throughout the Indian and Pacific oceans, from the Red Sea and East Africa across the Indo-Pacific to Micronesia, being a typical inhabitant of the Great Barrier Reef. Soft corals belonging to this taxonomic genus are an important resource of secondary metabolites, mainly terpenoids, which are endowed with unique structures and a wide range of biological activities [85,86]. Among them, clavukoellian A and clavukoellian D-related 4-O-deacetylparalemnolin D exerted an anti-locomotory effect on human umbilical vein endothelial (HUVEC) cells in wound healing assays, thus showing interesting anti-angiogenetic activity that merits further investigation [87].
Cyanea capillata (Linnaeus, 1758; Scyphozoa, Semaeostomeae: Cyaneidae; Figure 13), commonly known as lion’s mane jellyfish, populates the cold waters of the more northerly areas of the North and Arctic seas and the Pacific and Atlantic oceans, also finding itself along the coasts of Southeast China [88,89,90]. This organism represents the largest jellyfish known to date, with an umbrella from which originate a large number of reddish-brown or yellowish tentacles long up to 30 m and wide up to one meter, endowed with considerable amounts of nematocysts that release toxins dangerous for humans [91]. It is widely acknowledged that jellyfishes are important sources of many different bioactive molecules. This species, in particular, is a good producer of antioxidants conceivably aimed to protect its supercoiled DNA from oxidative damage. In fact, the molecular cloning and recombinant protein production of peroxiredoxin-, thioredoxin-, and Cu/Zn superoxide dismutase (SOD) homologues was successfully performed starting from cDNA libraries of C. capillata tentacles [92,93,94]. In addition, while using the same molecular source a recombinant protein with potent anti-bacterial activity was expressed and purified [95]. Wang et al. [96] demonstrated the promoting effect of C. capillata’s tentacle extracts on the migratory behavior of human umbilical vein endothelial cells in scratch wound healing assays, thus considering this mixture a good candidate as wound healing regulator, improving tissue repair in unfavorable conditions. In a following study, they isolated and purified a C. capillata-specific granulin, the 5782 Da-bioactive component of the extract, able to trigger cell locomotion, and also up-regulation of cyclin B1 and D1 and cell growth, via the activation of ERK1/2 and MAPK signal cascade [97].
Rhopilema esculentum (Kishinouye, 1891; Scyphozoa, Rhizostomeae: Rhizostomatidae; Figure 14) is a jellyfish species that inhabits pelagic and tropical environments, in particular populating the Northwestern Pacific area, the coasts of China and Japan, the Yellow Sea and the Bohai Sea. It can achieve the diameter of several cm and the weight of 30 kg. This organism is commonly known as flame jellyfish due to its mouth-arms that are endowed with abundant filamentous and large spindle-shaped appendices bearing undefined terminal clubs. R. esculentum is an eatable species, widespread in the market for its pleasant flavour and its lipid profile rich of fatty acids and glycerophospholipids that makes it a potential healthy food for human consumption and an aliment that is suitable for some commercial fishes and crustaceans [98,99]. In addition, significant insecticidal properties were demonstrated in the full proteinous venom of this invertebrate, thereby being a potential component of environmentally-friendly insecticides [100], whereas peptides that were obtained from R. esculentum’s hydrolysates exhibited angiotensin I-converting enzyme (ACE)-inhibitory and, therefore, anti-hypertensive and anti-oxidant properties [101,102]. Research interest has recently been focused on the tissue repair properties of the collagen component extracted from this organism. Felician et al. [103] demonstrated the ability of preparations of collagen peptides, which were obtained through either collagenase II- or papain digestion, to induce the motility of human vein umbilical cells in scratch wound healing assays. Moreover, the same preparations were applied to the back of wounded mice and the results that were obtained in vivo confirmed their regenerative properties determining a significant decrease of the wounded area, especially in case of treatment with collagenase II-digested samples. Immunohistochemical analyses of the areas of the in-progress tissue reconstitution showed the up-regulation of TGFβ1 and β-FGF, known factors that are involved in the inflammatory and angiogenetic phases of skin repair whose acceleration by the peptide mixture makes it a promising product for wound repair. Noteworthy, the collagen component extracted from R. esculentum has also been the object of studies showing its remarkable anti-photoaging and hemostatic properties, and its suitability and safety for use as a scaffold supporting the chondrogenic differentiation of adult stem cells in cartilage tissue engineering, which merits further exploitation [104,105,106].

4. Mollusca

Uroteuthis (Doryteuthis) singhalensis (Ortmann, 1891; Cephalopoda, Myopsida: Loliginidae, Figure 15A) is a demersal species, which is typical of subtropical and tropical areas, especially of the Indo-West Pacific zone, representing one of the commercially-important squid species in India. This cephalopod has been used as a source of the polysaccharide chitosan, which exhibits powerful antioxidant and anticoagulant properties, and of invertebrate collagen, which is a good alternative to the mammalian counterpart being the animal’s skin available in large amount as fish markets’ left-over [107,108]. Veeruraj et al. [109] recently reported the application of a biocomposite matrix that is constituted by the radical scavenging-carotenoid astaxhantin ((6S)-6-hydroxy-3-[(1E,3E,5E,7E,9E,11E,13E,15E,17E)-18-[(4S)-4-hydroxy-2,6,6- trimethyl-3- oxocyclohexen-1-yl]3,7,,12,16-tetramethyloctadeca-1,3,5,7,9,11,13,15,17-nonaenyl]-2,4,4-trimethylcyclo hex-2-en-1-one; Figure 15B incorporated into a scaffold of collagen, both extracted from U. singhalensis, for the healing of dermal wounds of rats in vivo. The obtained data demonstrated a higher wound contraction rate and tensile strenght and a faster epithelization time at the excision site, with an increased deposition of newly synthesized collagen in the damaged area following fibroblast proliferation and activation. Histopathological observations confirmed the remarkable healing properties of the material, additionally putting the onset of a pronounced and accelerated revascularization of the tissue in evidence. The obtained cumulative results highlighted the potentiality of this composite sponge in providing a superior-quality wound dressing, also in terms of stability, low-cost, biodegradability, and biocompatibility, which may have additional applications in tissue engineering and regenerative medicine.
Sepia kobiensis (Hoyle, 1885; Cephalopoda, Sepiida: Sepiidae; Figure 16), also known as kobi cuttlefish, is a benthic species distributed in the Indo-West Pacific area from Japan to the Persian Gulf. It is an eatable organism with a nutritional value that is very close to that of fin fish, although the various parameters tested (i.e., protein, moisture and ash contents, and total lipid, cholesterol and triglyceride levels) vary in the different parts of the body, head, arm, tentacle, and coat [110]. Additionally, chitosan and collagen of this cephalopod attracted keen research interest. The first component showed a number of therapeutically-interesting properties, i.e., anti-bacterial and anti-microbial, as well as anti-oxidant and anti-lipidemic activities, the latter being successfully applied to in vivo rat models of carbon tetrachloride-induced hepatic damage [111,112,113,114]. Ramasamy and Shanmugan [115] prepared a bioscaffold with collagen and chitosan extracted from S. kobiensis and tested it in an in vivo rat wound healing model. They found that the hybrid film determined an increased wound reduction and fibroblast proliferation and differentiation towards myofibroblasts, thereby determining the up-regulation of collagen deposition in the damaged tissue. Moreover, the presence of chitosan also guaranteed a free radical-scavenging action that protected the proliferating cytotypes, i.e., epithelial cells and fibroblasts, from the cytotoxic influence of reactive oxygen species (ROS). Thus, the merging of the beneficial effects of the two macromolecules makes this material a promising candidate as a dressing that supports a faster wound repair process.
Successful results were also achieved utilizing collagen preparations from the local cuttlefish Sepia officinalis (Linnaeus, 1758; Cephalopoda, Sepiida: Sepiidae; Figure 17), an invertebrate that is widely distributed in the Western Mediterranean Sea, which can be also found along the West African coasts. Collagen hydrogels were applied to the excised skin of female Wistar rats and proven to be a good quality dressing promoting tissue restoration to levels that are comparable to those of a commercial vegetable extract-based emulsion used as reference. In particular, S. officinalis’s collagen films induced a quick contraction and closure of the wound, with an absence of inflammation signs and, on the other hand, evidence of neo-vessel growth and active ECM deposition at the histological analysis [116]. Of note, collagen preparations of this cephalopod are widely used for diverse application, such as gelling agent, thickener, foaming agent, and emulsifier in food industries or natural bioactive filler promoting implant osteointegration [117,118].
Mytilus galloprovincialis (Lamarck, 1819; Bivalvia, Mytilida: Mytilidae; Figure 18A) is a widely distributed bivalve mollusk populating the Mediterranean and the Black Sea, the eastern Atlantic ocean from Morocco to the British Isles, but also present in Japan, California, South Africa, southern Australia, and New Zealand. It is a lamellibranch mollusk, which is, equipped with lamellar gills that absorb oxygen and simultaneously retain food for feeding. Therefore, being a filtering organism, it plays an indispensable role at ecological level, as marine purifier and it is widely used to evaluate the effects of stress environmental condition [119,120]. These invertebrates are endowed with an excellent nutritional value, owing to their remarkable richness in proteins, carbohydrates, essential vitamins, minerals, and different varieties of fatty acids that play beneficial roles for human health and, for this reason, they are in great demand on the international market [121,122,123]. Rapana venosa (Valenciennes, 1846; Gastropoda, Neogastropoda: Muricidae; Figure 18B) is a predatory benthic species of tropical environments typical of the Northwest Pacific area and introduced in the Atlantic, Mediterranean, and Black Sea [124,125]. It shows a variable color from grey to orange-brown or blonde, with darker brown dashes on the spiral ribs, and it is a widely consumed and commercially valuable mollusk in China. Recently, considerable interest has been paid to the concentrations of metals or pollutants, even radioactive, which R. venosa might contain, for an assessment of environmental monitoring and also the protection of human health, since this species is eatable and frequently consumed [126,127]. The anti-viral, anti-bacterial, and anti-cancer potentials of the hemocyanins obtained from the snail hemolymph have been extensively analyzed and demonstrated, thereby being promising components for formulations of anti-microbial and anti-tumoral vaccines [128,129,130]. Moreover, R. venosa’s shell was utilized to fabricate bionic coupling-layered ceramic/Al composites with improved lightweight and mechanical strength properties [131]. Being the gastropod a common predator of the mollusk M. galloprovincialis, Badiu and coworkers [132,133] examined in parallel the effect of lipid extracts and aminoacid preparations from M. galloprovincialis and R. venosa in in vivo wound healing tests.
Lipid extracts, containing slightly different complex mixtures with a high cholesterol content and good although varying amounts of several saturated and polyunsaturated fatty acids (PUFAs), including ω-3 eicosapentaenoic ((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17- pentaenoic acid, Figure 18C) and docosahexenoic acids, ((4Z,7Z,10Z,13Z,16Z,19Z)-docosa- 4,7,10,13,16,19-hexaenoic acid, Figure 18D) and, only for R.venosa, ω-6 arachidonic acid ((5Z,8Z,11Z,14Z)-icosa-5,8,11,14-tetraenoic acid, Figure 18E), were applied to skin burns in in vivo rat models in order to examine their potential healing activity. The results showed an acceleration of healing time when compared to a commercial fish oil-containing ointment used as reference, with complete re-epithelization and the deposition of dense ECM, re-vascularization, and absence of edema and inflammation Preparations from M. galloprovincialis exhibited a slightly faster recovery, thus suggesting the more active involvement of eicosapentanoic and docosahexenoic acids, which are abundant in its extract, in the wound healing process [132]. The same experimental study was also performed after application of aminoacid mixtures from protein extracts of M. galloprovincialis and R. venosa, which showed significant differences regarding aminoacidic distribution and content. Additionally, in this case, the correct formation of new epithelium and well-structured vessels, the improved collagen deposition and the reduced burn edema and leukocyte infiltration were observed [133]. This is likely due to the preponderance of thiol-containing aminoacids in the mixture, which exert anti-oxidant action preventing excessive lipid peroxidation and other aspects of the oxidative stress that are linked to the healing process. Of note, the damaged rat tissues healed faster when treated with R. venosa’s extracts, probably because of the beneficial effects of Leu, Lys, Thr, and Pro essential aminoacids, only present in these preparations.
Pinctada martensii (Dunker, 1880; Bivalvia, Ostreida: Margaritidae; Figure 19), also known as Pinctada imbricata (Röding, 1798), is a mollusk that is characteristic of benthic environments in the Atlantic and Indo-West Pacific areas and in the intertidal rocky regions of the Mumbai coast, to date listed among the most invasive species in the Mediterranean Sea [134]. It is one of the most important bivalves for the production of marine pearls all over the world and especially of the round «South China Sea pearls» that are typical of southern China. Despite the remarkable performance observed in the 90s [135], the rearing of this species subsequently decreased also due to environmental deterioration [136]. For this reason, this organism is commonly used to study the levels of environmental pollution, the effects of pollutants on the health of marine organisms, and the influence of various environmental stressors, such as changes in water temperature due to climate modification, on aquaculture yields. Proteins extracted from the meat and the nacre of this invertebrate have been shown to exert various functions, such as anti-microbial, anti-hypertensive, and osteogenetic [137,138,139]. Yang et al. [140] studied the effect of preparations of small molecular peptides from P. martensii’s mantle, i.e., the shell-producing organ, on skin wound repair in in vivo rat models. They demonstrated that the treatment was able to promote robust collagen deposition and re-epithelialization, thus accelerating wound healing, via the up-regulation of EGF, TGF-β1, and cyclin D1. The increase of CD31+ cytotypes in the tissue was indicative of enhanced neovascularization, whereas the up-regulation of IL-10 and the down-regulation of IL-6 was a sign of inhibition of the inflammatory response. A hypothetical explanation of the observed effect was based upon the high content in glutamic acid, glycine, cysteine, and phenylalanine of the preparations, which guarantee the nitrogen balance that is necessary for the continuous protein synthesis during angiogenesis, fibroblast growth, collagen deposition, and wound remodeling. In addition, Gln-Leu and Asp-Leu pairs, which are very common in the peptides, might act as radical scavengers, and Arg is known to promote collagen accumulation. Additionally, hydrophobic aminoacids, that improve immunity, accounted for more than half of the total aminoacids in the peptides. Interestingly, a similar ameliorated healing quality was obtained by the gastric administration of P. martensii’s mantle peptide preparations to mouse skin wound models [140].
Euchelus asper (Gmelin, 1791; Gastropoda, Seguenziida: Chilodontaidae; Figure 20) is an eatable and shell-oriented benthic mollusc that is typical of tropical areas and distributed in the Indo-West Pacific region, populating the west coast and south-east coast of India and the rocky beaches near the low tide points [141]. It is characterized by a thick and conoidal dull shell with brown, rosy, and black dots. Ether-soluble fractions of this organism were able to induce immunostimulated phagocytosis in vitro and immunosuppressive activity in vivo [142,143], whereas methanol extracts have been shown to exert an anti-osteoporotic effect in vivo, as indicated by the prominent anti-osteoclastogenic activity and reduction in bone loss induced by the lack of estrogen in a ovariectomised mice model [144]. Agrawal et al. [145] demonstrated that methanol preparations from E. asper displayed strong anti-angiogenic potential in CAM assays due to their inhibitory role on mesodermal blood vessel migration and compartmentalization resulted, at least in part, from a decreased ECM remodelling by MMPs. As a confirmation to this hypothesis, the methanol extracts were also found to down-regulate the activity of MMP-2 and -9 in A549 lung cancer cells and, since these enzymes are involved in the process of cell migration and invasion, A549 cell motility in scratch wound healing assays was consistently reduced.
Haliotis diversicolor (Reeve, 1846; Gastropoda, Lepetellida: Haliotidae; Figure 21) is a sea snail that can be found in temperate north-western Pacific Ocean off Japan, Taiwan, Vietnam, western Australia, Indonesian waters, and off New Caledonia [146]. Its coloration is very variable, normally reddish-brown, scarlet, and green in the external part, while the inner surface is silvery with light green and red reflections. It is one of the most important commercial species of molluscs, especially in the southern areas of China. Like most mollusks, H. diversicolor is widely used as a bioindicator for the study of the effects of environmental pollutants or other types of environmental stressors on marine organisms at the cellular and/or molecular level [147,148,149]. The shell of this species, also called “shijueming”, has been considered to be a traditional Chinese medicine used in the past for eye diseases and skin lesions. Xu et al. reported an effective action of this traditional medicine towards the survival rate of human lens epithelial cells damaged by condition of oxidative stress [150], demonstrating its antioxidant and anti-inflammatory properties. Based on those, more recently Chen et al. [151] tested the effect of H. diversicolor’s shell powder on inflammation and healing rates of burn injured rats. Their study revealed that the preparation induced a noticeable decrease in neutrophil infiltration, probably due to the down-regulation of nitric oxid synthase, and a significant improvement in the wound healing process with the formation of a well-arranged collagen matrix stimulated by the increase of TGF-β1 content during the tissue remodeling phase.
Euphausia superba (Dana, 1850; Malacostraca, Euphausiacea: Euphausiidae; Figure 22) is a pelagic krill that is distributed in the circumpolar belt between the Antarctic continent and the Polar Front [152]. This crustacean is considered to be an economic and ecological keystone [153] for its active involvement in carbon sequestration, iron and nitrogen recycling, ammonium absorption, and conversion of phytoplankton into animal proteins by connecting the different trophic levels [154,155]. The study of the cold adaptation mechanisms of E. superba at a transcriptome level has given interesting results that are useful for the design of new and improved recombinant enzyme variants that are endowed with adaptability to cold temperatures [156]. Much interest has been focused on the high nutritional content of this organism, due to the presence of proteins showing a higher biological value than milk or other animal counterparts, a remarkable amount of long-chain, in particular, ω-3, PUFAs, which makes it the second most popular source after fish oil, and all nine essential amino acids thus meeting the FAO/WHO requirements for human health [157,158]. For this reason, krill oil, which is rich in eicosapentaenoic and docosahexaenoic acids, vitamin A and E, and the carotenoid astaxanthin [159], is widely used as a food supplement also in light of its ability to reduce the risks of different pathologies, such as coronary heart diseases, diabetes, hypertension, osteoporosis, inflammation, autoimmune diseases, metabolic syndrome, tumors, and neurological diseases [160,161,162,163]. Costanzo et al. [164] have tested the ability of krill oil to ameliorate the state of intestinal inflammation in an in vitro model while using HT-29 and Caco-2 cell lines. In particular, when confluent cells, mimicking the intact barrier of the gut epithelium, were submitted to scratch wound healing assay in the presence of a mixture of the pro-inflammatory tumour necrosis factor-α and interferon-γ cytokines, the co-exposure to krill oil determined an increase of the rate of cell layer reconstitution. This result, complemented by the evidence of a better recovery of intercellular adhesion systems with a reduction of inflammation-related actin stress fibers, a decrease of cell death, and an impairment of adhesiveness and invasiveness of luminal bacteria on cells, strongly supports the value of krill oil as a prospective drug against human chronic intestinal inflammation. The beneficial effect of krill oil was supposed to be mostly based upon its richness in this lipid component due to the absence of ω-3 PUFAs in corn oil, used as a negative control for the study.

5. Echinodermata

The phylum Echinodermata, gathering invertebrates that display a radial symmetry, incorporates about 6000 living marine species around the globe. In recent years, echinoderms have attracted much attention as producers of biomolecules with promising anti-microbial, anti-protozoal, anti-fungal, and anticancer properties, making them potential candidates for drug development and future therapeutic applications [165,166,167,168]. Astropecten indicus (Döderlein, 1888; Asteroidea, Paxillosida: Astropectinidae; Figure 23), also called comb star or sand star, is a starfish that is distributed in the tropical environments of the Indo-Pacific area, and in particular of the United Arab Emirates, Qatar, Thailand, Gulf of Mannar, and Palk Bay region. It is a benthic echinoderm that is typically found in the sediments of the intertidal and subtidal regions, where it feeds using infaunal molluscs, and represents a prototypic marine organism to study the effects of environmental pollution [169,170]. Recently, Baveja et al. [171] isolated the AiP1 metalloproteinase from the coelomic fluid of A. indicus and demonstrated its fibrinolytic potential that is likely mediated by the von Willebrand factor (vWf)-like domain located in its central β sheet. Moreover, the ability of AiP1 to modulate the motile attitude of HaCaT keratinocytes and HEK293 embryonic kidney epithelial cells was checked through scratch wound healing assays demonstrating the additional AiP1-promoting effect on cell migration since vWf is known to sustain cell locomotory behavior. This result was confirmed by the evidence of the up-regulation of N-cadherin, indicative of EMT, in HEK293 cells. The wound healing activity exerted by AiP1, coupled with the increased cell proliferation reported in the same paper, prompt a more detailed study of the still unidentified mechanism of action of this component of A. indicus’s coelomic fluid to devise a novel treatment agent for tissue repair.
Himerometra magnipinna (AH Clark, 1908; Crinoidea, Comatulida: Himerometridae; Figure 24A), also known as Himerometra robustipinna (Carpenter, 1881) or red feather star, is a sessile comatulid that lives on hard corals and hard substrates in the Indian and Pacific Oceans, being abundant in the coastal areas of Vietnam and in Nha Trang Bay in the South China Sea [172]. Usually, it has reddish maroon arms and feeds with phytoplankton and zooplankton on detritus. Given that crinoids can regrow arms, internal organs, and entire viscera, the species of this order, including H. magnipinna, were the object of considerable studies on the cellular basis of the regeneration process with particular focus on the mechanisms of transdifferentiation [173,174,175,176,177]. A recent paper by Tseng et al. [178] focused on the effect of the H. magnipinna-derived anthraquinone (+)-rhodoptilometrin (1,3,8-trihydroxy-6-[(1S)-1-hydroxy propyl]anthracene-9,10- dione; Figure 24B) on the migratory behavior of gingival and oral mucosa fibroblasts through scratch wound healing and transwell migration assays. In particular, they found an increased number of migrating cells and improved wound repair rate in the presence of the compound, albeit only limited to gingival fibroblasts. This was coupled to the up-regulation of FAK, fibronectin, and type I collagen, selected as migration-associated ECM-related markers. In the same paper, increased cell viability and glycolysis/oxidative phosphorylation activity, with the latter known to sustain cell mobility functions, were found in rhodoptilometrin-treated human gingival cells, thereby strongly suggesting the potential of the molecule as a beneficial additive in formulations for dental health care and gingival recession treatment.
Stichopus japonicus (Selenka, 1867; Holothuroidea, Synallactida: Stichopodidae; Figure 25), also known as Apostichopus japonicus (Selenka, 1867) or Japanese sea cucumber, is a benthic species distributed in the Northwestern Pacific area. It may show three different colors, either red, green, or black, and it populates the shallow coasts from the intertidal zone to depths greater than 100 m. This echinoderm is one of the most important maricultured species in Korea, China, Japan, and several Southeast Asian countries, due to its high market value for human feeding [179]. It is widely recognized that most bioactive properties of sea cucumbers derive from components extracted from their body wall. For example, in the case of S. japonicus total extracts exhibited anti-oxidant characteristics, and isolated molecules, such as unsaturated fatty acids and a fucosylated chondroitin sulfate glycosaminoglycan, were proven to exert α-glucosidase inhibitory, immunostimulatory, anti-coagulant, and hypolipidemic activities [180,181,182,183,184]. Collagen, which is the major component of the body wall of S. japonicus [185], has been studied for its potential as an additive and functional food ingredient that is capable of maintaining food quality thanks to its gelling properties [186,187]. Park et al. [188] examined the effect of S. japonicus’s collagen on keratinocyte biology in vitro. When submitted to scratch wound healing assay, in the presence of this collagen preparation, the HaCaT cell migration rate was higher than that obtained in both bovine gelatin-coated and non-coated plates, being similar to that shown when the cells were seeded onto rat type I collagen substrates. This was accompanied by an up-regulation of fibronectin, as shown by RT-PCR, whose synthesis and extracellular deposition is a recognized fundamental event in the first steps of wound repair. In addition, S. japonicus’s collagen increased keratinocyte proliferation positively acting on cell entry in the mitotic phase. Thus, this sea cucumber appears as a valid alternative source with respect to other fish and mammalian species for the extraction of bulky amounts of collagen for biomedical applications, also in light of the documented ability of this collagen to improve skin barrier functions and inhibit melanogenesis, and to play anti-aging, anti-allergic, and anti-wrinkle roles [189,190,191].
Isostichopus badionotus (Selenka, 1867; Holothuroidea, Synallactida: Stichopodidae; Figure 26) is a resource of great economic importance in the south-east of the Gulf of Mexico, Central America and the Caribbean zone. In fact, being rich in fatty acids important for human health [192], it is used as a functional food capable of sustaining growth, improving the lipid profile, and reducing the levels of cholesterol, lipoproteins, and triglycerides in both serum and the liver where it was proven to down-regulate the expression of genes that are associated with dyslipidemia [193]. The most important molecules for biomedical purposes obtained from this sea cucumber, also responsible of the mentioned effects, are fucosylate chondroitin sulphate and fucoidan [194] whose additional properties as anti-coagulant, anti-thrombotic, and anti-malarial compounds have been acknowledged [195,196,197]. Li et al. [198] reported the ability of small molecule oligopeptides that were obtained from I. badionotus by enzymatic hydrolysis to induce the healing of wounds in a diabetic rat model in vivo after back skin section. In particular, in the peptide-treated individuals the wound status displayed a better histological appearance with a higher tensile strength, and the microscopic observations showed increased angiogenesis, collagen deposition, and epithelial reconstitution confirmed by the higher expression of vascular endothelial growth factor (VEGF) in the tissue and more elevated levels of NO and stromal cell-derived factor 1α, both being involved in the secretion of angiogenic factors, in animal sera. Moreover, the peptide treatment favoured the enhancement of antioxidant levels and the reduction of the pro-inflammatory interleukin-6 and -8, tumor necrosis factor-α, chemokine (C-C motif) ligand 2 and C-reactive protein, counteracted by the up-regulation of the anti-inflammatory interleukin-10. This indicates that the natural peptide mixture from I. badionotus sea cucumber might have promising applications for the treatment of diabetic wound healing, which still represents a significant clinical problem.
Ophiocoma erinaceus (Müller & Troschel, 1842; Ophiuroidea, Ophiacanthida: Ophiocomidae; Figure 27), also known as brittle star, is typical of tropical- and reef-associated environments and is distributed in the whole Indo-Pacific area (except for Pakistan and western India) being dominant in the Persian Gulf and in the coast of South Africa [172,199]. In the search of novel anti-angiogenic compounds, O. erinaceus’s alcoholic extracts, which were endowed with anti-oxidant properties, were administered to A2780cp ovarian tumor cells in culture and an in vivo CAM model, demonstrating a significant inhibition of cell growth and a down-regulation of formation of new blood vessels, coupled to the decrease of VEGF, TGF-β, and FGF-β mRNA expression in endothelial cells [200,201]. Among the components of the extract, an isolated polysaccharide and a saponin fraction were subjected to further studies. In fact, the first compound was found able to exert the previously-mentioned effects, and also decrease the viability of HUVEC cells, their expression of paxillin and MMP-9 mRNAs, whose products are involved in metastatization, and migration in scratch wound healing assays [202]. More recently, the brittle star polysaccharide was also proven to accelerate the wound healing in a male Wistar rat model in vivo [203]. On the other hand, the brittle star saponins appeared to be responsible for the radical scavenging function of the extract via increase of the SOD level in exposed cervical cancer HeLa cells. The survival rate of this cell line was also decreased due to the triggering of the intrinsic apoptotic pathway, differently from that found with non-tumoral fibroblasts. Moreover, saponins impaired cell locomotion in wound healing assay in vitro, parallelly reducing the adhesion of cells to matrigel substrates and inducing the down-regulation of MMP-2 and MMP-9 [204,205]. Of note, brittle star saponins also exhibited an anti-inflammatory ability both in in vitro assays and on macrophages that were differentiated from THP-1/M monocytes, as evidenced by the observed suppression of pro-inflammatory cytokines gene expression and production [206]. Thus, the active principles present in extracts from O. erinaceus appear to be promising candidates against human cervical carcinoma and in the prevention of angiogenesis-related disorders.
Stichopus herrmanni (Semper, 1868; Olothuroidea, Synallactida: Stichopodidae; Figure 28), also known as curryfish herrmanni, is a benthic species distributed in the Indo-West Pacific region, being dominant in tropical shallow waters, which represents an important fishery commodity. This orgamism, previously known as Stichopus variegatus in Indonesia and Malaysia, has found use for food consumption and preparation of traditional medicines in Asian countries, and for environmental monitoring, e.g., the quantification of marine pollution by polycyclic aromatic hydrocarbons [207,208]. The tegument of this invertebrate is known to contain high amounts of collagen and glycosaminoglycans that have been the object of researches. Biocomposites of hydroxyapatite and S. herrmannii’s collagen have been proven to increase the number of neo-formed osteoblasts during bone healing and regeneration, with no toxic side effects [209]. Zohdi et al. [210] prepared a S. hermannii’s extract-containing hydrogel dressing (“Gamat Hydrogel”), which, when applied to burn skin lesions in rats, improved wound contraction and closure, aided by a reduced inflammatory reaction, with signs of more efficient re-epithelization at the histological observation. The glycosaminoglycan component in S. herrmannii extracts was shown to play a role as a positive modulator of inflammation also in the healing process of oral mucous traumatic ulcers in rats [211]. More recently, Arundina et al. [212] demonstrated the stimulating effects of the extracts, which are also rich sources of soluble mediators, on the growth behavior and osteogenic differentiation of mesenchymal stem cells, thereby confirming that preparations from S. herrmannii may have beneficial effects on the acceleration of the wound healing and tissue regeneration processes.

6. Tunicata

Tunicates (subphylum Chordata) are marine organisms that are covered by a “tunic”, i.e., a tough body shield which mainly incorporates a highly crystalline cellulose nanofiber called tunicin [213,214], and proteins that contain 3,4,5-trihydroxyphenylalanine (TOPA) and pyrogallol amino acid [215,216]. Tunicates are known to be among the richest sources of biologically-active compounds together with sponges and bryozoans [217,218] and possess a very efficient mechanism of healing allowing for the repair of tunic injuries in sea water, a wet and salty environment similar to human body fluid [213]. In particular, at the pH of marine water (8.2) pyrogallol is oxidized and provides the key adhesion functionality forming covalent cross-links with tunicin, which has 120 gigapascals of stiffness and contributes to the mechanical support of the tunic, other TOPAs, or proteins [214]. These pyrogallol-mediated bonds favor the seal of the torn tunic and the control of bleeding. In light of such observations, a limited number of studies have been addressed to the identification of tunicate compounds active on cell migration in vitro and in vivo and tissue repair.
Phallusia arabica (Savigny, 1816; Ascidiacea, Phlebobranchia: Ascidiidae; Figure 29A) is a dark grey large solitary ascidian that populates the Red Sea, and the North Atlantic and South Pacific oceans. Ethanolic extracts of P. arabica exhibited a positive activity on the skin repair rate in excision rat models in vivo and a significant enhancement of the contraction in incision wounds with a faster re-epithelization and collagen deposition period [219]. It is supposed that the beneficial effect of the ascidian extract lies in the abundant presence of flavonoids, whose anti-inflammatory, anti-fungal, anti-oxidant, and wound healing properties are acknowledged, and the anti-oxidant n-hexadecanoic (palmitic) acid [220] (Figure 29B).
Phallusia nigra (Savigny, 1816; Ascidiacea, Phlebobranchia: Ascidiidae; Figure 30) is a velvet black large solitary ascidian, very common in the warm waters of the western Atlantic, ranging from Bermuda to Brazil, as well as in other seas like the Red Sea, Gulf of Aden, and Gulf of Guinea. It occurs as the major component of fouling communities on the hull of ships, piers, pilings and harbour installations. Also the methanolic extract of P. nigra exhibited a beneficial effect towards wound healing in in vivo rat models, similar to those that were obtained with P. arabica extracts, and also in this case the positive influence on tissue repair speed and quality was ascribed to the flavonoid and n-hexadecanoic acid components of the preparation [221,222].
Styela clava (Herdman, 1881; Ascidiacea, Stolidobranchia: Styelidae; Figure 31), also known as stalked sea squirt, is a solitary ascidian displaying a brown or yellow rough and wrinkled surface which is native of the Northwestern Pacific area and it has become a conspicuous member of fouling communities in Northwestern European waters. The cellulose films manufactured from S. clava’s tunic (SCT) have been the object of research [223]. These films exhibited good biocompatibility and degradability [224] and, when regeneration and angiogenesis in the surgical wounds of Sprague–Dawley rats were examined after the application of three different preparations of SCT-cellulose hydrogels, the one regenerated from freeze-dried membranes appeared to be a better dressing support when compared to regenerated plain and alginate-supplemented hydrogels [225]. This was due to the highest strength and lowest elongation level being exhibited by this preparation and the consequent amelioration of granulation tissue formation, angiogenesis, and skin reconstitution in the absence of toxic effects. At the molecular level, the reconstituted freeze-dried membranes appeared to up-regulate the levels of VEGF, collagen and TGF-β1, with activation of p-Smad 2/3 signalization, in the damaged tissue. More recently, Song et al. [226] studied the effect of selenium-loaded STC-cellulose films on cutaneous wounds of diabetic rats, showing, also in this experimental system, the acceleration of the healing process with the complete recovery of re-epithelization and skin repair following the activation of the downstream signaling pathways of VEGF, angiopoietin-2/1, and insulin, and the restoration of anti-oxidative status. These data demonstrate a potential use of the S. clava-derived material as a beneficial supplementary compound in formulations for topical application to cutaneous wounds.

7. Conclusions

An assortment of key events in development, health, and disease depends on collective cell motility which represents a common theme joining morphogenesis, tissue regeneration, and tumor biology [1]. It is widely acknowledged that several signal transduction and molecular mechanisms underlying and critical for cell migration and re-epithelization in wound repair are highly similar to those that are involved in epithelial tumor ingrowth and metastasis. As for medical treatments, it is known that, in some cases, anticancer agents may show interference with wound healing and, in turn, drugs used for non-healing ulcers of diabetic patients may promote carcinogenesis. Thus, there are many molecular and physiologic points in common between wound closure and tumor progression [227]. On the other hand, over the years, the literature showed the rapid rising of the prevalence, severity, and complexity of both cancer and wound healing pathologies, with the consequent remarkable increase in healthcare costs. It is imperative for modern society the procurement of alternative sources of bioactive compounds to replace overexploited resources and, on the other hand, improve treatments and the quality of life of patients via novel and more efficacious drugs. Marine invertebrates are a still poorly exploited valuable resource of natural products that may significantly improve the process of skin regeneration and restrain tumor cell migration, as documented by the in vitro and in vivo studies presented in this review and summarized in a synoptic way in Table 1. Only a very limited number of compounds and methods of extraction have been patented and/or introduced for treatment schemes to date, i.e., krill oil preparations [228], cellulose films from S. clava (patent KR20150117748A South Korea), S. herrmanni’s collagen for biocomposites [229], and S. japonicus’s fucosylated chondroitin sulfate (patent CN102367285A China). In conclusion, there is still an enormous potential offered by marine invertebrates towards a variety of applications that may significantly improve human health and the well-being of future generations. Therefore, the advancement of knowledge in this direction will result in new approaches that involve the identification of the most promising candidates for the utilization as new extracts/molecules, also in combination with existing drugs, which are targeted for biomedical translation.

Author Contributions

C.L., M.M., V.L. and M.V. collected the references; C.L. and M.V. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by The University of Palermo (Italy), grant Fondo Finalizzato alla Ricerca (FFR) 2018 to C.L. and M.V.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Li, L.; He, Y.; Zhao, M.; Jiang, J. Collective cell migration: Implications for wound healing and cancer invasion. Burns Trauma 2013, 1, 21–26. [Google Scholar] [CrossRef] [Green Version]
  2. Camley, B.A.; Rappel, W.-J. Physical models of collective cell motility: From cell to tissue. J. Phys. D Appl. Phys. 2017, 50, 113002. [Google Scholar] [CrossRef] [PubMed]
  3. Ladoux, B.; Mège, R.-M. Mechanobiology of collective cell behaviours. Nat. Rev. Mol. Cell Biol. 2017, 18, 743–757. [Google Scholar] [CrossRef] [PubMed]
  4. Eelen, G.; de Zeeuw, P.; Simons, M.; Carmeliet, P. Endothelial Cell Metabolism in Normal and Diseased Vasculature. Circ. Res. 2015, 116, 1231–1244. [Google Scholar] [CrossRef] [PubMed]
  5. Potente, M.; Gerhardt, H.; Carmeliet, P. Basic and Therapeutic Aspects of Angiogenesis. Cell 2011, 146, 873–887. [Google Scholar] [CrossRef] [Green Version]
  6. Shaw, T.J.; Martin, P. Wound repair at a glance. J. Cell Sci. 2009, 122, 3209–3213. [Google Scholar] [CrossRef] [Green Version]
  7. Eming, S.A.; Martin, P.; Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Translat. Med. 2014, 6, 265sr6. [Google Scholar] [CrossRef] [Green Version]
  8. Greaves, N.S.; Ashcroft, K.J.; Baguneid, M.; Bayat, A. Current understanding of molecular and cellular mechanisms in fibroplasia and angiogenesis during acute wound healing. J. Dermatol. Sci. 2013, 72, 206–217. [Google Scholar] [CrossRef]
  9. Arwert, E.N.; Hoste, E.; Watt, F.M. Epithelial stem cells, wound healing and cancer. Nat. Rev. Cancer 2012, 12, 170–180. [Google Scholar] [CrossRef]
  10. Plikus, M.V.; Gay, D.L.; Treffeisen, E.; Wang, A.; Supapannachart, R.J.; Cotsarelis, G. Epithelial stem cells and implications for wound repair. Semin. Cell Dev. Biol. 2012, 23, 946–953. [Google Scholar] [CrossRef] [Green Version]
  11. Stefonek-Puccinelli, T.J.; Masters, K.S. Regulation of cell signaling and function via changes in growth factor presentation. In Proceedings of the 2009 Annual International Conference of the IEEE Engineering in Medicine and Biology Society, Minneapolis, MN, USA, 3–6 September 2009; IEEE: Piscataway, NJ, USA, 2009; pp. 1167–1171. [Google Scholar]
  12. Hiroyasu, S.; Colburn, Z.T.; Jones, J.C.R. A hemidesmosomal protein regulates actin dynamics and traction forces in motile keratinocytes. FASEB J. 2016, 30, 2298–2310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kiwanuka, E.; Junker, J.P.; Eriksson, E. Transforming growth factor β1 regulates the expression of CCN2 in human keratinocytes via Smad-ERK signalling: TGF-β1-induced CCN2 expression in keratinocytes. Int. Wound J. 2017, 14, 1006–1018. [Google Scholar] [CrossRef] [PubMed]
  14. Boudra, R.; Ramsey, M.R. Understanding Transcriptional Networks Regulating Initiation of Cutaneous Wound Healing. Yale J. Biol. Med. 2020, 93, 161–173. [Google Scholar]
  15. Jonkman, J.E.N.; Cathcart, J.A.; Xu, F.; Bartolini, M.E.; Amon, J.E.; Stevens, K.M.; Colarusso, P. An introduction to the wound healing assay using live-cell microscopy. Cell Adhes. Migrat. 2014, 8, 440–451. [Google Scholar] [CrossRef] [Green Version]
  16. Albini, A.; Iwamoto, Y.; Kleinman, H.K.; Martin, G.R.; Aaronson, S.A.; Kozlowski, J.M.; McEwan, R.N. A rapid in vitro assay for quantitating the invasive potential of tumor cells. Cancer Res. 1987, 47, 3239–3245. [Google Scholar] [PubMed]
  17. Luparello, C. PTHrP regulates the expression of stress proteins in breast cancer cells inducing modifications in urokinase-plasminogen activator and MMP-1 expression. J. Cell Sci. 2003, 116, 2421–2430. [Google Scholar] [CrossRef] [Green Version]
  18. Luparello, C. Effect of Manganese Chloride and of Cotreatment with Cadmium Chloride on the In vitro Proliferative, Motile and Invasive Behavior of MDA-MB231 Breast Cancer Cells. Molecules 2019, 24, 1205. [Google Scholar] [CrossRef] [Green Version]
  19. Ribatti, D. The chick embryo chorioallantoic membrane (CAM) assay. Reprod. Toxicol. 2017, 70, 97–101. [Google Scholar] [CrossRef]
  20. Naik, M.; Brahma, P.; Dixit, M. A Cost-Effective and Efficient Chick Ex-Ovo CAM Assay Protocol to Assess Angiogenesis. Methods Protoc. 2018, 1, 19. [Google Scholar] [CrossRef] [Green Version]
  21. João De Masi, E.C.D.; Campos, A.C.L.; João De Masi, F.D.; Ratti, M.A.S.; Ike, I.S.; João De Masi, R.D. The influence of growth factors on skin wound healing in rats. Braz. J. Otorhinolaryngol. 2016, 82, 512–521. [Google Scholar] [CrossRef] [Green Version]
  22. Cai, E.Z.; Ang, C.H.; Raju, A.; Tan, K.B.; Hing, E.C.H.; Loo, Y.; Wong, Y.C.; Lee, H.; Lim, J.; Moochhala, S.M.; et al. Creation of Consistent Burn Wounds: A Rat Model. Arch. Plast. Surg. 2014, 41, 317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Weber, B.; Lackner, I.; Haffner-Luntzer, M.; Palmer, A.; Pressmar, J.; Scharffetter-Kochanek, K.; Knöll, B.; Schrezenemeier, H.; Relja, B.; Kalbitz, M. Modeling trauma in rats: Similarities to humans and potential pitfalls to consider. J. Transl. Med. 2019, 17, 305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Parnell, L.K.S.; Volk, S.W. The Evolution of Animal Models in Wound Healing Research: 1993–2017. Adv. Wound Care 2019, 8, 692–702. [Google Scholar] [CrossRef] [PubMed]
  25. Krafts, K.P. Tissue repair: The hidden drama. Organogenesis 2010, 6, 225–233. [Google Scholar] [CrossRef] [Green Version]
  26. Lindequist, U. Marine-Derived Pharmaceuticals-Challenges and Opportunities. Biomol. Ther. 2016, 24, 561–571. [Google Scholar] [CrossRef] [Green Version]
  27. Simpson, T.L. The Cell Biology of Sponges; Springer New York: New York, NY, USA, 1984; ISBN 978-1-4612-9740-6. [Google Scholar]
  28. Pozzolini, M.; Gallus, L.; Ghignone, S.; Ferrando, S.; Candiani, S.; Bozzo, M.; Bertolino, M.; Costa, G.; Bavestrello, G.; Scarfì, S. Insights into the evolution of metazoan regenerative mechanisms: Roles of TGF superfamily members in tissue regeneration of the marine sponge Chondrosia reniformis. J. Exp. Biol. 2019, 222. [Google Scholar] [CrossRef] [Green Version]
  29. Swatschek, D.; Schatton, W.; Kellermann, J.; Müller, W.E.G.; Kreuter, J. Marine sponge collagen: Isolation, characterization and effects on the skin parameters surface-pH, moisture and sebum. Eur. J. Pharm. Biopharm. 2002, 53, 107–113. [Google Scholar] [CrossRef]
  30. Kreuter, J.; Muller, W.; Swatschek, D.; Schatton, W.; Schatton, M.; Wolfgang S. Inc. Method for Isolating Sponge Collagen and Producing Nanoparticulate Collagen, and the Use. U.S. Patent 20030032601A1, 13 February 2003. [Google Scholar]
  31. Pozzolini, M.; Scarfì, S.; Gallus, L.; Castellano, M.; Vicini, S.; Cortese, K.; Gagliani, M.; Bertolino, M.; Costa, G.; Giovine, M. Production, Characterization and Biocompatibility Evaluation of Collagen Membranes Derived from Marine Sponge Chondrosia reniformis Nardo, 1847. Mar. Drugs 2018, 16, 111. [Google Scholar] [CrossRef] [Green Version]
  32. Fassini, D.; Duarte, A.R.; Reis, R.; Silva, T. Bioinspiring Chondrosia reniformis (Nardo, 1847) Collagen-Based Hydrogel: A New Extraction Method to Obtain a Sticky and Self-Healing Collagenous Material. Mar. Drugs 2017, 15, 380. [Google Scholar] [CrossRef] [Green Version]
  33. Pozzolini, M.; Millo, E.; Oliveri, C.; Mirata, S.; Salis, A.; Damonte, G.; Arkel, M.; Scarfì, S. Elicited ROS Scavenging Activity, Photoprotective, and Wound-Healing Properties of Collagen-Derived Peptides from the Marine Sponge Chondrosia reniformis. Mar. Drugs 2018, 16, 465. [Google Scholar] [CrossRef] [Green Version]
  34. Tsuda, M.; Ishibashi, M.; Agemi, K.; Sasaki, T.; Kobayashi, J. Stelliferins A–F, new antineoplastic isomalabaricane triterpenes from the Okinawan marine sponge Jaspis stellifera. Tetrahedron 1991, 47, 2181–2194. [Google Scholar] [CrossRef]
  35. Kobayashi, J.; Yuasa, K.; Kobayashi, T.; Sasaki, T.; Tsuda, M. Jaspiferals A ∼ G, new cytotoxic isomalabaricane-type nortriterpenoids from Okinawan marine sponge Jaspis stellifera. Tetrahedron 1996, 52, 5745–5750. [Google Scholar] [CrossRef]
  36. Tang, S.-A.; Zhou, Q.; Guo, W.-Z.; Qiu, Y.; Wang, R.; Jin, M.; Zhang, W.; Li, K.; Yamori, T.; Dan, S.; et al. In vitro Antitumor Activity of Stellettin B, a Triterpene from Marine Sponge Jaspis stellifera, on Human Glioblastoma Cancer SF295 Cells. Mar. Drugs 2014, 12, 4200–4213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Jin, D.-J.; Tang, S.-A.; Xing, G.-S.; Zhao, W.-J.; Zhao, C.; Duan, H.-Q.; Lin, W.-H. Jaspiferins C–F, four new isomalabaricane-type triterpenoids from the South China Sea sponge Jaspis stellifera. J. Asian Nat. Prod. Res. 2014, 16, 427–433. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, R.; Zhang, Q.; Peng, X.; Zhou, C.; Zhong, Y.; Chen, X.; Qiu, Y.; Jin, M.; Gong, M.; Kong, D. Stellettin B Induces G1 Arrest, Apoptosis and Autophagy in Human Non-small Cell Lung Cancer A549 Cells via Blocking PI3K/Akt/mTOR Pathway. Sci. Rep. 2016, 6, 27071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Xu, W.-G.; Wang, J.; Xing, G.-S.; Xu, J.-J.; Qiao, W.; Zhao, C.; Tang, S.-A. Jaspiferin G, a new isomalabaricane-type triterpenoid from the sponge Jaspis stellifera. Z. Nat. C 2016, 71, 111–114. [Google Scholar] [CrossRef]
  40. Xu, W.; Wang, J.; Qiao, W.; Zhao, C.; Tang, S. Jaspiferins H–J, New Isomalabaricane-Type Terpenoids from the South China Sea Marine Sponge Jaspis stellifera. Chem. Nat. Compd. 2018, 54, 84–87. [Google Scholar] [CrossRef]
  41. Chen, Y.; Zhou, Q.; Zhang, L.; Zhong, Y.; Fan, G.; Zhang, Z.; Wang, R.; Jin, M.; Qiu, Y.; Kong, D. Stellettin B induces apoptosis in human chronic myeloid leukemia cells via targeting PI3K and Stat5. Oncotarget 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  42. Cheng, S.-Y.; Chen, N.-F.; Lin, P.-Y.; Su, J.-H.; Chen, B.-H.; Kuo, H.-M.; Sung, C.-S.; Sung, P.-J.; Wen, Z.-H.; Chen, W.-F. Anti-Invasion and Antiangiogenic Effects of Stellettin B through Inhibition of the Akt/Girdin Signaling Pathway and VEGF in Glioblastoma Cells. Cancers 2019, 11, 220. [Google Scholar] [CrossRef] [Green Version]
  43. Gu, F.; Wang, L.; He, J.; Liu, X.; Zhang, H.; Li, W.; Fu, L.; Ma, Y. Girdin, an actin-binding protein, is critical for migration, adhesion, and invasion of human glioblastoma cells. J. Neurochem. 2014, 131, 457–469. [Google Scholar] [CrossRef]
  44. Kashman, Y.; Groweiss, A.; Shmueli, U. Latrunculin, a new 2-thiazolidinone macrolide from the marine sponge. Tetrahedron Lett. 1980, 21, 3629–3632. [Google Scholar] [CrossRef]
  45. Sabanay, I.; Tian, B.; Gabelt, B.T.; Geiger, B.; Kaufman, P.L. Latrunculin B effects on trabecular meshwork and corneal endothelial morphology in monkeys. Exp. Eye Res. 2006, 82, 236–246. [Google Scholar] [CrossRef] [PubMed]
  46. Lu, Y.-Y.; Lin, Y.-K.; Wen, Z.-H.; Chen, Y.-C.; Chen, S.-A.; Chen, Y.-J. Latrunculin B modulates electrophysiological characteristics and arrhythmogenesis in pulmonary vein cardiomyocytes. Clin. Sci. 2016, 130, 721–732. [Google Scholar] [CrossRef] [PubMed]
  47. El Sayed, K.A.; Youssef, D.T.A.; Marchetti, D. Bioactive Natural and Semisynthetic Latrunculins. J. Nat. Prod. 2006, 69, 219–223. [Google Scholar] [CrossRef] [PubMed]
  48. Sayed, K.A.E.; Khanfar, M.A.; Shallal, H.M.; Muralidharan, A.; Awate, B.; Youssef, D.T.A.; Liu, Y.; Zhou, Y.-D.; Nagle, D.G.; Shah, G. Latrunculin A and Its C-17-O-Carbamates Inhibit Prostate Tumor Cell Invasion and HIF-1 Activation in Breast Tumor Cells. J. Nat. Prod. 2008, 71, 396–402. [Google Scholar] [CrossRef] [Green Version]
  49. Li, M.; Wang, Y.X.; Luo, Y.; Zhao, J.; Li, Q.; Zhang, J.; Jiang, Y. Hypoxia inducible factor-1α-dependent epithelial to mesenchymal transition under hypoxic conditions in prostate cancer cells. Oncol. Rep. 2016, 36, 521–527. [Google Scholar] [CrossRef] [Green Version]
  50. Khanfar, M.A.; Youssef, D.T.A.; El Sayed, K.A. Semisynthetic Latrunculin Derivatives as Inhibitors of Metastatic Breast Cancer: Biological Evaluations, Preliminary Structure-Activity Relationship and Molecular Modeling Studies. ChemMedChem 2010, 5, 274–285. [Google Scholar] [CrossRef] [Green Version]
  51. Badr, J.M.; Shaala, L.A.; Abou-Shoer, M.I.; Tawfik, M.K.; Habib, A.-A.M. Bioactive Brominated Metabolites from the Red Sea Sponge Pseudoceratina arabica. J. Nat. Prod. 2008, 71, 1472–1474. [Google Scholar] [CrossRef]
  52. Shaala, L.; Asfour, H.; Youssef, D.; Żółtowska-Aksamitowska, S.; Wysokowski, M.; Tsurkan, M.; Galli, R.; Meissner, H.; Petrenko, I.; Tabachnick, K.; et al. New Source of 3D Chitin Scaffolds: The Red Sea Demosponge Pseudoceratina arabica (Pseudoceratinidae, Verongiida). Mar. Drugs 2019, 17, 92. [Google Scholar] [CrossRef] [Green Version]
  53. Chen, C.-L.; Kao, Y.-C.; Yang, P.-H.; Sung, P.-J.; Wen, Z.-H.; Chen, J.-J.; Huang, Y.-B.; Chen, P.-Y. A Small Dibromotyrosine Derivative Purified from Pseudoceratina Sp. Suppresses TGF-β Responsiveness by Inhibiting TGF-β Type I Receptor Serine/Threonine Kinase Activity: S PONGE -D ERIVED D IBROMOTYROSINE M ETABOLITE I NHIBITS TGF-β R ESPONSIVENESS. J. Cell. Biochem. 2016, 117, 2800–2814. [Google Scholar] [CrossRef]
  54. Shaala, L.; Youssef, D.; Sulaiman, M.; Behery, F.; Foudah, A.; Sayed, K. Subereamolline A as a Potent Breast Cancer Migration, Invasion and Proliferation Inhibitor and Bioactive Dibrominated Alkaloids from the Red Sea Sponge Pseudoceratina arabica. Mar. Drugs 2012, 10, 2492–2508. [Google Scholar] [CrossRef] [PubMed]
  55. Abou-Shoer, M.I.; Shaala, L.A.; Youssef, D.T.A.; Badr, J.M.; Habib, A.-A.M. Bioactive Brominated Metabolites from the Red Sea Sponge Suberea mollis. J. Nat. Prod. 2008, 71, 1464–1467. [Google Scholar] [CrossRef] [PubMed]
  56. Shaala, L.A.; Bamane, F.H.; Badr, J.M.; Youssef, D.T.A. Brominated Arginine-Derived Alkaloids from the Red Sea Sponge Suberea mollis. J. Nat. Prod. 2011, 74, 1517–1520. [Google Scholar] [CrossRef] [PubMed]
  57. Abbas, A.T.; El-Shitany, N.A.; Shaala, L.A.; Ali, S.S.; Azhar, E.I.; Abdel-dayem, U.A.; Youssef, D.T.A. Red Sea Suberea mollis Sponge Extract Protects against CCl 4 -Induced Acute Liver Injury in Rats via an Antioxidant Mechanism. Evid.-Based Compl. Altern. Med. 2014, 2014, 1–9. [Google Scholar] [CrossRef] [PubMed]
  58. Youssef, D.T.A.; Ibrahim, A.K.; Khalifa, S.I.; Mesbah, M.K.; Mayer, A.M.S.; van Soest, R.W.M. New anti-inflammatory sterols from the Red Sea sponges Scalarispongia aqabaensis and Callyspongia siphonella. Nat. Prod. Commun. 2010, 5, 27–31. [Google Scholar]
  59. Sobahi, T.R.A.; Ayyad, S.-E.N.; Abdel-Lateff, A.; Algandaby, M.M.; Alorfi, H.S.; Abdel-Naim, A.B. Cytotoxic metabolites from Callyspongia siphonella display antiproliferative activity by inducing apoptosis in HCT-116 cells. Phcog. Mag. 2017, 13, 37. [Google Scholar] [CrossRef]
  60. El-Hawary, S.S.; Sayed, A.M.; Mohammed, R.; Hassan, H.M.; Rateb, M.E.; Amin, E.; Mohammed, T.A.; El-Mesery, M.; Bin Muhsinah, A.; Alsayari, A.; et al. Bioactive Brominated Oxindole Alkaloids from the Red Sea Sponge Callyspongia siphonella. Mar. Drugs 2019, 17, 465. [Google Scholar] [CrossRef] [Green Version]
  61. Szakács, G.; Paterson, J.K.; Ludwig, J.A.; Booth-Genthe, C.; Gottesman, M.M. Targeting multidrug resistance in cancer. Nat. Rev. Drug Discov. 2006, 5, 219–234. [Google Scholar] [CrossRef]
  62. Foudah, A.I.; Jain, S.; Busnena, B.A.; El Sayed, K.A. Optimization of Marine Triterpene Sipholenols as Inhibitors of Breast Cancer Migration and Invasion. ChemMedChem 2013, 8, 497–510. [Google Scholar] [CrossRef]
  63. Ito, T.; Aimaiti, S.; Win, N.N.; Kodama, T.; Morita, H. New sesquiterpene lactones, vernonilides A and B, from the seeds of Vernonia anthelmintica in Uyghur and their antiproliferative activities. Bioorganic Med. Chem. Lett. 2016, 26, 3608–3611. [Google Scholar] [CrossRef]
  64. Akl, M.; Foudah, A.; Ebrahim, H.; Meyer, S.; Sayed, K. The Marine-Derived Sipholenol A-4-O-3′,4′-Dichlorobenzoate Inhibits Breast Cancer Growth and Motility in vitro and in vivo through the Suppression of Brk and FAK Signaling. Mar. Drugs 2014, 12, 2282–2304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kobayashi, M.; Kawazoe, K.; Kitagawa, I. Araguspongines B, C, D, E, F, G, H, and J, new vasodilative bis-1-oxaquinolizidine alkaloids from an Okinawan marine sponge, Xestospongia sp. Chem. Pharm. Bull. 1989, 37, 1676–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Dai, J.; Sorribas, A.; Yoshida, W.Y.; Kelly, M.; Williams, P.G. Xestosaprols from the Indonesian Marine Sponge Xestospongia sp. J. Nat. Prod. 2010, 73, 1188–1191. [Google Scholar] [CrossRef] [Green Version]
  67. Zhou, X.; Lu, Y.; Lin, X.; Yang, B.; Yang, X.; Liu, Y. Brominated aliphatic hydrocarbons and sterols from the sponge Xestospongia testudinaria with their bioactivities. Chem. Phys. Lipids 2011, 164, 703–706. [Google Scholar] [CrossRef]
  68. Ankisetty, S.; Slattery, M. Antibacterial Secondary Metabolites from the Cave Sponge Xestospongia sp. Mar. Drugs 2012, 10, 1037–1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Ecoy, G.A.U.; Chamni, S.; Suwanborirux, K.; Chanvorachote, P.; Chaotham, C. Jorunnamycin A from Xestospongia sp. Suppresses Epithelial to Mesenchymal Transition and Sensitizes Anoikis in Human Lung Cancer Cells. J. Nat. Prod. 2019, 82, 1861–1873. [Google Scholar] [CrossRef]
  70. Murtihapsari, M.; Salam, S.; Kurnia, D.; Darwati, D.; Kadarusman, K.; Abdullah, F.F.; Herlina, T.; Husna, M.H.; Awang, K.; Shiono, Y.; et al. A new antiplasmodial sterol from Indonesian marine sponge, Xestospongia sp. Nat. Prod. Res. 2019, 1–8. [Google Scholar] [CrossRef]
  71. Halim, H.; Chunhacha, P.; Suwanborirux, K.; Chanvorachote, P. Anticancer and antimetastatic activities of Renieramycin M, a marine tetrahydroisoquinoline alkaloid, in human non-small cell lung cancer cells. Anticancer Res. 2011, 31, 193–201. [Google Scholar]
  72. Satyanarayana, S.; Satyavati, D.; Rao, D.V. Hypoglycaemic activity of extracts from soft corals of Andaman and Nicobar coasts in rats. Indian J. Exp. Biol. 2000, 38, 180–181. [Google Scholar]
  73. Liang, C.-H.; Wang, G.-H.; Liaw, C.-C.; Lee, M.-F.; Wang, S.-H.; Cheng, D.-L.; Chou, T.-H. Extracts from Cladiella australis, Clavularia viridis and Klyxum simplex (Soft Corals) are Capable of Inhibiting the Growth of Human Oral Squamous Cell Carcinoma Cells. Mar. Drugs 2008, 6, 595–606. [Google Scholar] [CrossRef]
  74. Wu, S.-Y.; Wang, H.-M.; Wen, Y.-S.; Liu, W.; Li, P.-H.; Chiu, C.-C.; Chen, P.-C.; Huang, C.-Y.; Sheu, J.-H.; Wen, Z.-H. 4-(Phenylsulfanyl)butan-2-One Suppresses Melanin Synthesis and Melanosome Maturation In vitro and In vivo. IJMS 2015, 16, 20240–20257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Wen, Z.-H.; Chao, C.-H.; Wu, M.-H.; Sheu, J.-H. A neuroprotective sulfone of marine origin and the in vivo anti-inflammatory activity of an analogue. Eur. J. Med. Chem. 2010, 45, 5998–6004. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, S.-C.; Chien, Y.-C.; Pan, C.-H.; Sheu, J.-H.; Chen, C.-Y.; Wu, C.-H. Inhibitory Effect of Dihydroaustrasulfone Alcohol on the Migration of Human Non-Small Cell Lung Carcinoma A549 Cells and the Antitumor Effect on a Lewis Lung Carcinoma-Bearing Tumor Model in C57BL/6J Mice. Mar. Drugs 2014, 12, 196–213. [Google Scholar] [CrossRef] [PubMed]
  77. Chen, Y.-C.; Wen, Z.-H.; Lee, Y.-H.; Chen, C.-L.; Hung, H.-C.; Chen, C.-H.; Chen, W.-F.; Tsai, M.-C. Dihydroaustrasulfone Alcohol Inhibits PDGF-Induced Proliferation and Migration of Human Aortic Smooth Muscle Cells through Inhibition of the Cell Cycle. Mar. Drugs 2015, 13, 2390–2406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Li, P.-C.; Sheu, M.-J.; Ma, W.-F.; Pan, C.-H.; Sheu, J.-H.; Wu, C.-H. Anti-Restenotic Roles of Dihydroaustrasulfone Alcohol Involved in Inhibiting PDGF-BB-Stimulated Proliferation and Migration of Vascular Smooth Muscle Cells. Mar. Drugs 2015, 13, 3046–3060. [Google Scholar] [CrossRef] [Green Version]
  79. Hassan, H.M.; Khanfar, M.A.; Elnagar, A.Y.; Mohammed, R.; Shaala, L.A.; Youssef, D.T.A.; Hifnawy, M.S.; El Sayed, K.A. Pachycladins A−E, Prostate Cancer Invasion and Migration Inhibitory Eunicellin-Based Diterpenoids from the Red Sea Soft Coral Cladiella pachyclados. J. Nat. Prod. 2010, 73, 848–853. [Google Scholar] [CrossRef]
  80. Lin, W.-Y.; Chen, B.-W.; Huang, C.-Y.; Wen, Z.-H.; Sung, P.-J.; Su, J.-H.; Dai, C.-F.; Sheu, J.-H. Bioactive Cembranoids, Sarcocrassocolides P–R, from the Dongsha Atoll Soft Coral Sarcophyton crassocaule. Mar. Drugs 2014, 12, 840–850. [Google Scholar] [CrossRef]
  81. Su, C.-C.; Su, J.-H.; Lin, J.-J.; Chen, C.-C.; Hwang, W.-I.; Huang, H.H.; Wu, Y.-J. An Investigation into the Cytotoxic Effects of 13-Acetoxysarcocrassolide from the Soft Coral Sarcophyton crassocaule on Bladder Cancer Cells. Mar. Drugs 2011, 9, 2622–2642. [Google Scholar] [CrossRef]
  82. Su, C.-C.; Chen, J.; Din, Z.-H.; Su, J.-H.; Yang, Z.-Y.; Chen, Y.-J.; Wang, R.; Wu, Y.-J. 13-Acetoxysarcocrassolide Induces Apoptosis on Human Gastric Carcinoma Cells Through Mitochondria-Related Apoptotic Pathways: p38/JNK Activation and PI3K/AKT Suppression. Mar. Drugs 2014, 12, 5295–5315. [Google Scholar] [CrossRef] [Green Version]
  83. Liu, C.-I.; Chen, C.-C.; Chen, J.-C.; Su, J.-H.; Huang, H.H.; Chen, J.Y.-F.; Wu, Y.-J. Proteomic Analysis of Anti-Tumor Effects of 11-Dehydrosinulariolide on CAL-27 Cells. Mar. Drugs 2011, 9, 1254–1272. [Google Scholar] [CrossRef] [Green Version]
  84. Su, T.-R.; Lin, J.-J.; Chiu, C.-C.; Chen, J.Y.-F.; Su, J.-H.; Cheng, Z.-J.; Hwang, W.-I.; Huang, H.H.; Wu, Y.-J. Proteomic investigation of anti-tumor activities exerted by sinularin against A2058 melanoma cells: Proteomics and 2DE. Electrophoresis 2012, 33, 1139–1152. [Google Scholar] [CrossRef]
  85. Iguchi, K.; Fukaya, T.; Takahashi, H.; Watanabe, K. Stolonilactone, a novel terpenoid-related compound, isolated from the Okinawan soft coral Clavularia koellikeri. J. Org. Chem. 2004, 69, 4351–4355. [Google Scholar] [CrossRef] [PubMed]
  86. Iguchi, K.; Fukaya, T.; Yasumoto, A.; Watanabe, K. New Marine Sesquiterpenoids and Diterpenoids from the Okinawan Soft Coral Clavularia koellikeri. J. Nat. Prod. 2004, 67, 577–583. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, Q.; Hu, Z.; Luo, X.; Liu, J.; Li, G.; Cao, S.; Liu, Q. Clavukoellians A–F, Highly Rearranged Nardosinane Sesquiterpenoids with Antiangiogenic Activity from Clavularia koellikeri. J. Nat. Prod. 2019, 82, 1331–1337. [Google Scholar] [CrossRef]
  88. Båmstedt, U. Trophodynamics of the two scyphozoan jellyfishes, Aurelia aurita and Cyanea capillata, in western Norway. ICES J. Mar. Sci. 1994, 51, 369–382. [Google Scholar] [CrossRef]
  89. Holst, S.; Jarms, G. Effects of low salinity on settlement and strobilation of scyphozoa (Cnidaria): Is the lion’s mane Cyanea capillata (L.) able to reproduce in the brackish Baltic Sea? Hydrobiologia 2010, 645, 53–68. [Google Scholar] [CrossRef]
  90. Xiao, L.; Zhang, J.; Wang, Q.; He, Q.; Liu, S.; Li, Y.; Zhang, L. In vitro and in vivo haemolytic studies of tentacle-only extract from jellyfish Cyanea capillata. Toxicol. In Vitro 2010, 24, 1203–1207. [Google Scholar] [CrossRef]
  91. Doyle, T.; Headlam, J.; Wilcox, C.; MacLoughlin, E.; Yanagihara, A. Evaluation of Cyanea capillata Sting Management Protocols Using Ex Vivo and In vitro Envenomation Models. Toxins 2017, 9, 215. [Google Scholar] [CrossRef] [Green Version]
  92. Ruan, Z.; Liu, G.; Guo, Y.; Zhou, Y.; Wang, Q.; Chang, Y.; Wang, B.; Zheng, J.; Zhang, L. First Report of a Thioredoxin Homologue in Jellyfish: Molecular Cloning, Expression and Antioxidant Activity of CcTrx1 from Cyanea capillata. PLoS ONE 2014, 9, e97509. [Google Scholar] [CrossRef] [Green Version]
  93. Ruan, Z.; Liu, G.; Wang, B.; Zhou, Y.; Lu, J.; Wang, Q.; Zhao, J.; Zhang, L. First Report of a Peroxiredoxin Homologue in Jellyfish: Molecular Cloning, Expression and Functional Characterization of CcPrx4 from Cyanea capillata. Mar. Drugs 2014, 12, 214–231. [Google Scholar] [CrossRef]
  94. Wang, B.; Liu, G.; Wang, C.; Ruan, Z.; Wang, Q.; Wang, B.; Qiu, L.; Zou, S.; Zhang, X.; Zhang, L. Molecular cloning and functional characterization of a Cu/Zn superoxide dismutase from jellyfish Cyanea capillata. Int. J. Biol. Macromol. 2020, 144, 1–8. [Google Scholar] [CrossRef] [PubMed]
  95. Zhou, Y.; Liu, G.; Cheng, X.; Wang, Q.; Wang, B.; Wang, B.; Zhang, H.; He, Q.; Zhang, L. Antimicrobial activity of a newly identified Kazal-type serine proteinase inhibitor, CcKPI1, from the jellyfish Cyanea capillata. Int. J. Biol. Macromol. 2018, 107, 1945–1955. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, B.; Liu, D.; Wang, C.; Wang, Q.; Zhang, H.; Liu, G.; He, Q.; Zhang, L. Tentacle extract from the jellyfish Cyanea capillata increases proliferation and migration of human umbilical vein endothelial cells through the ERK1/2 signaling pathway. PLoS ONE 2017, 12, e0189920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Wang, C.; Wang, B.; Wang, B.; Wang, Q.; Liu, G.; Fan, C.; Zhang, L. A novel granulin homologue isolated from the jellyfish Cyanea capillata promotes proliferation and migration of human umbilical vein endothelial cells through the ERK1/2-signaling pathway. Int. J. Biol. Macromol. 2019, 135, 212–225. [Google Scholar] [CrossRef]
  98. Liu, C.; Chen, S.; Zhuang, Z.; Yan, J.; Liu, C.; Cui, H. Potential of utilizing jellyfish as food in culturing Pampus argenteus juveniles. Hydrobiologia 2015, 754, 189–200. [Google Scholar] [CrossRef]
  99. Zhu, S.; Ye, M.; Xu, J.; Guo, C.; Zheng, H.; Hu, J.; Chen, J.; Wang, Y.; Xu, S.; Yan, X. Lipid Profile in Different Parts of Edible Jellyfish Rhopilema esculentum. J. Agric. Food Chem. 2015, 63, 8283–8291. [Google Scholar] [CrossRef]
  100. Yu, H.; Liu, X.; Dong, X.; Li, C.; Xing, R.; Liu, S.; Li, P. Insecticidal activity of proteinous venom from tentacle of jellyfish Rhopilema esculentum Kishinouye. Bioorganic Med. Chem. Lett. 2005, 15, 4949–4952. [Google Scholar] [CrossRef]
  101. Yu, H.; Liu, X.; Xing, R.; Liu, S.; Li, C.; Li, P. Radical scavenging activity of protein from tentacles of jellyfish Rhopilema esculentum. Bioorganic Med. Chem. Lett. 2005, 15, 2659–2664. [Google Scholar] [CrossRef]
  102. Yu, H.; Liu, X.; Xing, R.; Liu, S.; Guo, Z.; Wang, P.; Li, C.; Li, P. In vitro determination of antioxidant activity of proteins from jellyfish Rhopilema esculentum. Food Chem. 2006, 95, 123–130. [Google Scholar] [CrossRef]
  103. Felician, F.F.; Yu, R.-H.; Li, M.-Z.; Li, C.-J.; Chen, H.-Q.; Jiang, Y.; Tang, T.; Qi, W.-Y.; Xu, H.-M. The wound healing potential of collagen peptides derived from the jellyfish Rhopilema esculentum. Chin. J. Traumatol. 2019, 22, 12–20. [Google Scholar] [CrossRef]
  104. Hoyer, B.; Bernhardt, A.; Lode, A.; Heinemann, S.; Sewing, J.; Klinger, M.; Notbohm, H.; Gelinsky, M. Jellyfish collagen scaffolds for cartilage tissue engineering. Acta Biomater. 2014, 10, 883–892. [Google Scholar] [CrossRef] [PubMed]
  105. Pustlauk, W.; Paul, B.; Brueggemeier, S.; Gelinsky, M.; Bernhardt, A. Modulation of chondrogenic differentiation of human mesenchymal stem cells in jellyfish collagen scaffolds by cell density and culture medium: Chondrogenic differentiation of hMSCs in jellyfish collagen scaffolds. J. Tissue Eng. Regen. Med. 2017, 11, 1710–1722. [Google Scholar] [CrossRef] [PubMed]
  106. Sewing, J.; Klinger, M.; Notbohm, H. Jellyfish collagen matrices conserve the chondrogenic phenotype in two- and three-dimensional collagen matrices: Jellyfish collagen matrices conserve the chondrogenic phenotype. J. Tissue Eng. Regen. Med. 2017, 11, 916–925. [Google Scholar] [CrossRef] [PubMed]
  107. Veeruraj, A.; Arumugam, M.; Ajithkumar, T.; Balasubramanian, T. Isolation and characterization of collagen from the outer skin of squid (Doryteuthis singhalensis). Food Hydrocoll. 2015, 43, 708–716. [Google Scholar] [CrossRef]
  108. Ramasamy, P.; Subhapradha, N.; Thinesh, T.; Selvin, J.; Selvan, K.M.; Shanmugam, V.; Shanmugam, A. Characterization of bioactive chitosan and sulfated chitosan from Doryteuthis singhalensis (Ortmann, 1891). Int. J. Biol. Macromol. 2017, 99, 682–691. [Google Scholar] [CrossRef]
  109. Veeruraj, A.; Liu, L.; Zheng, J.; Wu, J.; Arumugam, M. Evaluation of astaxanthin incorporated collagen film developed from the outer skin waste of squid Doryteuthis singhalensis for wound healing and tissue regenerative applications. Mater. Sci. Eng. C 2019, 95, 29–42. [Google Scholar] [CrossRef]
  110. Ramasamy, P.; Subhapradha, N.; Sudharsan, S.; Seedevi, P.; Shanmugam, V.; Shanmugam, A. Nutritional evaluation of the different body parts of cuttlefish Sepia kobiensis Hoyle, 1885. Afr. J. Food Sci. 2012, 6, 535–538. [Google Scholar] [CrossRef]
  111. Ramasamy, P.; Subhapradha, N.; Srinivasan, A.; Shanmugam, V.; Krishnamoorthy, J.; Shanmugam, A. In vitro evaluation of antimicrobial activity of methanolic extract from selected species of Cephalopods on clinical isolates. Afr. J. Microbiol. Res. 2011, 5, 3884–3889. [Google Scholar] [CrossRef] [Green Version]
  112. Ramasamy, P.; Subhapradha, N.; Shanmugam, V.; Shanmugam, A. Protective effect of chitosan from Sepia kobiensis (Hoyle 1885) cuttlebone against CCl4 induced hepatic injury. Int. J. Biol. Macromol. 2014, 65, 559–563. [Google Scholar] [CrossRef]
  113. Ramasamy, P.; Subhapradha, N.; Shanmugam, V.; Shanmugam, A. Extraction, characterization and antioxidant property of chitosan from cuttlebone Sepia kobiensis (Hoyle 1885). Int. J. Biol. Macromol. 2014, 64, 202–212. [Google Scholar] [CrossRef]
  114. Shanmugam, A.; Kathiresan, K.; Nayak, L. Preparation, characterization and antibacterial activity of chitosan and phosphorylated chitosan from cuttlebone of Sepia kobiensis (Hoyle, 1885). Biotechnol. Rep. 2016, 9, 25–30. [Google Scholar] [CrossRef] [Green Version]
  115. Ramasamy, P.; Shanmugam, A. Characterization and wound healing property of collagen–chitosan film from Sepia kobiensis (Hoyle, 1885). Int. J. Biol. Macromol. 2015, 74, 93–102. [Google Scholar] [CrossRef] [PubMed]
  116. Jridi, M.; Lassoued, I.; Kammoun, A.; Nasri, R.; Chaâbouni, M.; Nasri, M.; Souissi, N. Screening of factors influencing the extraction of gelatin from the skin of cuttlefish using supersaturated design. Food Bioprod. Proc. 2015, 94, 525–535. [Google Scholar] [CrossRef]
  117. García-Enriquez, S.; Guadarrama, H.E.R.; Reyes-González, I.; Mendizábal, E.; Jasso-Gastinel, C.F.; García-Enriquez, B.; Rembao-Bojórquez, D.; Pane-Pianese, C. Mechanical Performance and In vivo Tests of an Acrylic Bone Cement Filled with Bioactive Sepia Officinalis Cuttlebone. J. Biomater. Sci. Poly. Ed. 2010, 21, 113–125. [Google Scholar] [CrossRef] [PubMed]
  118. Kchaou, H.; Jridi, M.; Abdelhedi, O.; Nasreddine, B.; Karbowiak, T.; Nasri, M.; Debeaufort, F. Development and characterization of cuttlefish (Sepia officinalis) skin gelatin-protein isolate blend films. Int. J. Biol. Macromol. 2017, 105, 1491–1500. [Google Scholar] [CrossRef]
  119. Parisi, M.G.; Mauro, M.; Sarà, G.; Cammarata, M. Temperature increases, hypoxia, and changes in food availability affect immunological biomarkers in the marine mussel Mytilus galloprovincialis. J. Comp. Physiol. B 2017, 187, 1117–1126. [Google Scholar] [CrossRef]
  120. Vazzana, M.; Ceraulo, M.; Mauro, M.; Papale, E.; Dioguardi, M.; Mazzola, S.; Arizza, V.; Chiaramonte, M.; Buscaino, G. Effects of acoustic stimulation on biochemical parameters in the digestive gland of Mediterranean mussel Mytilus galloprovincialis (Lamarck, 1819). J. Acoust. Soc. Amer. 2020, 147, 2414–2422. [Google Scholar] [CrossRef]
  121. Pogoda, B.; Buck, B.H.; Saborowski, R.; Hagen, W. Biochemical and elemental composition of the offshore-cultivated oysters Ostrea edulis and Crassostrea gigas. Aquaculture 2013, 400–401, 53–60. [Google Scholar] [CrossRef] [Green Version]
  122. Prato, E.; Biandolino, F.; Parlapiano, I.; Giandomenico, S.; Denti, G.; Calò, M.; Spada, L.; Di Leo, A. Proximate, fatty acids and metals in edible marine bivalves from Italian market: Beneficial and risk for consumers health. Sci. Total Environ. 2019, 648, 153–163. [Google Scholar] [CrossRef]
  123. Prato, E.; Fanelli, G.; Parlapiano, I.; Biandolino, F. Bioactive fatty acids in seafood from Ionian Sea and relation to dietary recommendations. Int. J. Food Sci. Nutr. 2020, 1–13. [Google Scholar] [CrossRef]
  124. Chandler, E.A.; McDOWELL, J.R.; Graves, J.E. Genetically monomorphic invasive populations of the rapa whelk, Rapana venosa. Mol. Ecol. 2008, 17, 4079–4091. [Google Scholar] [CrossRef]
  125. Lanfranconi, A.; Hutton, M.; Brugnoli, E.; Muniz, P. New record of the alien mollusc Rapana venosa (Valenciennes 1846) in the Uruguayan coastal zone of Río de la Plata. Pan-Am. J. Aquat. Sci. 2009, 4, 216–221. [Google Scholar]
  126. Dalgic, G.; Kiris, E.; Baltas, H.; Sirin, M. Determination of radiological hazard parameters in sea snails (Rapana venosa) in the East Black Sea Coast of Turkey. Mar. Pollut. Bull. 2018, 135, 441–445. [Google Scholar] [CrossRef] [PubMed]
  127. Zhelyazkov, G.; Yankovska-Stefanova, T.; Mineva, E.; Stratev, D.; Vashin, I.; Dospatliev, L.; Valkova, E.; Popova, T. Risk assessment of some heavy metals in mussels (Mytilus galloprovincialis) and veined rapa whelks (Rapana venosa) for human health. Mar. Pollut. Bull. 2018, 128, 197–201. [Google Scholar] [CrossRef] [PubMed]
  128. Dolashka-Angelova, P.; Lieb, B.; Velkova, L.; Heilen, N.; Sandra, K.; Nikolaeva-Glomb, L.; Dolashki, A.; Galabov, A.S.; Beeumen, J.V.; Stevanovic, S.; et al. Identification of Glycosylated Sites in Rapana Hemocyanin by Mass Spectrometry and Gene Sequence, and Their Antiviral Effect. Bioconjugate Chem. 2009, 20, 1315–1322. [Google Scholar] [CrossRef] [PubMed]
  129. Boyanova, O.; Dolashka, P.; Toncheva, D.; Rammensee, H.-G.; Stevanović, S. In vitro effect of molluscan hemocyanins on CAL-29 and T-24 bladder cancer cell lines. Biomed. Rep. 2013, 1, 235–238. [Google Scholar] [CrossRef] [Green Version]
  130. Dolashka, P.; Dolashki, A.; Van Beeumen, J.; Floetenmeyer, M.; Velkova, L.; Stevanovic, S.; Voelter, W. Antimicrobial Activity of Molluscan Hemocyanins from Helix and Rapana Snails. CPB 2016, 17, 263–270. [Google Scholar] [CrossRef]
  131. Zhao, Q.; Liang, Y.; Liu, Q.; Zhang, Z.; Yu, Z.; Ren, L. Study on the Mechanical Properties of Bionic Coupling Layered B4C/5083Al Composite Materials. Materials 2018, 11, 680. [Google Scholar] [CrossRef] [Green Version]
  132. Badiu, D.L.; Balu, A.M.; Barbes, L.; Luque, R.; Nita, R.; Radu, M.; Tanase, E.; Rosoiu, N. Physico-Chemical Characterisation of Lipids from Mytilus galloprovincialis (L.) and Rapana venosa and their Healing Properties on Skin Burns. Lipids 2008, 43, 829–841. [Google Scholar] [CrossRef]
  133. Badiu, D.L.; Luque, R.; Dumitrescu, E.; Craciun, A.; Dinca, D. Amino Acids from Mytilus galloprovincialis (L.) and Rapana venosa Molluscs Accelerate Skin Wounds Healing via Enhancement of Dermal and Epidermal Neoformation. Protein J. 2010, 29, 81–92. [Google Scholar] [CrossRef]
  134. Gavrilović, A.; Piria, M.; Guo, X.-Z.; Jug-Dujaković, J.; Ljubučić, A.; Krkić, A.; Iveša, N.; Marshall, B.A.; Gardner, J.P.A. First evidence of establishment of the rayed pearl oyster, Pinctada imbricata radiata (Leach, 1814), in the eastern Adriatic Sea. Mar. Pollut. Bull. 2017, 125, 556–560. [Google Scholar] [CrossRef] [PubMed]
  135. Yang, C.; Wang, Q.; Hao, R.; Liao, Y.; Du, X.; Deng, Y. Effects of replacing microalgae with an artificial diet on pearl production traits and mineralization-related gene expression in pearl oyster Pinctada fucata martensii. Aquac. Res. 2017, 48, 5331–5337. [Google Scholar] [CrossRef]
  136. Qiu, Y.; Lu, H.; Zhu, J.; Chen, X.; Wang, A.; Wang, Y. Characterization of novel EST-SSR markers and their correlations with growth and nacreous secretion traits in the pearl oyster Pinctada martensii (Dunker). Aquaculture 2014, 420–421, S92–S97. [Google Scholar] [CrossRef]
  137. Xu, Z.-Y.; Liu, Y.-L.; Lin, J.-B.; Cheng, K.-L.; Wang, Y.-G.; Yao, H.-L.; Wei-Peng; Wu, H.-Y.; Su, W.-W.; Shaw, P.-C.; et al. Preparative expression and purification of a nacreous protein N16 and testing its effect on osteoporosis rat model. Int. J. Biol. Macromol. 2018, 111, 440–445. [Google Scholar] [CrossRef] [PubMed]
  138. Liu, P.; Lan, X.; Yaseen, M.; Wu, S.; Feng, X.; Zhou, L.; Sun, J.; Liao, A.; Liao, D.; Sun, L. Purification, Characterization and Evaluation of Inhibitory Mechanism of ACE Inhibitory Peptides from Pearl Oyster (Pinctada fucata martensii) Meat Protein Hydrolysate. Mar. Drugs 2019, 17, 463. [Google Scholar] [CrossRef] [Green Version]
  139. He, J.; Liang, H.; Zhu, J.; Fang, X. Separation, identification and gene expression analysis of PmAMP-1 from Pinctada fucata martensii. Fish. Shellfish Immunol. 2019, 92, 728–735. [Google Scholar] [CrossRef]
  140. Yang, F.; Qin, X.; Zhang, T.; Lin, H.; Zhang, C. Evaluation of Small Molecular Polypeptides from the Mantle of Pinctada Martensii on Promoting Skin Wound Healing in Mice. Molecules 2019, 24, 4231. [Google Scholar] [CrossRef] [Green Version]
  141. Apte, D. Field Guide to the Marine Life of India; Animesh Apte: Mumbai, India, 2012; ISBN 978-93-5067-144-3. [Google Scholar]
  142. Ponkshe, C.A.; Indap, M.M. In vivo and in vitro evaluation for immunomodulatory activity of three marine animal extracts with reference to phagocytosis. Indian J. Exp. Biol. 2002, 40, 1399–1402. [Google Scholar]
  143. Akerkar, A.S.; Ponkshe, C.A.; Indap, M.M. Evaluation of immunomodulatory activity of extracts from marine animals. Indian J. Mar. Sci. 2009, 38, 6. [Google Scholar]
  144. Balakrishnan, B.; Chiplunkar, S.V.; Indap, M.M. Methanol Extract of Euchelus asper Prevents Bone Resorption in Ovariectomised Mice Model. J. Osteoporos. 2014, 2014, 1–9. [Google Scholar] [CrossRef] [Green Version]
  145. Agrawal, S.; Chaugule, S.; More, S.; Rane, G.; Indap, M. Methanolic extract of Euchelus asper exhibits in-ovo anti-angiogenic and in vitro anti-proliferative activities. Biol. Res. 2017, 50, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Lee, Y.-H.; Vacquier, V.D. Evolution and systematics in Haliotidae (Mollusca: Gastropoda): Inferences from DNA sequences of sperm lysin. Mar. Biol. 1995, 124, 267–278. [Google Scholar] [CrossRef]
  147. Lu, J.; Feng, J.; Cai, S.; Chen, Z. Metabolomic responses of Haliotis diversicolor to organotin compounds. Chemosphere 2017, 168, 860–869. [Google Scholar] [CrossRef] [PubMed]
  148. Sun, Y.; Zhang, X.; Wang, Y.; Day, R.; Yang, H.; Zhang, Z. Immunity-related genes and signaling pathways under hypoxic stresses in Haliotis diversicolor: A transcriptome analysis. Sci. Rep. 2019, 9, 19741. [Google Scholar] [CrossRef] [PubMed]
  149. Fang, Z.; Sun, Y.; Zhang, X.; Wang, G.; Li, Y.; Wang, Y.; Zhang, Z. Responses of HSP70 Gene to Vibrio parahaemolyticus Infection and Thermal Stress and Its Transcriptional Regulation Analysis in Haliotis diversicolor. Molecules 2019, 24, 162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Xu, G.; Cui, L.; Lin, Y.; Wu, Y. Protective effect of haliotidis on the oxidative damage in the human lens epithelial cells. Zhonghua Yan Ke Za Zhi 2013, 49, 817–821. [Google Scholar]
  151. Chen, Z.-C.; Wu, S.-Y.S.; Su, W.-Y.; Lin, Y.-C.; Lee, Y.-H.; Wu, W.-H.; Chen, C.-H.; Wen, Z.-H. Anti-inflammatory and burn injury wound healing properties of the shell of Haliotis diversicolor. BMC Complement. Altern. Med. 2016, 16, 487. [Google Scholar] [CrossRef] [Green Version]
  152. Nicol, S.; Constable, A.J.; Pauly, T. Estimates of Circumpolar Abundance of Antarctic Krill Based on Recent Acoustic Density Measurements. CCAMLR Sci. 2000, 7. [Google Scholar]
  153. Liu, Z.-J.; Xu, L.-X.; Zhu, G.-P. Behavioral and physiological ecology of Antarctic krill (Euphausia superba): A review. Ying Yong Sheng Tai Xue Bao 2019, 30, 4344–4352. [Google Scholar] [CrossRef]
  154. Schmidt, K.; Atkinson, A.; Steigenberger, S.; Fielding, S.; Lindsay, M.C.M.; Pond, D.W.; Tarling, G.A.; Klevjer, T.A.; Allen, C.S.; Nicol, S.; et al. Seabed foraging by Antarctic krill: Implications for stock assessment, bentho-pelagic coupling, and the vertical transfer of iron. Limnol. Oceanogr. 2011, 56, 1411–1428. [Google Scholar] [CrossRef]
  155. Murphy, E.J.; Hofmann, E.E.; Watkins, J.L.; Johnston, N.M.; Piñones, A.; Ballerini, T.; Hill, S.L.; Trathan, P.N.; Tarling, G.A.; Cavanagh, R.A.; et al. Comparison of the structure and function of Southern Ocean regional ecosystems: The Antarctic Peninsula and South Georgia. J. Mar. Syst. 2013, 109–110, 22–42. [Google Scholar] [CrossRef]
  156. Zhou, T.; Wang, X.; Yan, J.; Li, Y. Gene analysis and structure prediction for the cold-adaption mechanism of trypsin from the krill Euphausia superba (Dana, 1852): Gene analysis and structure prediction for the cold-adaption mechanism. J. Sci. Food Agric. 2018. [Google Scholar] [CrossRef] [PubMed]
  157. Winther, B.; Hoem, N.; Berge, K.; Reubsaet, L. Elucidation of Phosphatidylcholine Composition in Krill Oil Extracted from Euphausia superba. Lipids 2011, 46, 25–36. [Google Scholar] [CrossRef] [Green Version]
  158. Backes, J.M.; Howard, P.A. Krill Oil for Cardiovascular Risk Prevention: Is it for Real? Hosp. Pharm. 2014, 49, 907–912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Ierna, M.; Kerr, A.; Scales, H.; Berge, K.; Griinari, M. Supplementation of diet with krill oil protects against experimental rheumatoid arthritis. BMC Musculoskelet. Disord. 2010, 11, 136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Kwantes, J.M.; Grundmann, O. A Brief Review of Krill Oil History, Research, and the Commercial Market. J. Diet. Suppl. 2015, 12, 23–35. [Google Scholar] [CrossRef] [PubMed]
  161. Choi, J.; Jang, J.; Son, D.; Im, H.-S.; Kim, J.; Park, J.; Choi, W.; Han, S.-B.; Hong, J. Antarctic Krill Oil Diet Protects against Lipopolysaccharide-Induced Oxidative Stress, Neuroinflammation and Cognitive Impairment. IJMS 2017, 18, 2554. [Google Scholar] [CrossRef] [Green Version]
  162. Ursoniu, S.; Sahebkar, A.; Serban, M.-C.; Antal, D.; Mikhailidis, D.P.; Cicero, A.; Athyros, V.; Rizzo, M.; Rysz, J.; Banach, M. Lipid-modifying effects of krill oil in humans: Systematic review and meta-analysis of randomized controlled trials. Nutr. Rev. 2017, 75, 361–373. [Google Scholar] [CrossRef]
  163. Mao, L.; Wang, F.; Li, Y.; Dai, Y.; Liu, Y.; Wang, J.; Xue, C. Oil from Antarctic krill (Euphausia superba) facilitates bone formation in dexamethasone-treated mice. Food Sci. Biotechnol. 2019, 28, 539–545. [Google Scholar] [CrossRef]
  164. Costanzo, M.; Cesi, V.; Prete, E.; Negroni, A.; Palone, F.; Cucchiara, S.; Oliva, S.; Leter, B.; Stronati, L. Krill oil reduces intestinal inflammation by improving epithelial integrity and impairing adherent-invasive Escherichia coli pathogenicity. Dig. Liver Dis. 2016, 48, 34–42. [Google Scholar] [CrossRef]
  165. Schillaci, D.; Cusimano, M.; Cunsolo, V.; Saletti, R.; Russo, D.; Vazzana, M.; Vitale, M.; Arizza, V. Immune mediators of sea-cucumber Holothuria tubulosa (Echinodermata) as source of novel antimicrobial and anti-staphylococcal biofilm agents. AMB Express 2013, 3, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Lazzara, V.; Arizza, V.; Luparello, C.; Mauro, M.; Vazzana, M. Bright Spots in The Darkness of Cancer: A Review of Starfishes-Derived Compounds and Their Anti-Tumor Action. Mar. Drugs 2019, 17, 617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Luparello, C.; Ragona, D.; Asaro, D.M.L.; Lazzara, V.; Affranchi, F.; Celi, M.; Arizza, V.; Vazzana, M. Cytotoxic Potential of the Coelomic Fluid Extracted from the Sea Cucumber Holothuria tubulosa against Triple-Negative MDA-MB231 Breast Cancer Cells. Biology 2019, 8, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Luparello, C.; Ragona, D.; Asaro, D.M.L.; Lazzara, V.; Affranchi, F.; Arizza, V.; Vazzana, M. Cell-Free Coelomic Fluid Extracts of the Sea Urchin Arbacia lixula Impair Mitochondrial Potential and Cell Cycle Distribution and Stimulate Reactive Oxygen Species Production and Autophagic Activity in Triple-Negative MDA-MB231 Breast Cancer Cells. JMSE 2020, 8, 261. [Google Scholar] [CrossRef] [Green Version]
  169. Coteur, G.; Gosselin, P.; Wantier, P.; Chambost-Manciet, Y.; Danis, B.; Pernet, P.; Warnau, M.; Dubois, P. Echinoderms as Bioindicators, Bioassays, and Impact Assessment Tools of Sediment-Associated Metals and PCBs in the North Sea. Arch. Environ. Contam. Toxicol. 2003, 45, 190–202. [Google Scholar] [CrossRef]
  170. Vazzana, M.; Mauro, M.; Ceraulo, M.; Dioguardi, M.; Papale, E.; Mazzola, S.; Arizza, V.; Beltrame, F.; Inguglia, L.; Buscaino, G. Underwater high frequency noise: Biological responses in sea urchin Arbacia lixula (Linnaeus, 1758). Comp. Biochem. Physiol. Part A Mol. Integr. Physiol. 2020, 242, 110650. [Google Scholar] [CrossRef]
  171. Baveja, M.; Sarkar, A.; Mondal, S.; Pathan, J.; Chakrabarty, D. AiP1, a protein from the coelomic fluid of sea star Astropecten indicus promotes wound healing and fibrinogenolysis in vitro. JoBAZ 2019, 80, 50. [Google Scholar] [CrossRef] [Green Version]
  172. Clark, A.M.; Rowe, F.W.E. Monograph of Shallow-Water Indo-West. Pacific Echinoderms; British Museum (Natural History): London, UK, 1971; ISBN 978-0-565-00690-7. [Google Scholar]
  173. Candia Carnevali, M. Regeneration in Echinoderms: Repair, regrowth, cloning. ISJ 2006, 3, 64–76. [Google Scholar]
  174. Mozzi, D.; Dolmatov, I.; Bonasoro, F.; Carnevali, M. Visceral regeneration in the crinoid Antedon mediterranea: Basic mechanisms, tissues and cells involved in gut regrowth. Open Life Sci. 2006, 1, 609–635. [Google Scholar] [CrossRef] [Green Version]
  175. Gahn, F.J.; Baumiller, T.K. Evolutionary History of Regeneration in Crinoids (Echinodermata). Integr. Comp. Biol. 2010, 50, 514. [Google Scholar] [CrossRef] [PubMed]
  176. Bobrovskaya, N.V.; Dolmatov, I.Y. Autotomy of the Visceral Mass in the Feather Star Himerometra robustipinna (Crinoidea, Comatulida). Biol. Bull. 2014, 226, 81–91. [Google Scholar] [CrossRef] [PubMed]
  177. Kalacheva, N.V.; Eliseikina, M.G.; Frolova, L.T.; Dolmatov, I.Y. Regeneration of the digestive system in the crinoid Himerometra robustipinna occurs by transdifferentiation of neurosecretory-like cells. PLoS ONE 2017, 12, e0182001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Tseng, C.-C.; Lai, Y.-C.; Kuo, T.-J.; Su, J.-H.; Sung, P.-J.; Feng, C.-W.; Lin, Y.-Y.; Chen, P.-C.; Tai, M.-H.; Cheng, S.-Y.; et al. Rhodoptilometrin, a Crinoid-Derived Anthraquinone, Induces Cell Regeneration by Promoting Wound Healing and Oxidative Phosphorylation in Human Gingival Fibroblast Cells. Mar. Drugs 2019, 17, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Xia, B.; Ren, Y.; Wang, F.; Yu, D.; Cui, G.; Chen, J. A comparative study on growth, protein turnover and energy budget of green and white color morphs of sea cucumber Apostichopus japonicus (Selenka). Aquacult. Environ. Interact. 2017, 9, 405–414. [Google Scholar] [CrossRef] [Green Version]
  180. Husni, A.; Shin, I.-S.; You, S.; Chung, D. Antioxidant Properties of Water and Aqueous Ethanol Extracts and Their Crude Saponin Fractions from a Far-eastern Sea Cucumber. Food Sci. Biotechnol. 2009, 18, 419–424. [Google Scholar]
  181. Nguyen, T.H.; Um, B.H.; Kim, S.M. Two Unsaturated Fatty Acids with Potent α-Glucosidase Inhibitory Activity Purified from the Body Wall of Sea Cucumber (Stichopus japonicus). J. Food Sci. 2011, 76, H208–H214. [Google Scholar] [CrossRef]
  182. Guan, R.; Peng, Y.; Zhou, L.; Zheng, W.; Liu, X.; Wang, P.; Yuan, Q.; Gao, N.; Zhao, L.; Zhao, J. Precise Structure and Anticoagulant Activity of Fucosylated Glycosaminoglycan from Apostichopus japonicus: Analysis of Its Depolymerized Fragments. Mar. Drugs 2019, 17, 195. [Google Scholar] [CrossRef] [Green Version]
  183. Wang, H.; Xu, L.; Yu, M.; Wang, Y.; Jiang, T.; Yang, S.; Lv, Z. Glycosaminoglycan from Apostichopus japonicus induces immunomodulatory activity in cyclophosphamide-treated mice and in macrophages. Int. J. Biol. Macromol. 2019, 130, 229–237. [Google Scholar] [CrossRef]
  184. Zhu, Z.; Dong, X.; Yan, C.; Ai, C.; Zhou, D.; Yang, J.; Zhang, H.; Liu, X.; Song, S.; Xiao, H.; et al. Structural Features and Digestive Behavior of Fucosylated Chondroitin Sulfate from Sea Cucumbers Stichopus japonicus. J. Agric. Food Chem. 2019, 67, 10534–10542. [Google Scholar] [CrossRef]
  185. Saito, M.; Kunisaki, N.; Urano, N.; Kimura, S. Collagen as the Major Edible Component of Sea Cucumber (Stichopus japonicus). J. Food Sci. 2002, 67, 1319–1322. [Google Scholar] [CrossRef]
  186. Bordbar, S.; Anwar, F.; Saari, N. High-Value Components and Bioactives from Sea Cucumbers for Functional Foods—A Review. Mar. Drugs 2011, 9, 1761–1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Zhang, X.; Sun, L.; Yuan, J.; Sun, Y.; Gao, Y.; Zhang, L.; Li, S.; Dai, H.; Hamel, J.-F.; Liu, C.; et al. The sea cucumber genome provides insights into morphological evolution and visceral regeneration. PLoS Biol. 2017, 15, e2003790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Park, S.-Y.; Lim, H.K.; Lee, S.; Hwang, H.C.; Cho, S.K.; Cho, M. Pepsin-solubilised collagen (PSC) from Red Sea cucumber (Stichopus japonicus) regulates cell cycle and the fibronectin synthesis in HaCaT cell migration. Food Chem. 2012, 132, 487–492. [Google Scholar] [CrossRef] [PubMed]
  189. Duan, J.; Ishida, M.; Aida, K.; Tsuduki, T.; Zhang, J.; Manabe, Y.; Hirata, T.; Sugawara, T. Dietary Cerebroside from Sea Cucumber (Stichopus japonicus): Absorption and Effects on Skin Barrier and Cecal Short-Chain Fatty Acids. J. Agric. Food Chem. 2016, 64, 7014–7021. [Google Scholar] [CrossRef] [PubMed]
  190. Mou, J.; Li, Q.; Qi, X.; Yang, J. Structural comparison, antioxidant and anti-inflammatory properties of fucosylated chondroitin sulfate of three edible sea cucumbers. Carbohyd. Poly. 2018, 185, 41–47. [Google Scholar] [CrossRef]
  191. Kwon, T.-R.; Oh, C.T.; Bak, D.-H.; Kim, J.H.; Seok, J.; Lee, J.H.; Lim, S.H.; Yoo, K.H.; Kim, B.J.; Kim, H. Effects on skin of Stichopus japonicus viscera extracts detected with saponin including Holothurin A: Down-regulation of melanin synthesis and up-regulation of neocollagenesis mediated by ERK signaling pathway. J. Ethnopharmacol. 2018, 226, 73–81. [Google Scholar] [CrossRef]
  192. Pastrana, O.J.; Santafé, G.G.; Torres, O.L. Perfil de Ácidos Grasos y Evaluación de las Actividades Antioxidante y Antifúngica del Holotureo Isostichopus badionotus. Inf. Tecnol. 2016, 27, 03–10. [Google Scholar] [CrossRef]
  193. Olivera-Castillo, L.; Davalos, A.; Grant, G.; Valadez-Gonzalez, N.; Montero, J.; Barrera-Perez, H.A.M.; Chim-Chi, Y.; Olvera-Novoa, M.A.; Ceja-Moreno, V.; Acereto-Escoffie, P.; et al. Diets Containing Sea Cucumber (Isostichopus badionotus) Meals Are Hypocholesterolemic in Young Rats. PLoS ONE 2013, 8, e79446. [Google Scholar] [CrossRef] [Green Version]
  194. Chen, S.; Xue, C.; Yin, L.; Tang, Q.; Yu, G.; Chai, W. Comparison of structures and anticoagulant activities of fucosylated chondroitin sulfates from different sea cucumbers. Carbohydr. Poly. 2011, 83, 688–696. [Google Scholar] [CrossRef]
  195. Chen, S.; Hu, Y.; Ye, X.; Li, G.; Yu, G.; Xue, C.; Chai, W. Sequence determination and anticoagulant and antithrombotic activities of a novel sulfated fucan isolated from the sea cucumber Isostichopus badionotus. Biochim. Biophys. Acta (BBA) Gen. Subj. 2012, 1820, 989–1000. [Google Scholar] [CrossRef]
  196. Li, J.; Li, S.; Zhi, Z.; Yan, L.; Ye, X.; Ding, T.; Yan, L.; Linhardt, R.; Chen, S. Depolymerization of Fucosylated Chondroitin Sulfate with a Modified Fenton-System and Anticoagulant Activity of the Resulting Fragments. Mar. Drugs 2016, 14, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Marques, J.; Vilanova, E.; Mourão, P.A.S.; Fernàndez-Busquets, X. Marine organism sulfated polysaccharides exhibiting significant antimalarial activity and inhibition of red blood cell invasion by Plasmodium. Sci. Rep. 2016, 6, 24368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Li, S.; Li, J.; Mao, G.; Hu, Y.; Ye, X.; Tian, D.; Linhardt, R.J.; Chen, S. Fucosylated chondroitin sulfate oligosaccharides from Isostichopus badionotus regulates lipid disorder in C57BL/6 mice fed a high-fat diet. Carbohydr. Polym. 2018, 201, 634–642. [Google Scholar] [CrossRef] [PubMed]
  199. Clark, A.M.; Courtman-Stock, J. The Echinoderms of Southern Africa; British Museum (Natural History): London, UK, 1976; ISBN 978-0-565-00776-8. [Google Scholar]
  200. Baharara, J.; Amini, E. Phytochemical Screening, Antioxidant Effect and Down Regulation of TGF-β Induced by Ophiocoma erinaceus Brittle Star Crude Extract. Zahedan J. Res. Med. Sci. 2015, 17. [Google Scholar] [CrossRef]
  201. Kachooei, S.A.; Rahmani, R.; Zareh, N.; Donyadideh, F.; Kachooei, S.A.; Nabiuni, M.; Yazdansetad, S. Down-regulation of TGF-β, VEGF, and bFGF in vascular endothelial cells of chicken induced by a brittle star (Ophiocoma erinaceus) extract. Heliyon 2020, 6, e03199. [Google Scholar] [CrossRef] [Green Version]
  202. Baharara, J.; Amini, E.; Musavi, M. Anti-Vasculogenic Activity of a Polysaccharide Derived from Brittle Star via Inhibition of VEGF, Paxillin and MMP-9. Iran. J. Biotechnol. 2017, 15, 179–185. [Google Scholar] [CrossRef] [Green Version]
  203. Abolhasani, I.; Baharara, J.; Mahdavi Shahri, N.; Amini, E. The regenerative properties of the extracted polysaccharide from Brittle star (Ophiocoma erinaceus) on cutaneous wound in male Wistar rat. nbr 2020, 7, 9–18. [Google Scholar]
  204. Amini, E.; Nabiuni, M.; Baharara, J.; Parivar, K.; Asili, J. Metastatic Inhibitory and Radical Scavenging Efficacies of Saponins Extracted from the Brittle Star (Ophiocoma erinaceus). Asian Pac. J. Cancer Prev. 2015, 16, 4751–4758. [Google Scholar] [CrossRef] [Green Version]
  205. Amini, E.; Nabiuni, M.; Baharara, J.; Parivar, K.; Asili, J. In-vitro Pro Apoptotic Effect of Crude Saponin from Ophiocoma erinaceus against Cervical Cancer. Iran. J. Pharm. Res. 2017, 16, 266–276. [Google Scholar]
  206. Baharara, J.; Amini, E.; Salek-Abdollahi, F. Anti-inflammatory properties of saponin fraction from Ophiocoma erinaceus. Iran. J. Fish. Sci. 2019. [Google Scholar] [CrossRef]
  207. Khazaali, A.; Kunzmann, A.; Bastami, K.D.; Baniamam, M. Baseline of polycyclic aromatic hydrocarbons in the surface sediment and sea cucumbers (Holothuria leucospilota and Stichopus hermanni) in the northern parts of Persian Gulf. Mar. Pollut. Bull. 2016, 110, 539–545. [Google Scholar] [CrossRef] [PubMed]
  208. Pangestuti, R.; Arifin, Z. Medicinal and health benefit effects of functional sea cucumbers. J. Tradit. Complement. Med. 2018, 8, 341–351. [Google Scholar] [CrossRef] [PubMed]
  209. Wahyuningtyas, E.; Sugiatno, E. Stichopus Hermanni Collagen with Local Hydroxyapatite as Bone Substitute Material Toward Osteoclast Number and Toxicity. In Proceedings of the 2018 1st International Conference on Bioinformatics, Biotechnology, and Biomedical Engineering Bioinformatics and Biomedical Engineering, Yogyakarta, Indonesia, 19–20 October 2018 ; IEEE: Piscataway, NJ, USA, 2018; pp. 1–4. [Google Scholar]
  210. Zohdi, R.M.; Zakaria, Z.A.B.; Yusof, N.; Mustapha, N.M.; Abdullah, M.N.H. Sea cucumber (Stichopus hermanii) based hydrogel to treat burn wounds in rats. J. Biomed. Mater. Res. 2011, 98B, 30–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Arundina, I.; Yuliati, Y.; Soesilawati, P.; Damaiyanti, D.W.; Maharani, D. The effects of golden sea cucumber extract (Stichopus hermanii) on the number of lymphocytes during the healing process of traumatic ulcer on wistar rat’s oral mucous. Dent. J. (Majalah Kedokteran Gigi) 2015, 48, 100. [Google Scholar] [CrossRef] [Green Version]
  212. Arundina, I.; Suardita, K.; Setiabudi, H.; Dwi Ariani, M. Golden Sea Cucumbers (Stichopus Hermanii) as Growth Factors of Stem Cells. J. Int. Dental Med. Res. 2016, 9, 242–248. [Google Scholar]
  213. Kimura, S.; Ohshima, C.; Hirose, E.; Nishikawa, J.; Itoh, T. Cellulose in the house of the appendicularian Oikopleura rufescens. Protoplasma 2001, 216, 71–74. [Google Scholar] [CrossRef]
  214. Capadona, J.R.; Shanmuganathan, K.; Tyler, D.J.; Rowan, S.J.; Weder, C. Stimuli-Responsive Polymer Nanocomposites Inspired by the Sea Cucumber Dermis. Science 2008, 319, 1370–1374. [Google Scholar] [CrossRef]
  215. Kime-Hunt, E.; Spartalian, K.; Holmes, S.; Mohan, M.; Carrano, C.J. Vanadium metabolism in tunicates: The coordination chemistry of V(III), V(IV), and V(V) with models for the tunichromes. J. Inorg. Biochem. 1991, 41, 125–141. [Google Scholar] [CrossRef]
  216. Taylor, S.W.; Ross, M.M.; Waite, J.H. Novel 3,4-Di- and 3,4,5-Trihydroxyphenylalanine-Containing Polypeptides from the Blood Cells of the Ascidians Ascidia ceratodes and Molgula manhattensis. Arch. Biochem. Biophy. 1995, 324, 228–240. [Google Scholar] [CrossRef]
  217. Davis, R.A.; Carroll, A.R.; Quinn, R.J. Longithorones J and K, Two New Cyclofarnesylated Quinone Derived Metabolites from the Australian Ascidian Aplidium longithorax. J. Nat. Prod. 1999, 62, 158–160. [Google Scholar] [CrossRef]
  218. Mauro, M.; Lazzara, V.; Punginelli, D.; Arizza, V.; Vazzana, M. Antitumoral compounds from vertebrate sister group: A review of Mediterranean ascidians. Dev. Comp. Immunol. 2020, 108, 103669. [Google Scholar] [CrossRef] [PubMed]
  219. Christy, H.K.S.; Margret, R.J.; Meenakshi, K. Evaluation of wound healing activity of Phallusia Arabica. World J. Pharm. Res. 2015, 4, 13. [Google Scholar]
  220. Christy, H.K.S.; Margret, R.J.; Meenakshi, V.K. Infrared and gas chromatogram-mass spectral studies of the ethanolic extract of Phallusia arabica Savigny, 1816. Arch. Appl. Sci. Res. 2013, 5, 17–23. [Google Scholar]
  221. Gopalakrishnan, S.; Meenakshi, V.K.; Shanmuga Priya, D. Chemical Investigation Of The Simple Ascidian Phallusia Nigra Savigny, 1816 Of Tuticorin Coast By Gc-Ms. Int. J. Pharm. Bio. Sci. 2011, 2, 382–387. [Google Scholar]
  222. Gopalakrishnan, S.B.; Priya, D.S.; Meenakshi, V. Wound healing activity of the methanolic extract of Phallusia nigra Sav. Int. J. Chem. Pharm. Sci. 2012, 3, 8. [Google Scholar]
  223. Jung, Y.J. Properties of Regenerated Cellulose Films Prepared from the Tunicate Styela clava. Korean J. Fish. Aquat. Sci. 2008, 41, 237–242. [Google Scholar] [CrossRef]
  224. Song, S.H.; Kim, J.E.; Lee, Y.J.; Kwak, M.H.; Sung, G.Y.; Kwon, S.H.; Son, H.J.; Lee, H.S.; Jung, Y.J.; Hwang, D.Y. Cellulose film regenerated from Styela clava tunics have biodegradability, toxicity and biocompatibility in the skin of SD rats. J. Mater. Sci. Mater. Med. 2014, 25, 1519–1530. [Google Scholar] [CrossRef]
  225. Song, S.H.; Seong, K.Y.; Kim, J.E.; Go, J.; Koh, E.K.; Sung, J.E.; Son, H.J.; Jung, Y.J.; Kim, H.S.; Hong, J.T.; et al. Effects of different cellulose membranes regenerated from Styela clava tunics on wound healing. Int. J. Mol. Med. 2017, 39, 1173–1187. [Google Scholar] [CrossRef] [Green Version]
  226. Song, S.H.; Kim, J.E.; Koh, E.K.; Sung, J.E.; Lee, H.A.; Yun, W.B.; Hong, J.T.; Hwang, D.Y. Selenium-loaded cellulose film derived from Styela clava tunic accelerates the healing process of cutaneous wounds in streptozotocin-induced diabetic Sprague–Dawley rats. J. Dermatol. Treat. 2018, 29, 606–616. [Google Scholar] [CrossRef]
  227. Sundaram, G.M.; Quah, S.; Sampath, P. Cancer: The dark side of wound healing. FEBS J. 2018, 285, 4516–4534. [Google Scholar] [CrossRef]
  228. Xie, D.; Gong, M.; Wei, W.; Jin, J.; Wang, X.; Wang, X.; Jin, Q. Antarctic Krill (Euphausia superba) Oil: A Comprehensive Review of Chemical Composition, Extraction Technologies, Health Benefits, and Current Applications: Antarctic krill (Euphausia superba) oil. Comprehen. Rev. Food Sci. Food Saf. 2019, 18, 514–534. [Google Scholar] [CrossRef] [Green Version]
  229. Wahyuningtyas, E.; Hsu, L.-C.; Lan, W.-C.; Wen, S.-C.; Ou, K.-L.; Chou, H.-H.; Huang, M.-S.; Sugiatno, E. Application of a Promising Bone Graft Substitute in Bone Tissue Regeneration: Characterization, Biocompatibility, and In vivo Animal Study. BioMed. Res. Int. 2019, 2019, 1–7. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) Phase-contrast microscopic image of a scratch created in a monolayer of HEPG2 liver carcinoma cells. (B) Light microscopic image of MDA-MB231 breast carcinoma cells migrated through the pores of a polycarbonate filter in a Boyden chamber assay, fixed with 95% ethanol, and stained with 0.02 g toluidine blue/mL. Bar = 10 μm.
Figure 1. (A) Phase-contrast microscopic image of a scratch created in a monolayer of HEPG2 liver carcinoma cells. (B) Light microscopic image of MDA-MB231 breast carcinoma cells migrated through the pores of a polycarbonate filter in a Boyden chamber assay, fixed with 95% ethanol, and stained with 0.02 g toluidine blue/mL. Bar = 10 μm.
Molecules 25 02471 g001
Figure 2. A specimen of Chondrosia reniformis demosponge. © Bernard Picton (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Figure 2. A specimen of Chondrosia reniformis demosponge. © Bernard Picton (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Molecules 25 02471 g002
Figure 3. (A) Image of a paraffin section of Jaspis stellifera demosponge. Photo by D. Drew (YPM IZ 087028). Courtesy of the Peabody Museum of Natural History, Division of Vertebrate Paleontology, Yale University; peabody.yale.edu. (B) 2D structure of stellettin B; https://pubchem.ncbi.nlm.nih.gov/compound/5352082#section=2D-Structure.
Figure 3. (A) Image of a paraffin section of Jaspis stellifera demosponge. Photo by D. Drew (YPM IZ 087028). Courtesy of the Peabody Museum of Natural History, Division of Vertebrate Paleontology, Yale University; peabody.yale.edu. (B) 2D structure of stellettin B; https://pubchem.ncbi.nlm.nih.gov/compound/5352082#section=2D-Structure.
Molecules 25 02471 g003
Figure 4. (A) A specimen of Negombata magnifica demosponge. Photo © Alexander Vasenin (CC BY-SA 3.0) https://en.wikipedia.org/. (B) Two-dimensional (2D) structure of latrunculin B; https://pubchem.ncbi.nlm.nih.gov/compound/Latrunculin-B#section=2D-Structure. (C) 2D structure of latrunculin A; https://pubchem.ncbi.nlm.nih.gov/compound/Latrunculin-a#section=2D-Structure.
Figure 4. (A) A specimen of Negombata magnifica demosponge. Photo © Alexander Vasenin (CC BY-SA 3.0) https://en.wikipedia.org/. (B) Two-dimensional (2D) structure of latrunculin B; https://pubchem.ncbi.nlm.nih.gov/compound/Latrunculin-B#section=2D-Structure. (C) 2D structure of latrunculin A; https://pubchem.ncbi.nlm.nih.gov/compound/Latrunculin-a#section=2D-Structure.
Molecules 25 02471 g004
Figure 5. (A) A specimen of Pseudoceratina arabica demosponge © Danièle Heitz; https://nomadica.jimdofree.com/. (B) 2D structure of moloka’iamine; https://pubchem.ncbi.nlm.nih.gov/compound/Moloka_Iamine#section=Structures. (C) 2D structure of hydroxymoloka’iamine; https://pubchem.ncbi.nlm.nih.gov/compound/25016152. (D) A specimen of Suberea mollis demosponge; Abbas et al., 2014 (CC BY 3.0).
Figure 5. (A) A specimen of Pseudoceratina arabica demosponge © Danièle Heitz; https://nomadica.jimdofree.com/. (B) 2D structure of moloka’iamine; https://pubchem.ncbi.nlm.nih.gov/compound/Moloka_Iamine#section=Structures. (C) 2D structure of hydroxymoloka’iamine; https://pubchem.ncbi.nlm.nih.gov/compound/25016152. (D) A specimen of Suberea mollis demosponge; Abbas et al., 2014 (CC BY 3.0).
Molecules 25 02471 g005
Figure 6. (A) A specimen of Siphonocalina siphonella demosponge © Alexander Vasenin (CC BY-SA 3.0); https://commons.wikimedia.org/. (B) 2D structure of sipholenol A; https://pubchem.ncbi.nlm.nih.gov/compound/Sipholenol-A#section=Structures.
Figure 6. (A) A specimen of Siphonocalina siphonella demosponge © Alexander Vasenin (CC BY-SA 3.0); https://commons.wikimedia.org/. (B) 2D structure of sipholenol A; https://pubchem.ncbi.nlm.nih.gov/compound/Sipholenol-A#section=Structures.
Molecules 25 02471 g006
Figure 7. Examples of specimens of Xestospongia sp. (A) X. muta © Renaud Houdinet; https://flickr.com/; (B) X. testudinaria, photo by Albert Kok (Public domain) https://commons.wikimedia.org/.
Figure 7. Examples of specimens of Xestospongia sp. (A) X. muta © Renaud Houdinet; https://flickr.com/; (B) X. testudinaria, photo by Albert Kok (Public domain) https://commons.wikimedia.org/.
Molecules 25 02471 g007
Figure 8. (A) A specimen of Cladiella australis soft coral © David (CC BY-NC-SA 4.0) https://www.marinespecies.org/. (B) 2D structure of austrasulfone; https://pubchem.ncbi.nlm.nih.gov/compound/50907657#section=Structures.
Figure 8. (A) A specimen of Cladiella australis soft coral © David (CC BY-NC-SA 4.0) https://www.marinespecies.org/. (B) 2D structure of austrasulfone; https://pubchem.ncbi.nlm.nih.gov/compound/50907657#section=Structures.
Molecules 25 02471 g008
Figure 9. (A) A specimen of Cladiella pachyclados soft coral © Danièle Heitz; https://nomadica.jimdofree.com/. (B) Two-dimensional (2D) structure of pachycladin A; https://pubchem.ncbi.nlm.nih.gov/compound/Pachycladin-A#section=2D-Structure. (C) 2D structure of sclerophytin F methyl ether; https://pubchem.ncbi.nlm.nih.govcompound/21635655. (D) 2D structure of polyanthelin A; https://pubchem.ncbi.nlm.nih.gov/compound/12168107. (E) 2D structure of sclerophytin A; https://pubchem.ncbi.nlm.nih.gov/compound/Sclerophytin-A#section=2D-Structure.
Figure 9. (A) A specimen of Cladiella pachyclados soft coral © Danièle Heitz; https://nomadica.jimdofree.com/. (B) Two-dimensional (2D) structure of pachycladin A; https://pubchem.ncbi.nlm.nih.gov/compound/Pachycladin-A#section=2D-Structure. (C) 2D structure of sclerophytin F methyl ether; https://pubchem.ncbi.nlm.nih.govcompound/21635655. (D) 2D structure of polyanthelin A; https://pubchem.ncbi.nlm.nih.gov/compound/12168107. (E) 2D structure of sclerophytin A; https://pubchem.ncbi.nlm.nih.gov/compound/Sclerophytin-A#section=2D-Structure.
Molecules 25 02471 g009
Figure 10. (A) A specimen of Sarcophyton crassocaule soft coral © Leen Van Ofwegen (CC BY-NC-SA 4.0); https://www.marinespecies.org/ (B) 2D structure of 13-acetoxysarcocrassolide; https://pubchem.ncbi.nlm.nih.gov/compound/21775908.
Figure 10. (A) A specimen of Sarcophyton crassocaule soft coral © Leen Van Ofwegen (CC BY-NC-SA 4.0); https://www.marinespecies.org/ (B) 2D structure of 13-acetoxysarcocrassolide; https://pubchem.ncbi.nlm.nih.gov/compound/21775908.
Molecules 25 02471 g010
Figure 11. (A) 2D structure of 11-dehydrosinulariolide; http://www.chemspider.com/Chemical-Structure.10470882.html. (B) 2D structure of sinularin; https://pubchem.ncbi.nlm.nih.gov/compound/Sinularin#section=2D-Structure.
Figure 11. (A) 2D structure of 11-dehydrosinulariolide; http://www.chemspider.com/Chemical-Structure.10470882.html. (B) 2D structure of sinularin; https://pubchem.ncbi.nlm.nih.gov/compound/Sinularin#section=2D-Structure.
Molecules 25 02471 g011
Figure 12. An image of Clavularia koellikeri; Wang et al. (2019) [87].
Figure 12. An image of Clavularia koellikeri; Wang et al. (2019) [87].
Molecules 25 02471 g012
Figure 13. An indivividual of Cyanea capillata © Alexander Semenov; https://www.flickr.com/.
Figure 13. An indivividual of Cyanea capillata © Alexander Semenov; https://www.flickr.com/.
Molecules 25 02471 g013
Figure 14. A specimen of Rhopilema esculentum. © Bill Abbott (CC BY-SA 2.0); https://en.wikipedia.org/.
Figure 14. A specimen of Rhopilema esculentum. © Bill Abbott (CC BY-SA 2.0); https://en.wikipedia.org/.
Molecules 25 02471 g014
Figure 15. (A) A specimen of Uroteuthis singhalensis; http://www.borneomolluscs.com. (B) 2D structure of astaxanthin; https://pubchem.ncbi.nlm.nih.gov/compound/Astaxanthin#section=2D-Structure.
Figure 15. (A) A specimen of Uroteuthis singhalensis; http://www.borneomolluscs.com. (B) 2D structure of astaxanthin; https://pubchem.ncbi.nlm.nih.gov/compound/Astaxanthin#section=2D-Structure.
Molecules 25 02471 g015
Figure 16. A specimen of Sepia kobiensis. © Maynard Hogg (CC BY-NC-SA 2.0); https://www.flickr.com/.
Figure 16. A specimen of Sepia kobiensis. © Maynard Hogg (CC BY-NC-SA 2.0); https://www.flickr.com/.
Molecules 25 02471 g016
Figure 17. A specimen of Sepia officinalis. © Hans Hillewaert (CC BY-SA 4.0); http://www.marinespecies.org/.
Figure 17. A specimen of Sepia officinalis. © Hans Hillewaert (CC BY-SA 4.0); http://www.marinespecies.org/.
Molecules 25 02471 g017
Figure 18. (A) A group of Mytilus galloprovincialis bivalves. © Pillon Robert (CC BY-NC-SA 4.0); http://www.marinespecies.org/. (B) Images of the shell of Rapana venosa. © H. Zell (CC BY-SA 3.0); https://en.wikipedia.org/. (C) 2D structure of eicosapentaenoic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Eicosapentaenoic-acid#section=2D-Structure. (D) 2D structure of docosahexenoic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Docosahexaenoic-acid#section=Structures. (E) 2D structure of arachidonic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Arachidonic-acid#section=2D-Structure.
Figure 18. (A) A group of Mytilus galloprovincialis bivalves. © Pillon Robert (CC BY-NC-SA 4.0); http://www.marinespecies.org/. (B) Images of the shell of Rapana venosa. © H. Zell (CC BY-SA 3.0); https://en.wikipedia.org/. (C) 2D structure of eicosapentaenoic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Eicosapentaenoic-acid#section=2D-Structure. (D) 2D structure of docosahexenoic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Docosahexaenoic-acid#section=Structures. (E) 2D structure of arachidonic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Arachidonic-acid#section=2D-Structure.
Molecules 25 02471 g018
Figure 19. Images of the shell of Pinctada imbricata. © Natural History Museum Rotterdam (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Figure 19. Images of the shell of Pinctada imbricata. © Natural History Museum Rotterdam (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Molecules 25 02471 g019
Figure 20. Images of the shell of Euchelus asper. © H. Zell (CC BY-SA 3.0); https://commons.wikimedia.org.
Figure 20. Images of the shell of Euchelus asper. © H. Zell (CC BY-SA 3.0); https://commons.wikimedia.org.
Molecules 25 02471 g020
Figure 21. Images of the shell of Haliotis diversicolor squamata. © Natural History Museum Rotterdam (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Figure 21. Images of the shell of Haliotis diversicolor squamata. © Natural History Museum Rotterdam (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Molecules 25 02471 g021
Figure 22. Specimens of Euphausia superba. © Brett Wilks; http://www.antarctica.gov.au/.
Figure 22. Specimens of Euphausia superba. © Brett Wilks; http://www.antarctica.gov.au/.
Molecules 25 02471 g022
Figure 23. An individual of Astropecten indicus. © Ria Tan (CC BY-NC-ND 2.0); https://www.flickr.com/.
Figure 23. An individual of Astropecten indicus. © Ria Tan (CC BY-NC-ND 2.0); https://www.flickr.com/.
Molecules 25 02471 g023
Figure 24. (A) An individual of Himerometra magnipinna. © Andrew Green; https://reeflifesurvey.com/. (B) 2D structure of rhodoptilometrin; https://pubchem.ncbi.nlm.nih.gov/compound/Rhodoptilometrin#section=2D-Structure.
Figure 24. (A) An individual of Himerometra magnipinna. © Andrew Green; https://reeflifesurvey.com/. (B) 2D structure of rhodoptilometrin; https://pubchem.ncbi.nlm.nih.gov/compound/Rhodoptilometrin#section=2D-Structure.
Molecules 25 02471 g024
Figure 25. A specimen of Stichopus japonicus. © Alexander Semenov; https://www.flickr.com.
Figure 25. A specimen of Stichopus japonicus. © Alexander Semenov; https://www.flickr.com.
Molecules 25 02471 g025
Figure 26. A specimen of Isostichopus badionotus. © Dan Hershman (CC BY 2.0); https://commons.wikimedia.org/.
Figure 26. A specimen of Isostichopus badionotus. © Dan Hershman (CC BY 2.0); https://commons.wikimedia.org/.
Molecules 25 02471 g026
Figure 27. An individual of Ophiocoma erinaceus. © Patrick Randall (CC BY-NC-SA 2.0); https://www.flickr.com/.
Figure 27. An individual of Ophiocoma erinaceus. © Patrick Randall (CC BY-NC-SA 2.0); https://www.flickr.com/.
Molecules 25 02471 g027
Figure 28. A specimen of Stichopus herrmanni. © Andrew Green; https://reeflifesurvey.com/.
Figure 28. A specimen of Stichopus herrmanni. © Andrew Green; https://reeflifesurvey.com/.
Molecules 25 02471 g028
Figure 29. (A) An image of Phallusia arabica. © Frédéric André; https://doris.ffessm.fr/. (B) 2D structure of n-hexadecanoic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Palmitic-acid#section=2D-Structure.
Figure 29. (A) An image of Phallusia arabica. © Frédéric André; https://doris.ffessm.fr/. (B) 2D structure of n-hexadecanoic acid; https://pubchem.ncbi.nlm.nih.gov/compound/Palmitic-acid#section=2D-Structure.
Molecules 25 02471 g029
Figure 30. An image of Phallusia nigra. © Rosana M. Rocha; http://invasions.si.edu/.
Figure 30. An image of Phallusia nigra. © Rosana M. Rocha; http://invasions.si.edu/.
Molecules 25 02471 g030
Figure 31. An image of Styela clava. © Claude Nozères (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Figure 31. An image of Styela clava. © Claude Nozères (CC BY-NC-SA 4.0); http://www.marinespecies.org/.
Molecules 25 02471 g031
Table 1. Anti-cancer and wound healing-promoting properties of molecules and crude extracts from marine invertebrates.
Table 1. Anti-cancer and wound healing-promoting properties of molecules and crude extracts from marine invertebrates.
EffectsPhylumSpeciesCompoundsReferences
Anti-cell migration/invasion propertiesPoriferaJaspis stellifera
Negombata magnifica
Pseudoceratina arabica
Suberea mollis
Siphonocalina (Callyspongia) siphonella
Xestospongia sp.
Stellettin B
Latrunculin B and 15-O-methyl-latrunculin B
Latrunculin A
Bromotyrosine derivative (1’R,5’S,6’S)-2-(3’,5’-dibromo-1’,6’ -dihydroxy-4’-oxocyclohex-2’-enyl) acetonitrile
Moloka’iamine
Ceratinin A
Ceratinin D hydroxymoloka’iamine
subereamolline A
Sipholenol A and its aliphatic polar and aromatic ester derivatives
Sipholenol A-4-O-3′,4′-dichlorobenzoate derivative
Ranieramycins (i.e ranieramycin M)
[42]
[47]
[48]
[53]
[54]
[62,64]
[71]
CnidariaCladiella australis
Cladiella pachyclados
Sarcophyton crassocaule
Sinularia nanolobata
Clavularia koellikeri
Dihydroaustrasulfone alcohol (synthetic precursor of Austrasulfone)
Pachycladin A
Sclerophytin F methyl ether
Polyanthelin A
Sclerophytin A
Cembrenolide diterpene 13-acetoxysarcocrassolide
11-dehydrosinulariolide
Sinularin
Clavukoellian A and clavukoellian D-related 4-O-Deacetylparalemnolin D
[76,77,78]
[79]
[81,82]
[83,84]
[87]
MolluscaEuchelus asperMethanol extracts[145]
EchinodermataOphiocoma erinaceusSaponins[204,205]
Pro wound-healing propertiesPoriferaChondrosia reniformisFraction enriched in hydroxyproline- containing collagenous peptides[33]
CnidariaCyanea capillata
Rhopilema esculentum
Tentacle extracts, Granulin
Collagen peptides
[96,97]
[103]
MolluscaUroteuthis (Doryteuthis) singhalensis
Sepia kobiensis
Sepia officinalis
Mytilus galloprovincialis
and Rapana venosa
Pinctada martensii
Haliotis diversicolor
Euphausia superba
Radical scavenging-carotenoid astaxhantin
Chitosan and collagen
Collagen
Lipid extracts; Aminoacid mixture from protein extract
Peptides extracted from P. martensii’s mantle
H. diversicolor’s shell powder
Krill oil
[109]
[115]
[116]
[132,133]
[140]
[151]
[164]
EchinodermataAstropecten indicus
Himerometra magnipinna
Stichopus japonicus
Isostichopus badionotus
Ophiocoma erinaceus
Stichopus herrmanni
AiP1 metalloproteinase
Anthraquinone (+)-rhodoptilometrin
Collagen
Oligopeptides
Polysaccharides
“Gamat Hydrogel” (S. hermannii’s extract-containing hydrogel dressing)
[171]
[178]
[188]
[198]
[203]
[210]
TunicataPhallusia arabica
Phallusia nigra
Styela clava
Ethanolic extracts
Methanolic extract
Cellulose films manufactured from S. clava’s tunic (SCT)
selenium-loaded STC-cellulose films
[219]
[221,222]
[225,226]

Share and Cite

MDPI and ACS Style

Luparello, C.; Mauro, M.; Lazzara, V.; Vazzana, M. Collective Locomotion of Human Cells, Wound Healing and Their Control by Extracts and Isolated Compounds from Marine Invertebrates. Molecules 2020, 25, 2471. https://doi.org/10.3390/molecules25112471

AMA Style

Luparello C, Mauro M, Lazzara V, Vazzana M. Collective Locomotion of Human Cells, Wound Healing and Their Control by Extracts and Isolated Compounds from Marine Invertebrates. Molecules. 2020; 25(11):2471. https://doi.org/10.3390/molecules25112471

Chicago/Turabian Style

Luparello, Claudio, Manuela Mauro, Valentina Lazzara, and Mirella Vazzana. 2020. "Collective Locomotion of Human Cells, Wound Healing and Their Control by Extracts and Isolated Compounds from Marine Invertebrates" Molecules 25, no. 11: 2471. https://doi.org/10.3390/molecules25112471

Article Metrics

Back to TopTop