Next Article in Journal
Potential of Marine Bacterial Metalloprotease A69 in the Preparation of Peanut Peptides with Angiotensin-Converting Enzyme (ACE)-Inhibitory and Antioxidant Properties
Previous Article in Journal
Streptomyces-Fungus Co-Culture Enhances the Production of Borrelidin and Analogs: A Genomic and Metabolomic Approach
Previous Article in Special Issue
New Insights into the Mechanism of Ulva pertusa on Colitis in Mice: Modulation of the Pain and Immune System
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunomodulatory Compounds from the Sea: From the Origins to a Modern Marine Pharmacopoeia

by
Edoardo Andrea Cutolo
1,*,†,
Rosanna Campitiello
2,3,†,
Roberto Caferri
1,
Vittorio Flavio Pagliuca
1,
Jian Li
4,
Spiros Nicolas Agathos
4,5 and
Maurizio Cutolo
2,3
1
Laboratory of Photosynthesis and Bioenergy, Department of Biotechnology, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
2
Laboratory of Experimental Rheumatology and Academic, Division of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
3
IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
4
Qingdao Innovation and Development Base, Harbin Engineering University, No. 1777 Sansha Road, Qingdao 150001, China
5
Bioengineering Laboratory, Earth and Life Institute, Catholic University of Louvain, B-1348 Louvain-la-Neuve, Belgium
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Mar. Drugs 2024, 22(7), 304; https://doi.org/10.3390/md22070304
Submission received: 27 May 2024 / Revised: 24 June 2024 / Accepted: 26 June 2024 / Published: 28 June 2024
(This article belongs to the Special Issue Marine Immunomodulatory Compounds)

Abstract

:
From sea shores to the abysses of the deep ocean, marine ecosystems have provided humanity with valuable medicinal resources. The use of marine organisms is discussed in ancient pharmacopoeias of different times and geographic regions and is still deeply rooted in traditional medicine. Thanks to present-day, large-scale bioprospecting and rigorous screening for bioactive metabolites, the ocean is coming back as an untapped resource of natural compounds with therapeutic potential. This renewed interest in marine drugs is propelled by a burgeoning research field investigating the molecular mechanisms by which newly identified compounds intervene in the pathophysiology of human diseases. Of great clinical relevance are molecules endowed with anti-inflammatory and immunomodulatory properties with emerging applications in the management of chronic inflammatory disorders, autoimmune diseases, and cancer. Here, we review the historical development of marine pharmacology in the Eastern and Western worlds and describe the status of marine drug discovery. Finally, we discuss the importance of conducting sustainable exploitation of marine resources through biotechnology.

1. Why Does the Sea Matter for Human Health?

From the birth of human civilization to the rise of the modern global economy, the ocean has been a core element for development, providing waterways for exploration, cultural exchanges, and trade [1]. From the philosophers of the classical antiquity to present-day oceanographic expeditions, the scientific study of the sea has been a constant human endeavour stimulated by the fascination with its biological diversity [2].
The classification of marine life, however, is still far from offering a conclusive picture. According to initial estimates, of the ~8.75 million species inhabiting the planet, ~2.2 million live in the oceans, ~91% of which still await description [3]. Recent reconsiderations, however, suggest that marine biodiversity, particularly of fungi, protists, and prokaryotes, has been significantly underestimated, now projected to the millions [4,5].
The latest census of marine life, the World Register of Marine Species, contains ~242,000 species. Despite growing quickly (on average, 2332 new species every year), this repository is expected to include the remaining 1–2 million undescribed species, thus entirely covering marine life, only several hundred years from now [6].
Oceans are still largely underexplored sources of lead compounds [7]. From coral reefs to hydrothermal vents, the ocean is characterized by diverse habitats, including extreme environments such as the deep-sea benthic zones [8], where unique ecosystems thrive. Therefore, the sea is an untapped source of chemodiversity to look for secondary metabolites and bioactive compounds with potential applications in human health, some having already entered clinical practice [9,10,11,12,13]. Immune-mediated inflammatory diseases are a significant burden for national healthcare systems, especially in high-income countries, with an increasing incidence registered over the last three decades [14,15]. Moreover, in nearly all forms of cancer, chronic inflammation is involved in disease development [16]. Pharmacological immunomodulation is, therefore, crucial to restore the homeostasis of the immune system in situations of both over- and under-reaction [17,18].
Virtually all marine phyla, from phytoplankton to invertebrates, produce bioactive compounds with pharmacological potential [19,20,21,22], including anti-inflammatory and immunomodulatory properties [23,24,25,26,27]. Although marine pharmacology has its roots in antiquity, the ocean is witnessing a scientific renaissance propelled by interdisciplinary drug discovery research assisted by powerful, high-throughput technologies like untargeted metagenomics and metabolomics [28,29,30,31].

2. Marine Pharmacology in the Mists of Time

From time immemorial, marine flora and fauna have been used in folk and traditional medicine (TM) [32]. However, in most cases, this knowledge is orally transmitted; therefore, ethnomedicinal approaches of drug discovery are not always straightforward. Nonetheless, since TM is the mainstay of healthcare delivery in 80% of African and Asian countries [33,34], this heritage is a valuable resource for the identification of new bioactive compounds [35,36].
Since the dawn of mankind, seaweeds (macroalgae) have been consumed in the diet and for medicinal purposes [37], as revealed by archaeological records from 2500 BC suggesting the trading of kelp (Laminariales) between coastal and mountainous areas of the Peruvian and Chilean Andes [38,39,40]. Similarly, the Incas from the Andean lakes of Peru and the Aztecs in the Valley of Mexico consumed the cyanobacterial species Nostoc spp., Phormidium tenue, and Chroococcus turgidus [41,42].
Marine zootherapy is still practiced in West Africa, Central and South America, and East Asia [43,44,45,46,47,48]. Recent ethnopharmacological studies described at least 300 TM uses of marine animals globally [49,50], of which, however, only invertebrates were confirmed sources of therapeutic compounds [51]. The overexploitation of marine animals in TM, particularly of mammals, hawksbill sea turtles (Eretmochelys imbricata), manta rays, and devil rays (Mobulidae), is a recognized risk factor for wildlife population decline and species extinction [52,53,54], and, in turn, precludes the identification of new, potentially therapeutic compounds [55]. Therefore, international regulatory policies are urged to restrict the hunting and marketing of threatened species and to promote the conservation of fragile marine ecosystems [56,57].

3. The Origins of Marine Pharmacology and Immunology in the West

Descriptions of medical uses of marine animals are found in Greek and Byzantine texts from the classical antiquity (fifth century BC–7th century AD) [58,59], including the De Materia Medica by the father of Western pharmacognosy, Pedanius Dioscorides (40–90 AD) [60], and the De natura animalium by the Roman author Claudius Aelianus (175–235) [61]. Similarly, seaweeds are mentioned in the Corpus Hippocraticum by Hippocrates of Kos (460–370 BC), in the Historia Plantarum by Theophrastus (350–287 BC), and in the Naturalis Historia by Gaius Plinius Secundus (Pliny the Elder, 23–79 AD) [62,63]. Moreover, around this time, the Roman author Aulus Cornelius Celsus (25 BC–50 AD) formulated the oldest recorded definition of inflammation: “Notae vero inflammationis sunt quatuor: rubor et tumor cum calore et dolore” (the signs of inflammation are four: redness, swelling, fever, and pain) [64]. From the second century BC onwards, technology and science [65,66] constantly flowed between the Far East and the Mediterranean via the Silk Road (Sichou zhi lu, 丝绸之路), a network of land and sea trading posts connecting the Greco-Roman world with Mongolia and China via the Middle East, Eurasia, Persia, and India. Besides primarily serving geopolitical interests [67], the Silk Road promoted the reciprocal exhange of medical and pharmacological knowledge between the Far East and the Western world [68,69].
During the Age of Discovery in the fifteenth century, European countries assembled vast collections of flora from overseas, de facto establishing global bioprospecting, although ethnobotanical knowledge was also lost due to the forced conversion of the indigenous peoples to Christianity, especially by the Conquistadores [70]. Nonetheless, exotic species incessantly flowed from the overseas colonies in the four corners of the world to the main cultural centers of Europe, resulting in new discoveries in pharmacology [71] and Western medicine eventually being united in 1948 under the aegis of the International Pharmacopoeia of the World Health Organization [72].
In the 1960s, Western science achieved a deeper understanding of immunity and inflammation with the elucidation of the structure of antibodies and their generation via genetic recombination, as well as the identification of antibody-producing B cells, regulatory T-lymphocytes, and dendritic cells as antigen-presenting cells. Ironically, the concept of autoimmunity—the condition whereby toxic autoantibodies recognise self-antigens, causing chronic inflammation—was formulated in 1892 by German physician Paul Ehrlich, although it was rejected as “physiologically inconceivable” and referred to as “horror autotoxicus” [73]. Only in 1965 was autoimmunity recognized as a common immunological disorder underlying the pathogenesis of chronic inflammatory diseases [74]. Eventually, the invention of monoclonal antibodies and their application in clinical practice, as well as the discovery of cellular checkpoint control, paved the way for cancer immunotherapy and targeted therapies for autoimmune diseases [75]. Today, a plethora of inflammatory mediators are known, including sub-populations of immune cell types, their released soluble factors (cytokines, antibodies), and the intracellular genetic and molecular mechanisms which sustain inflammatory disease [76].

4. The Evolution of Marine Materia Medica in the Far East

Written records of marine flora appear in the 2500-year-old Chinese book the Classic of Poetry (Shijing, 诗经) [77,78,79,80]. The main pharmacological heritage in the Far East, however, is the Bencao, a series of compendia of materia medica produced over 2000 years [81]. Among the earliest versions, the Xinxiu Bencao (新修本草, Newly Revised Materia Medica) is the first pharmacopoeia commissioned by Imperial order during the Tang dinasty (618–907) and the oldest example of national codex, describing 850 medicinals, many of which are still used in Chinese TM [82]. Under the Tang rule, China became a cosmopolitan society by incorporating foreign cultural elements introduced via the Silk Road [83], like those found in the Haiyao Bencao (海藥本草, Overseas Pharmacopoeia or Pharmacopoeia of Foreign Drugs) compiled by Li Xun (855-930), a Chinese-born Persian physician [84]. The subsequent Zheng Lei Ben Cao (證類本草, Materia Medica Arranged According to Pattern) was compiled in 1108 AD during the Song Dynasty (960–1279) and included 1746 medicinals, among which are two marine macroalgae: Gloiopeltis furcata (Rhodophyta) and Laminaria (Saccharina) japonica (or sweet kelp, Phaeophyceae) [85,86]. The Bencao Tujing (圖經本草, Illustrated Classic of Materia Medica, 1061) and the Zhu Fan Zhi (诸蕃志, A Description of Barbarian Nations, 1225), also published under the Song rule, reflected the impact of the Maritime Silk Road seafaring on the evolution of Chinese medical knowledge [87,88,89]. Under the Mongol-led Yuan Dynasty (1271 to 1368), scientific ideas circulated inside the Empire via the Indian Ocean trade routes [90], and, as a result, the Islamic formulary Huihui Yaofang (回回藥方考釋, Muslim Medicinal Recipes) became a reference Imperial medical text [91,92]. During the Great Ming Dynasty (1368–1644), the court physician Li Shizhen (1518–1593) compiled the Bencao Gangmu (本草綱目, Compendium of Materia Medica), an outstanding piece of scientific literature describing 1892 medicinals. This text includes a detailed description of marine fauna and flora and stood until the nineteenth century as a reference for taxonomical classification in East Asia [93]. Notably, the Bencao Gangmu contains medical prescriptions based on seaweeds of the Ulvophyceae, Phaeophyceae, Florideohyceae, Trebouxiophyceae, and Bangiophyceae classes; cyanobacteria of the Nostoc genus; and seahorses [94,95].
The species Hippocampus kuda is known to produce an antitumor peptide with inhibitory activity on major intracellular signalling cascades: the nuclear factor kB (NF-kB)-mediated pathway, the Janus kinase 2/Signal Transducers and Activators of Transcription 3 (JAK2/STAT3) pathways, and the Jun N-terminal kinase (JNK)/p38 mitogen-activated Protein Kinase (p38 MAPK) pathway [96,97,98,99,100,101,102]. Seahorses are heavily used in TM, with an estimated annual consumption of approximately 250 tons in China and Hong Kong [103]. Such overexploitation, however, is posing a severe extinction threat to several Hippocampus species in both East Asia and Latin America [104], which are now included in the Appendix II of the Convention on International Trade in Endangered Species of Wild Fauna and Flora (CITES) [105] and in the Red List of the International Union for Conservation of Nature (IUCN) [106].
During the last imperial Dynasty of the Qing (1644–1911), European missionaries visited and established themselves in China, introducing the Christian faith and other Western cultural elements, including cartography. At the court of the Qing Emperor Shengzu, the Flemish Jesuit and astronomer Ferdinand Verbiest (1623–1688) published the Kunyu Quantu (坤舆全图, Full Map of the World) in 1674, one of several Chinese world maps produced in that era. Geography offered a glimpse of the outer world, attracting the attention of the traditionally self-centered and self-isolated Chinese civilization towards Western science (Figure 1) [107]. The Qing era was characterized by profound and irreversible changes in the Chinese society caused by violent political upheaval and by Western colonization, forcing the opening to foreign trade [108]. Furthermore, the end of the Imperial era was followed by the adoption of a universal “modern” science, although TM will never be fully replaced [109].
The opening of the Marine Biological Station of Amoy University in the 1930s under the guidance of foreign-trained Chinese biologists [110] marked a major leap forward in Chinese exploration of national marine biodiversity. These endeavours continue to the present day, with extensive bioprospecting activity being conducted in the South China Sea, leading to newly discovered marine lead compounds [111].
Much credit for the development of Chinese mariculture in the second half of the twentieth century goes to the US-trained marine botanist Cheng Kui Tseng (1909–2005), who worked at the Institute of Oceanology at Qingdao (Shandong province). This unsung hero of the 1940s modernization movement known as “Saving the Country by Means of Science” (科学救国) [112] contributed to the breaking of records of seaweed productivity achieved in the northern coastal Shandong Province [113].
Standing out from the crowd, China boasts a Modern Marine Materia Medica, a scientific encyclopaedia of marine medicinal organisms and of chemicals curated by the Key Laboratory of Marine Drugs of the Ministry of Education at the Ocean University of China in Qingdao. Besides holding tremendous scientific value, this project, supported by the special program “Project 908” (Comprehensive investigation and evaluation on the offshore oceans of China, Zhong guo jin hai hai yang zong he diao cha yu ping jia, 中国近海海洋综合调查与评价) [114], offers a privileged insight into one of the oldest surviving human civilizations, reflecting the uninterrupted continuity of Chinese culture throughout the millennia [115].

The Yin Yang Dialectic of Autoimmunity

With the introduction of Western medicine into China in the middle seventeenth century, traditional Chinese medicine (TCM) started to evolve in the constant struggle between traditionalism and modernization [116]. In TCM, spiritual and scientific concepts coexist in a holistic discipline [117], which is in stark contrast with the reductionist Western approach. However, some TCM principles reflect key features of the immune system: balance, defense, holism, and circadian rhythms [118]. According to TCM, healthy immune functions require a harmonious equilibrium of Yin reserves (阴, organs, tissues, cells, and body fluids) and Yang (阳, physiological functions). The two are opposing forces constantly trying to win over one another. Yin and Yang are kept in a constant dynamic balance to preserve the Qi (气), the body’s vital energy [119], a concept analogous to Pneuma (breath of life, spirit) from classical antiquity [120]. A persistent Yin deficiency causes Shanghuo (上火), or heat syndrome, eventually creating an “excess of pathogenesis caused by deficiency” [121]. In a healthy individual, apoptosis regulates tissue homeostasis, maintaining Yin and Yang balance. When the capacity to remove apoptotic cells is overwhelmed by tissue degradation, the excessive exposure of auto-antigens to the immune system breaks immunological tolerance, triggering autoimmunity. Remarkably, several immune components embody the Yin Yang dialectic [122]. For instance, CD4+ T helper cells undergo dynamic functional specialization via cytokine-mediated signaling feedback [123]. Initially, a Th1 differentiation program is activated by interleukin (IL)-12 and interferon gamma (IFN-γ). Th1 cells later switch to producing the Th2-program cytokine IL-4 to prevent unrestrained Th1 proliferation.
Similarly, dendritic cells undergo divergent differentiation programs in response to chemical stimuli [124]. Instead, regulatory T cells—a lymphocyte population which suppresses immune responses and maintains self-tolerance—can convert to inflammatory Th17 cells [125]. Finally, individual cytokines can be either Yin or Yang elements, i.e., IL-6 is endowed with both pro- and anti-inflammatory functions depending on the context [126]. Strikingly, many TCM practices either stimulate or suppress immune functions [127,128,129]; therefore, this ancient medical art predates the discovery of the immune system and its complex functioning.

5. The Birth of Marine Microbiology: Sailing on the Ocean of Chemodiversity

The invention of optical instruments in the eighteenth century by Dutch lensmaker Antonie van Leeuwenhoeck and English polymath Robert Hooke enabled the discovery of marine microbes, marking a fundamental breakthrough in the appreciation of ocean biodiversity [130,131]. The ensuing scientific excitement stimulated the publication of Die Bakterien des Meeres (Bacteria of the Sea) in 1894 by German biologist Bernhard Fischer [132] and, later, of the treatise Marine Microbiology: A Monograph on Hydrobacteriology by American microbiologist Claude ZoBell [133]. Decades later, the discovery of marine cyanobacteria by John Waterbury and Sallie Chisholm [134,135] provided new insights into ocean primary productivity and revolutionized marine ecophysiology.
In the wake of the genomic and post-genomic eras, the exploration of the ocean by means of molecular techniques has revealed the staggering complexity of microbial biodiversity [136]. Metagenomics was first introduced in marine sciences by The Sorcerer II Global Ocean Sampling expedition led by American biotechnologist Craig Venter (2004–2006) [137,138,139]. The subsequent Malaspina (2010) and Tara Oceans expeditions (2009–2013) further revealed the richness of microbial life from the deep-sea environments [140,141,142]. The picture of the global ocean microbial genome was recently assembled in the KAUST Metagenome Analysis Platform (KMAP) Global Ocean Gene Catalog 1.0. This database contains 308.6 million gene clusters assembled from 2.102 metagenomes and represents an invaluable resource for the functional discovery of microbial metabolic pathways [143].

5.1. Prokaryotes and Metazoan-Associated Microbiota

Marine bacteria produce an arsenal of secondary metabolites used for inter- and intra-specific communication [144]. During evolution, key genes encoding polyketide synthases and non-ribosomal peptide synthetases have undergone extensive reshuffling within operons, creating a remarkable diversification of metabolic pathways [145]. A recent analysis of marine prokaryotic genomes revealed an astonishing complexity of biosynthetic gene clusters, although only a tiny fraction of secondary metabolites has been studied [146]. A major obstacle to microbial drug discovery, however, lies in the recalcitrance to culturability of marine isolates, since most species naturally grow in consortia [147,148]. Moreover, many secondary metabolites originally believed to be produced by marine invertebrates derive from the associated microbiota [149]. Mutualistic relationships between fungi and bacteria and metazoans (holobionts) are found in corals and sponges [150]. Strikingly, nearly all bioactive polyketides and peptides isolated from the Theonella swinhoei (porifera) holobiont are produced by the filamentous bacterial symbiont Entotheonella spp. [151,152,153]. Similarly, the anticancer molecule trabectedin was isolated from Candidatus Endoecteinascidia frumentensis, the bacterial symbiont of the sea squirt Ecteinascidia turbinata [154]. However, the study of secondary metabolites from mutualistic relationships is complicated by the strict host dependency of endosymbionts and the alteration of holobiont composition upon ex situ cultivation [155].

5.2. Fungi and Protists

Several Talaromyces fungal symbionts of algae and sponges [156] produce polyketides, alkaloids, terpenoids, peptides, and lipids, with reported anti-inflammatory potential [157,158]. Saprotrophic protists are emerging biofactories of immunomodulatory lipids, including the omega-3 polyunsaturated docosahexaenoic (DHA, C22:6 ω-3) and eicosapentaenoic acids (EPA, C20:5 ω-3) produced by the Thraustochytrid Schizochytrium sp. (reviewed in [159,160]), which is amenable to fermentation at a large scale in seawater and wastewater [161]. Notably, the anti-inflammatory properties of DHA compounds were recently reported in a clinical trial involving patients with rheumatoid arthritis [162].

5.3. Marine Eukaryotic Microalgae and Cyanoprokaryotes

Eukaryotic phytoplankton (hereafter microalgae) is potentially the richest resource for drug discovery and a promising platform for large-scale and low-cost production of high-value metabolites [163]. Microalgae comprise a vast group of photosynthetic microbes producing a huge repertoire of anti-inflammatory and immunomodulatory pigments and lipids [159,164,165,166,167].
Carotenoids are lipophilic pigments [168,169,170,171] designated as carotenes (lycopene and α- and β-carotene), which contribute to light-harvesting, and the oxygenated derivatives xanthophylls (or ketocarotenoids: astaxanthin, fucoxanthin and lutein), which are mainly involved in the detoxification of reactive oxidative species (ROS) generated by photosynthetic reactions. The ketocarotenoid astaxanthin is the microalgal pigment of greatest pharmacological value, being endowed with strong antioxidant capacity (extensively reviewed in [159]). The biological production of astaxanthin, however, is restrained by the slow growth of its native producer, the freshwater Chlorophyte Haematococcus lacustris (previously named Haematococcus pluvialis) [172]. Accordingly, the establishment of optimal cultivation strategies of H. lacustris and the domestication of high-yielding strains are key to accruing astaxanthin accumulation [173,174,175,176].
In response to stress, several microalgae synthesize DHA, EPA [177], while the chlorophyte Tetraselmis chui accumulates monogalactosyldiacylglycerols, which inhibit nitric oxide production [178,179,180,181]. The haptophyte Tisochrysis lutea (formerly known as Isochrysis affinis galbana) is the main DHA producer, although the eustigmatophytes Microchloropsis salina, Nannochloropsis oceanica, and Microchloropsis gaditana are emerging EPA producers, and species of the Pavlovophyceae family are sources of both carotenoids and functional lipids [182,183,184]. Diatoms are widely distributed eukaryotic phytoplankton [185] which produce valuable immunomodulatory pigments and lipids [186], particularly the genus Thalassiosira, which accumulates human-like prostaglandins in response to environmental stress [187,188,189,190].
At present, China is the major world producer of microalgal biomass for human consumption [191], while the US and several European countries are at the forefront of microalgal biotechnology research [192,193]. Bioprospecting for industrially relevant microalgae, instead, is conducted worldwide, mainly in inhospitable habitats, since extremophiles hyper-accumulate bioactive compounds [194] and display robust growth phenotypes under co-cultivation [195,196,197].
The full biotechnological exploitation of non-conventional microalgae, however, requires improvements in biomass yield and downstream processing to retrieve target metabolites [198,199].
Finally, marine cyanoprokaryotes are emerging sources of bioactive metabolites [200], several endowed with anti-inflammatory and immunomodulatory properties, including polysaccharides, phenols, flavonoids, and phycobiliproteins [201,202,203,204]. Notably, the light-harvesting pigment C-phycocyanin is a selective inhibitor of the enzyme cyclooxigenase-2 producing the pro-inflammatory mediator prostaglandin E2 [205,206,207,208].

6. Mining the Seabed for Novel Bioactive Compounds

Considered an azoic zone until the late nineteenth century [209], the deep sea has always stimulated the curiosity of scientists interested in the search for life in this mysterious environment. Starting with the British-led oceanographic dredging cruises of the H.M. SS. Porcupine and Lightning (1868–1870) [210,211] and the ensuing Challenger Atlantic voyage led by marine zoologist Sir Charles Wyville Thomson (1872–1876) [212], the abysses revealed remarkable biodiversity. These were followed by the French Travailleur and Talisman expeditions (1880–1883), during which barophilic microbes were collected and cultivated by Adolph-Adrien Certes, a disciple of Louis Pasteur [213,214].
The excitement for these newly discovered ecosystems was felt internationally, prompting the establishment of zoological stations dedicated to the study of marine biology and ecology [215]. Among these, the Stazione Zoologica of Naples (Italy), founded in 1872 by German zoologist Anton Dohrn, rapidly acquired international prestige and, today, is a leading institution in the field of marine bioprospecting and drug discovery research [216]. Between 1950 and 1952, the Danish-led Galathea Deep Sea Expedition (1950–1952) reached sampling depths of 10,000 m below sea level, revealing the existence of extremely barophilic bacteria [217,218] and prompting their cultivation in the laboratory [219]. In 1979, the manned submersible vessel Alvin enabled the discovery of hydrothermal vents in the Galápagos Rift of the East Pacific Ocean and of their associated communities of extremophile microbes [220].
Arguably, the deep sea is the last uncharted frontier for bioprospecting [221,222]. This extreme environment is home to thermophile, halophile, alkalophile, psychrophile, piezophile, and polyextremophile microorganisms, which produce a panoply of secondary metabolites (the chemical structures of recently identified lead compounds from deep-sea organisms are shown in Figure 2, while in Table 1, their biological activity and half-maximal inhibitory concentration, IC50, values are provided) [223,224,225,226]. Deep-sea biodiversity and chemodiversity are currently being investigated using approaches combining imaging, sampling, and genomics analysis with the creation of dedicated repositories, such as the MArine Bioprospecting PATent (MABPAT) Database, which provides free access to this constantly expanding biological landscape [227,228,229].

6.1. Deep-Sea Prokaryotes

Despite their physiological adaptation to extreme environmental conditions, deep-sea microbes can be easily cultured under laboratory conditions, enabling detailed investigation of their secondary metabolites and, recently, genetic engineering [230,231]. Bioprospecting for anti-inflammatory compounds of deep-sea bacteria resulted in the identification of a macrolactin derivative (7,13-epoxyl-macrolactin A, a 24-membered ring lactone, Figure 2.1) from Bacillus subtilis B5, which strongly inhibited pro-inflammatory gene expression and prevented the production of the inflammatory mediators interleukin-1β and IL-6 [232]. Another example of a bacterium-derived compound with immunomodulatory properties is the exopolysaccharide from Planococcus rifietoensis (described in Section 8.2) [233]. As in the case of shallow-water species of Porifera, symbioses between microbes and invertebrates are equally common in the deep sea, although less heterogeneous, as revealed by recent investigations into the sponge-associated microbiome [234]. Therefore, deep-sea holobionts are potential new sources of bioactive compounds awaiting characterization.

6.2. Deep-Sea Fungi

The advancement of deep-sea bioprospecting is reflected by the increasing number of newly identified bioactive compounds from fungal species [235], with the Microbacterium, Dermacoccus, Streptomyces, and Verrucosispora of the phylum actinomycota being the most studied genera [236]. Several ascomycota species also produce anti-inflammatory compounds with inhibitory activity against nitric oxide release. These include cyclopenol (a 7-membered 2,5-dioxopiperazine alkaloid, Figure 2.2), derived from Aspergillus sp.; the fusaric acid derivatives hepialiamides (Figure 2.3); and one novel hybrid polyketide hepialide (Figure 2.4) from Samsoniella hepiali. The latter also produces uridine, ergosterol, walterolactone A, (4R, 5S)-5-hydroxyhexan-4-olide, and myrothecol (Figure 2.5–10). Furthermore, Acremonium sp. and Eutypella sp. accumulate eremophilane-like sesquiterpenoids (Figure 2.11) [237,238,239,240,241], while the Ascomycetes Penicillium oxalicum and chrysogenum produce alkaloids and chrysamides (Figure 2.12–15), respectively, capable of suppressing the synthesis of pro-inflammatory mediators, including the potent cytokine IL-17 in vitro in the case of P. chrysogenum [242,243]. Finally, the Basidiomycete Cystobasidium laryngis has been shown to produce diphenazine derivatives with anti-neuroinflammatory properties (Figure 2.16) [244].
Table 1. Recently discovered anti-inflammatory and immunomodulatory compounds from deep-sea microorganisms.
Table 1. Recently discovered anti-inflammatory and immunomodulatory compounds from deep-sea microorganisms.
MoleculeSource Organism(s)Biological Activity—Half Maximal Inhibitory Concentration (IC50)Development StageRef.
7,13-epoxyl-macrolactin ABacillus subtilis B5
(Gram-positive bacterium)
Suppression of inducible nitric oxide synthase, IL-1β, and IL-6 expression in cultured activated murine macrophages (IC50 N.D.).Preclinical trial[232]
Extracellular ExopolysaccharidePlanococcus rifietoensis AP-5
(Gram-positive bacterium)
Stimulation of IL-10, IL-6, IL-1β, and TNF-α production by human cultured monocytes (IC50 N.D.).Preclinical trial[233]
Cyclopenol
(7-membered 2,5-dioxopiperazine alkaloid)
Aspergillus sp.
(Ascomycota)
Suppression of nitric oxide release by cultured activated murine macrophages via inhibition of the NF-κB pathway. Down-regulation of inducible nitric oxide synthase, IL-1β and IL-6 in cultured activated murine microglia (IC50 30 µM).Preclinical trial[237]
Hepialiamides (fusaric acid derivatives)Samsoniella hepiali W7
(Ascomycota)
Suppression of nitric oxide release by cultured activated murine microglia (IC50 1 µM).Preclinical trial[238]
Polyketide hepialideSamsoniella hepiali W7
(Ascomycota)
Suppression of nitric oxide release by cultured activated murine microglia (IC50 1 µM).Preclinical trial[238]
5′-O-acetyladenosine, uridine, ergosterol, walterolactone ASamsoniella hepiali W7
(Ascomycota)
Suppression of nitric oxide release by cultured activated murine microglia (IC50 1 µM).Preclinical trial[238]
(4R,5S)-5-hydroxyhexan-4-olideSamsoniella hepiali W7
(Ascomycota)
Suppression of nitric oxide release by cultured activated murine microglia (IC50 426 nM).Preclinical trial[238]
2-benzoyl tetrahydrofuran
enantiomers
(−)-1S-myrothecol, (+)-1R-myrothecol
Myrothecium sp.
(Ascomycota)
Suppression of nitric oxide release by cultured activated murine macrophages (IC50 1.20 and 1.41 µgmL−1).Preclinical trial[239]
Acremeremophilanes
Eremophilane-Type
Sesquiterpenoids
Acremonium sp.
(Ascomycota)
Suppression of nitric oxide release by cultured activated murine macrophages (IC50 8 to 45 μM).Preclinical trial[240]
Eremophilane-Type
Sesquiterpenoids
Eutypella sp.
(Ascomycota)
Suppression of nitric oxide production by cultured activated murine macrophages (IC50 8 to >50 μM).Preclinical trial[241]
Oxaline (A), isorhodoptilometrin (B), and 5-hydroxy-7-(2′-hydroxypropyl)-2-methyl-chromone (C).Penicillium oxalicum
(Ascomycota)
Suppression of nitric oxide and prostaglandin E2 production by cultured murine microglia cells.
Down-regulation of inducible nitric oxide synthase and cyclo-oxygenase-2 expression. Inhibition of TNF-α, IL-1β, IL-6, and IL-12 production via interference with the NF-κB and MAPK pathways (IC50 A 9, B 15, and C 75 μM).
Preclinical trial[242]
Dimeric nitrophenyl trans-epoxyamides
Chrysamides A-C
Penicillium chrysogenum
(Ascomycota)
Suppression pro-inflammatory cytokine IL-17 production by cultured murine naïve T cells (IC50 C 75 μM).Preclinical trial[243]
Phenazostatins
(Diphenazine derivatives)
Cystobasidium laryngis
(Basidiomycota)
Suppression of nitric oxide and IL-6 production by activated murine macrophages in vitro via inhibition of NF-κB pathway. Suppression of IL-1β, IL-6, and inducible nitric oxide synthase expression in cultured murine microglia cells (IC50 0,30–170 μM).Preclinical trial[244]
Figure 2. Chemical structures of recently identified immunomodulatory and anti-inflammatory compounds from deep-sea organisms described in Table 1. (1) 7,13-epoxyl-macrolactin A from Bacillus subtilis B5; (2) cyclopenol from Aspergillus sp. [237]; (39) hepialiamide, polyketide hepialide, 5′-O-Acetyladenosine, uridine, ergosterol, walterolactone A, and (4R, 5S)-5-hydroxyhexan-4-olide from Samsoniella hepiali W7; (10) myrothecol from Myrothecium sp. [239]; (11) eremophilane from Acremonium sp. and Eutypella sp. [240,241] (1214) oxaline, isorhodoptilometrin, and 5-hydroxy-7-(2′-hydroxypropyl)-2-methyl-chromone from Penicillium oxalicum [242]; (15) chrysamide from Penicillium chrysogenum [243]; and (16) phenazostatin from Cystobasidium laryngis [244].
Figure 2. Chemical structures of recently identified immunomodulatory and anti-inflammatory compounds from deep-sea organisms described in Table 1. (1) 7,13-epoxyl-macrolactin A from Bacillus subtilis B5; (2) cyclopenol from Aspergillus sp. [237]; (39) hepialiamide, polyketide hepialide, 5′-O-Acetyladenosine, uridine, ergosterol, walterolactone A, and (4R, 5S)-5-hydroxyhexan-4-olide from Samsoniella hepiali W7; (10) myrothecol from Myrothecium sp. [239]; (11) eremophilane from Acremonium sp. and Eutypella sp. [240,241] (1214) oxaline, isorhodoptilometrin, and 5-hydroxy-7-(2′-hydroxypropyl)-2-methyl-chromone from Penicillium oxalicum [242]; (15) chrysamide from Penicillium chrysogenum [243]; and (16) phenazostatin from Cystobasidium laryngis [244].
Marinedrugs 22 00304 g002

7. Emerging Marine Immunomodulatory Lead Compounds

7.1. Seaweeds (Macroalgae)

Seaweeds, or macroalgae, are plant-like multicellular phototrophs found in intertidal waters attached to rocks or floating free in the open sea [194]. The edible red seaweed Porphyra dentata (Rhodophyta, Bangiales) is widely used worldwide in TM and contains phenolic compounds which inhibit the synthesis of the pro-inflammatory signaling molecule nitric oxide (NO) and suppress the NF-kB pathway in vitro [245].
Several species are farmed in the ocean due to their highly nutritious composition and content of bioactive compounds, mainly sulfated polysaccharides [246,247,248]. For instance, porphyran, a sulfated galactan from red seaweeds (Porphyra), stimulates the immune function and apoptotic/autophagic processes, and is considered a candidate anti-cancer drug (IC50 20 μg/mL) [249]. Similarly, the polysaccharide fraction from Lithothamnion muelleri (Hapalidiaceae) displayed immunomodulatory activity by inhibiting the synthesis of pro-inflammatory chemokines in an animal model of arthritis [250]. Moreover, fucoidan and ulvan extracted from the brown macroalga (kelp) Undaria pinnatifida and the green alga Ulva lactuca (IC50 623.58–785.48 µg/mL), respectively, are potent immunostimulators and antioxidants, and thus, have potential applications to reduce the side effects of immunosuppressive therapies [251,252]. Anti-inflammatory effects were reported for polysaccharide extracts from Halimeda tuna (Ulvophyceae) [10] and Posidonia oceanica (Alismatales) [253], and for carrageenans derived from different red seaweeds (Chondrus crispus, Ahnfeltiopsis devoniensis, Sarcodiotheca gaudichaudii, and Palmaria palmata) [254]. Finally, polysaccharide fucosterol and phlorotannins from the brown macroalgae Sargassum wightii and Eisenia bicyclis (eckol, dieckol and 7-phloroeckol; IC50 52.86, 51.42 and 26.87 μg/mL, respectively) suppressed the release of pro-inflammatory mediators in animal models of arthritis [255,256].

7.2. Invertebrates

Several marine invertebrate phyla are known producers of anti-inflammatory and immunomodulatory compounds [257,258]. Corals are colonial organisms of the class Anthozoa (Cnidaria), typically found in reef ecosystems in tropical and sub-tropical water. Coral bioprospecting is mainly focussed on the octocorallia order Alcyonacea (Gorgonians or soft corals), which is known to produce a vast repertoire of secondary metabolites of pharmacological relevance [259], mainly immunomodulatory lipid derivatives and terpenoids [260,261]. For instance, the sesquiterpenes (C15H24) capnellenes, isolated from the species Capnella imbricata, inhibited the expression of the pro-inflammatory enzymes inducible nitric oxide synthase and cyclooxygenase-2 in cultured macrophages (used at 10 μM) [262,263]. The Caribbean gorgonian Plexaura homomalla, instead, emerged as the highest natural producer of mammalian-like prostaglandins, which are hormone-like oxygenated metabolites of C20 fatty acids involved in the modulation and resolution of inflammation [264,265]. Similarly, diterpenes (C20H32) isolated from the Formosan gorgonians Briareum excavatum (excavatolide, or excavatoid B used at 50 μM, E and F with ED50 > 40 μg/mL) and Sinularia querciformis (11-epi-sinulariolide acetate; IC50 50 μM) inhibited the synthesis of several pro-inflammatory mediators in arthritis animal models [266,267,268]. Lastly, peptides isolated from the venom of the jellyfish Pelagia noctiluca (Pelagiidae) strongly suppressed nitric oxide production in vitro (used at 50 μg/mL) [269].
Ascidians (Chordata, subphylum Tunicata) have emerged as novel sources of bioactive compounds, mainly derived from their innate immune system [270]. Recently, a synthetic peptide derived from the sea squirt Styela clava was shown to exert both antimicrobial and immunomodulatory activities in animal models. In particular, the clavanin-MO peptide (used at 2 μM) promoted the synthesis of the anti-inflammatory cytokine IL-10 while suppressing the release of the pro-inflammatory factors IL-12 and tumor necrosis factor-α (TNF-α) upon bacterial infection [271]. Another study described the in vitro inhibitory effects of chemical inhibitors based on the structure of metabolites from Herdmania momus against multiple pro-inflammatory enzymes and the release of pro-inflammatory cytokines in activated macrophages (IC50 between 7.59 and 39.20 μM) [272]. Finally, although the chemical nature of the bioactive compound(s) still awaits characterization, extracts of the Indonesian ascidian Polycarpa aurata acted as hydrogen sulfide donors in vitro, suppressing the pro-inflammatory response of cultured macrophages (used at 50 µg/mL) [273].
Gastropods of the family Muricidae (Mollusca) are known to produce mucus-containing bioactive molecules. A recent study showed the immunomodulatory effects of the mucus of Bolinus brandaris, suggesting that a still-uncharacterized compound could trigger the immune system against cancerous cells by inducing monocyte differentiation (IC50 ranging between ≤1 and ~10 µg/mL) [274]. Moreover, lipids extracted from the mussel Mytilus coruscus exerted a strong anti-inflammatory effect in a murine arthritis model, reducing the levels of the pro-inflammatory mediators leukotriene B₄, prostaglandin E₂, and thromboxane B₂, but also of the cytokines IL-1β, IL-6, interferon-γ, and tumor necrosis factor-α [275]. Notably, a similar preparation was tested in a clinical trial involving rheumatoid arthritis patients with similar outcomes [276]. Finally, the two sea hares Aplysia fasciata and Aplysia punctata (Anaspidea) were shown to produce immunomodulatory lipids which suppressed the activity of pro-inflammatory enzymes and nitric oxide production in vitro (IC50 77 and 74 µg/mL, respectively) [277].
Echinodermata are another phylum of marine animals which synthesize bioactive compounds with immunomodulatory properties [278]. Echinozoa, or sea urchins, are the best-studied group, with a recent example of an anti-inflammatory lead compound identified in Scaphechinus mirabilis (described in detail in Section 8.1) [279]. Another example includes Isostichopus badionotus, whose extracts suppress the expression of pro-inflammatory genes in vivo [280]. Moreover, a recent study described the anti-inflammatory activity of the protein cargo of extracellular vesicles of the sea cucumber (Holothuroidea) Stichopus japonicus, showing a strong inhibition of the release of pro-inflammatory cytokines by cultured synoviocytes, a cell type involved in the pathogenesis of osteoarthritis (used at 10 µg/mL) [281].
Lastly, the phylum Porifera contains several species of marine sponges, especially of the genus Hyrtios, which are sources of bioactive compounds, mainly alkaloids and terpenoids [282]. Early studies have reported the in vitro and in vivo anti-inflammatory activity of scalaristerol (5alpha,8alpha-dihydroxycholest-6-en-3beta-ol) and callysterol (ergosta-5,11-dien-3beta-ol), isolated from Scalarispongia aqabaensis (Thorectidae) and Callyspongia siphonella (Callyspongidae), respectively [283], and from Aplysina caissara (Aplysinidae), Haliclona sp. (Chaliunidae), and Dragmacidon reticulatum (Axinellidae), although the chemical nature of their bioactive compounds could not be identified [284]. Recently, fistularins (bromotyrosine acids) isolated from Ecionemia acervus (Ancorinidae) strongly inhibited the activity of pro-inflammatory enzymes and the release of inflammatory cytokines from cultured macrophages (tested range between 5 and μM) [285]. Similarly, the brominated alkaloid aeroplysinin derived from Aplysina aerophoba displayed inhibitory effects on cultured vascular endothelial cells by suppressing the NF-kB pathway (IC50 3 µM) [286]. Furthermore, the norditerpene dihydrogracilin A, derived from the Antarctic sponge Dendrilla membranosa (Darwinellidae), suppressed the NF-kB pathway in cultured human peripheral blood mononuclear cells and dampened the production of the pro-inflammatory cytokine IL-6 (tested range between 0.3 and 10 µM) [287]. Finally, lipids extracted from Halichondria sitiens exerted immunomodulatory effects on cultured dendritic cells (used at 10 µg/mL) by suppressing the secretion on the pro-inflammatory cytokines IL-12 and Il-6, but also prevented the production of IFN-γ by CD4+ T lymphocytes, thus blocking the so-called Th1-type immune response (explained in detail in Section 8.2) [288].

7.3. Mangrove Habitats

Mangrove forests are threatened coastal habitats at the interface between terrestrial and marine tropical environments in which salt-tolerant plants (halophytes) create unique ecosystems hosting a plethora of interacting microorganisms [289]. Mangroves are widely consumed in the TM of Southern India [290,291,292,293], and recent ethnopharmacological studies have isolated several phytochemicals with anti-inflammatory and immunomodulatory properties from Aegiceras corniculatum [294], Rhizophora mucronata [295], and Sonneratia apetala [296]. Notably, the leaf extracts from A. corniculatum inhibited the production of pro-inflammatory cytokines (TNF-α, IL-6, and IL-12) by in vitro cultured immune cells [295], while the extracts from Lumnitzera racemosa displayed anti-angiogenic properties (IC50 ranging between 2,57 and 4,95 µM) [297]. Moreover, agalloide terpenoids from Ceriops decandra (tested at 100 μM), and, mainly, Excoecaria agallocha, suppressed NF-kB pathway activation [298,299,300]. Besides providing new phytopharmaceuticals, mangrove forests are suitable habitats for bioprospecting microbial compounds [301]. For instance, two new sesquiterpenoid derivatives (elgonenes M and N used at 5 and 20 μM, respectively) were identified in the fungus Roussoella sp. after isolation from a mangrove sediment, which inhibited the synthesis of pro-inflammatory cytokines by cultured immune cells [302].

8. Marine Pharmacology, Quo Vadis?

Several marine lead compounds are currently being assessed in pre-clinical and clinical studies, while seven marine drugs have already received “first-in-class” status, i.e., are endowed with “new and unique mechanism(s) of action” [303,304,305,306]. Most marine drugs in clinical use find application in cancer immunotherapy, as antibody–drug conjugates and in the management of chronic inflammatory conditions (reviewed in detail in [307,308]). Two examples of recently identified marine molecules are provided in the following paragraphs.

8.1. The Sea Urchin Echinochrome A and Its Applications in Systemic Sclerosis

Systemic sclerosis (SSc) is a rare immune-mediated connective tissue disease characterized by microvascular damage followed by aberrant autoimmune responses of the skin and internal organs, including the gastrointestinal tract, kidneys, lungs, and heart [309]. Fibrosis is a hallmark of SSc pathogenesis. This process is driven by activated pro-fibrotic myofibroblasts, highly differentiated cells which produce contractile proteins such as alpha-smooth muscle actin, resulting in excessive extracellular matrix deposition [310]. Although myofibroblasts contribute to the physiological process of wound healing in damaged tissues, their aberrant activation contributes to diffuse fibrosis and chronic inflammation [311,312]. Innate immune cells—particularly monocytes and macrophages—are established mediators of the fibrotic process in SSc [313]. Therefore, the discovery of novel immunomodulatory and anti-fibrotic molecules is of great clinical relevance for slowing SSc progression.
The marine compound Echinochrome A (6-ethyl-2,3,5,7,8-pentahydroxy-1,4-naphthoquinone, EchA) is a natural pigment from the echinoderm Scaphechinus mirabilis [314] endowed with antioxidant anti-fibrotic properties [315,316,317,318]. Recently, it was reported that EchA reduced collagen deposition and alleviated dermal thickness in an SSc animal model [279]. In this study, bleomycin was inoculated in the mouse skin for three weeks to induce dermal injury and to activate pro-inflammatory immune cells. The administration of EchA suppressed different mechanisms involved in the fibrotic process, including fibroblast activation and myofibroblast maturation; tumor growth factor (TGF)-β1-mediated expression of smooth muscle actin; phosphorylation of pro-fibrotic transcription factors in skin fibroblasts; and, finally, differentiation of macrophages into both M1 and M2 cells (Figure 3). These effects resulted in lower serum concentrations of pro-inflammatory cytokines TNF-α and IFN-γ. Overall, EchA is a promising marine lead compound with anti-fibrotic properties and, thus, potential application in the clinical management of SSc. Recently, a novel administration system based on polymeric nanofibers was developed to improve the water solubility of EchA. This pharmacological advance is expected to promote the controlled release of the drug, enhancing its bioavailability [319].

8.2. Deep-Sea Bacteria Exopolysaccharides and Their Applications in Cancer Immunotherapy

The interplay between immunity and tumorigenesis is a cornerstone of cancer biology, since the immune system exerts a multifaceted influence in terms of thwarting tumor initiation, progression, and metastasis [320]. Cancer immunotherapy emerged in the late twentieth century with the observation by American physician William Coley that sarcomas shrunk following inoculation of the tumor mass with killed bacteria [321]. It is now well established that tumor recognition and rejection by the immune system involve a complex dialogue between adaptive and innate immune cell types, including CD8+ cytotoxic T cells, CD4+ helper T (Th) cells (Th1, Th2, and Th17 lineages), regulatory T cells (Tregs), and myeloid-derived suppressor cells [322]. Moreover, macrophages—highly adaptable phagocytic immune cells [323]—can act as antigen-presenting cells (APCs) and differentiate into classically activated (M1) and alternatively activated (M2) types, with the former promoting inflammation and the latter fostering tissue repair [324]. M1 and M2, however, are the extremes of a broader cell type spectrum [325,326,327], since macrophages are known to interfere with tumorigenesis by influencing angiogenesis, fibrosis, and tumor cell phagocytosis. Moreover, macrophages orchestrate immunosurveillance by expressing costimulatory molecules like CD86 (B7-2) or T cell inhibitory molecules and promote the recruitment of immunosuppressive T-reg cells [328].
CD86 presented by APCs can bind either to cytotoxic T lymphocyte antigen 4 (CTLA-4) or CD28 on the surface of CD4+ and CD8+ T cells, causing their inhibition or activation, respectively [329]. Notably, upon cytokine signaling, APCs mediate the “immune synapse”, activating cytotoxic CD8+ T cells thanks the stimulatory and inhibitory receptors programmed cell death 1 (PD-1) and CTLA-4 [330]. Despite immune surveillance, neoplastic cells can still escape the immune system defense mechanisms [331]. Immunotherapy aims at overcoming this phenomenon by unleashing the host immune system against malignant cells. At present, immunotherapy approaches have been successful in promoting positive clinical responses across multiple cancer types [331].
The discovery of immune checkpoint inhibitors by American physician D. R. Leach prompted the use of antibodies to block CTLA-4 and trigger robust immune responses to achieve tumor shrinkage [332]. Currently, immune checkpoint inhibitors such as anti-CTLA-4, anti-PD-1, and anti-PD-L1 are regularly used in clinical practice to target regulatory pathways in T cells, essentially to reactivate the immune response against malignant cells [333,334]. However, the consequence of excessive activation of the immune system is the onset of autoimmune diseases such as rheumatic polymyalgia or serum-negative arthritis [335]. In this context, the dysregulation of the delicate balance between M1/M2 macrophages contributes to the pathogenesis of autoimmune diseases such as rheumatoid arthritis [336]. The understanding of the complex interplay between cancer and autoimmunity represents a continuously advancing area of study. Therefore, the identification of novel immunoactive molecules is crucial to developing new therapeutic strategies.
A recent study described the immunostimulating effect of a marine exopolysaccharide (EPS) produced by the deep-sea psychrotolerant Gram-positive bacterium Planococcus rifietoensis AP-5 [233]. This compound was tested on in vitro-cultured THP-1 monocytes differentiated into macrophage-like cells and treated with different EPS concentrations (5, 10, 20, 50, and 100 μg/mL). The authors reported negligible cell toxicity at low dosages, but increased phagocytic activity and high cytokine (IL-10, IL-6, IL-1β, and TNF-α) production, suggesting strong immunoregulatory properties of EPS on innate immune responses and, thus, a potential application of this marine polysaccharide as a complementary agent in cancer immunotherapy (Figure 4).

8.3. Synthetic Biology and Molecular Pharming in Microalgae

The genetic manipulation of microalgal genomes represents a booming field in biotechnology, projecting photosynthetic microbes as viable alternatives to conventional heterotrophic hosts (bacteria and yeasts) for the production of high-value recombinant therapeutics [337,338]. Advanced genetic tools are constantly being developed to: (i) achieve high-expression of foreign DNA sequences coupled to synthetic cis-acting regulatory elements [339,340]; (ii) introduce multigene expression constructs [341]; (iii) conduct iterative editing interventions in the nuclear genome [342]; and (iv) exploit the chloroplast genome for metabolic engineering and production of recombinant proteins [343,344]. Microalgae are suitable platforms for producing recombinant protein-based therapeutics since they perform eukaryotic post-translational modifications and can be engineered to secrete heterologous products in the cultivation media [345]. Different classes of recombinant therapeutics can be produced in microalgae, including full-length antibodies [346], anti-cancer cytokines [347], and immune receptors [348].
Furthermore, genetic egineering can be employed to enhance the yield of functional metabolites. For instance, endogenous metabolic circuits can be rewired to hyperaccumulate specific pathway intermediates or modified to accumulate non-native metabolites. Alternatively, entirely new biosynthetic pathways can be introduced to produce exotic metabolites starting from endogenous substrates [349,350]. Metabolic engineering in the model freshwater chlorophyte Chlamydomonas reinhardtii resulted in the synthesis of the non-native ketocarotenoid astaxanthin via overexpression of two heterologous biosynthetic genes [351] and via CRISPR-Cas9-based gene inactivation coupled to transgenesis [352]. In this respect, the recent genome annotation of Haematococcus lacustris is expected to facilitate the genetic engineering of astaxanthin accumulation, both in the native producer and in heterologous hosts [353].
Indeed, although most genetic tools have been established in Chlamydomonas reinhardtii, engineering strategies are currently being tested in non-conventional strains, including the marine rhodophyte Porphyridium purpureum, in which the expression of a glycosylated viral antigen was recently reported [354]. This is expected to significantly expand the range of therapeutic uses of microalgae, with respect to both recombinant protein expression [355] and hyper-accumulation of high-value pigments and lipids [356,357,358,359]. Finally, current developments of synthetic biology in marine cyanobacteria are expected to introduce significant novelties into the biomanufacturing of therapeutics in these highly productive photosynthetic microbes [360,361].

8.4. The Marine Viral Dark Matter and Its Potential for Medical Biotechnology

Despite not finding a place in the tree of life, viruses are the undisputed engines of evolution in the marine biosphere and are major drivers of its biogeochemical cycles [362,363]. The genetic complexity of the “marine viral dark matter” is just beginning to surface through recent studies [364], and is expected to bring about not only new insights into ecophysiological dynamics, but also innovations in biomedicine [365,366]. Historically, viruses have been a source of inspiration in the development of biomedical applications like vaccine production, cellular transfection, and phage therapy, to name a few. Of particular interest are phytoplankton-infecting viruses like Phycodnaviridae [367] and Cyanophages [368], infecting marine eukaryotic microalgae and cyanoprokaryotes, respectively. Moreover, certain structural features of viral proteins have broader relevance for biotechnology. One example is inteins, self-cleavable protein splicing elements enriched in marine viral genomes, which have been engineered into valuable biotechnological tools [369,370]. However, due to the scarcity of reports describing the use of marine viruses in biotechnology, at present, it is difficult to predict their impact on future developments of biomedical applications.

9. Conclusions

Marine pharmacology has come a long way, from superstitious practices to present-day high-throughput drug discovery pipelines. During the last three decades, the bioprospecting of marine environments has identified hundreds of lead compounds with potential applications in the clinical management of chronic inflammatory diseases and cancer. Several of these will likely be implemented as complementary agents in the clinical practice along with already established therapeutics, such as anti-cytokines, monoclonal antibodies, and immune checkpoint inhibitors.
It should be noted, however, that the developing “ocean blue economy” is threatening marine biodiversity due to the intense activities of shipping, transportation, fisheries, tourism, and renewable energy production. Among these, seabed mining has been proposed as a severe cause of biodiversity erosion [371,372,373,374], and the ecological impact of this industrial activity was revealed by metagenomics analysis showing a reduction of deep-sea microbial biodiversity [375]. Indeed, in contrast with land environments, the high seas are still a largely ungoverned and vast “no man’s land” lacking sustainable planning for resource management [376].
Moreover, anthropogenic climate change has manifold impacts on marine ecophysiology. It should be noted that the distribution of global marine plankton follows latitudinal gradients, with a steady decline towards the poles. On the one hand, ocean warming is expected to cause a tropicalization of plankton diversity in temperate and polar waters, putting at risk these under-explored fragile ecosystems and, thus, precluding future bioprospecting endeavours [377]. On the other hand, ocean acidification, a direct consequence of rising atmospheric CO2 levels, is suggested to interfere with the metabolism of marine flora, particularly of seaweeds, affecting their polysaccharide, fatty acid, and secondary metabolite composition and profile and potentially altering their reported pharmacological uses [378]. Therefore, the ultimate frontier of marine pharmacology lies in the development of sustainable biomanufacturing platforms of therapeutic compounds, away from low-yielding and threatened natural producers, through synthetic biology approaches in photosynthetic microbes.

Author Contributions

Conceptualization, E.A.C.; writing—original draft preparation, E.A.C., R.C. (Rosanna Campitiello); writing—review and editing, M.C., S.N.A., R.C. (Roberto Caferri), V.F.P., J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We wish to thank Arianna (Cai Hang) for assistance with the revision of Chinese writing in the text.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Paine, L. The Sea and Civilization: A Maritime History of the World; Knopf Doubleday Publishing Group: New York, NY, USA, 2013. [Google Scholar]
  2. Deacon, M. Scientists and the Sea, 1650–1900: A Study of Marine Science; Academic Press: Cambridge, MA, USA, 1971. [Google Scholar]
  3. Mora, C.; Tittensor, D.P.; Adl, S.; Simpson, A.G.; Worm, B. How many species are there on Earth and in the ocean? PLoS Biol. 2011, 9, e1001127. [Google Scholar] [CrossRef] [PubMed]
  4. Louca, S.; Mazel, F.; Doebeli, M.; Parfrey, L.W. A census-based estimate of Earth’s bacterial and archaeal diversity. PLoS Biol. 2019, 17, e3000106. [Google Scholar] [CrossRef] [PubMed]
  5. Baldrian, P.; Větrovský, T.; Lepinay, C.; Kohout, P. High-throughput sequencing view on the magnitude of global fungal diversity. Fungal Divers. 2022, 114, 539–547. [Google Scholar] [CrossRef]
  6. Bouchet, P.; Decock, W.; Lonneville, B.; Vanhoorne, B.; Vandepitte, L. Marine biodiversity discovery: The metrics of new species descriptions. Front. Mar. Sci. 2023, 10, 929989. [Google Scholar] [CrossRef]
  7. Wainwright, C.L.; Teixeira, M.M.; Adelson, D.L.; Braga, F.C.; Buenz, E.J.; Campana, P.R.V.; David, B.; Glaser, K.B.; Harata-Lee, Y.; Howes, M.-J.R.; et al. Future directions for the discovery of natural product-derived immunomodulating drugs: An IUPHAR positional review. Pharmacol. Res. 2022, 177, 106076. [Google Scholar] [CrossRef] [PubMed]
  8. Moles, J.; Torrent, A.; Alcaraz, M.J.; Ruhí, R.; Avila, C. Anti-inflammatory activity in selected Antarctic benthic organisms. Front. Mar. Sci. 2014, 1, 24. [Google Scholar] [CrossRef]
  9. Lindequist, U. Marine-Derived Pharmaceuticals—Challenges and Opportunities. Biomol. Ther. 2016, 24, 561–571. [Google Scholar] [CrossRef] [PubMed]
  10. Kraiem, M.; Ben Hamouda, S.; Eleroui, M.; Ajala, M.; Feki, A.; Dghim, A.; Boujhoud, Z.; Bouhamed, M.; Badraoui, R.; Pujo, J.M.; et al. Anti-Inflammatory and Immunomodulatory Properties of a Crude Polysaccharide Derived from Green Seaweed Halimeda tuna: Computational and Experimental Evidences. Mar. Drugs 2024, 22, 85. [Google Scholar] [CrossRef] [PubMed]
  11. Haque, N.; Parveen, S.; Tang, T.; Wei, J.; Huang, Z. Marine Natural Products in Clinical Use. Mar. Drugs 2022, 20, 528. [Google Scholar] [CrossRef] [PubMed]
  12. Mayer, A.M.; Pierce, M.; Reji, M.; Wu, A.C.; Jekielek, K.K.; Le, H.Q.; Howe, K.; Butt, M.; Seo, S.; Newman, D.J.; et al. Marine-Derived Pharmaceuticals in Clinical Trials in 2022. J. Pharmacol. Exp. Ther. 2023, 385, 299. [Google Scholar]
  13. Carroll, A.R.; Copp, B.R.; Grkovic, T.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2024, 41, 162–207. [Google Scholar] [CrossRef] [PubMed]
  14. Wu, D.; Jin, Y.; Xing, Y.; Abate, M.D.; Abbasian, M.; Abbasi-Kangevari, M.; Abbasi-Kangevari, Z.; Abd-Allah, F.; Abdelmasseh, M.; Abdollahifar, M.-A.; et al. Global, regional, and national incidence of six major immune-mediated inflammatory diseases: Findings from the global burden of disease study 2019. eClinicalMedicine 2023, 64, 102193. [Google Scholar] [CrossRef] [PubMed]
  15. Miller, F.W. The increasing prevalence of autoimmunity and autoimmune diseases: An urgent call to action for improved understanding, diagnosis, treatment, and prevention. Curr. Opin. Immunol. 2023, 80, 102266. [Google Scholar] [CrossRef] [PubMed]
  16. Michels, N.; Aart, C.v.; Morisse, J.; Huybrechts, I. Chronic Inflammation Toward Cancer Incidence: An Epidemiological Systematic Review. J. Glob. Oncol. 2018, 4, 24s. [Google Scholar] [CrossRef]
  17. Strzelec, M.; Detka, J.; Mieszczak, P.; Sobocińska, M.K.; Majka, M. Immunomodulation—A general review of the current state-of-the-art and new therapeutic strategies for targeting the immune system. Front. Immunol. 2023, 14, 1127704. [Google Scholar] [CrossRef] [PubMed]
  18. Wraith, D.C. The Future of Immunotherapy: A 20-Year Perspective. Front. Immunol. 2017, 8, 1668. [Google Scholar] [CrossRef] [PubMed]
  19. Banday, A.H.; Azha, N.u.; Farooq, R.; Sheikh, S.A.; Ganie, M.A.; Parray, M.N.; Mushtaq, H.; Hameed, I.; Lone, M.A. Exploring the potential of marine natural products in drug development: A comprehensive review. Phytochem. Lett. 2024, 59, 124–135. [Google Scholar] [CrossRef]
  20. Karthikeyan, A.; Joseph, A.; Nair, B.G. Promising bioactive compounds from the marine environment and their potential effects on various diseases. J. Genet. Eng. Biotechnol. 2022, 20, 14. [Google Scholar] [CrossRef] [PubMed]
  21. Malve, H. Exploring the ocean for new drug developments: Marine pharmacology. J. Pharm. Bioallied Sci. 2016, 8, 83–91. [Google Scholar] [CrossRef] [PubMed]
  22. Bisaria, K.; Sinha, S.; Srivastava, A.; Singh, R. Marine Pharmacognosy: An Overview of Marine-Derived Pharmaceuticals. In Marine Niche: Applications in Pharmaceutical Sciences: Translational Research; Nathani, N.M., Mootapally, C., Gadhvi, I.R., Maitreya, B., Joshi, C.G., Eds.; Springer: Singapore, 2020; pp. 361–381. [Google Scholar]
  23. Li, C.-Q.; Ma, Q.-Y.; Gao, X.-Z.; Wang, X.; Zhang, B.-L. Research Progress in Anti-Inflammatory Bioactive Substances Derived from Marine Microorganisms, Sponges, Algae, and Corals. Mar. Drugs 2021, 19, 572. [Google Scholar] [CrossRef] [PubMed]
  24. Mayer, A.M.S.; Pierce, M.L.; Howe, K.; Rodríguez, A.D.; Taglialatela-Scafati, O.; Nakamura, F.; Fusetani, N. Marine pharmacology in 2018: Marine compounds with antibacterial, antidiabetic, antifungal, anti-inflammatory, antiprotozoal, antituberculosis and antiviral activities; affecting the immune and nervous systems, and other miscellaneous mechanisms of action. Pharmacol. Res. 2022, 183, 106391. [Google Scholar] [CrossRef]
  25. Ahmad, B.; Shah, M.; Choi, S. Oceans as a Source of Immunotherapy. Mar. Drugs 2019, 17, 282. [Google Scholar] [CrossRef] [PubMed]
  26. Montuori, E.; de Pascale, D.; Lauritano, C. Recent Discoveries on Marine Organism Immunomodulatory Activities. Mar. Drugs 2022, 20, 422. [Google Scholar] [CrossRef] [PubMed]
  27. Khursheed, M.; Ghelani, H.; Jan, R.K.; Adrian, T.E. Anti-Inflammatory Effects of Bioactive Compounds from Seaweeds, Bryozoans, Jellyfish, Shellfish and Peanut Worms. Mar. Drugs 2023, 21, 524. [Google Scholar] [CrossRef] [PubMed]
  28. Bekiari, M. Marine Bioprospecting: Understanding the Activity and Some Challenges Related to Environmental Protection, Scientific Research, Ethics, and the Law. In Blue Planet Law: The Ecology of our Economic and Technological World; Garcia, M.d.G., Cortês, A., Eds.; Springer International Publishing: Cham, Switzerland, 2023; pp. 237–252. [Google Scholar]
  29. Reverter, M.; Rohde, S.; Parchemin, C.; Tapissier-Bontemps, N.; Schupp, P.J. Metabolomics and Marine Biotechnology: Coupling Metabolite Profiling and Organism Biology for the Discovery of New Compounds. Front. Mar. Sci. 2020, 7, 613471. [Google Scholar] [CrossRef]
  30. Bayona, L.M.; de Voogd, N.J.; Choi, Y.H. Metabolomics on the study of marine organisms. Metabolomics 2022, 18, 17. [Google Scholar] [CrossRef] [PubMed]
  31. Thukral, M.; Allen, A.E.; Petras, D. Progress and challenges in exploring aquatic microbial communities using non-targeted metabolomics. ISME J. 2023, 17, 2147–2159. [Google Scholar] [CrossRef] [PubMed]
  32. Antunes, E.M.; Beukes, D.R.; Caro-Diaz, E.J.E.; Narchi, N.E.; Tan, L.T.; Gerwick, W.H. Chapter 5—Medicines from the sea. In Oceans and Human Health (Second Edition); Fleming, L.E., Alcantara Creencia, L.B., Gerwick, W.H., Goh, H.C., Gribble, M.O., Maycock, B., Solo-Gabriele, H., Eds.; Academic Press: San Diego, CA, USA, 2023; pp. 103–148. [Google Scholar] [CrossRef]
  33. Oyebode, O.; Kandala, N.-B.; Chilton, P.J.; Lilford, R.J. Use of traditional medicine in middle-income countries: A WHO-SAGE study. Health Policy Plan. 2016, 31, 984–991. [Google Scholar] [CrossRef]
  34. World Health Organization. WHO Global Report on Traditional and Complementary Medicine 2019; World Health Organization: Geneva, Switzerland, 2019. [Google Scholar]
  35. Jansen, C.; Baker, J.D.; Kodaira, E.; Ang, L.; Bacani, A.J.; Aldan, J.T.; Shimoda, L.M.N.; Salameh, M.; Small-Howard, A.L.; Stokes, A.J.; et al. Medicine in motion: Opportunities, challenges and data analytics-based solutions for traditional medicine integration into western medical practice. J. Ethnopharmacol. 2021, 267, 113477. [Google Scholar] [CrossRef] [PubMed]
  36. Buenz, E.J.; Verpoorte, R.; Bauer, B.A. The Ethnopharmacologic Contribution to Bioprospecting Natural Products. Annu. Rev. Pharmacol. Toxicol. 2018, 58, 509–530. [Google Scholar] [CrossRef] [PubMed]
  37. Pérez-Lloréns, J.L.; Mouritsen, O.G.; Rhatigan, P.; Cornish, M.L.; Critchley, A.T. Seaweeds in mythology, folklore, poetry, and life. J. Appl. Phycol. 2020, 32, 3157–3182. [Google Scholar] [CrossRef]
  38. Moseley, M.E. The Maritime Foundations of Andean Civilization; Springer: Berlin/Heidelberg, Germany, 1975. [Google Scholar]
  39. Dillehay, T.D.; Ramírez, C.; Pino, M.; Collins, M.B.; Rossen, J.; Pino-Navarro, J.D. Monte Verde: Seaweed, food, medicine, and the peopling of South America. Science 2008, 320, 784–786. [Google Scholar] [CrossRef] [PubMed]
  40. Déléris, P.; Nazih, H.; Bard, J.M. Chapter 10—Seaweeds in Human Health. In Seaweed in Health and Disease Prevention; Fleurence, J., Levine, I., Eds.; Academic Press: San Diego, CA, USA, 2016; pp. 319–367. [Google Scholar] [CrossRef]
  41. Ruiz Vigo, W.M. Importancia de las Cyanophytas. Rev. Perspect. 2019, 20, 240–244. [Google Scholar] [CrossRef]
  42. Godínez-Ortega, J.L.; Ortega, M.M.; Garduño-Solórzano, G.; Oliva, M.G.; Vilaclara, G. Traditional knowledge of Mexican continental algae. J. Ethnobiol. 2001, 21, 57–88. [Google Scholar]
  43. Sowunmi, A.A. Fin-fishes in Yorùbá natural healing practices from southwest Nigeria. J. Ethnopharmacol. 2007, 113, 72–78. [Google Scholar] [CrossRef] [PubMed]
  44. Begossi, A.; Ramires, M. Fish Folk Medicine of Caiçara (Atlantic Coastal Forest) and Caboclo (Amazon Forest) Communities. In Animals in Traditional Folk Medicine: Implications for Conservation; Alves, R.R.N., Rosa, I.L., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 91–108. [Google Scholar]
  45. El-Deir, A.C.A.; Collier, C.A.; Almeida Neto, M.S.d.; Silva, K.M.d.S.; Policarpo, I.d.S.; Araujo, T.A.S.; Alves, R.R.N.; de Albuquerque, U.P.; Moura, G.J.B.d. Ichthyofauna used in traditional medicine in Brazil. Evid.-Based Complement. Altern. Med. 2012, 2012, 474716. [Google Scholar] [CrossRef] [PubMed]
  46. Alves, R.R.; Rosa, I.L. From cnidarians to mammals: The use of animals as remedies in fishing communities in NE Brazil. J. Ethnopharmacol. 2006, 107, 259–276. [Google Scholar] [CrossRef] [PubMed]
  47. Luo, B.; Nong, Y.; Zhang, T.; Zhang, S.; Hu, R. The use and sustainable development of marine animal drugs by the Kinh people in Beibu Gulf. Front. Mar. Sci. 2023, 10, 1175316. [Google Scholar] [CrossRef]
  48. Alino, P.M.; Cajipe, G.J.B.; Ganzon-Fortes, E.T.; Licuanan, W.R.Y.; Montano, N.E.; Tupas, L.M. The Use of Marine Organisms in Folk Medicine and Horticulture: A Preliminary Study; Seaweed Information Center (SICEN), University of the Philippines: Quezon City, Philippines, 1990. [Google Scholar]
  49. Salleh, H.; Mohamed, W.N.; Mat, N.H.N.; Yusof, Y.; Syamimi, N. Traditional medicines from marine resources: Understanding the consumer’s knowledge and perceptions. Int. J. Adv. Appl. Sci. 2020, 7, 110–118. [Google Scholar] [CrossRef]
  50. Mahomoodally, M.F.; Sanaa, D.A.; Zengin, G.; Gallo, M.; Montesano, D. Traditional Therapeutic Uses of Marine Animal Parts and Derived Products as Functional Foods—A Systematic Review. Food Rev. Int. 2023, 39, 827–857. [Google Scholar] [CrossRef]
  51. Guo, Y.; Ding, Y.; Xu, F.; Liu, B.; Kou, Z.; Xiao, W.; Zhu, J. Systems pharmacology-based drug discovery for marine resources: An example using sea cucumber (Holothurians). J. Ethnopharmacol. 2015, 165, 61–72. [Google Scholar] [CrossRef] [PubMed]
  52. Miller, E.A.; McClenachan, L.; Uni, Y.; Phocas, G.; Hagemann, M.E.; Van Houtan, K.S. The historical development of complex global trafficking networks for marine wildlife. Sci. Adv. 2019, 5, eaav5948. [Google Scholar] [CrossRef] [PubMed]
  53. Siddiqui, S.A.; Baruah, S.; Wu, Y.S.; Yuansah, S.C.; Castro-Muñoz, R.; Szymkowiak, A.; Kulawik, P. Investigating the sustainability, utilisation, consumption and conservation of sea mammals—A systematic review. Sustain. Prod. Consum. 2024, 46, 400–417. [Google Scholar] [CrossRef]
  54. O’Malley, M.p.; Townsend, K.A.; Hilton, P.; Heinrichs, S.; Stewart, J.D. Characterization of the trade in manta and devil ray gill plates in China and South-east Asia through trader surveys. Aquat. Conserv. Mar. Freshw. Ecosyst. 2017, 27, 394–413. [Google Scholar] [CrossRef]
  55. Huxtable, R.J. The pharmacology of extinction. J. Ethnopharmacol. 1992, 37, 1–11. [Google Scholar] [CrossRef] [PubMed]
  56. Porter, L.; Lai, H.Y. Marine Mammals in Asian Societies; Trends in Consumption, Bait, and Traditional Use. Front. Mar. Sci. 2017, 4, 47. [Google Scholar] [CrossRef]
  57. Nurse, A. Preventing marine wildlife crime: An evaluation of legal protection and enforcement perspectives. Front. Conserv. Sci. 2023, 3, 1102823. [Google Scholar] [CrossRef]
  58. Müller, W.E.; Batel, R.; Schröder, H.C.; Müller, I.M. Traditional and Modern Biomedical Prospecting: Part I-the History: Sustainable Exploitation of Biodiversity (Sponges and Invertebrates) in the Adriatic Sea in Rovinj (Croatia). Evid. Based Complement. Altern. Med. 2004, 1, 71–82. [Google Scholar] [CrossRef] [PubMed]
  59. Voultsiadou, E. Therapeutic properties and uses of marine invertebrates in the ancient Greek world and early Byzantium. J. Ethnopharmacol. 2010, 130, 237–247. [Google Scholar] [CrossRef] [PubMed]
  60. Quave, C.L.; Pieroni, A. Mediterranean Zootherapy: A Historical to Modern Perspective. In Animals in Traditional Folk Medicine: Implications for Conservation; Alves, R.R.N., Rosa, I.L., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 303–316. [Google Scholar]
  61. Papadopoulos, D. Medical Knowledge and the Aquatic Animals in Claudius Aelianus’s on the Characteristics of Animals. Available online: https://journals.openedition.org/rursuspicae/2357 (accessed on 26 May 2024).
  62. Pérez-Lloréns, J.L.; Critchley, A.T.; Cornish, M.L.; Mouritsen, O.G. Saved by seaweeds (II): Traditional knowledge, home remedies, medicine, surgery, and pharmacopoeia. J. Appl. Phycol. 2023, 35, 2049–2068. [Google Scholar] [CrossRef]
  63. Buckley, S.; Hardy, K.; Hallgren, F.; Kubiak-Martens, L.; Miliauskienė, Ž.; Sheridan, A.; Sobkowiak-Tabaka, I.; Subirà, M.E. Human consumption of seaweed and freshwater aquatic plants in ancient Europe. Nat. Commun. 2023, 14, 6192. [Google Scholar] [CrossRef]
  64. Cavaillon, J.M. Once upon a time, inflammation. J. Venom. Anim. Toxins Incl. Trop. Dis. 2021, 27, e20200147. [Google Scholar] [CrossRef] [PubMed]
  65. Millward, J.A.; Millward, J.A. The biological silk road. In The Silk Road: A Very Short Introduction; Oxford University Press: Oxford, UK, 2013; p. 39. [Google Scholar] [CrossRef]
  66. Millward, J.A.; Millward, J.A. The technological silk road. In The Silk Road: A Very Short Introduction; Oxford University Press: Oxford, UK, 2013; p. 64. [Google Scholar] [CrossRef]
  67. Peters, M.A. The ancient Silk Road and the birth of merchant capitalism. Educ. Philos. Theory 2021, 53, 955–961. [Google Scholar] [CrossRef]
  68. Bradley, S. The Silk Road and Sources of Chinese Medicine Expansion: Part 2—Formularies. Chin. Med. Cult. 2018, 1, 68–70. [Google Scholar] [CrossRef]
  69. Bradley, S. The Silk Road and Sources of Chinese Medicine Expansion: Part 1—Materia Medica. Chin. Med. Cult. 2018, 1, 29–31. [Google Scholar] [CrossRef]
  70. Díaz, J.L. Ethnopharmacology of sacred psychoactive plants used by the Indians of Mexico. Annu. Rev. Pharmacol. Toxicol. 1977, 17, 647–675. [Google Scholar] [CrossRef]
  71. Anderson, T.C. Medicine and Travel in the Colonies (1600–1750). In Encyclopedia of Early Modern Philosophy and the Sciences; Jalobeanu, D., Wolfe, C.T., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 1–11. [Google Scholar]
  72. World Health Organization. The International Pharmacopoeia; World Health Organization: Geneva, Switzerland, 2022; Online Version. [Google Scholar]
  73. Silverstein, A.M. Autoimmunity versus horror autotoxicus: The struggle for recognition. Nat. Immunol. 2001, 2, 279–281. [Google Scholar] [CrossRef] [PubMed]
  74. Mackay, I.R. Travels and travails of autoimmunity: A historical journey from discovery to rediscovery. Autoimmun. Rev. 2010, 9, A251–A258. [Google Scholar] [CrossRef] [PubMed]
  75. Kaufmann, S.H.E. Immunology’s Coming of Age. Front. Immunol. 2019, 10, 684. [Google Scholar] [CrossRef] [PubMed]
  76. Plytycz, B.; Seljelid, R. From inflammation to sickness: Historical perspective. Arch. Immunol. Ther. Exp. 2003, 51, 105–109. [Google Scholar]
  77. Tilden, J.E. The marine and fresh water algae of China. Lingnan Sci. J. 1929, 7, 349–398. [Google Scholar]
  78. Gao, K. Chinese studies on the edible blue-green alga, Nostoc flagelliforme: A review. J. Appl. Phycol. 1998, 10, 37–49. [Google Scholar] [CrossRef]
  79. Chase, F.M. Useful Algae; Government Printing Office: Washington, DC, USA, 1942. [Google Scholar]
  80. Tseng, C.K. The Past, Present and Future of Phycology in China; Springer: Dordrecht, The Netherlands, 2004; pp. 11–20. [Google Scholar]
  81. Zhao, Z.; Guo, P.; Brand, E. A concise classification of bencao (materia medica). Chin. Med. 2018, 13, 18. [Google Scholar] [CrossRef]
  82. Pan, M.H.; Chiou, Y.S.; Tsai, M.L.; Ho, C.T. Anti-inflammatory activity of traditional Chinese medicinal herbs. J. Tradit. Complement. Med. 2011, 1, 8–24. [Google Scholar] [CrossRef] [PubMed]
  83. Rong, X.; Church, S.K.; Galambos, I.; Li, H.; Zheng, C. The Silk Road and Cultural Exchanges between East and West; Brill: Leiden, The Netherlands, 2022; pp. 93–123. [Google Scholar] [CrossRef]
  84. Ming, C. The Transmission of Foreign Medicine via the Silk Roads in Medieval China: A Case Study of Haiyao Bencao. Asian Med. 2007, 3, 241–264. [Google Scholar] [CrossRef]
  85. Chen, X.; Ni, L.; Fu, X.; Wang, L.; Duan, D.; Huang, L.; Xu, J.; Gao, X. Molecular Mechanism of Anti-Inflammatory Activities of a Novel Sulfated Galactofucan from Saccharina japonica. Mar. Drugs 2021, 19, 430. [Google Scholar] [CrossRef] [PubMed]
  86. Pan, L.; Fu, T.; Cheng, H.; Mi, J.; Shang, Q.; Yu, G. Polysaccharide from edible alga Gloiopeltis furcata attenuates intestinal mucosal damage by therapeutically remodeling the interactions between gut microbiota and mucin O-glycans. Carbohydr. Polym. 2022, 278, 118921. [Google Scholar] [CrossRef] [PubMed]
  87. Peng, H.S.; Huang, L.Q. Foreign medicinal materials in Bencao Tujing. Zhonghua Yi Shi Za Zhi 2021, 51, 15–23. [Google Scholar] [CrossRef] [PubMed]
  88. Xiu, L.-L.; Xu, S.-C.; Wang, Q.-X.; Chen, S.-H.; Liu, H.-Y.; Zhong, G.-S. Achievements of the Compendium Bencao Tujing (Illustrated Classic of Materia Medica): A Preliminary Study. Chin. Med. Cult. 2022, 5, 84–90. [Google Scholar] [CrossRef]
  89. Zhilin, Y.; ZHANG, Y. Zhu Fan Zhi and the Maritime Road of Aromatic Medicine in the Song Dynasty. Chin. Med. Cult. 2023, 6, 349–356. [Google Scholar] [CrossRef]
  90. Buell, P.D. Eurasia, Medicine and Trade: Arabic Medicine in East Asia—How It Came to Be There and How It Was Supported, Including Possible Indian Ocean Connections for the Supply of Medicinals. In Early Global Interconnectivity across the Indian Ocean World, Volume II: Exchange of Ideas, Religions, and Technologies; Schottenhammer, A., Ed.; Springer International Publishing: Cham, Switzerland, 2019; pp. 261–293. [Google Scholar]
  91. Buell, P.D.; Anderson, E.N. Arabic Medicine in China: Tradition, Innovation, and Change; Brill: Leiden, The Netherlands, 2021. [Google Scholar] [CrossRef]
  92. Shi, J.; Yang, Y.; Zhou, X.; Zhao, L.; Li, X.; Yusuf, A.; Hosseini, M.; Sefidkon, F.; Hu, X. The current status of old traditional medicine introduced from Persia to China. Front. Pharmacol. 2022, 13, 953352. [Google Scholar] [CrossRef] [PubMed]
  93. Marcon, F.; Marcon, F. The Bencao gangmu and the World It Created. In The Knowledge of Nature and the Nature of Knowledge in Early Modern Japan; University of Chicago Press: Chicago, IL, USA, 2015; p. 28. [Google Scholar] [CrossRef]
  94. Yang, L.-E.; Lu, Q.-Q.; Brodie, J. A review of the bladed Bangiales (Rhodophyta) in China: History, culture and taxonomy. Eur. J. Phycol. 2017, 52, 251–263. [Google Scholar] [CrossRef]
  95. May, B.T. Takako Seahorses in the Ben cao gang mu and contemporary Chinese medicine. J. Aust. Chin. Med. Educ. Res. Counc. 2002, 7, 2–13. [Google Scholar]
  96. Qian, Z.-J.; Ryu, B.; Kim, M.-M.; Kim, S.-K. Free radical and reactive oxygen species scavenging activities of the extracts from seahorse, Hippocampus kuda Bleeler. Biotechnol. Bioprocess Eng. 2008, 13, 705–715. [Google Scholar] [CrossRef]
  97. Ryu, B.; Qian, Z.J.; Kim, S.K. Purification of a peptide from seahorse, that inhibits TPA-induced MMP, iNOS and COX-2 expression through MAPK and NF-kappaB activation, and induces human osteoblastic and chondrocytic differentiation. Chem. Biol. Interact. 2010, 184, 413–422. [Google Scholar] [CrossRef] [PubMed]
  98. Ryu, B.; Qian, Z.J.; Kim, S.K. SHP-1, a novel peptide isolated from seahorse inhibits collagen release through the suppression of collagenases 1 and 3, nitric oxide products regulated by NF-kappaB/p38 kinase. Peptides 2010, 31, 79–87. [Google Scholar] [CrossRef]
  99. Malemud, C.J. The role of the JAK/STAT signal pathway in rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis. 2018, 10, 117–127. [Google Scholar] [CrossRef] [PubMed]
  100. Brown, K.D.; Claudio, E.; Siebenlist, U. The roles of the classical and alternative nuclear factor-kappaB pathways: Potential implications for autoimmunity and rheumatoid arthritis. Arthritis Res. Ther. 2008, 10, 212. [Google Scholar] [CrossRef]
  101. Li, Y.; Qian, Z.J.; Kim, S.K. Cathepsin B inhibitory activities of three new phthalate derivatives isolated from seahorse, Hippocampus Kuda Bleeler. Bioorg Med. Chem. Lett. 2008, 18, 6130–6134. [Google Scholar] [CrossRef] [PubMed]
  102. Schett, G.; Zwerina, J.; Firestein, G. The p38 mitogen-activated protein kinase (MAPK) pathway in rheumatoid arthritis. Ann. Rheum. Dis. 2008, 67, 909–916. [Google Scholar] [CrossRef] [PubMed]
  103. Law, S. Dried seahorse in traditional medicine: A narrative review. Infect. Dis. Herb. Med. 2021, 2, 158. [Google Scholar] [CrossRef]
  104. Rosa, I.L.; Defavari, G.R.; Alves, R.R.N.; Oliveira, T.P.R. Seahorses in Traditional Medicines: A Global Overview. In Animals in Traditional Folk Medicine: Implications for Conservation; Alves, R.R.N., Rosa, I.L., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 207–240. [Google Scholar]
  105. Nakamura, J.K. Blaise: Implementing the Convention on International Trade in Endangered Species of Wild Fauna and Flora (CITES) through National Fisheries Legal Frameworks: A Study and a Guide. FAO Legal Guide; FAO: Rome, Italy, 2020. [Google Scholar]
  106. IUCN. The IUCN Red List of Threatened Species; IUCN: Gland, Switzerland, 2023. [Google Scholar]
  107. Walravens, H. Father Verbiest’s Chinese World Map (1674). Imago Mundi 1991, 43, 31–47. [Google Scholar] [CrossRef]
  108. Wang, Z.; Wu, Q. Restructuring Trade: Circulation of Medicinal Materials in East Asia in the 18th Century. Uisahak 2023, 32, 279–319. [Google Scholar] [CrossRef]
  109. Elshakry, M. When Science Became Western: Historiographical Reflections. Isis 2010, 101, 98–109. [Google Scholar] [CrossRef] [PubMed]
  110. Luk, C.Y.L. The First Marine Biological Station in Modern China: Amoy University and Amphioxus. In Why Study Biology by the Sea? Matlin, K.S., Maienschein, J., Ankeny, R.A., Eds.; University of Chicago Press: Chicago, IL, USA, 2020; p. 68. [Google Scholar] [CrossRef]
  111. Liu, J.; Gu, Y.-c.; Su, M.-z.; Guo, Y.-w. Chemistry and bioactivity of secondary metabolites from South China Sea marine fauna and flora: Recent research advances and perspective. Acta Pharmacol. Sin. 2022, 43, 3062–3079. [Google Scholar] [CrossRef] [PubMed]
  112. Neushul, P.; Wang, Z. Between the Devil and the Deep Sea: C. K. Tseng, Mariculture, and the Politics of Science in Modern China. Isis 2000, 91, 59–88. [Google Scholar] [CrossRef]
  113. Liang, Y.; Cheng, X.; Zhu, H.; Shutes, B.; Yan, B.; Zhou, Q.; Yu, X. Historical Evolution of Mariculture in China During Past 40 Years and Its Impacts on Eco-environment. Chin. Geogr. Sci. 2018, 28, 363–373. [Google Scholar] [CrossRef]
  114. Fu, X.-M.; Zhang, M.-Q.; Shao, C.-L.; Li, G.-Q.; Bai, H.; Dai, G.-L.; Chen, Q.-W.; Kong, W.; Fu, X.-J.; Wang, C.-Y. Chinese Marine Materia Medica Resources: Status and Potential. Mar. Drugs 2016, 14, 46. [Google Scholar] [CrossRef] [PubMed]
  115. Cao, W.; Liu, J.; Dai, Y.; Zhou, Y.; Li, R.; Yu, P. Bibliometric Analysis of Marine Traditional Chinese Medicine in Pharmacopoeia of the People’s Republic of China: Development, Differences, and Trends Directions. Evid.-Based Complement. Altern. Med. 2022, 2022, 3971967. [Google Scholar] [CrossRef] [PubMed]
  116. Lei, S.H.-l. Neither Donkey nor Horse: Medicine in the Struggle over China’s Modernity; University of Chicago Press: Chicago, IL, USA, 2014. [Google Scholar] [CrossRef]
  117. Wang, Y. The scientific nature of traditional Chinese medicine in the post-modern era. J. Tradit. Chin. Med. Sci. 2019, 6, 195–200. [Google Scholar] [CrossRef]
  118. Ju, L.; Jiang, J.; Jin, Y.; Armand, J.-P.; Charron, D. Modern immunology is crucial to revealing the biological mechanisms of traditional Chinese medicine. J. Tradit. Chin. Med. Sci. 2023, 10, 383–394. [Google Scholar] [CrossRef]
  119. Jaeger, S. A Geomedical Approach to Chinese Medicine: The Origin of the Yin-Yang Symbol; IntechOpen: London, UK, 2012. [Google Scholar]
  120. Verbeke, G. L’evolution de la Doctrine du Pneuma: Du Stoicisme a S. Augustin; Desclee de Brouwer: Bilbao, Spain, 1945. [Google Scholar]
  121. Li, Y.M.; Kelley, K.W. Traditional Chinese medicine: What does modern immunology have to do with it? Brain Behav. Immun. Integr. 2024, 5, 100044. [Google Scholar] [CrossRef]
  122. Bottaccioli, F.; Bottaccioli, A.G. The suggestions of ancient Chinese philosophy and medicine for contemporary scientific research, and integrative care. Brain Behav. Immun. Integr. 2024, 5, 100024. [Google Scholar] [CrossRef]
  123. Sun, L.; Su, Y.; Jiao, A.; Wang, X.; Zhang, B. T cells in health and disease. Signal Transduct. Target. Ther. 2023, 8, 235. [Google Scholar] [CrossRef] [PubMed]
  124. Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef] [PubMed]
  125. Lee, G.R. The Balance of Th17 versus Treg Cells in Autoimmunity. Int. J. Mol. Sci. 2018, 19, 730. [Google Scholar] [CrossRef] [PubMed]
  126. Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef] [PubMed]
  127. Ma, H.D.; Deng, Y.R.; Tian, Z.; Lian, Z.X. Traditional Chinese medicine and immune regulation. Clin. Rev. Allergy Immunol. 2013, 44, 229–241. [Google Scholar] [CrossRef] [PubMed]
  128. Yang, C.; Li, D.; Ko, C.-N.; Wang, K.; Wang, H. Active ingredients of traditional Chinese medicine for enhancing the effect of tumor immunotherapy. Front. Immunol. 2023, 14, 1133050. [Google Scholar] [CrossRef] [PubMed]
  129. Jakobsson, P.-J.; Robertson, L.; Welzel, J.; Zhang, M.; Zhihua, Y.; Kaixin, G.; Runyue, H.; Zehuai, W.; Korotkova, M.; Göransson, U. Where traditional Chinese medicine meets Western medicine in the prevention of rheumatoid arthritis. J. Intern. Med. 2022, 292, 745–763. [Google Scholar] [CrossRef] [PubMed]
  130. Leeuwenhoek, A.V. Observations, communicated to the publisher by Mr. Antony van Leewenhoeck, in a dutch letter of the 9th Octob. 1676. here English’d: Concerning little animals by him observed in rain-well-sea- and snow water; as also in water wherein pepper had lain infused. Philos. Trans. R. Soc. Lond. 1677, 12, 821–831. [Google Scholar] [CrossRef]
  131. Hooke, R. Micrographia, or, Some Physiological Descriptions of Minute Bodies Made by Magnifying Glasses: With Observations and Inquiries Thereupon. Available online: https://www.biodiversitylibrary.org/bibliography/904 (accessed on 24 May 2024).
  132. Fischer, B. Die Bakterien des Meeres Nach den Untersuchungen der Plankton-Expedition: Unter Gleichzeitiger Berücksichtigung Einiger Älterer und Neuerer Untersuchungen; Lipsius & Tischer: Chicago, IL, USA, 1894. [Google Scholar]
  133. ZoBell, C.E. Marine Microbiology: A Monograph on Hydrobacteriology; Chronica Botanica Company: Waltham, MA, USA, 1946. [Google Scholar]
  134. Waterbury, J.B.; Watson, S.W.; Guillard, R.R.L.; Brand, L.E. Widespread occurrence of a unicellular, marine, planktonic, cyanobacterium. Nature 1979, 277, 293–294. [Google Scholar] [CrossRef]
  135. Chisholm, S.W.; Olson, R.J.; Zettler, E.R.; Goericke, R.; Waterbury, J.B.; Welschmeyer, N.A. A novel free-living prochlorophyte abundant in the oceanic euphotic zone. Nature 1988, 334, 340–343. [Google Scholar] [CrossRef]
  136. Petersen, L.-E.; Kellermann, M.Y.; Schupp, P.J. Secondary Metabolites of Marine Microbes: From Natural Products Chemistry to Chemical Ecology. In YOUMARES 9—The Oceans: Our Research, Our Future: Proceedings of the 2018 Conference for YOUng MArine RESearcher in Oldenburg, Germany; Jungblut, S., Liebich, V., Bode-Dalby, M., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 159–180. [Google Scholar]
  137. Venter, J.C.; Remington, K.; Heidelberg, J.F.; Halpern, A.L.; Rusch, D.; Eisen, J.A.; Wu, D.; Paulsen, I.; Nelson, K.E.; Nelson, W. Environmental genome shotgun sequencing of the Sargasso Sea. Science 2004, 304, 66–74. [Google Scholar] [CrossRef] [PubMed]
  138. Nicholls, H. Sorcerer II: The search for microbial diversity roils the waters. PLoS Biol. 2007, 5, e74. [Google Scholar] [CrossRef] [PubMed]
  139. Rusch, D.B.; Halpern, A.L.; Sutton, G.; Heidelberg, K.B.; Williamson, S.; Yooseph, S.; Wu, D.; Eisen, J.A.; Hoffman, J.M.; Remington, K.; et al. The Sorcerer II Global Ocean Sampling expedition: Northwest Atlantic through eastern tropical Pacific. PLoS Biol. 2007, 5, e77. [Google Scholar] [CrossRef] [PubMed]
  140. Duarte, C.M. Seafaring in the 21St Century: The Malaspina 2010 Circumnavigation Expedition. Limnol. Oceanogr. Bull. 2015, 24, 11–14. [Google Scholar] [CrossRef]
  141. Vincent, F.; Ibarbalz, F.M.; Bowler, C. Chapter 15—Global marine phytoplankton revealed by the Tara Oceans expedition. In Advances in Phytoplankton Ecology; Clementson, L.A., Eriksen, R.S., Willis, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 531–561. [Google Scholar] [CrossRef]
  142. Acinas, S.G.; Sánchez, P.; Salazar, G.; Cornejo-Castillo, F.M.; Sebastián, M.; Logares, R.; Royo-Llonch, M.; Paoli, L.; Sunagawa, S.; Hingamp, P.; et al. Deep ocean metagenomes provide insight into the metabolic architecture of bathypelagic microbial communities. Commun. Biol. 2021, 4, 604. [Google Scholar] [CrossRef] [PubMed]
  143. Laiolo, E.; Alam, I.; Uludag, M.; Jamil, T.; Agusti, S.; Gojobori, T.; Acinas, S.G.; Gasol, J.M.; Duarte, C.M. Metagenomic probing toward an atlas of the taxonomic and metabolic foundations of the global ocean genome. Front. Sci. 2024, 1, 1038696. [Google Scholar] [CrossRef]
  144. Um, S.; Pyee, Y.; Kim, E.-H.; Lee, S.K.; Shin, J.; Oh, D.-C. Thalassospiramide G, a New γ-Amino-Acid-Bearing Peptide from the Marine Bacterium Thalassospira sp. Mar. Drugs 2013, 11, 611–622. [Google Scholar] [CrossRef] [PubMed]
  145. Giordano, D.; Coppola, D.; Russo, R.; Denaro, R.; Giuliano, L.; Lauro, F.M.; di Prisco, G.; Verde, C. Chapter Four—Marine Microbial Secondary Metabolites: Pathways, Evolution and Physiological Roles. In Advances in Microbial Physiology; Poole, R.K., Ed.; Academic Press: Cambridge, MA, USA, 2015; Volume 66, pp. 357–428. [Google Scholar]
  146. Wei, B.; Hu, G.-A.; Zhou, Z.-Y.; Yu, W.-C.; Du, A.-Q.; Yang, C.-L.; Yu, Y.-L.; Chen, J.-W.; Zhang, H.-W.; Wu, Q.; et al. Global analysis of the biosynthetic chemical space of marine prokaryotes. Microbiome 2023, 11, 144. [Google Scholar] [CrossRef] [PubMed]
  147. Rodrigues, C.J.C.; de Carvalho, C.C.C.R. Cultivating marine bacteria under laboratory conditions: Overcoming the “unculturable” dogma. Front. Bioeng. Biotechnol. 2022, 10, 964589. [Google Scholar] [CrossRef]
  148. Mu, D.-S.; Ouyang, Y.; Chen, G.-J.; Du, Z.-J. Strategies for culturing active/dormant marine microbes. Mar. Life Sci. Technol. 2021, 3, 121–131. [Google Scholar] [CrossRef] [PubMed]
  149. Andryukov, B.; Mikhailov, V.; Besednova, N. The Biotechnological Potential of Secondary Metabolites from Marine Bacteria. J. Mar. Sci. Eng. 2019, 7, 176. [Google Scholar] [CrossRef]
  150. Liu, L.; Zheng, Y.-Y.; Shao, C.-L.; Wang, C.-Y. Metabolites from marine invertebrates and their symbiotic microorganisms: Molecular diversity discovery, mining, and application. Mar. Life Sci. Technol. 2019, 1, 60–94. [Google Scholar] [CrossRef]
  151. Wilson, M.C.; Mori, T.; Rückert, C.; Uria, A.R.; Helf, M.J.; Takada, K.; Gernert, C.; Steffens, U.A.E.; Heycke, N.; Schmitt, S.; et al. An environmental bacterial taxon with a large and distinct metabolic repertoire. Nature 2014, 506, 58–62. [Google Scholar] [CrossRef] [PubMed]
  152. Mori, T.; Cahn, J.K.B.; Wilson, M.C.; Meoded, R.A.; Wiebach, V.; Martinez, A.F.C.; Helfrich, E.J.N.; Albersmeier, A.; Wibberg, D.; Dätwyler, S.; et al. Single-bacterial genomics validates rich and varied specialized metabolism of uncultivated Entotheonella sponge symbionts. Proc. Natl. Acad. Sci. USA 2018, 115, 1718–1723. [Google Scholar] [CrossRef] [PubMed]
  153. Lackner, G.; Peters, E.E.; Helfrich, E.J.N.; Piel, J. Insights into the lifestyle of uncultured bacterial natural product factories associated with marine sponges. Proc. Natl. Acad. Sci. USA 2017, 114, E347–E356. [Google Scholar] [CrossRef] [PubMed]
  154. Rath, C.M.; Janto, B.; Earl, J.; Ahmed, A.; Hu, F.Z.; Hiller, L.; Dahlgren, M.; Kreft, R.; Yu, F.; Wolff, J.J.; et al. Meta-omic Characterization of the Marine Invertebrate Microbial Consortium That Produces the Chemotherapeutic Natural Product ET-743. ACS Chem. Biol. 2011, 6, 1244–1256. [Google Scholar] [CrossRef] [PubMed]
  155. Knobloch, S.; Jóhannsson, R.; Marteinsson, V. Co-cultivation of the marine sponge Halichondria panicea and its associated microorganisms. Sci. Rep. 2019, 9, 10403. [Google Scholar] [CrossRef]
  156. Kamat, S.; Kumar, S.; Philip, S.; Kumari, M. Chapter 10—Secondary metabolites from marine fungi: Current status and application. In Microbial Biomolecules; Kumar, A., Bilal, M., Ferreira, L.F.R., Kumari, M., Eds.; Academic Press: Cambridge, MA, USA, 2023; pp. 181–209. [Google Scholar] [CrossRef]
  157. Xu, J.; Yi, M.; Ding, L.; He, S. A Review of Anti-Inflammatory Compounds from Marine Fungi, 2000-2018. Mar. Drugs 2019, 17, 636. [Google Scholar] [CrossRef] [PubMed]
  158. Nicoletti, R.; Bellavita, R.; Falanga, A. The Outstanding Chemodiversity of Marine-Derived Talaromyces. Biomolecules 2023, 13, 1021. [Google Scholar] [CrossRef] [PubMed]
  159. Cutolo, E.A.; Caferri, R.; Campitiello, R.; Cutolo, M. The Clinical Promise of Microalgae in Rheumatoid Arthritis: From Natural Compounds to Recombinant Therapeutics. Mar. Drugs 2023, 21, 630. [Google Scholar] [CrossRef] [PubMed]
  160. dos Reis, G.A.; Rodrigues, C.; Wiatek, A.M.; de Melo Pereira, G.V.; de Carvalho, J.C.; Martinez-Burgos, W.J.; Karp, S.G.; Soccol, V.T.; Letti, L.A.J.; Soccol, C.R. Exploring the potential of Thraustochytrids and other microorganisms for sustainable omega-3 production: A comprehensive review of patents, perspectives, and scale-up strategies. Syst. Microbiol. Biomanuf. 2024, 4, 448–462. [Google Scholar] [CrossRef]
  161. Chen, Z.-L.; Yang, L.-H.; He, S.-J.; Du, Y.-H.; Guo, D.-S. Development of a green fermentation strategy with resource cycle for the docosahexaenoic acid production by Schizochytrium sp. Bioresour. Technol. 2023, 385, 129434. [Google Scholar] [CrossRef]
  162. Dawczynski, C.; Dittrich, M.; Neumann, T.; Goetze, K.; Welzel, A.; Oelzner, P.; Völker, S.; Schaible, A.M.; Troisi, F.; Thomas, L.; et al. Docosahexaenoic acid in the treatment of rheumatoid arthritis: A double-blind, placebo-controlled, randomized cross-over study with microalgae vs. sunflower oil. Clin. Nutr. 2018, 37, 494–504. [Google Scholar] [CrossRef] [PubMed]
  163. Barone, G.D.; Cernava, T.; Ullmann, J.; Liu, J.; Lio, E.; Germann, A.T.; Nakielski, A.; Russo, D.A.; Chavkin, T.; Knufmann, K.; et al. Recent developments in the production and utilization of photosynthetic microorganisms for food applications. Heliyon 2023, 9, e14708. [Google Scholar] [CrossRef] [PubMed]
  164. Levasseur, W.; Perré, P.; Pozzobon, V. A review of high value-added molecules production by microalgae in light of the classification. Biotechnol. Adv. 2020, 41, 107545. [Google Scholar] [CrossRef]
  165. Riccio, G.; Lauritano, C. Microalgae with Immunomodulatory Activities. Mar. Drugs 2019, 18, 2. [Google Scholar] [CrossRef] [PubMed]
  166. Ghallab, D.S.; Ibrahim, R.S.; Mohyeldin, M.M.; Shawky, E. Marine algae: A treasure trove of bioactive anti-inflammatory compounds. Mar. Pollut. Bull. 2024, 199, 116023. [Google Scholar] [CrossRef] [PubMed]
  167. Dullius, A.; Buffon, G.; Junior, M.F.; Giuliatti, S. Artificial Intelligence in Phycochemicals Recognition. In Value-Added Products from Algae: Phycochemical Production and Applications; Abomohra, A., Ende, S., Eds.; Springer International Publishing: Cham, Switzerland, 2024; pp. 97–122. [Google Scholar]
  168. Galasso, C.; Corinaldesi, C.; Sansone, C. Carotenoids from Marine Organisms: Biological Functions and Industrial Applications. Antioxidants 2017, 6, 96. [Google Scholar] [CrossRef] [PubMed]
  169. Muñoz-Miranda, L.A.; Iñiguez-Moreno, M. An extensive review of marine pigments: Sources, biotechnological applications, and sustainability. Aquat. Sci. 2023, 85, 68. [Google Scholar] [CrossRef] [PubMed]
  170. Generalić Mekinić, I.; Šimat, V.; Rathod, N.B.; Hamed, I.; Čagalj, M. Algal Carotenoids: Chemistry, Sources, and Application. Foods 2023, 12, 2768. [Google Scholar] [CrossRef] [PubMed]
  171. Razzak, S.A. Comprehensive overview of microalgae-derived carotenoids and their applications in diverse industries. Algal Res. 2024, 78, 103422. [Google Scholar] [CrossRef]
  172. Chekanov, K.; Lukyanov, A.; Boussiba, S.; Aflalo, C.; Solovchenko, A. Modulation of photosynthetic activity and photoprotection in Haematococcus pluvialis cells during their conversion into haematocysts and back. Photosynth. Res. 2016, 128, 313–323. [Google Scholar] [CrossRef] [PubMed]
  173. Samhat, K.; Kazbar, A.; Takache, H.; Gonçalves, O.; Drouin, D.; Ismail, A.; Pruvost, J. Optimization of continuous astaxanthin production by Haematococcus pluvialis in nitrogen-limited photobioreactor. Algal Res. 2024, 80, 103529. [Google Scholar] [CrossRef]
  174. Liaqat, F.; Khazi, M.I.; Bahadar, A.; He, L.; Aslam, A.; Liaquat, R.; Agathos, S.N.; Li, J. Mixotrophic cultivation of microalgae for carotenoid production. Rev. Aquac. 2023, 15, 35–61. [Google Scholar] [CrossRef]
  175. Li, X.; Wang, X.; Duan, C.; Yi, S.; Gao, Z.; Xiao, C.; Agathos, S.N.; Wang, G.; Li, J. Biotechnological production of astaxanthin from the microalga Haematococcus pluvialis. Biotechnol. Adv. 2020, 43, 107602. [Google Scholar] [CrossRef]
  176. McQuillan, J.L.; Cutolo, E.A.; Evans, C.; Pandhal, J. Proteomic characterization of a lutein-hyperaccumulating Chlamydomonas reinhardtii mutant reveals photoprotection-related factors as targets for increasing cellular carotenoid content. Biotechnol. Biofuels Bioprod. 2023, 16, 166. [Google Scholar] [CrossRef] [PubMed]
  177. Sousa, S.; Carvalho, A.P.; Gomes, A.M. Factors impacting the microbial production of eicosapentaenoic acid. Appl. Microbiol. Biotechnol. 2024, 108, 368. [Google Scholar] [CrossRef]
  178. Riccio, G.; De Luca, D.; Lauritano, C. Monogalactosyldiacylglycerol and Sulfolipid Synthesis in Microalgae. Mar. Drugs 2020, 18, 237. [Google Scholar] [CrossRef] [PubMed]
  179. Lopes, D.; Aveiro, S.S.; Conde, T.; Rey, F.; Couto, D.; Melo, T.; Moreira, A.S.P.; Domingues, M.R. Chapter 6—Algal lipids: Structural diversity, analysis and applications. In Functional Ingredients from Algae for Foods and Nutraceuticals, 2nd ed.; Dominguez, H., Pereira, L., Kraan, S., Eds.; Woodhead Publishing: Sawston, UK, 2023; pp. 335–396. [Google Scholar] [CrossRef]
  180. Conde, T.A.; Zabetakis, I.; Tsoupras, A.; Medina, I.; Costa, M.; Silva, J.; Neves, B.; Domingues, P.; Domingues, M.R. Microalgal Lipid Extracts Have Potential to Modulate the Inflammatory Response: A Critical Review. Int. J. Mol. Sci. 2021, 22, 9825. [Google Scholar] [CrossRef] [PubMed]
  181. Banskota, A.H.; Gallant, P.; Stefanova, R.; Melanson, R.; O’Leary, S.J. Monogalactosyldiacylglycerols, potent nitric oxide inhibitors from the marine microalga Tetraselmis chui. Nat. Prod. Res. 2013, 27, 1084–1090. [Google Scholar] [CrossRef] [PubMed]
  182. Maciel, F.; Madureira, L.; Geada, P.; Teixeira, J.A.; Silva, J.; Vicente, A.A. The potential of Pavlovophyceae species as a source of valuable carotenoids and polyunsaturated fatty acids for human consumption. Biotechnol. Adv. 2024, 74, 108381. [Google Scholar] [CrossRef]
  183. Thurn, A.-L.; Schobel, J.; Weuster-Botz, D. Photoautotrophic Production of Docosahexaenoic Acid- and Eicosapentaenoic Acid-Enriched Biomass by Co-Culturing Golden-Brown and Green Microalgae. Fermentation 2024, 10, 220. [Google Scholar] [CrossRef]
  184. García-García, P.; Ospina, M.; Señoráns, F.J. Tisochrysis lutea as a source of omega-3 polar lipids and fucoxanthin: Extraction and characterization using green solvents and advanced techniques. J. Appl. Phycol. 2024. [Google Scholar] [CrossRef]
  185. Pierella Karlusich, J.J.; Cosnier, K.; Zinger, L.; Henry, N.; Nef, C.; Bernard, G.; Scalco, E.; Dvorak, E.; Jimenez Vieira, F.R.; Delage, E.; et al. Patterns and drivers of diatom diversity and abundance in the global ocean. bioRxiv 2024. [Google Scholar] [CrossRef]
  186. Peng, K.; Amenorfenyo, D.K.; Rui, X.; Huang, X.; Li, C.; Li, F. Effect of Iron Concentration on the Co-Production of Fucoxanthin and Fatty Acids in Conticribra weissflogii. Mar. Drugs 2024, 22, 106. [Google Scholar] [CrossRef] [PubMed]
  187. Mogal, A.; Kukreja, S.; Gautam, S. Sustainable Production of Diatom-Based Omega-3 Fatty Acids. In Biotechnological Processes for Green Energy, and High Value Bioproducts by Microalgae, and Cyanobacteria Cultures; Martínez-Roldán, A.d.J., Ed.; Springer International Publishing: Cham, Switzerland, 2024; pp. 131–137. [Google Scholar]
  188. Di Dato, V.; Orefice, I.; Amato, A.; Fontanarosa, C.; Amoresano, A.; Cutignano, A.; Ianora, A.; Romano, G. Animal-like prostaglandins in marine microalgae. ISME J. 2017, 11, 1722–1726. [Google Scholar] [CrossRef] [PubMed]
  189. Di Costanzo, F.; Di Dato, V.; Ianora, A.; Romano, G. Prostaglandins in Marine Organisms: A Review. Mar. Drugs 2019, 17, 428. [Google Scholar] [CrossRef] [PubMed]
  190. Di Dato, V.; Barbarinaldi, R.; Amato, A.; Di Costanzo, F.; Fontanarosa, C.; Perna, A.; Amoresano, A.; Esposito, F.; Cutignano, A.; Ianora, A.; et al. Variation in prostaglandin metabolism during growth of the diatom Thalassiosira rotula. Sci. Rep. 2020, 10, 5374. [Google Scholar] [CrossRef] [PubMed]
  191. Chen, J.; Wang, Y.; Benemann, J.R.; Zhang, X.; Hu, H.; Qin, S. Microalgal industry in China: Challenges and prospects. J. Appl. Phycol. 2016, 28, 715–725. [Google Scholar] [CrossRef]
  192. Araújo, R.; Vázquez Calderón, F.; Sánchez López, J.; Azevedo, I.C.; Bruhn, A.; Fluch, S.; Garcia Tasende, M.; Ghaderiardakani, F.; Ilmjärv, T.; Laurans, M.; et al. Current Status of the Algae Production Industry in Europe: An Emerging Sector of the Blue Bioeconomy. Front. Mar. Sci. 2021, 7, 626389. [Google Scholar] [CrossRef]
  193. Garrido-Cardenas, J.A.; Manzano-Agugliaro, F.; Acien-Fernandez, F.G.; Molina-Grima, E. Microalgae research worldwide. Algal Res. 2018, 35, 50–60. [Google Scholar] [CrossRef]
  194. Zhang, L.; Liao, W.; Huang, Y.; Wen, Y.; Chu, Y.; Zhao, C. Global seaweed farming and processing in the past 20 years. Food Prod. Process. Nutr. 2022, 4, 23. [Google Scholar] [CrossRef]
  195. Selvam, J.; Mal, J.; Singh, S.; Yadav, A.; Giri, B.S.; Pandey, A.; Sinha, R. Bioprospecting marine microalgae as sustainable bio-factories for value-added compounds. Algal Res. 2024, 79, 103444. [Google Scholar] [CrossRef]
  196. Rojas-Villalta, D.; Gómez-Espinoza, O.; Murillo-Vega, F.; Villalta-Romero, F.; Guerrero, M.; Guillén-Watson, R.; Núñez-Montero, K. Insights into Co-Cultivation of Photosynthetic Microorganisms for Novel Molecule Discovery and Enhanced Production of Specialized Metabolites. Fermentation 2023, 9, 941. [Google Scholar] [CrossRef]
  197. Badary, A.; Hidasi, N.; Ferrari, S.; Mayfield, S.P. Isolation and characterization of microalgae strains able to grow on complex biomass hydrolysate for industrial application. Algal Res. 2024, 78, 103381. [Google Scholar] [CrossRef]
  198. Occhipinti, P.S.; Russo, N.; Foti, P.; Zingale, I.M.; Pino, A.; Romeo, F.V.; Randazzo, C.L.; Caggia, C. Current challenges of microalgae applications: Exploiting the potential of non-conventional microalgae species. J. Sci. Food Agric. 2024, 104, 3823–3833. [Google Scholar] [CrossRef] [PubMed]
  199. Diankristanti, P.A.; Hei Ernest Ho, N.; Chen, J.-H.; Nagarajan, D.; Chen, C.-Y.; Hsieh, Y.-M.; Ng, I.S.; Chang, J.-S. Unlocking the potential of microalgae as sustainable bioresources from up to downstream processing: A critical review. Chem. Eng. J. 2024, 488, 151124. [Google Scholar] [CrossRef]
  200. Menon, R.; Thomas, R.; Sood, N.; Gokhale, T. Marine Phytoplankton: Bioactive Compounds and Their Applications in Medicine. In Marine Bioactive Molecules for Biomedical and Pharmacotherapeutic Applications; Veera Bramhachari, P., Berde, C.V., Eds.; Springer Nature: Singapore, 2023; pp. 251–282. [Google Scholar]
  201. Tan, L.T.; Phyo, M.Y. Marine Cyanobacteria: A Source of Lead Compounds and their Clinically-Relevant Molecular Targets. Molecules 2020, 25, 2197. [Google Scholar] [CrossRef] [PubMed]
  202. Gómez, P.I.; Mayorga, J.; Flaig, D.; Castro-Varela, P.; Jaupi, A.; Ulloa, P.A.; Soto-Bartierra, J.; Henríquez, V.; Rojas, V. Looking beyond Arthrospira: Comparison of antioxidant and anti-inflammatory properties of ten cyanobacteria strains. Algal Res. 2023, 74, 103182. [Google Scholar] [CrossRef]
  203. Cruz, J.D.; Delattre, C.; Felpeto, A.B.; Pereira, H.; Pierre, G.; Morais, J.; Petit, E.; Silva, J.; Azevedo, J.; Elboutachfaiti, R.; et al. Bioprospecting for industrially relevant exopolysaccharide-producing cyanobacteria under Portuguese simulated climate. Sci. Rep. 2023, 13, 13561. [Google Scholar] [CrossRef] [PubMed]
  204. Jha, S.; Singh, V.K.; Singh, A.P.; Gupta, A.; Rana, P.; Sinha, R.P. The Radiant World of Cyanobacterial Phycobiliproteins: Examining Their Structure, Functions, and Biomedical Potentials. Targets 2024, 2, 32–51. [Google Scholar] [CrossRef]
  205. Ismail, M.M.; El-Fakharany, E.M.; Hegazy, G.E. Purification and fractionation of phycobiliproteins from Arthrospira platensis and Corallina officinalis with evaluating their biological activities. Sci. Rep. 2023, 13, 14270. [Google Scholar] [CrossRef] [PubMed]
  206. Reddy, M.C.; Subhashini, J.; Mahipal, S.V.; Bhat, V.B.; Srinivas Reddy, P.; Kiranmai, G.; Madyastha, K.M.; Reddanna, P. C-Phycocyanin, a selective cyclooxygenase-2 inhibitor, induces apoptosis in lipopolysaccharide-stimulated RAW 264.7 macrophages. Biochem. Biophys. Res. Commun. 2003, 304, 385–392. [Google Scholar] [CrossRef]
  207. Chen, H.; Qi, H.; Xiong, P. Phycobiliproteins—A Family of Algae-Derived Biliproteins: Productions, Characterization and Pharmaceutical Potentials. Mar. Drugs 2022, 20, 450. [Google Scholar] [CrossRef] [PubMed]
  208. Khazi, M.I.; Li, C.; Liaqat, F.; Malec, P.; Li, J.; Fu, P. Acclimation and Characterization of Marine Cyanobacterial Strains Euryhalinema and Desertifilum for C-Phycocyanin Production. Front. Bioeng. Biotechnol. 2021, 9, 752024. [Google Scholar] [CrossRef] [PubMed]
  209. Forbes, E.; Godwin-Austen, R.A.C. The Natural History of the European Seas; By the Late Prof. Edw. Forbes; John Van Voorst: London, UK, 1859. [Google Scholar]
  210. Carpenter, W.B.; Jeffreys, J.G.; Thomson, C.T.W. Preliminary report of the scientific exploration of the deep sea in H. M. surveying-vessel ‘Porcupine’, during the summerof 1869. Proc. R. Soc. Lond. 1870, 18, 397–492. [Google Scholar] [CrossRef]
  211. Thomson, C.W.S.; Carpenter, W.B.; Jeffreys, J.G. The Depths of the Sea: An Account of the General Results of the Dredging Cruises of H.M. SS. Porcupine and Lightning during the Summers of 1868, 1869 and 1870, under the Scientific Direction of Dr. Carpenter, F.R.S., J. Gwyn Jeffreys, F.R.S., and Dr. Wyville Thomson, F.R.S.; Macmillan and Co: London, UK, 1873. [Google Scholar]
  212. Thomson, C.W.S. The Voyage of the “Challenger”: The Atlantic: A Preliminary Account of the General Results of the Exploring Voyage of H.M.S. “Challenger” during the Year 1873 and the Early Part of the Year 1876; Harper & Brothers: New York, NY, USA, 1878; Volume 1. [Google Scholar]
  213. Certes, A.-A. Sur la culture, à l’abri des germes atmosphériques, des eaux et des sédiments rapportés par les expéditions du Travailleur et du Talisman. In Comptes Rendue Hebdomadaire des Séances de l’Académie des Sciences; Comptes rendus de l’Académie des Sciences: Paris, France, 1884; pp. 690–693. [Google Scholar]
  214. Certes, A.-A. Note relative à l’action des hautes pressions sur la vitalité des microorganismes d’eau douce et d’eau de mer. Comptes Rendue Hebd. Des Séances De L’académie Des Sci. 1884, 36, 385–388. [Google Scholar]
  215. Dohrn, A. The Foundation of Zoological Stations. Nature 1872, 5, 277–280. [Google Scholar] [CrossRef]
  216. Groeben, C. The Stazione Zoologica Anton Dohrn as a place for the circulation of scientific ideas: Vision and management. In Proceedings of the 31st Annual Conference of IAMSLIC, Rome, Italy, 10–14 October 2005. [Google Scholar]
  217. Danske dybhavsekspedition jorden, R.; Bruun, A.F. The Galathea Deep Sea Expedition, 1950–1952, Described by Members of the Expedition; Macmillan: New York, NY, USA, 1956. [Google Scholar]
  218. Zobell, C.E. Bacterial Life at the Bottom of the Philippine Trench. Science 1952, 115, 507–508. [Google Scholar] [CrossRef] [PubMed]
  219. Yayanos, A.A.; Dietz, A.S.; Van Boxtel, R. Obligately barophilic bacterium from the Mariana trench. Proc. Natl. Acad. Sci. USA 1981, 78, 5212–5215. [Google Scholar] [CrossRef] [PubMed]
  220. Corliss, J.B.; Dymond, J.; Gordon, L.I.; Edmond, J.M.; von Herzen, R.P.; Ballard, R.D.; Green, K.; Williams, D.; Bainbridge, A.; Crane, K.; et al. Submarine Thermal Springs on the Galápagos Rift. Science 1979, 203, 1073–1083. [Google Scholar] [CrossRef] [PubMed]
  221. Poli, A.; Finore, I.; Romano, I.; Gioiello, A.; Lama, L.; Nicolaus, B. Microbial Diversity in Extreme Marine Habitats and Their Biomolecules. Microorganisms 2017, 5, 25. [Google Scholar] [CrossRef] [PubMed]
  222. Cario, A.; Oliver, G.C.; Rogers, K.L. Exploring the Deep Marine Biosphere: Challenges, Innovations, and Opportunities. Front. Earth Sci. 2019, 7, 225. [Google Scholar] [CrossRef]
  223. Sun, C.; Mudassir, S.; Zhang, Z.; Feng, Y.; Chang, Y.; Che, Q.; Gu, Q.; Zhu, T.; Zhang, G.; Li, D. Secondary Metabolites from Deep-Sea Derived Microorganisms. Curr. Med. Chem. 2020, 27, 6244–6273. [Google Scholar] [CrossRef] [PubMed]
  224. Saide, A.; Lauritano, C.; Ianora, A. A Treasure of Bioactive Compounds from the Deep Sea. Biomedicines 2021, 9, 1556. [Google Scholar] [CrossRef] [PubMed]
  225. Zhang, C.; Peng, Y.; Liu, X.; Wang, J.; Dong, X. Deep-sea microbial genetic resources: New frontiers for bioprospecting. Trends Microbiol. 2024, 32, 321–324. [Google Scholar] [CrossRef] [PubMed]
  226. Wang, Y.N.; Meng, L.H.; Wang, B.G. Progress in Research on Bioactive Secondary Metabolites from Deep-Sea Derived Microorganisms. Mar. Drugs 2020, 18, 614. [Google Scholar] [CrossRef] [PubMed]
  227. Burns, J.A.; Becker, K.P.; Casagrande, D.; Daniels, J.; Roberts, P.; Orenstein, E.; Vogt, D.M.; Teoh, Z.E.; Wood, R.; Yin, A.H.; et al. An in situ digital synthesis strategy for the discovery and description of ocean life. Sci. Adv. 2024, 10, eadj4960. [Google Scholar] [CrossRef] [PubMed]
  228. Zhivkoplias, E.; Pranindita, A.; Dunshirn, P.; Jouffray, J.-B.; Blasiak, R. Novel Database Reveals Growing Prominence Of Deep-Sea Life for Marine Bioprospecting; Research Square: Durham, NC, USA, 2023. [Google Scholar]
  229. Li, G.; Wong, T.-W.; Shih, B.; Guo, C.; Wang, L.; Liu, J.; Wang, T.; Liu, X.; Yan, J.; Wu, B.; et al. Bioinspired soft robots for deep-sea exploration. Nat. Commun. 2023, 14, 7097. [Google Scholar] [CrossRef]
  230. Pettit, R.K. Culturability and secondary metabolite diversity of extreme microbes: Expanding contribution of deep sea and deep-sea vent microbes to natural product discovery. Mar. Biotechnol. 2011, 13, 1–11. [Google Scholar] [CrossRef] [PubMed]
  231. Zhang, Y.; Li, X.; Bartlett, D.H.; Xiao, X. Current developments in marine microbiology: High-pressure biotechnology and the genetic engineering of piezophiles. Curr. Opin. Biotechnol. 2015, 33, 157–164. [Google Scholar] [CrossRef] [PubMed]
  232. Yan, X.; Zhou, Y.-X.; Tang, X.-X.; Liu, X.-X.; Yi, Z.-W.; Fang, M.-J.; Wu, Z.; Jiang, F.-Q.; Qiu, Y.-K. Macrolactins from Marine-Derived Bacillus subtilis B5 Bacteria as Inhibitors of Inducible Nitric Oxide and Cytokines Expression. Mar. Drugs 2016, 14, 195. [Google Scholar] [CrossRef] [PubMed]
  233. Hu, X.; Zhao, S.; Li, F.; Zhang, X.; Pan, Y.; Lu, J.; Li, Y.; Bao, M. The structure, characterization and immunomodulatory potential of exopolysaccharide produced by Planococcus rifietoensis AP-5 from deep-sea sediments of the Northwest Pacific. Int. J. Biol. Macromol. 2023, 245, 125452. [Google Scholar] [CrossRef] [PubMed]
  234. Busch, K.; Slaby, B.M.; Bach, W.; Boetius, A.; Clefsen, I.; Colaço, A.; Creemers, M.; Cristobo, J.; Federwisch, L.; Franke, A.; et al. Biodiversity, environmental drivers, and sustainability of the global deep-sea sponge microbiome. Nat. Commun. 2022, 13, 5160. [Google Scholar] [CrossRef]
  235. Wang, Z.; Qader, M.; Wang, Y.; Kong, F.; Wang, Q.; Wang, C. Progress in the discovery of new bioactive substances from deep-sea associated fungi during 2020-2022. Front. Mar. Sci. 2023, 10, 1232891. [Google Scholar] [CrossRef]
  236. Kamjam, M.; Sivalingam, P.; Deng, Z.; Hong, K. Deep Sea Actinomycetes and Their Secondary Metabolites. Front. Microbiol. 2017, 8, 760. [Google Scholar] [CrossRef]
  237. Wang, L.; Li, M.; Lin, Y.; Du, S.; Liu, Z.; Ju, J.; Suzuki, H.; Sawada, M.; Umezawa, K. Inhibition of cellular inflammatory mediator production and amelioration of learning deficit in flies by deep sea Aspergillus-derived cyclopenin. J. Antibiot. 2020, 73, 622–629. [Google Scholar] [CrossRef] [PubMed]
  238. Zou, Z.-B.; Wu, T.-Z.; Yang, L.-H.; He, X.-W.; Liu, W.-Y.; Zhang, K.; Xie, C.-L.; Xie, M.-M.; Zhang, Y.; Yang, X.-W.; et al. Hepialiamides A–C: Aminated Fusaric Acid Derivatives and Related Metabolites with Anti-Inflammatory Activity from the Deep-Sea-Derived Fungus Samsoniella hepiali W7. Mar. Drugs 2023, 21, 596. [Google Scholar] [CrossRef] [PubMed]
  239. Lu, X.; He, J.; Wu, Y.; Du, N.; Li, X.; Ju, J.; Hu, Z.; Umezawa, K.; Wang, L. Isolation and characterization of new anti-inflammatory and antioxidant components from deep marine-derived fungus Myrothecium sp. Bzo-l062. Mar. Drugs 2020, 18, 597. [Google Scholar] [CrossRef] [PubMed]
  240. Cheng, Z.; Zhao, J.; Liu, D.; Proksch, P.; Zhao, Z.; Lin, W. Eremophilane-type sesquiterpenoids from an Acremonium sp. fungus isolated from deep-sea sediments. J. Nat. Prod. 2016, 79, 1035–1047. [Google Scholar] [CrossRef] [PubMed]
  241. Niu, S.; Liu, D.; Shao, Z.; Proksch, P.; Lin, W. Eremophilane-type sesquiterpenoids in a deep-sea fungus Eutypella sp. activated by chemical epigenetic manipulation. Tetrahedron 2018, 74, 7310–7325. [Google Scholar] [CrossRef]
  242. Kim, D.-C.; Quang, T.H.; Tien, N.T.; Kim, K.-W.; Kim, Y.-C.; Ngan, N.T.T.; Cuong, N.X.; Nam, N.H.; Oh, H. Anti-neuroinflammatory effect of oxaline, isorhodoptilometrin, and 5-hydroxy-7-(2′-hydroxypropyl)-2-methyl-chromone obtained from the marine fungal strain Penicillium oxalicum CLC-MF05. Arch. Pharmacal Res. 2022, 45, 90–104. [Google Scholar] [CrossRef] [PubMed]
  243. Chen, S.; Wang, J.; Lin, X.; Zhao, B.; Wei, X.; Li, G.; Kaliaperumal, K.; Liao, S.; Yang, B.; Zhou, X. Chrysamides A–C, three dimeric nitrophenyl trans-epoxyamides produced by the deep-sea-derived fungus Penicillium chrysogenum SCSIO41001. Org. Lett. 2016, 18, 3650–3653. [Google Scholar] [CrossRef] [PubMed]
  244. Lee, H.-S.; Kang, J.S.; Cho, D.-Y.; Choi, D.-K.; Shin, H.J. Isolation, Structure Determination, and Semisynthesis of Diphenazine Compounds from a Deep-Sea-Derived Strain of the Fungus Cystobasidium laryngis and Their Biological Activities. J. Nat. Prod. 2022, 85, 857–865. [Google Scholar] [CrossRef] [PubMed]
  245. Kazłowska, K.; Hsu, T.; Hou, C.C.; Yang, W.C.; Tsai, G.J. Anti-inflammatory properties of phenolic compounds and crude extract from Porphyra dentata. J. Ethnopharmacol. 2010, 128, 123–130. [Google Scholar] [CrossRef] [PubMed]
  246. Xu, J.; Liao, W.; Liu, Y.; Guo, Y.; Jiang, S.; Zhao, C. An overview on the nutritional and bioactive components of green seaweeds. Food Prod. Process. Nutr. 2023, 5, 18. [Google Scholar] [CrossRef]
  247. Souza, C.R.M.; Bezerra, W.P.; Souto, J.T. Marine Alkaloids with Anti-Inflammatory Activity: Current Knowledge and Future Perspectives. Mar. Drugs 2020, 18, 147. [Google Scholar] [CrossRef] [PubMed]
  248. Pereira, L.; Valado, A. Harnessing the power of seaweed: Unveiling the potential of marine algae in drug discovery. Explor. Drug Sci. 2023, 1, 475–496. [Google Scholar] [CrossRef]
  249. Pradhan, B.; Rout, L.; Ki, J.-S. Immunomodulatory and anti-inflammatory and anticancer activities of porphyran, a sulfated galactan. Carbohydr. Polym. 2023, 301, 120326. [Google Scholar] [CrossRef] [PubMed]
  250. Costa, V.V.; Amaral, F.A.; Coelho, F.M.; Queiroz-Junior, C.M.; Malagoli, B.G.; Gomes, J.H.; Lopes, F.; Silveira, K.D.; Sachs, D.; Fagundes, C.T.; et al. Lithothamnion muelleri treatment ameliorates inflammatory and hypernociceptive responses in antigen-induced arthritis in mice. PLoS ONE 2015, 10, e0118356. [Google Scholar] [CrossRef] [PubMed]
  251. Yoo, H.J.; You, D.-J.; Lee, K.-W. Characterization and Immunomodulatory Effects of High Molecular Weight Fucoidan Fraction from the Sporophyll of Undaria pinnatifida in Cyclophosphamide-Induced Immunosuppressed Mice. Mar. Drugs 2019, 17, 447. [Google Scholar] [CrossRef] [PubMed]
  252. Pradhan, B.; Bhuyan, P.P.; Ki, J.-S. Immunomodulatory, Antioxidant, Anticancer, and Pharmacokinetic Activity of Ulvan, a Seaweed-Derived Sulfated Polysaccharide: An Updated Comprehensive Review. Mar. Drugs 2023, 21, 300. [Google Scholar] [CrossRef] [PubMed]
  253. Vasarri, M.; Leri, M.; Barletta, E.; Ramazzotti, M.; Marzocchini, R.; Degl’Innocenti, D. Anti-inflammatory properties of the marine plant Posidonia oceanica (L.) Delile. J. Ethnopharmacol. 2020, 247, 112252. [Google Scholar] [CrossRef] [PubMed]
  254. Premarathna, A.D.; Ahmed, T.A.E.; Rjabovs, V.; Hammami, R.; Critchley, A.T.; Tuvikene, R.; Hincke, M.T. Immunomodulation by xylan and carrageenan-type polysaccharides from red seaweeds: Anti-inflammatory, wound healing, cytoprotective, and anticoagulant activities. Int. J. Biol. Macromol. 2024, 260, 129433. [Google Scholar] [CrossRef] [PubMed]
  255. Sarithakumari, C.H.; Renju, G.L.; Kurup, G.M. Anti-inflammatory and antioxidant potential of alginic acid isolated from the marine algae, Sargassum wightii on adjuvant-induced arthritic rats. Inflammopharmacology 2013, 21, 261–268. [Google Scholar] [CrossRef]
  256. Jung, H.A.; Jin, S.E.; Ahn, B.R.; Lee, C.M.; Choi, J.S. Anti-inflammatory activity of edible brown alga Eisenia bicyclis and its constituents fucosterol and phlorotannins in LPS-stimulated RAW264.7 macrophages. Food Chem. Toxicol. 2013, 59, 199–206. [Google Scholar] [CrossRef] [PubMed]
  257. Pereira, D.M.; Valentão, P.; Andrade, P.B. Chapter 7—Lessons from the Sea: Distribution, SAR, and Molecular Mechanisms of Anti-inflammatory Drugs from Marine Organisms. In Studies in Natural Products Chemistry; Atta ur, R., Ed.; Elsevier: Amsterdam, The Netherlands, 2013; Volume 40, pp. 205–228. [Google Scholar]
  258. Senthilkumar, K.; Kim, S.-K. Marine Invertebrate Natural Products for Anti-Inflammatory and Chronic Diseases. Evid.-Based Complement. Altern. Med. Ecam 2013, 2013, 572859. [Google Scholar] [CrossRef] [PubMed]
  259. Dokalahy, E.E.; El-Seedi, H.R.; Farag, M.A. Soft Coral Biodiversity in the Red Sea Family Alcyoniidae: A Biopharmaceutical and Ecological Perspective. In Biodiversity and Chemotaxonomy; Ramawat, K.G., Ed.; Springer International Publishing: Cham, Switzerland, 2019; pp. 55–85. [Google Scholar]
  260. Imbs, A.B.; Dembitsky, V.M. Coral Lipids. Mar. Drugs 2023, 21, 539. [Google Scholar] [CrossRef] [PubMed]
  261. Bautista, C.A.; Puentes, C.A.; Vargas-Peláez, C.M.; Santos-Acevedo, M.; Ramos, F.A.; Gómez-León, J.; Castellanos Hernández, L. The state of the art of marine natural products in Colombia. Rev. Colomb. De Química 2022, 51, 24–39. [Google Scholar] [CrossRef]
  262. Lai, K.-H.; Fan, Y.-C.; Peng, B.-R.; Wen, Z.-H.; Chung, H.-M. Capnellenes from Capnella imbricata: Deciphering Their Anti-Inflammatory-Associated Chemical Features. Pharmaceuticals 2023, 16, 916. [Google Scholar] [CrossRef]
  263. González, Y.; Torres-Mendoza, D.; Jones, G.E.; Fernandez, P.L. Marine Diterpenoids as Potential Anti-Inflammatory Agents. Mediat. Inflamm. 2015, 2015, 263543. [Google Scholar] [CrossRef]
  264. Mustonen, A.-M.; Nieminen, P. Fatty Acids and Oxylipins in Osteoarthritis and Rheumatoid Arthritis—A Complex Field with Significant Potential for Future Treatments. Curr. Rheumatol. Rep. 2021, 23, 41. [Google Scholar] [CrossRef] [PubMed]
  265. Valmsen, K.; Järving, I.; Boeglin, W.E.; Varvas, K.; Koljak, R.; Pehk, T.; Brash, A.R.; Samel, N. The origin of 15R-prostaglandins in the Caribbean coral Plexaura homomalla: Molecular cloning and expression of a novel cyclooxygenase. Proc. Natl. Acad. Sci. USA 2001, 98, 7700–7705. [Google Scholar] [CrossRef] [PubMed]
  266. Lin, Y.Y.; Lin, S.C.; Feng, C.W.; Chen, P.C.; Su, Y.D.; Li, C.M.; Yang, S.N.; Jean, Y.H.; Sung, P.J.; Duh, C.Y.; et al. Anti-Inflammatory and Analgesic Effects of the Marine-Derived Compound Excavatolide B Isolated from the Culture-Type Formosan Gorgonian Briareum excavatum. Mar. Drugs 2015, 13, 2559–2579. [Google Scholar] [CrossRef] [PubMed]
  267. Sung, P.J.; Chen, B.Y.; Lin, M.R.; Hwang, T.L.; Wang, W.H.; Sheu, J.H.; Wu, Y.C. Excavatoids E and F: Discovery of two new briaranes from the cultured octocoral Briareum excavatum. Mar. Drugs 2009, 7, 472–482. [Google Scholar] [CrossRef] [PubMed]
  268. Lin, Y.Y.; Jean, Y.H.; Lee, H.P.; Chen, W.F.; Sun, Y.M.; Su, J.H.; Lu, Y.; Huang, S.Y.; Hung, H.C.; Sung, P.J.; et al. A soft coral-derived compound, 11-epi-sinulariolide acetate suppresses inflammatory response and bone destruction in adjuvant-induced arthritis. PLoS ONE 2013, 8, e62926. [Google Scholar] [CrossRef] [PubMed]
  269. Ayed, Y.; Sghaier, R.M.; Laouini, D.; Bacha, H. Evaluation of anti-proliferative and anti-inflammatory activities of Pelagia noctiluca venom in Lipopolysaccharide/Interferon-γ stimulated RAW264.7 macrophages. Biomed. Pharmacother. 2016, 84, 1986–1991. [Google Scholar] [CrossRef] [PubMed]
  270. La Paglia, L.; Vazzana, M.; Mauro, M.; Urso, A.; Arizza, V.; Vizzini, A. Bioactive Molecules from the Innate Immunity of Ascidians and Innovative Methods of Drug Discovery: A Computational Approach Based on Artificial Intelligence. Mar. Drugs 2024, 22, 6. [Google Scholar] [CrossRef] [PubMed]
  271. Silva, O.N.; de la Fuente-Núñez, C.; Haney, E.F.; Fensterseifer, I.C.M.; Ribeiro, S.M.; Porto, W.F.; Brown, P.; Faria-Junior, C.; Rezende, T.M.B.; Moreno, S.E.; et al. An anti-infective synthetic peptide with dual antimicrobial and immunomodulatory activities. Sci. Rep. 2016, 6, 35465. [Google Scholar] [CrossRef] [PubMed]
  272. Ju, Z.; Su, M.; Hong, J.; La Kim, E.; Moon, H.R.; Chung, H.Y.; Kim, S.; Jung, J.H. Design of balanced COX inhibitors based on anti-inflammatory and/or COX-2 inhibitory ascidian metabolites. Eur. J. Med. Chem. 2019, 180, 86–98. [Google Scholar] [CrossRef]
  273. Casertano, M.; Esposito, E.; Bello, I.; Indolfi, C.; Putra, M.Y.; Di Cesare Mannelli, L.; Ghelardini, C.; Menna, M.; Sorrentino, R.; Cirino, G.; et al. Searching for Novel Sources of Hydrogen Sulfide Donors: Chemical Profiling of Polycarpa aurata Extract and Evaluation of the Anti-Inflammatory Effects. Mar. Drugs 2023, 21, 641. [Google Scholar] [CrossRef]
  274. Sansone, C.; Balestra, C.; Pistelli, L.; Del Mondo, A.; Osca, D.; Brunet, C.; Crocetta, F. A Comparative Analysis of Mucus Immunomodulatory Properties from Seven Marine Gastropods from the Mediterranean Sea. Cells 2022, 11, 2340. [Google Scholar] [CrossRef] [PubMed]
  275. Li, G.; Fu, Y.; Zheng, J.; Li, D. Anti-inflammatory activity and mechanism of a lipid extract from hard-shelled mussel (Mytilus coruscus) on chronic arthritis in rats. Mar. Drugs 2014, 12, 568–588. [Google Scholar] [CrossRef] [PubMed]
  276. Fu, Y.; Li, G.; Zhang, X.; Xing, G.; Hu, X.; Yang, L.; Li, D. Lipid extract from hard-shelled mussel (Mytilus coruscus) improves clinical conditions of patients with rheumatoid arthritis: A randomized controlled trial. Nutrients 2015, 7, 625–645. [Google Scholar] [CrossRef] [PubMed]
  277. Pereira, R.B.; Taveira, M.; Valentão, P.; Sousa, C.; Andrade, P.B. Fatty acids from edible sea hares: Anti-inflammatory capacity in LPS-stimulated RAW 264.7 cells involves iNOS modulation. RSC Adv. 2015, 5, 8981–8987. [Google Scholar] [CrossRef]
  278. Ghelani, H.; Khursheed, M.; Adrian, T.E.; Jan, R.K. Anti-Inflammatory Effects of Compounds from Echinoderms. Mar. Drugs 2022, 20, 693. [Google Scholar] [CrossRef] [PubMed]
  279. Park, G.-T.; Yoon, J.-W.; Yoo, S.-B.; Song, Y.-C.; Song, P.; Kim, H.-K.; Han, J.; Bae, S.-J.; Ha, K.-T.; Mishchenko, N.P.; et al. Echinochrome A Treatment Alleviates Fibrosis and Inflammation in Bleomycin-Induced Scleroderma. Mar. Drugs 2021, 19, 237. [Google Scholar] [CrossRef] [PubMed]
  280. Olivera-Castillo, L.; Grant, G.; Kantún-Moreno, N.; Acevedo-Fernández, J.J.; Puc-Sosa, M.; Montero, J.; Olvera-Novoa, M.A.; Negrete-León, E.; Santa-Olalla, J.; Ceballos-Zapata, J.; et al. Sea cucumber (Isostichopus badionotus) body-wall preparations exert anti-inflammatory activity in vivo. PharmaNutrition 2018, 6, 74–80. [Google Scholar] [CrossRef]
  281. Jo, S.-H.; Kim, C.; Park, S.-H. Novel Marine Organism-Derived Extracellular Vesicles for Control of Anti-Inflammation. Tissue Eng. Regen. Med. 2021, 18, 71–79. [Google Scholar] [CrossRef] [PubMed]
  282. Shady, N.H.; El-Hossary, E.M.; Fouad, M.A.; Gulder, T.A.M.; Kamel, M.S.; Abdelmohsen, U.R. Bioactive Natural Products of Marine Sponges from the Genus Hyrtios. Molecules 2017, 22, 781. [Google Scholar] [CrossRef] [PubMed]
  283. Youssef, D.T.; Ibrahim, A.K.; Khalifa, S.I.; Mesbah, M.K.; Mayer, A.M.; van Soest, R.W. New anti-inflammatory sterols from the Red Sea sponges Scalarispongia aqabaensis and Callyspongia siphonella. Nat. Prod. Commun. 2010, 5, 27–31. [Google Scholar] [PubMed]
  284. Hort, M.A.; Silva Júnior, F.M.R.d.; Garcia, E.M.; Peraza, G.G.; Soares, A.; Lerner, C.; Muccillo-Baisch, A.L. Antinociceptive and Anti-inflammatory Activities of Marine Sponges Aplysina Caissara, Haliclona sp. and Dragmacidon Reticulatum. Braz. Arch. Biol. Technol. 2018, 61, e18180104. [Google Scholar] [CrossRef]
  285. Ji, Y.K.; Lee, S.M.; Kim, N.-H.; Tu, N.V.; Kim, Y.N.; Heo, J.D.; Jeong, E.J.; Rho, J.-R. Stereochemical Determination of Fistularins Isolated from the Marine Sponge Ecionemia acervus and Their Regulatory Effect on Intestinal Inflammation. Mar. Drugs 2021, 19, 170. [Google Scholar] [CrossRef] [PubMed]
  286. Vidal, I.; Castilla, L.; Marrero, A.D.; Bravo-Ruiz, I.; Bernal, M.; Manrique, I.; Quesada, A.R.; Medina, M.; Martínez-Poveda, B. The Sponge-Derived Brominated Compound Aeroplysinin-1 Impairs the Endothelial Inflammatory Response through Inhibition of the NF-κB Pathway. Mar. Drugs 2022, 20, 605. [Google Scholar] [CrossRef] [PubMed]
  287. Ciaglia, E.; Malfitano, A.M.; Laezza, C.; Fontana, A.; Nuzzo, G.; Cutignano, A.; Abate, M.; Pelin, M.; Sosa, S.; Bifulco, M. Immuno-modulatory and anti-inflammatory effects of dihydrogracilin A, a terpene derived from the marine sponge Dendrilla membranosa. Int. J. Mol. Sci. 2017, 18, 1643. [Google Scholar] [CrossRef] [PubMed]
  288. Di, X.; Oskarsson, J.T.; Omarsdottir, S.; Freysdottir, J.; Hardardottir, I. Lipophilic fractions from the marine sponge Halichondria sitiens decrease secretion of pro-inflammatory cytokines by dendritic cells and decrease their ability to induce a Th1 type response by allogeneic CD4(+) T cells. Pharm. Biol. 2017, 55, 2116–2122. [Google Scholar] [CrossRef] [PubMed]
  289. Palit, K.; Rath, S.; Chatterjee, S.; Das, S. Microbial diversity and ecological interactions of microorganisms in the mangrove ecosystem: Threats, vulnerability, and adaptations. Environ. Sci. Pollut. Res. 2022, 29, 32467–32512. [Google Scholar] [CrossRef] [PubMed]
  290. Nabeelah Bibi, S.; Fawzi, M.M.; Gokhan, Z.; Rajesh, J.; Nadeem, N.; Rengasamy Kannan, R.R.; Albuquerque, R.D.D.G.; Pandian, S.K. Ethnopharmacology, Phytochemistry, and Global Distribution of Mangroves―A Comprehensive Review. Mar. Drugs 2019, 17, 231. [Google Scholar] [CrossRef] [PubMed]
  291. Obón, C.; Rivera, D.; Verde, A.; Alcaraz, F. Ethnopharmacology and Medicinal Uses of Extreme Halophytes. In Handbook of Halophytes: From Molecules to Ecosystems towards Biosaline Agriculture; Grigore, M.-N., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 1–29. [Google Scholar]
  292. Murugesan, M.; Theivendram, T.; Thangapandian, R.; Murugesan, A. Survey of the ethnomedicinal use of marine halophytes among the indigenes of coastal regions of Tamil Nadu, India. Phytocoenologia 2020, 50, 173–186. [Google Scholar] [CrossRef]
  293. Das, S.K.; Das, B.; Jena, A.B.; Pradhan, C.; Sahoo, G.; Dandapat, J. Therapeutic Potential and Ethnopharmacology of Dominant Mangroves of Bhitarkanika National Park, Odisha, India. Chem. Biodivers. 2022, 19, e202100857. [Google Scholar] [CrossRef]
  294. Roome, T.; Dar, A.; Ali, S.; Naqvi, S.; Choudhary, M.I. A study on antioxidant, free radical scavenging, anti-inflammatory and hepatoprotective actions of Aegiceras corniculatum (stem) extracts. J. Ethnopharmacol. 2008, 118, 514–521. [Google Scholar] [CrossRef] [PubMed]
  295. Kumari, C.S.; Yasmin, N.; Hussain, M.R.; Babuselvam, M. Invitro anti-inflammatory and anti-artheritic property of Rhizopora mucronata leaves. Int. J. Pharma Sci. Res. 2015, 6, 482–485. [Google Scholar]
  296. Islam, M.T. Chemical profile and biological activities of Sonneratia apetala (Buch.-Ham.). Adv. Tradit. Med. 2020, 20, 123–132. [Google Scholar] [CrossRef]
  297. Yu, S.-Y.; Wang, S.-W.; Hwang, T.-L.; Wei, B.-L.; Su, C.-J.; Chang, F.-R.; Cheng, Y.-B. Components from the leaves and twigs of mangrove Lumnitzera racemosa with anti-angiogenic and anti-inflammatory effects. Mar. Drugs 2018, 16, 404. [Google Scholar] [CrossRef] [PubMed]
  298. Jiang, Z.-P.; Yu, Y.; Shen, L. Agallolides AM, including two rearranged ent-atisanes featuring a bicyclo [3.2. 1] octane motif, from the Chinese Excoecaria agallocha. Bioorganic Chem. 2020, 104, 104206. [Google Scholar] [CrossRef] [PubMed]
  299. Deepa, M.; Darsan, M.B.; Ramalingam, C. In Vitro Evaluation of the antioxidant, anti-inflammatory and antiproliferative activities of the leaf extracts of Excoecaria agallocha L. Int. J. Pharm. Sci. 2015, 7, 346–352. [Google Scholar]
  300. Babuselvam, M.; Ravikumar, S.; Farook, K.M.; Abideen, S.; Mohamed, M.P.; Uthiraselvam, M. Evaluation of anti-inflammatory and analgesic effects on the extracts of different parts of Excoecaria agallocha L. J. Appl. Pharm. Sci. 2012, 2, 108–112. [Google Scholar] [CrossRef]
  301. Cavalcante, S.B.; dos Santos Biscaino, C.; Kreusch, M.G.; da Silva, A.F.; Duarte, R.T.D.; Robl, D. The hidden rainbow: The extensive biotechnological potential of Antarctic fungi pigments. Braz. J. Microbiol. 2023, 54, 1675–1687. [Google Scholar] [CrossRef] [PubMed]
  302. Xiao, Z.; Cai, J.; Chen, T.; Wang, Y.; Chen, Y.; Zhu, Y.; Chen, C.; Yang, B.; Zhou, X.; Tao, H. Two New Sesquiterpenoids and a New Shikimic Acid Metabolite from Mangrove Sediment-Derived Fungus Roussoella sp. SCSIO 41427. Mar. Drugs 2024, 22, 103. [Google Scholar] [CrossRef] [PubMed]
  303. Daniotti, S.; Re, I. Marine Biotechnology: Challenges and Development Market Trends for the Enhancement of Biotic Resources in Industrial Pharmaceutical and Food Applications. A Statistical Analysis of Scientific Literature and Business Models. Mar. Drugs 2021, 19, 61. [Google Scholar] [CrossRef] [PubMed]
  304. Papon, N.; Copp, B.R.; Courdavault, V. Marine drugs: Biology, pipelines, current and future prospects for production. Biotechnol. Adv. 2022, 54, 107871. [Google Scholar] [CrossRef] [PubMed]
  305. Shikov, A.N.; Flisyuk, E.V.; Obluchinskaya, E.D.; Pozharitskaya, O.N. Pharmacokinetics of Marine-Derived Drugs. Mar. Drugs 2020, 18, 557. [Google Scholar] [CrossRef] [PubMed]
  306. Malaweera, D.B.O.; Wijesundara, N.M. Trends in Development of Marine-based Anti-inflammatory Pharmaceuticals. In Encyclopedia of Marine Biotechnology; Wiley: Hoboken, NJ, USA, 2020; pp. 2471–2485. [Google Scholar] [CrossRef]
  307. Cappello, E.; Nieri, P. From Life in the Sea to the Clinic: The Marine Drugs Approved and under Clinical Trial. Life 2021, 11, 1390. [Google Scholar] [CrossRef] [PubMed]
  308. Kapoor, S.; Nailwal, N.; Kumar, M.; Barve, K. Recent Patents and Discovery of Anti-inflammatory Agents from Marine Source. Recent Pat. Inflamm. Allergy Drug Discov. 2019, 13, 105–114. [Google Scholar] [CrossRef] [PubMed]
  309. Volkmann, E.R.; Andréasson, K.; Smith, V. Systemic sclerosis. Lancet 2023, 401, 304–318. [Google Scholar] [CrossRef] [PubMed]
  310. Denton, C.P.; Khanna, D. Systemic sclerosis. Lancet 2017, 390, 1685–1699. [Google Scholar] [CrossRef] [PubMed]
  311. Micallef, L.; Vedrenne, N.; Billet, F.; Coulomb, B.; Darby, I.A.; Desmoulière, A. The myofibroblast, multiple origins for major roles in normal and pathological tissue repair. Fibrogenesis Tissue Repair 2012, 5, S5. [Google Scholar] [CrossRef] [PubMed]
  312. Corallo, C.; Cutolo, M.; Volpi, N.; Franci, D.; Aglianò, M.; Montella, A.; Chirico, C.; Gonnelli, S.; Nuti, R.; Giordano, N. Histopathological findings in systemic sclerosis-related myopathy: Fibrosis and microangiopathy with lack of cellular inflammation. Ther. Adv. Musculoskelet. Dis. 2017, 9, 3–10. [Google Scholar] [CrossRef] [PubMed]
  313. Cutolo, M.; Soldano, S.; Smith, V. Pathophysiology of systemic sclerosis: Current understanding and new insights. Expert. Rev. Clin. Immunol. 2019, 15, 753–764. [Google Scholar] [CrossRef] [PubMed]
  314. Anderson, H.A.; Mathieson, J.W.; Thomson, R.H. Distribution of spinochrome pigments in echinoids. Comp. Biochem. Physiol. 1969, 28, 333–345. [Google Scholar] [CrossRef] [PubMed]
  315. Sedova, K.; Bernikova, O.; Azarov, J.; Shmakov, D.; Vityazev, V.; Kharin, S. Effects of echinochrome on ventricular repolarization in acute ischemia. J. Electrocardiol. 2015, 48, 181–186. [Google Scholar] [CrossRef]
  316. Mohamed, A.S.; Sadek, S.A.; Hassanein, S.S.; Soliman, A.M. Hepatoprotective Effect of Echinochrome Pigment in Septic Rats. J. Surg. Res. 2019, 234, 317–324. [Google Scholar] [CrossRef] [PubMed]
  317. Lennikov, A.; Kitaichi, N.; Noda, K.; Mizuuchi, K.; Ando, R.; Dong, Z.; Fukuhara, J.; Kinoshita, S.; Namba, K.; Ohno, S.; et al. Amelioration of endotoxin-induced uveitis treated with the sea urchin pigment echinochrome in rats. Mol. Vis. 2014, 20, 171–177. [Google Scholar]
  318. Lebed’ko, O.A.; Ryzhavskii, B.Y.; Demidova, O.V. Effect of Antioxidant Echinochrome A on Bleomycin-Induced Pulmonary Fibrosis. Bull. Exp. Biol. Med. 2015, 159, 351–354. [Google Scholar] [CrossRef] [PubMed]
  319. Kikionis, S.; Papakyriakopoulou, P.; Mavrogiorgis, P.; Vasileva, E.A.; Mishchenko, N.P.; Fedoreyev, S.A.; Valsami, G.; Ioannou, E.; Roussis, V. Development of Novel Pharmaceutical Forms of the Marine Bioactive Pigment Echinochrome A Enabling Alternative Routes of Administration. Mar. Drugs 2023, 21, 250. [Google Scholar] [CrossRef] [PubMed]
  320. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
  321. Coley, W.B. The treatment of malignant tumors by repeated inoculations of erysipelas: With a report of ten original cases, 1893. Clin. Orthop. Relat. Res. 1991, 266, 3–11. [Google Scholar]
  322. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol. Immunol. 2020, 17, 807–821. [Google Scholar] [CrossRef] [PubMed]
  323. Gordon, S. Elie Metchnikoff: Father of natural immunity. Eur. J. Immunol. 2008, 38, 3257–3264. [Google Scholar] [CrossRef] [PubMed]
  324. Tardito, S.; Martinelli, G.; Soldano, S.; Paolino, S.; Pacini, G.; Patane, M.; Alessandri, E.; Smith, V.; Cutolo, M. Macrophage M1/M2 polarization and rheumatoid arthritis: A systematic review. Autoimmun. Rev. 2019, 18, 102397. [Google Scholar] [CrossRef]
  325. Toledo, D.M.; Pioli, P.A. Macrophages in Systemic Sclerosis: Novel Insights and Therapeutic Implications. Curr. Rheumatol. Rep. 2019, 21, 31. [Google Scholar] [CrossRef] [PubMed]
  326. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef] [PubMed]
  327. Long, K.B.; Beatty, G.L. Harnessing the antitumor potential of macrophages for cancer immunotherapy. Oncoimmunology 2013, 2, e26860. [Google Scholar] [CrossRef] [PubMed]
  328. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
  329. Brzostek, J.; Gascoigne, N.R.; Rybakin, V. Cell Type-Specific Regulation of Immunological Synapse Dynamics by B7 Ligand Recognition. Front. Immunol. 2016, 7, 24. [Google Scholar] [CrossRef] [PubMed]
  330. Amarnath, S.; Mangus, C.W.; Wang, J.C.; Wei, F.; He, A.; Kapoor, V.; Foley, J.E.; Massey, P.R.; Felizardo, T.C.; Riley, J.L.; et al. The PDL1-PD1 axis converts human TH1 cells into regulatory T cells. Sci. Transl. Med. 2011, 3, 111ra120. [Google Scholar] [CrossRef] [PubMed]
  331. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [PubMed]
  332. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef] [PubMed]
  333. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [PubMed]
  334. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutiérrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  335. Kostine, M.; Finckh, A.; Bingham, C.O.; Visser, K.; Leipe, J.; Schulze-Koops, H.; Choy, E.H.; Benesova, K.; Radstake, T.; Cope, A.P.; et al. EULAR points to consider for the diagnosis and management of rheumatic immune-related adverse events due to cancer immunotherapy with checkpoint inhibitors. Ann. Rheum. Dis. 2021, 80, 36–48. [Google Scholar] [CrossRef] [PubMed]
  336. Cutolo, M.; Campitiello, R.; Gotelli, E.; Soldano, S. The Role of M1/M2 Macrophage Polarization in Rheumatoid Arthritis Synovitis. Front. Immunol. 2022, 13, 867260. [Google Scholar] [CrossRef]
  337. Goold, H.D.; Moseley, J.L.; Lauersen, K.J. The synthetic future of algal genomes. Cell Genom. 2024, 4, 100505. [Google Scholar] [CrossRef]
  338. Li, J.; Agathos, S.N.; Gao, Z. Editorial: Emerging trends in genetic engineering of microalgae. Front. Bioeng. Biotechnol. 2024, 12, 1403711. [Google Scholar] [CrossRef]
  339. Torres-Tiji, Y.; Sethuram, H.; Gupta, A.; McCauley, J.; Dutra-Molino, J.V.; Pathania, R.; Saxton, L.; Kang, K.; Hillson, N.J.; Mayfield, S.P. Bioinformatic prediction and high throughput in vivo screening to identify cis-regulatory elements for the development of algal synthetic promoters. bioRxiv 2024. [Google Scholar] [CrossRef]
  340. McQuillan, J.L.; Berndt, A.J.; Sproles, A.E.; Mayfield, S.P.; Pandhal, J. Novel cis-regulatory elements as synthetic promoters to drive recombinant protein expression from the Chlamydomonas reinhardtii nuclear genome. New Biotechnol. 2022, 68, 9–18. [Google Scholar] [CrossRef] [PubMed]
  341. Jacobebbinghaus, N.; Lauersen, K.J.; Kruse, O.; Baier, T. Bicistronic expression of nuclear transgenes in Chlamydomonas reinhardtii. Plant J. 2024, 118, 1400–1412. [Google Scholar] [CrossRef] [PubMed]
  342. Ross, I.L.; Budiman, S.; Le, H.P.; Xiong, D.; Hemker, F.; Millen, E.A.; Oey, M.; Hankamer, B. Strategy for unlimited cycles of scarless oligonucleotide directed gene editing in Chlamydomonas reinhardtii. bioRxiv 2024. [Google Scholar] [CrossRef]
  343. Cutolo, E.A.; Mandalà, G.; Dall’Osto, L.; Bassi, R. Harnessing the Algal Chloroplast for Heterologous Protein Production. Microorganisms 2022, 10, 743. [Google Scholar] [CrossRef]
  344. Inckemann, R.; Chotel, T.; Brinkmann, C.K.; Burgis, M.; Andreas, L.; Baumann, J.; Sharma, P.; Klose, M.; Barret, J.; Ries, F.; et al. Advancing chloroplast synthetic biology through high-throughput plastome engineering of Chlamydomonas reinhardtii. bioRxiv 2024. [Google Scholar] [CrossRef]
  345. Xie, Z.; He, J.; Peng, S.; Zhang, X.; Kong, W. Biosynthesis of protein-based drugs using eukaryotic microalgae. Algal Res. 2023, 74, 103219. [Google Scholar] [CrossRef]
  346. Dubey, K.K.; Kumar, A.; Baldia, A.; Rajput, D.; Kateriya, S.; Singh, R.; Nikita; Tandon, R.; Mishra, Y.K. Biomanufacturing of glycosylated antibodies: Challenges, solutions, and future prospects. Biotechnol. Adv. 2023, 69, 108267. [Google Scholar] [CrossRef] [PubMed]
  347. Akram, M.; Khan, M.A.; Ahmed, N.; Bhatti, R.; Pervaiz, R.; Malik, K.; Tahir, S.; Abbas, R.; Ashraf, F.; Ali, Q. Cloning and expression of an anti-cancerous cytokine: Human IL-29 gene in Chlamydomonas reinhardtii. AMB Express 2023, 13, 23. [Google Scholar] [CrossRef]
  348. Torres-Tiji, Y.; Fields, F.J.; Yang, Y.; Heredia, V.; Horn, S.J.; Keremane, S.R.; Jin, M.M.; Mayfield, S.P. Optimized production of a bioactive human recombinant protein from the microalgae Chlamydomonas reinhardtii grown at high density in a fed-batch bioreactor. Algal Res. 2022, 66, 102786. [Google Scholar] [CrossRef]
  349. Stavridou, E.; Karapetsi, L.; Nteve, G.M.; Tsintzou, G.; Chatzikonstantinou, M.; Tsaousi, M.; Martinez, A.; Flores, P.; Merino, M.; Dobrovic, L.; et al. Landscape of microalgae omics and metabolic engineering research for strain improvement: An overview. Aquaculture 2024, 587, 740803. [Google Scholar] [CrossRef]
  350. Cao, K.; Cui, Y.; Sun, F.; Zhang, H.; Fan, J.; Ge, B.; Cao, Y.; Wang, X.; Zhu, X.; Wei, Z.; et al. Metabolic engineering and synthetic biology strategies for producing high-value natural pigments in Microalgae. Biotechnol. Adv. 2023, 68, 108236. [Google Scholar] [CrossRef]
  351. Sharma, A.; Nawkarkar, P.; Kapase, V.U.; Chhabra, M.; Kumar, S. Engineering of ketocarotenoid biosynthetic pathway in Chlamydomonas reinhardtii through exogenous gene expression. Syst. Microbiol. Biomanuf. 2024. [Google Scholar] [CrossRef]
  352. Kneip, J.S.; Kniepkamp, N.; Jang, J.; Mortaro, M.G.; Jin, E.; Kruse, O.; Baier, T. CRISPR/Cas9-Mediated Knockout of the Lycopene ε-Cyclase for Efficient Astaxanthin Production in the Green Microalga Chlamydomonas reinhardtii. Plants 2024, 13, 1393. [Google Scholar] [CrossRef] [PubMed]
  353. Marcolungo, L.; Bellamoli, F.; Cecchin, M.; Lopatriello, G.; Rossato, M.; Cosentino, E.; Rombauts, S.; Delledonne, M.; Ballottari, M. Haematococcus lacustris genome assembly and annotation reveal diploid genetic traits and stress-induced gene expression patterns. Algal Res. 2024, 80, 103567. [Google Scholar] [CrossRef]
  354. Hammel, A.; Cucos, L.-M.; Caras, I.; Ionescu, I.; Tucureanu, C.; Tofan, V.; Costache, A.; Onu, A.; Hoepfner, L.; Hippler, M.; et al. The red alga Porphyridium as a host for molecular farming: Efficient production of immunologically active hepatitis C virus glycoprotein. Proc. Natl. Acad. Sci. USA 2024, 121, e2400145121. [Google Scholar] [CrossRef] [PubMed]
  355. Hammel, A.; Neupert, J.; Bock, R. Optimized transgene expression in the red alga Porphyridium purpureum and efficient recombinant protein secretion into the culture medium. Plant Mol. Biol. 2024, 114, 18. [Google Scholar] [CrossRef] [PubMed]
  356. Ye, Y.; Liu, M.; Yu, L.; Sun, H.; Liu, J. Nannochloropsis as an Emerging Algal Chassis for Light-Driven Synthesis of Lipids and High-Value Products. Mar. Drugs 2024, 22, 54. [Google Scholar] [CrossRef] [PubMed]
  357. Mariam, I.; Bettiga, M.; Rova, U.; Christakopoulos, P.; Matsakas, L.; Patel, A. Ameliorating microalgal OMEGA production using omics platforms. Trends Plant Sci. 2024. [Google Scholar] [CrossRef] [PubMed]
  358. Græsholt, C.; Brembu, T.; Volpe, C.; Bartosova, Z.; Serif, M.; Winge, P.; Nymark, M. Zeaxanthin epoxidase 3 Knockout Mutants of the Model Diatom Phaeodactylum tricornutum Enable Commercial Production of the Bioactive Carotenoid Diatoxanthin. Mar. Drugs 2024, 22, 185. [Google Scholar] [CrossRef] [PubMed]
  359. Sun, H.; Wang, J.; Li, Y.; Yang, S.; Chen, D.D.; Tu, Y.; Liu, J.; Sun, Z. Synthetic biology in microalgae towards fucoxanthin production for pharmacy and nutraceuticals. Biochem. Pharmacol. 2024, 220, 115958. [Google Scholar] [CrossRef] [PubMed]
  360. Melis, A.; Hidalgo Martinez, D.A.; Betterle, N. Perspectives of cyanobacterial cell factories. Photosynth. Res. 2023. [Google Scholar] [CrossRef] [PubMed]
  361. Bourgade, B.; Stensjö, K. Synthetic biology in marine cyanobacteria: Advances and challenges. Front. Microbiol. 2022, 13, 994365. [Google Scholar] [CrossRef] [PubMed]
  362. Middelboe, M.; Brussaard, C.P.D. Marine Viruses: Key Players in Marine Ecosystems. Viruses 2017, 9, 302. [Google Scholar] [CrossRef] [PubMed]
  363. Zhang, R.; Li, Y.; Yan, W.; Wang, Y.; Cai, L.; Luo, T.; Li, H.; Weinbauer, M.G.; Jiao, N. Viral control of biomass and diversity of bacterioplankton in the deep sea. Commun. Biol. 2020, 3, 256. [Google Scholar] [CrossRef] [PubMed]
  364. Santiago-Rodriguez, T.M.; Hollister, E.B. Unraveling the viral dark matter through viral metagenomics. Front. Immunol. 2022, 13, 1005107. [Google Scholar] [CrossRef] [PubMed]
  365. Allen, M.J.; Cicéron, F.; Monier, A. The potential of nature’s unseen industrious heroes: Marine viruses. Biochem. 2022, 44, 18–21. [Google Scholar] [CrossRef]
  366. D’Adamo, S.; Kormelink, R.; Martens, D.; Barbosa, M.J.; Wijffels, R.H. Prospects for viruses infecting eukaryotic microalgae in biotechnology. Biotechnol. Adv. 2021, 54, 107790. [Google Scholar] [CrossRef] [PubMed]
  367. Wilson, W.H.; Van Etten, J.L.; Allen, M.J. The Phycodnaviridae: The story of how tiny giants rule the world. Curr. Top. Microbiol. Immunol. 2009, 328, 1–42. [Google Scholar] [CrossRef] [PubMed]
  368. Shitrit, D.; Hackl, T.; Laurenceau, R.; Raho, N.; Carlson, M.C.G.; Sabehi, G.; Schwartz, D.A.; Chisholm, S.W.; Lindell, D. Genetic engineering of marine cyanophages reveals integration but not lysogeny in T7-like cyanophages. ISME J. 2022, 16, 488–499. [Google Scholar] [CrossRef] [PubMed]
  369. Novikova, O.; Topilina, N.; Belfort, M. Enigmatic distribution, evolution, and function of inteins. J. Biol. Chem. 2014, 289, 14490–14497. [Google Scholar] [CrossRef]
  370. Allen, M.J.; Lanzén, A.; Bratbak, G. Characterisation of the coccolithovirus intein. Mar. Genom. 2011, 4, 1–7. [Google Scholar] [CrossRef] [PubMed]
  371. Rotter, A.; Barbier, M.; Bertoni, F.; Bones, A.M.; Cancela, M.L.; Carlsson, J.; Carvalho, M.F.; Cegłowska, M.; Chirivella-Martorell, J.; Conk Dalay, M.; et al. The Essentials of Marine Biotechnology. Front. Mar. Sci. 2021, 8, 629629. [Google Scholar] [CrossRef]
  372. Evans, L.S.; Buchan, P.M.; Fortnam, M.; Honig, M.; Heaps, L. Putting coastal communities at the center of a sustainable blue economy: A review of risks, opportunities, and strategies. Front. Political Sci. 2023, 4, 1032204. [Google Scholar] [CrossRef]
  373. Niner, H.J.; Ardron, J.A.; Escobar, E.G.; Gianni, M.; Jaeckel, A.; Jones, D.O.B.; Levin, L.A.; Smith, C.R.; Thiele, T.; Turner, P.J.; et al. Deep-Sea Mining With No Net Loss of Biodiversity—An Impossible Aim. Front. Mar. Sci. 2018, 5, 53. [Google Scholar] [CrossRef]
  374. Paulus, E. Shedding Light on Deep-Sea Biodiversity—A Highly Vulnerable Habitat in the Face of Anthropogenic Change. Front. Mar. Sci. 2021, 8, 667048. [Google Scholar] [CrossRef]
  375. Ferguson, D.K.; Li, C.; Chakraborty, A.; Gittins, D.A.; Fowler, M.; Webb, J.; Campbell, C.; Morrison, N.; MacDonald, A.; Hubert, C.R.J. Multi-year seabed environmental baseline in deep-sea offshore oil prospective areas established using microbial biodiversity. Mar. Pollut. Bull. 2023, 194, 115308. [Google Scholar] [CrossRef] [PubMed]
  376. Zhang, H.; Chen, S. Overview of research on marine resources and economic development. Mar. Econ. Manag. 2022, 5, 69–83. [Google Scholar] [CrossRef]
  377. Ibarbalz, F.M.; Henry, N.; Brandão, M.C.; Martini, S.; Busseni, G.; Byrne, H.; Coelho, L.P.; Endo, H.; Gasol, J.M.; Gregory, A.C.; et al. Global Trends in Marine Plankton Diversity across Kingdoms of Life. Cell 2019, 179, 1084–1097.e1021. [Google Scholar] [CrossRef] [PubMed]
  378. Kumar, A.; AbdElgawad, H.; Castellano, I.; Selim, S.; Beemster, G.T.S.; Asard, H.; Buia, M.C.; Palumbo, A. Effects of ocean acidification on the levels of primary and secondary metabolites in the brown macroalga Sargassum vulgare at different time scales. Sci. Total Environ. 2018, 643, 946–956. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Marine pharmacology has come a long way, from superstitious practices to present-day high-throughput drug discovery pipelines. During the last three decades, the bioprospecting of marine environments has identified hundreds of lead compounds with potential applications in the clinical management of chronic inflammatory diseases and cancer. Historically, the East and West established their own corpuses of marine materia medica. During the last Chinese Imperial Dynasty of the Qing (1644–1911), European missionaries visited and established themselves in China, introducing the Christian faith and other Western cultural elements, including cartography. At the court of the Qing Emperor Shengzu, the Flemish Jesuit and astronomer Ferdinand Verbiest (1623–1688) published the Kunyu Quantu (坤舆全图, Full Map of the World) in 1674, one of several Chinese world maps produced in that era. Geography offered a glimpse into the outer world, attracting the attention of the traditionally self-centered and self-isolated Chinese civilization towards Western science. In the modern era, these two distant cultural worlds began a cross-fertilization of knowledge and, today, they together contribute to advancing the applications of marine resources in human health. Reproduced from [107].
Figure 1. Marine pharmacology has come a long way, from superstitious practices to present-day high-throughput drug discovery pipelines. During the last three decades, the bioprospecting of marine environments has identified hundreds of lead compounds with potential applications in the clinical management of chronic inflammatory diseases and cancer. Historically, the East and West established their own corpuses of marine materia medica. During the last Chinese Imperial Dynasty of the Qing (1644–1911), European missionaries visited and established themselves in China, introducing the Christian faith and other Western cultural elements, including cartography. At the court of the Qing Emperor Shengzu, the Flemish Jesuit and astronomer Ferdinand Verbiest (1623–1688) published the Kunyu Quantu (坤舆全图, Full Map of the World) in 1674, one of several Chinese world maps produced in that era. Geography offered a glimpse into the outer world, attracting the attention of the traditionally self-centered and self-isolated Chinese civilization towards Western science. In the modern era, these two distant cultural worlds began a cross-fertilization of knowledge and, today, they together contribute to advancing the applications of marine resources in human health. Reproduced from [107].
Marinedrugs 22 00304 g001
Figure 3. Antifibrotic activity of EchA in SSc model. EchA reduces skin cell infiltration (M1 and M2 macrophages) and myofibroblast activation, ameliorating skin thickness. Cytokines (IL), cluster of differentiation (CD), reactive oxygen species (ROS), protein-coupled receptor signaling pathway (CCL), transforming growth factor (TGF-β), platelet-derived growth factor receptors (PDGF-Rs), fibroblast growth factor receptor (FGFRs), and Echinochrome A (EchA). Figure created with Biorender (accessed on 12 June 2024), based on [279].
Figure 3. Antifibrotic activity of EchA in SSc model. EchA reduces skin cell infiltration (M1 and M2 macrophages) and myofibroblast activation, ameliorating skin thickness. Cytokines (IL), cluster of differentiation (CD), reactive oxygen species (ROS), protein-coupled receptor signaling pathway (CCL), transforming growth factor (TGF-β), platelet-derived growth factor receptors (PDGF-Rs), fibroblast growth factor receptor (FGFRs), and Echinochrome A (EchA). Figure created with Biorender (accessed on 12 June 2024), based on [279].
Marinedrugs 22 00304 g003
Figure 4. Immune surveillance and immunotherapy with immune checkpoint inhibitors. Illustration of in vitro EPS immunostimulant effect on monocyte-derived macrophages. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), antigen-presenting cell (APC), cytotoxic T-Lymphocyte antigen 4 (CTLA4), T cell receptor (TCR), cytokines (IL), cluster of differentiation (CD), human monocytic cell line (THP-1), tumor necrosis factor (TNF), phorbol 12-myristate13-acetate (PMA), and extracellular polysaccharides s(EPS). Figure created with Biorender (accessed on 12 June 2024), based on [233].
Figure 4. Immune surveillance and immunotherapy with immune checkpoint inhibitors. Illustration of in vitro EPS immunostimulant effect on monocyte-derived macrophages. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), antigen-presenting cell (APC), cytotoxic T-Lymphocyte antigen 4 (CTLA4), T cell receptor (TCR), cytokines (IL), cluster of differentiation (CD), human monocytic cell line (THP-1), tumor necrosis factor (TNF), phorbol 12-myristate13-acetate (PMA), and extracellular polysaccharides s(EPS). Figure created with Biorender (accessed on 12 June 2024), based on [233].
Marinedrugs 22 00304 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cutolo, E.A.; Campitiello, R.; Caferri, R.; Pagliuca, V.F.; Li, J.; Agathos, S.N.; Cutolo, M. Immunomodulatory Compounds from the Sea: From the Origins to a Modern Marine Pharmacopoeia. Mar. Drugs 2024, 22, 304. https://doi.org/10.3390/md22070304

AMA Style

Cutolo EA, Campitiello R, Caferri R, Pagliuca VF, Li J, Agathos SN, Cutolo M. Immunomodulatory Compounds from the Sea: From the Origins to a Modern Marine Pharmacopoeia. Marine Drugs. 2024; 22(7):304. https://doi.org/10.3390/md22070304

Chicago/Turabian Style

Cutolo, Edoardo Andrea, Rosanna Campitiello, Roberto Caferri, Vittorio Flavio Pagliuca, Jian Li, Spiros Nicolas Agathos, and Maurizio Cutolo. 2024. "Immunomodulatory Compounds from the Sea: From the Origins to a Modern Marine Pharmacopoeia" Marine Drugs 22, no. 7: 304. https://doi.org/10.3390/md22070304

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop