Bioactive Molecules in Ascidians: Comparison
Please note this is a comparison between Version 2 by Sirius Huang and Version 1 by Laura La Paglia.

The study of bioactive molecules of marine origin has created an important bridge between biological knowledge and its applications in biotechnology and biomedicine. Studies in different research fields, such as biomedicine, aim to discover marine molecules characterized by biological activities that can be used to produce potential drugs for human use. Increasing attention has been paid to a particular group of marine invertebrates, the Ascidians, as they are a source of bioactive products. 

  • bioactive molecules
  • marine invertebrate
  • artificial intelligence
  • innate immunity

1. Introduction

Ascidians are tunicates, marine invertebrate chordates, considered the sister group of vertebrates [1,2,3,4][1][2][3][4]. They owe their name to the tunic, an epidermally secreted external layer that envelops the body. The tunic is composed of an ECM rich in collagen and tunicin (a form of cellulose) and also rich in immune cells [5,6,7,8,9][5][6][7][8][9]. The tunic also contains proteins with 3,4-dihydroxyphenylalanine (DOPA), with a catechol moiety involved in the first line of immune reaction [10] and wound healing [11[11][12],12], and 3,4,5-trihydroxyphenylalanine (TOPA) with a pyrogallol moiety [13]. Ascidians are the source of many bioactive molecules belonging to a wide variety of chemical categories [14,15][14][15] and with potential health applications, including cytotoxic, antimitotic, antiviral and antimicrobial compounds [16,17,18,19][16][17][18][19]. Most of the metabolites synthesized by ascidians contribute to creating the physico-chemical barrier preventing the entrance of foreign organisms into the internal fluids or the colonization of the tunic by encrusting organisms. The most represented chemical classes among the bioactive secondary metabolites isolated from tunicates are alkaloids, polyketides, and peptides [18]. Cytotoxicity against mammalian cell lines and anti-proliferative activity were the most frequently assigned bioactivities. Compounds with cytotoxic and antineoplastic properties isolated from ascidians belong to disparate chemical classes, and three of them have entered clinical trials [20]. Due to their key phylogenetic position in chordate evolution, the ascidians are a powerful model for studying innate immunity [4]. They possess an exclusively innate immune system, including inflammatory, humoral, and cellular responses. On an evolutionary level, inflammation is a highly conserved phenomenon and appears to be an essential first line of defense for both invertebrates and vertebrates. The innate immune system is the major contributor to acute inflammation [21[21][22],22], a rapid protective response to microbial infection, tissue injury, and insults [23], and the principal promoter of inflammatory responses often involves infection by microbial invaders or exposure to foreign particles/irritants/pollutants [24].
When host cells capable of innate immune responses encounter pathogenic microbes or other foreign or host irritants, the inflammatory response initiates within minutes. The host cells recognize the stimulus through various sensing mechanisms involving trans-membrane receptors. In Ciona robusta (previously Ciona intestinalis), these interactions transmit signals to the nucleus, resulting in the activation and regulation of numerous genes via both transcriptional and post-transcriptional mechanisms [21,25,26,27,28,29[21][25][26][27][28][29][30][31][32][33],30,31,32,33], such as antimicrobial peptides and complement factors [34,35][34][35] and proinflammatory cytokines and chemokines which activate endothelial cells and recruit immune system cells to the infection site [36,37,38][36][37][38].
The immune system is made up of a complex and dynamic network of cell subsets and mediators that promote host defense from infectious agents or tumor cells and maintain immunological tolerance in the organisms [39,40][39][40]. Vertebrate immunity is classically divided into innate and adaptive immune systems that act interdependently based on bidirectional crosstalk [41]. It is well known that the innate immune system provides the first line of defense in immune response and can induce and regulate many different adaptive immunity functions [42].

2. Bioactive Molecules in Ascidians

Bioactive peptides (BPs) are composed of protein fragments or peptides with beneficial metabolic and physiological functions that promote human health; thus, they are excellent molecules for studying human health and disease and potential therapeutics [43,44][43][44]. Most have similar structures, such as <20 amino acid residue lengths, and contain hydrophobic residues [45,46][45][46]. Based on their modes of action, different classes of BP are evidenced: anticancer (ACP), antiviral, antimicrobial (AMP), anti-oxidative stress, and immunomodulatory peptides [15,16,17,18,19,47,48][15][16][17][18][19][47][48]. In the following sections, only AMP and immunomodulatory peptides will be discussed, as they are the only bioactive molecules shown in ascidians.

2.1. Bioactive Molecules with Antimicrobial Activity

AMPs are a class of small peptides that exist widely in nature, and they are an important part of the innate immune system of different organisms. AMPs have a broad range of inhibitory effects against bacteria, fungi, parasites, and viruses. The emergence of antibiotic-resistant microorganisms and increasing concerns about the use of antibiotics have resulted in the development of AMPs, which have good application prospects in medicine, food, animal husbandry, agriculture, and aquaculture. Microorganism resistance to antimicrobials is becoming increasingly severe with the abuse of antibiotics in medicine, agriculture, and animal husbandry. The prevalence of vancomycin-resistant enterococcus (VRE) and methicillin-resistant Staphylococcus aureus (MRSA) is increasing in clinical medicine, so countermeasures are urgently needed to address these bacterial infections. Research on AMPs is continuously developing, and AMP databases store a considerable amount of data on AMPs. A massive variety of antimicrobials has been extracted from tunicates. They belong to disparate chemical classes, such as polysulfides, alkyl sulfates, terpenes, amino alcohols, spiroketals, alkaloids, furanones, peptides, and others [15]. Some of them are synthesized by symbiotic organisms colonizing the tunic or the internal fluids [49,50][49][50]. Most of the known AMPs are produced by ascidian-circulating cells, mainly immunocytes (i.e., cells involved in immune responses) for defense purposes [51,52,53,54,55,56,57,58,59,60][51][52][53][54][55][56][57][58][59][60] (Table 1).
Table 1.
Bioactive molecules in ascidians from immunocytes: antimicrobial and immunomodulator peptides.
In Halocynthia roretzi, the tetrapeptides halocyamines A and B are produced by cytotoxic morula cells (MCs) [51], and their cytotoxic activity is likely related to their diphenol rings, representing suitable substrates for the enzyme phenoloxidase (PO), which is also stored inside MCs. The enzyme induces oxidative stress by oxidizing phenols to quinones with the consequent production of ROS [64]. The hemocytes of species H. aurantium synthesize the peptide dicynthaurin and the cationic peptide halocidin [54]. The native peptide of halocidin has a mass of 3443 Da and comprises two different subunits containing 18 amino acid residues and 15 residues which are linked covalently by a single cystine disulfide bond. Two different monomers were separately synthesized to make three additional isoforms (15-residue homodimer, 18-residue homodimer, heterodimer). Antimicrobial assays performed with synthetic peptides of halocidin confirmed that congeners of the 18-residue monomer were more active than those of the 15-residue monomer MRSA and multidrug-resistant Pseudomonas aeruginosa. Hemocytes from the solitary tunicate Styela clava contained a family of four α-helical antimicrobial peptides that were purified, sequenced, and named clavanins A, B, C, and D. Clavanins A–D (histidine-rich, -helix peptides) [49] and clavaspirin are synthesized by Styela clava MCs [56]. In lysates of hemocytes of the same species, five cationic antimicrobial peptides, called styelins, were identified and isolated [57,58][57][58]. In hemocytes of Styela plicata, the octapeptide plicatamide was isolated [59]. In the tunicates Microcosmus sabatieri and Halocynthia papillosa, antimicrobial activities were detected, and two novel peptides, halocyntin and papillosin, were isolated and characterized. These molecules display antibacterial activity against Gram-positive and Gram-negative bacteria. A combination of Edman degradation and mass spectrometry obtained a complete peptide characterization. The mature molecules of halocyntin and papillosin comprise 26 and 34 amino acid residues, respectively [60]. The enormous quantity of genomic data has become a promising source of putative AMPs due to progress in bioinformatics [65,66,67,68][65][66][67][68]. In C. intestinalis, using genome and expressed sequence tag (EST) data, a putative gene family has been identified exhibiting several structural features typical of AMPs. The synthetic peptide exerted potent antimicrobial activity against various bacteria and against the yeast Candida albicans, but it was not cytolytic for mammalian erythrocytes. Using the synthetic peptide as an antigen, specific antibodies were generated, and the natural parent molecule was localized to a compartment of a distinct hemocyte type, the univacuolar refractile granulocytes [53]. Furthermore, a gene family coding for putative AMPs was identified in the EST database of C. intestinalis and subsequently identified and cloned from the Northern European Ciona subspecies. Molecular analysis revealed that the natural peptide is synthesized and stored in a distinct hemocyte type, the univacuolar non-refractile granulocytes, and that the expression of the gene is markedly upregulated in hemocytes after immune challenge. The peptide proved highly effective against Gram-negative and Gram-positive bacteria, including several human and marine pathogens, as well as the yeast C. albicans. Using two different methods, it was demonstrated that the peptide kills Gram-negative and Gram-positive bacteria by permeabilizing their cytoplasmic membranes. Circular Dichroism (CD) spectroscopy revealed that in the presence of liposomes composed of negatively charged phospholipids, the peptide undergoes a conformational change and adopts an alpha-helical structure. Moreover, the peptide was virtually non-cytolytic for mammalian erythrocytes. Hence, this designed AMP may represent a valuable template for developing novel antibiotics [52]. Ci-MAM-A24, a synthetic AMP derived from a peptide precursor from immune cells of C. intestinalis, is potently active against representatives of Gram-positive and Gram-negative bacteria by permeabilizing their cytoplasmic membrane. The activity of Ci-MAM-A24 against different bacterial pathogens which frequently cause therapeutic problems was tested. Fedders et al. tested the killing capacity of Ci-MAM-A24 against clinically important anaerobic bacteria as well as multiresistant aerobic strains such as MRSA, VRE, extended-spectrum α-lactamase-producers, and multiple-resistant Pseudomonas aeruginosa, and all strains proved to be highly susceptible to Ci-MAM-A24 at low concentrations [69]. Furthermore, an in silico screening method has been developed based on further criteria such as size, amphipathicity, and aggregation propensity, by which 22 potential LCAMP candidates in the Ciona genome were computationally predicted. Among these LCAMP candidates, five novel salt-resistant LCAMPs with broad-spectrum antimicrobial activity were experimentally confirmed. This strategy was also successfully applied to the Xenopus tropicalis genome, suggesting that this method could apply to the in silico screening of any genome [34]. Finally, Lu et al. [61] investigated the potential sORFs encoding AMPs in C. intestinalis, and over 180 peptides deduced from the sORFs were predicted to be AMPs. Among the ten peptides tested, six were found to have significant EST matches, providing strong evidence for gene expression; five were proved to be active against the bacterial strains.

2.2. Bioactive Molecules with Immunomodulatory Effects

A recent idea is to use invertebrates as a source of molecules with potential immunoregulatory activities to improve strategies for modulating human immune system responses [65,66][65][66]. The innate immune system is composed of many interdependent cell types and mediators. It is one of the most critical natural systems for protection against many harmful bacteria, viruses, parasites, and fungi in human health, and against autoimmune diseases, cancer, allergies, and infections [70,71][70][71]. Preliminary studies have shown evidence supporting a complex interaction between the immune system and tumors [72]. Several innate system immunomodulators have been identified; these include cytokines (interleukins, interferons, and chemokines), substances isolated from microorganisms and fungi (lipopolysaccharides; LPS), and substances isolated from plants (polysaccharides and phenolic compounds) [73]. Tumor cells secrete altered protein products that must be recognized as foreign by the immune effector cells such as B, T, natural killer and natural killer T cells, and type I and II interferons, and perforin which are able to destroy tumor cells [74,75][74][75]. Therefore, the enhancement of the host immune response is one of the most important methods for inhibiting tumor growth and maintaining cellular homeostasis without harming the host. The selective modulation of immunity is an emerging concept driven by the tremendous advances in our understanding of this crucial host defense system. Invertebrates have drawn researchers’ interest as potential sources of new bioactive molecules owing to their immunomodulatory activities. An LPS challenge in the ascidian C. intestinalis generates the transcript, Ci8 short, with cis-regulatory elements in the 3′ UTR region which is essential for shaping innate immune responses. The derived amino acidic sequence from in silico analysis showed specific binding to human major histocompatibility complex (MHC) class I and class II alleles. The role of Ci8 short peptide (Table 1) was investigated in a more evolved immune system using human peripheral blood mononuclear cells (PBMCs) as an in vitro model. The biological activities of this molecule include the activation of the 70 kDa TCR ζ chain associated protein kinase (ZAP-70) and T cell receptor (TCR) Vβ oligo clonal selection on CD4+ T lymphocytes as well as increased proliferation and IFN-γ secretion. Furthermore, Ci8 short affects CD4+/CD25high-induced regulatory T cells (iTreg) subset selection, which co-expressed the functional markers TGF-β1/latency-associated protein (LAP) and CD39/CD73 [62]. Furthermore, Colombo et al. [63] evaluated the 3D structure of the C8 short-derived C. robusta chemo-attractive peptide (CrCP) (Table 1) by homology modeling, which showed that CrCP displayed structural characteristics already reported for a short domain of the vertebrate CRK gene, suggesting its possible involvement in cell migration mechanisms. The biological activity of CrCP was studied in vitro using a primary human dermal cell line. In vitro assays demonstrated that CrCP could induce the motility of HuDe cells in both wound healing and chemo-attractive experiments. Furthermore, CrCP modulates the expression of the matrix metalloproteinase-7 (MMP-7) and E-cadherin genes, and it induces the activation of the NF-κB signaling pathway.

References

  1. Zeng, L.; Swalla, B.J. Molecular phylogeny of the protochordates: Chordate evolution. Can. J. Zool. 2005, 83, 24–33.
  2. Delsuc, F.; Brinkmann, H.; Chourrout, D.; Philippe, H. Tunicates and not cephalochordates are the closest living relatives of vertebrates. Nature 2006, 439, 965–968.
  3. Tsagkogeorga, G.; Turon, X.; Hopcroft, R.R.; Tilak, M.K.; Feldstein, T.; Shenkar, N.; Loya, Y.; Huchon, D.; Douzery, E.J.P.; Del-suc, F. An updated 18S rRNA phylogeny of tunicates based on mixture and secondary structure models. BMC Evol. Biol. 2009, 9, 187.
  4. Delsuc, F.; Philippe, H.; Tsagkogeorga, G.; Simion, P.; Tilak, M.-K.; Turon, X.; López-Legentil, S.; Piette, J.; Lemaire, P.; Douzery, E.J.P. A phylogenomic framework and timescale for comparative studies of tuni-cates. BMC Biol. 2018, 16, 39.
  5. Parrinello, N. The reaction of Ciona intestinalis L. to subcuticular erythrocyte and protein injection. Dev. Comp. Immunol. 1981, 5, 105–110.
  6. Parrinello, N.; Patricolo, E.; Canicattì, C. Inflammatory-like reaction in the tunic of Ciona intestinalis (Tunicata). Encapsulation and tissue injury I. Biol. Bull. 1984, 167, 229–237.
  7. Parrinello, N.; Patricolo, E.; Canicattì, C. Inflammatory-like reaction in the tunic of Ciona intestinalis (Tunicata). Encapsulation tissue injury II. Biol. Bull. 1984, 167, 238–250.
  8. Vizzini, A.; Pergolizzi, M.; Vazzana, M.; Salerno, G.; Di Sano, C.; Macaluso, P.; Arizza, V.; Parrinello, D.; Cammarata, M.; Parrinello, N. FACIT collagen (1alpha-chain) is expressed by hemocytes and epidermis during the inflammatory response of the ascidian Ciona intestinalis. Dev. Comp. Immunol. 2008, 32, 682–692.
  9. Zhao, Y.; Li, J. Excellent Chemical and Material Cellulose from Tunicates: Diversity in Cellulose Production Yield and Chemical and Morphological Structures from Different Tunicate Species. Cellulose 2014, 21, 3427–3441.
  10. Vizzini, A.; Parrinello, D.; Sanfratello, M.A.; Trapani, M.R.; Mangano, V.; Parrinello, N.; Cammarata, M. Upregulated transcription of phenoloxidase genes in the pharynx and endostyle of Ciona intestinalis in response to LPS. J. Invertebr. Pathol. 2015, 126C, 6–11.
  11. Zeng, F.; Wunderer, J.; Salvenmoser, W.; Ederth, T.; Rothbächer, U. Identifying Adhesive Components in a Model Tunicate. Philos. Trans. R. Soc. B Biol. Sci. 2019, 374, 20190197.
  12. Li, S.; Huang, X.; Chen, Y.; Li, X.; Zhan, A. Identification and Characterization of Proteins Involved in Stolon Adhesion in the Highly Invasive Fouling Ascidian Ciona Robusta. Biochem. Biophys. Res. Commun. 2019, 510, 91–96.
  13. Taylor, S.W.; Kammerer, B.; Bayer, E. New Perspectives in the Chemistry and Biochemistry of the Tunichromes and Related Compounds. Chem. Rev. 1997, 97, 333–346.
  14. Dou, X.; Dong, B. Origins and Bioactivities of Natural Compounds Derived from Marine Ascidians and Their Symbionts. Mar. Drugs 2019, 17, 670.
  15. Casertano, M.; Menna, M.; Imperatore, C. The Ascidian-Derived Metabolites with Antimicrobial Properties. Antibiotics 2020, 9, 510.
  16. Adrian, T.E. Novel Marine-Derived Anti-Cancer Agents. Curr. Pharm. Des. 2007, 13, 3417–3426.
  17. Ramesh, C.; Tulasi, B.R.; Raju, M.; Thakur, N.; Dufossé, L. Marine Natural Products from Tunicates and Their Associated Microbes. Mar. Drugs 2021, 19, 308.
  18. Sivonen, K.; Leikoski, N.; Fewer, D.P.; Jokela, J. Cyanobactins—Ribosomal Cyclic Peptides Produced by Cyanobacteria. Appl. Microbiol. Biotechnol. 2010, 86, 1213–1225.
  19. Zeng, F.; Wunderer, J.; Salvenmoser, W.; Hess, M.W.; Ladurner, P.; Rothbächer, U. Papillae Revisited and the Nature of the Adhesive Secreting Collocytes. Dev. Biol. 2019, 448, 183–198290.
  20. 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.
  21. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801.
  22. Beutler, B.; Jiang, Z.; Georgel, P.; Crozat, K.; Croker, B.; Rutschmann, S.; Du, X.; Hoebe, K. Genetic analysis of host resistance: Toll-like receptor signaling and immunity at large. Annu. Rev. Immunol. 2006, 24, 353–389.
  23. Nathan, C. Points of control in inflammation. Nature 2002, 420, 846–852.
  24. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435.
  25. Medzhitov, R. Recognition of microorganisms and activation of the immune response. Nature 2007, 449, 819–826.
  26. Ishii, K.J.; Koyama, S.; Nakagawa, A.; Coban, C.; Akira, S. Host innate immune receptors and beyond: Making sense of microbial infections. Cell Host Microbe 2008, 3, 352–363.
  27. Beutler, B.A. TLRs and innate immunity. Blood 2009, 113, 1399–1407.
  28. Vizzini, A.; Parisi, M.G.; Di Falco, F.; Cardinale, L.; Cammarata, M.; Arizza, V. Identification of CPE and GAIT elements in 3′UTR of macrophage migration inhibitory factor (MIF) involved in the inflammatory response induced by LPS in Ciona robusta. Mol. Immunol. 2018, 99, 66–74.
  29. Vizzini, A. Gene expression and regulation of molecules involved in pharynx inflammatory response induced by LPS in Ciona intestinalis. Invertebr. Surviv. J. 2017, 14, 119–128.
  30. Arizza, V.; Bonura, A.; La Paglia, L.; Urso, A.; Pinsino, A.; Vizzini, A. Transcriptional and in silico analyses of MIF cytokine and TLR signalling interplay in the LPS inflammatory response of Ciona robusta. Sci. Rep. 2020, 10, 11339.
  31. Vizzini, A.; Bonura, A.; La Paglia, L.; Fiannaca, A.; La Rosa, M.; Urso, A.; Arizza, V. ceRNA Network Regulation of TGF-β, WNT, FOXO, Hedgehog Pathways in the Pharynx of Ciona robusta. Int. J. Mol. Sci. 2021, 22, 3497.
  32. Vizzini, A.; Bonura, A.; La Paglia, L.; Fiannaca, A.; La Rosa, M.; Urso, A.; Mau-ro, M.; Vazzana, M.; Arizza, V. Transcriptomic Analyses Reveal 2 and 4 Family Members of Cytochromes P450 (CYP) Involved in LPS Inflammatory Response in Pharynx of Ciona robusta. Int. J. Mol. Sci. 2021, 22, 11141.
  33. Vizzini, A.; Bonura, A.; Parrinello, D.; Sanfratello, M.A.; Longo, V.; Colombo, P. LPS Challenge Regulates Gene Expression and Tissue Localization of a Ciona intestinalis Gene through an Alternative Polyadenylation Mechanism. PLoS ONE 2013, 8, e63235.
  34. Ohtsuka, Y.; Inagaki, H. In silico identification and functional validation of linear cationic α-helical antimicrobial peptides in the ascidian Ciona Intestinalis. Sci. Rep. 2020, 10, 12619.
  35. Giacomelli, S.; Melillo, D.; Lambris, J.D.; Pinto, M.R. Immune competence of the Ciona intestinalis pharynx: Complement system-mediated activity. Fish. Shellfish. Immunol. 2012, 33, 946–952.
  36. Parrinello, N.; Vizzini, A.; Arizza, V.; Salerno, G.; Parrinello, D.; Cammarata, M.; Giaramita, F.T.; Vazzana, M. Enhanced expression of a cloned and sequenced Ciona intestinalis TNF alpha like (CiTNF alpha) gene during the LPS-induced inflammatory response. Cell Tissue Res. 2008, 334, 305–317.
  37. Vizzini, A.; Di Falco, F.; Parrinello, D.; Sanfratello, M.A.; Cammarata, M. Transforming growth factor beta (CiTGF-beta) gene expression is induced inthe inflammatory reaction of Ciona intestinalis. Dev. Comp. Immunol. 2016, 55, 102–110.
  38. Vizzini, A.; Falco, F.D.; Parrinello, D.; Sanfratello, M.A.; Mazzarella, C.; Parrinello, N.; Cammarata, M. Ciona intestinalis interleukin 17-like genes expression is upregulated by LPS challenge. Dev. Comp. Immunol. 2015, 48, 129–137.
  39. Brodin, P.; Davis, M.M. Human immune system variation. Nat. Rev. Immunol. 2017, 17, 21–29.
  40. Parkin, J.; Cohen, B. An overview of the immune system. Lancet 2001, 357, 1777–1789.
  41. Chaplin, D.D. Overview of the immune response. J. Allergy Clin. Immunol. 2010, 125, S3–S23.
  42. Iwasaki, A.; Medzhitov, R. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 2004, 5, 987–995.
  43. Chakrabarti, S.; Guha, S.; Majumder, K. Food-Derived Bioactive Peptides in Human Health: Challenges and Opportunities. Nutrients 2018, 10, 1738.
  44. Segura-Campos, M.; Chel-Guerrero, L.; Betancur-Ancona, D.; Hernandez-Escalante, V.M. Bioavailability of Bioactive Peptides. Food Rev. Int. 2011, 27, 213–226.
  45. Manikkam, V.; Vasiljevic, T.; Donkor, O.N.; Mathai, M.L. A Review of Potential Marine-derived Hypotensive and Anti-obesity Peptides. Crit. Rev. Food Sci. Nutr. 2016, 56, 92–112.
  46. Kitts, D.D.; Weiler, K. Bioactive proteins and peptides from food sources. Applications of bioprocesses used in isolation and recovery. Curr. Pharm. Des. 2003, 9, 1309–1323.
  47. Fiat, A.M.; Migliore-Samour, D.; Jolles, P.; Drouet, L.; Bal dit Sollier, C.; Caen, J. Biologically active peptides from milk proteins with emphasis on two examples concerning antithrombotic and immunomodulating activities. J. Dairy Sci. 1993, 76, 301–310.
  48. Fagundes, P.C.; Ceotto, H.; Potter, A.; Vasconcelos de Paiva Brito, M.A.; Brede, D.; Nes, I.F.; Bastos Mdo, C. Hyicin; a bioactive peptide produced by Staphylococcus hyicus with potential applications for food preservation. Res. Microbiol. 2011, 162, 1052–1059.
  49. Ayuningrum, D.; Liu, Y.; Riyanti; Sibero, M.T.; Kristiana, R.; Asagabaldan, M.A.; Wuisan, Z.G.; Trianto, A.; Radjasa, O.K.; Sabdono, A.; et al. Tunicate-Associated Bacteria Show a Great Potential for the Discovery of Antimicrobial Compounds. PLoS ONE 2019, 14, e0213797.
  50. Chen, L.; Hu, J.-S.; Xu, J.-L.; Shao, C.-L.; Wang, G.-Y. Biological and Chemical Diversity of Ascidian-Associated Microorganisms. Mar. Drugs 2018, 16, 362.
  51. Azumi, K.; Yokosawa, H.; Ishii, S. Halocyamines: Novel Antimicrobial Tetrapeptide-like Substances Isolated from the Hemocytes of the Solitary Ascidian Halocynthia roretzi. Biochemistry 1990, 29, 159–165.
  52. Fedders, H.; Michalek, M.; Grötzinger, J.; Leippe, M. An Exceptional Salt-Tolerant Antimicrobial Peptide Derived from a Novel Gene Family of Haemocytes of the Marine Invertebrate Ciona intestinalis. Biochem. J. 2008, 416, 65–75.
  53. Fedders, H.; Leippe, M. A Reverse Search for Antimicrobial Peptides in Ciona intestinalis: Identification of a Gene Family Expressed in Hemocytes and Evaluation of Activity. Dev. Comp. Immunol. 2008, 32, 286–298.
  54. Jang, W.S.; Kim, K.N.; Lee, Y.S.; Nam, M.H.; Lee, I.H. Halocidin: A New Anti-microbial Peptide from Hemocytes of the Solitary Tunicate, Halocynthia aurantium. FEBS Lett. 2002, 521, 81–86.
  55. Menzel, L.P.; Lee, I.H.; Sjostrand, B.; Lehrer, R.I. Immunolocalization of Clavanins in Styela clava Hemocytes. Dev. Comp. Immunol. 2002, 26, 505–515.
  56. Lee, I.H.; Zhao, C.; Nguyen, T.; Menzel, L.; Waring, A.J.; Sherman, M.A.; Lehrer, R.I. Clavaspirin, an Antibacterial and Haemolytic Peptide from Styela clava. J. Pept. Res. 2001, 58, 445–456.
  57. Lee, I.H.; Cho, Y.; Lehrer, R.I. Styelins, Broad-Spectrum Antimicrobial Peptides from the Solitary Tunicate, Styela clava. Comp. Biochem. Physiol. Part B Biochem. Mol. Biol. 1997, 118, 515–521.
  58. Taylor, S.W.; Craig, A.G.; Fischer, W.H.; Park, M.; Lehrer, R.I.; Styelin, D. An Extensively Modified Antimicrobial Peptide from Ascidian Hemocytes. J. Biol. Chem. 2000, 275, 38417–38426.
  59. Tincu, J.A.; Menzel, L.P.; Azimov, R.; Sands, J.; Hong, T.; Waring, A.J.; Taylor, S.W.; Lehrer, R.I. Plicatamide, an Antimicrobial Octapeptide from Styela plicata Hemocytes. J. Biol. Chem. 2003, 278, 13546–13553.
  60. Galinier, R.; Roger, E.; Sautiere, P.E.; Aumelas, A.; Banaigs, B.; Mitta, G. Halocyntin and papillosin, two new antimicrobial peptides isolated from hemocytes of the solitary tunicate, Halocynthia papillosa. J. Pept. Sci. 2009, 15, 48–55.
  61. Lu, Y.; Zhuang, Y.; Liu, J. Mining antimicrobial peptides from small open reading frames in Ciona intestinalis. J. Pept. Sci. 2014, 20, 25–29.
  62. Bonura, A.; Vizzini, A.; Vlah, S.; Gervasi, F.; Longo, A.; Melis, M.R.; Schildberg, F.A.; Colombo, P. Ci8 short, a novel LPS-induced peptide from the ascidian Ciona intestinalis, modulates responses of the human immune system. Immunobiology 2018, 223, 210–219.
  63. Longo, V.; Longo, A.; Martorana, A.; Lauria, A.; Augello, G.; Azzolina, A.; Cervello, M.; Colombo, P. Identification of an LPS-Induced Chemo-Attractive Peptide from Ciona robusta. Mar. Drugs 2020, 18, 209.
  64. Ballarin, L.; Cima, F.; Floreani, M.; Sabbadin, A. Oxidative Stress Induces Cytotoxicity during Rejection Reaction in the Compound Ascidian Botryllus schlosseri. Comp. Biochem. Physiol. Part. C Toxicol. Pharmacol. 2002, 133, 411–418.
  65. Gruber, C.W.; Muttenthaler, M. Discovery of defense- and neuropeptides in social ants by genome-mining. PLoS ONE 2012, 7, e32559.
  66. Belarmino, L.C.; Benko-Iseppon, A.M. Databank based mining on the track of antimicrobial weapons in plant genomes. Curr. Protein Pept. Sci. 2010, 11, 195–198.
  67. Silverstein, K.A.; Graham, M.A.; Paape, T.D.; VandenBosch, K.A. Genome organization of more than 300 defensin-like genes in Arabidopsis. Plant Physiol. 2005, 138, 600–610.
  68. Giacomelli, L.; Nanni, V.; Lenzi, L.; Zhuang, J.; Dalla Serra, M.; Banfield, M.J.; Town, C.D.; Silverstein, K.A.; Baraldi, E.; Moser, C. Identification and characterization of the defensin-like gene family of grapevine. Mol. Plant Microbe Interact. 2012, 25, 1118–1131.
  69. Fedders, H.; Podschun, R.; Leippe, M. The antimicrobial peptide Ci-MAM-A24 is highly active against multidrug-resistant and anaerobic bacteria pathogenic for humans. Int. J. Antimicrob. Agents 2010, 36, 264.
  70. Arron, J.R.; Choi, Y. Osteoimmunology: Bone versus immune system. Nature 2000, 408, 535–536.
  71. Kim, J.; Bentley, P.J.; Aickelin, U.; Green-smith, J.; Tedesco, G.; Twycross, J. Immune system approaches to intrusion detection–a review. Nat. Comput. 2007, 6, 413–466.
  72. Marcus, A.; Gowen, B.G.; Thompson, T.W.; Iannello, A.; Ardolino, M.; Deng, W.; Wang, L.; Shifrin, N.; Raulet, D.H. Recognition of tumors by the innate immune system and natural killer cells. Adv. Immunol. 2014, 122, 91–128.
  73. Schepetkin, I.A.; Kirpotina, L.N.; Jakiw, L.; Khlebnikov, A.I.; Blaskovich, C.L.; Jutila, M.A.; Quinn, M.T. Immunomodulatory activity of oenothein B isolated from Epilobium angustifolium. J. Immunol. 2009, 183, 6754–6766.
  74. Kim, R.; Emi, M.; Tanabe, K. Cancer immunoediting from immune surveillance to immune escape. Immunology 2007, 121, 1–14.
  75. Hendry, S.A.; Farnsworth, R.H.; Solomon, B.; Achen, M.G.; Stacker, S.A.; Fox, S.B. The Role of the Tumor Vasculature in the Host Immune Response: Implications for Therapeutic Strategies Targeting the Tumor Microenvironment. Front. Immunol. 2016, 7, 621.
More
Video Production Service