ArticlePDF AvailableLiterature Review

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

MDPI
Marine Drugs
Authors:

Abstract and Figures

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.
This content is subject to copyright.
Citation: 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. hps://doi.org/
10.3390/md22070304
Academic Editors: Angelo Fontana
and Carmela Gallo
Received: 27 May 2024
Revised: 24 June 2024
Accepted: 26 June 2024
Published: 28 June 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Swierland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Aribution (CC BY) license (hps://
creativecommons.org/licenses/by/
4.0/).
marine drugs
Review
Immunomodulatory Compounds from the Sea: From the
Origins to a Modern Marine Pharmacopoeia
Edoardo Andrea Cutolo 1, *,, Rosanna Campitiello 2, 3, , Roberto Caferri 1, Viorio Flavio Pagliuca 1, Jian Li 4,
Spiros Nicolas Agathos 4, 5 and Maurizio Cutolo 2, 3
1Laboratory of Photosynthesis and Bioenergy, Department of Biotechnology, University of Verona, Strada le
Grazie 15, 37134 Verona, Italy
2Laboratory of Experimental Rheumatology and Academic, Division of Clinical Rheumatology, Department
of Internal Medicine, University of Genoa, 16132 Genoa, Italy
3IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
4Qingdao Innovation and Development Base, Harbin Engineering University, No. 1777 Sansha Road,
Qingdao 150001, China; jian.li@hrbeu.edu.cn (J.L.); spiros.agathos@hrbeu.edu.cn (S.N.A.)
5Bioengineering Laboratory, Earth and Life Institute, Catholic University of Louvain,
B‑1348 Louvain‑la‑Neuve, Belgium
*Correspondence: edoardoandrea.cutolo@univr.it
These authors contributed equally to this work.
Abstract: From sea shores to the abysses of the deep ocean, marine ecosystems have provided human‑
ity with valuable medicinal resources. The use of marine organisms is discussed in ancient pharma‑
copoeias of dierent 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 poten‑
tial. This renewed interest in marine drugs is propelled by a burgeoning research eld investigating
the molecular mechanisms by which newly identied compounds intervene in the pathophysiology
of human diseases. Of great clinical relevance are molecules endowed with anti‑inammatory and
immunomodulatory properties with emerging applications in the management of chronic inam‑
matory 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 re‑
sources through biotechnology.
Keywords: bioprospecting; inammation; autoimmunity; synthetic biology; drug discovery; genetic
engineering; immunomodulation; deep sea; systemic sclerosis; rheumatoid arthritis
1. Why Does the Sea Maer 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, cul‑
tural exchanges, and trade [1]. From the philosophers of the classical antiquity to present‑
day oceanographic expeditions, the scientic study of the sea has been a constant human
endeavour stimulated by the fascination with its biological diversity [2].
The classication of marine life, however, is still far from oering a conclusive picture.
According to initial estimates, of the ~8.75 million species inhabiting the planet, ~2.2 mil‑
lion 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 signicantly 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 en‑
tirely covering marine life, only several hundred years from now [6].
Mar. Drugs 2024,22, 304. https://doi.org/10.3390/md22070304 https://www.mdpi.com/journal/marinedrugs
Mar. Drugs 2024,22, 304 2 of 32
Oceans are still largely underexplored sources of lead compounds [7]. From coral
reefs to hydrothermal vents, the ocean is characterized by diverse habitats, including ex‑
treme 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 [913]. Immune‑mediated inammatory diseases
are a signicant burden for national healthcare systems, especially in high‑income coun‑
tries, with an increasing incidence registered over the last three decades [14,15]. Moreover,
in nearly all forms of cancer, chronic inammation 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 [1922], including anti‑inammatory and im‑
munomodulatory properties [2327]. Although marine pharmacology has its roots in an‑
tiquity, the ocean is witnessing a scientic renaissance propelled by interdisciplinary drug
discovery research assisted by powerful, high‑throughput technologies like untargeted
metagenomics and metabolomics [2831].
2. Marine Pharmacology in the Mists of Time
From time immemorial, marine ora and fauna have been used in folk and tradi‑
tional medicine (TM) [32]. However, in most cases, this knowledge is orally transmied;
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 identication 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 sug‑
gesting the trading of kelp (Laminariales) between coastal and mountainous areas of the
Peruvian and Chilean Andes [3840]. 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 [4348]. Recent ethnopharmacological studies described at least 300 TM uses
of marine animals globally [49,50], of which, however, only invertebrates were conrmed
sources of therapeutic compounds [51]. The overexploitation of marine animals in TM, par‑
ticularly 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 [5254], and, in turn, precludes the identication of new, potentially therapeu‑
tic 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 (fth 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 Hip‑
pocrates 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 denition of inammation: Notae vero inammationis sunt
quatuor: rubor et tumor cum calore et dolore (the signs of inammation are four: redness,
swelling, fever, and pain) [64]. From the second century BC onwards, technology and sci‑
Mar. Drugs 2024,22, 304 3 of 32
ence [65,66] constantly owed 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 fteenth century, European countries assembled
vast collections of ora 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 owed 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 West‑
ern medicine eventually being united in 1948 under the aegis of the International Pharma‑
copoeia of the World Health Organization [72].
In the 1960s, Western science achieved a deeper understanding of immunity and in‑
ammation with the elucidation of the structure of antibodies and their generation via ge‑
netic recombination, as well as the identication 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, caus‑
ing chronic inammation—was formulated in 1892 by German physician Paul Ehrlich, al‑
though it was rejected as “physiologically inconceivable” and referred to as horror auto‑
toxicus [73]. Only in 1965 was autoimmunity recognized as a common immunological
disorder underlying the pathogenesis of chronic inammatory 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 inammatory
mediators are known, including sub‑populations of immune cell types, their released solu‑
ble factors (cytokines, antibodies), and the intracellular genetic and molecular mechanisms
which sustain inammatory disease [76].
4. The Evolution of Marine Materia Medica in the Far East
Wrien records of marine ora appear in the 2500‑year‑old Chinese book the Classic
of Poetry (Shijing,诗经) [7780]. The main pharmacological heritage in the Far East, how‑
ever, 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 rst 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 cosmopoli‑
tan 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 Paern) 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 Bar‑
barian Nations, 1225), also published under the Song rule, reected the impact of the Mar‑
itime Silk Road seafaring on the evolution of Chinese medical knowledge [8789]. Under
the Mongol‑led Yuan Dynasty (1271 to 1368), scientic 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 scientic literature describing 1892 medicinals. This text includes a detailed de‑
Mar. Drugs 2024,22, 304 4 of 32
scription of marine fauna and ora and stood until the nineteenth century as a reference
for taxonomical classication in East Asia [93]. Notably, the Bencao Gangmu contains med‑
ical prescriptions based on seaweeds of the Ulvophyceae, Phaeophyceae, Florideohyceae,
Trebouxiophyceae, and Bangiophyceae classes; cyanobacteria of the Nostoc genus; and sea‑
horses [94,95].
The species Hippocampus kuda is known to produce an antitumor peptide with in‑
hibitory 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 [96102]. 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 Hippocam‑
pus species in both East Asia and Latin America [104], which are now included in the Ap‑
pendix 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 vis‑
ited and established themselves in China, introducing the Christian faith and other West‑
ern 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 pro‑
duced in that era. Geography oered a glimpse of the outer world, aracting the aention
of the traditionally self‑centered and self‑isolated Chinese civilization towards Western sci‑
ence (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 sci‑
entic 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 scientic value, this project, supported by the
special program “Project 908” (Comprehensive investigation and evaluation on the o‑
shore 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], oers a privileged insight into one of the oldest sur‑
viving human civilizations, reecting the uninterrupted continuity of Chinese culture
throughout the millennia [115].
Mar. Drugs 2024,22, 304 5 of 32
Mar. Drugs 2024, 22, 304 5 of 34
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 (, 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, at-
tracting the attention of the traditionally self-centered and self-isolated Chinese civilization to-
wards 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 [111].
The opening of the Marine Biological Station of Amoy University in the 1930s under
the guidance of foreign-trained Chinese biologists [114] 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 [115].
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 un-
sung hero of the 1940s modernization movement known as “Saving the Country by
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 ma‑
rine environments has identied hundreds of lead compounds with potential applications in the
clinical management of chronic inammatory 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, intro‑
ducing 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 oered a glimpse into the outer world, aracting the aen‑
tion 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].
The Yin Yang Dialectic of Autoimmunity
With the introduction of Western medicine into China in the middle seventeenth cen‑
tury, traditional Chinese medicine (TCM) started to evolve in the constant struggle be‑
tween traditionalism and modernization [116]. In TCM, spiritual and scientic concepts
coexist in a holistic discipline [117], which is in stark contrast with the reductionist Western
approach. However, some TCM principles reect key features of the immune system: bal‑
ance, defense, holism, and circadian rhythms [118]. According to TCM, healthy immune
functions require a harmonious equilibrium of Yin reserves (, organs, tissues, cells, and
body uids) and Yang (, physiological functions). The two are opposing forces constantly
Mar. Drugs 2024,22, 304 6 of 32
trying to win over one another. Yin and Yang are kept in a constant dynamic balance to pre‑
serve the Qi (), the body’s vital energy [119], a concept analogous to Pneuma (breath of life,
spirit) from classical antiquity [120]. A persistent Yin deciency causes Shanghuo (上火), or
heat syndrome, eventually creating an “excess of pathogenesis caused by deciency” [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 degrada‑
tion, the excessive exposure of auto‑antigens to the immune system breaks immunolog‑
ical tolerance, triggering autoimmunity. Remarkably, several immune components em‑
body the Yin Yang dialectic [122]. For instance, CD4+ T helper cells undergo dynamic
functional specialization via cytokine‑mediated signaling feedback [123]. Initially, a Th1
dierentiation 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 dierentiation programs in response to
chemical stimuli [124]. Instead, regulatory T cells—a lymphocyte population which sup‑
presses immune responses and maintains self‑tolerance—can convert to inammatory
Th17 cells [125]. Finally, individual cytokines can be either Yin or Yang elements, i.e., IL‑6 is
endowed with both pro‑ and anti‑inammatory functions depending on the context [126].
Strikingly, many TCM practices either stimulate or suppress immune functions [127129];
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 scientic 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 Ameri‑
can microbiologist Claude ZoBell [133]. Decades later, the discovery of marine cyanobac‑
teria 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 bio‑
diversity [136]. Metagenomics was rst introduced in marine sciences by The Sorcerer II
Global Ocean Sampling expedition led by American biotechnologist Craig Venter
(2004–2006) [137139]. The subsequent Malaspina (2010) and Tara Oceans expeditions
(2009–2013) further revealed the richness of microbial life from the deep‑sea
environments [140142]. The picture of the global ocean microbial genome was recently as‑
sembled in the KAUST Metagenome Analysis Platform (KMAP) Global Ocean Gene Cata‑
log 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‑
specic communication [144]. During evolution, key genes encoding polyketide synthases
and non‑ribosomal peptide synthetases have undergone extensive reshuing within oper‑
ons, creating a remarkable diversication of metabolic pathways [145]. A recent analysis of
marine prokaryotic genomes revealed an astonishing complexity of biosynthetic gene clus‑
ters, 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
Mar. Drugs 2024,22, 304 7 of 32
from the associated microbiota [149]. Mutualistic relationships between fungi and bacte‑
ria and metazoans (holobionts) are found in corals and sponges [150]. Strikingly, nearly
all bioactive polyketides and peptides isolated from the Theonella swinhoei (porifera) holo‑
biont are produced by the lamentous bacterial symbiont Entotheonella spp. [151153]. Sim‑
ilarly, the anticancer molecule trabectedin was isolated from Candidatus Endoecteinascidia
frumentensis, the bacterial symbiont of the sea squirt Ecteinascidia turbinata [154]. How‑
ever, 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‑inammatory
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 seawa‑
ter and wastewater [161]. Notably, the anti‑inammatory 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‑inammatory and immunomodulatory pigments and
lipids [159,164167].
Carotenoids are lipophilic pigments [168171] 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 detoxication of reactive oxidative species (ROS) generated by photosyn‑
thetic reactions. The ketocarotenoid astaxanthin is the microalgal pigment of greatest phar‑
macological 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 (previ‑
ously named Haematococcus pluvialis) [172]. Accordingly, the establishment of optimal cul‑
tivation strategies of H. lacustris and the domestication of high‑yielding strains are key to
accruing astaxanthin accumulation [173176].
In response to stress, several microalgae synthesize DHA, EPA[177], while the chloro‑
phyte Tetraselmis chui accumulates monogalactosyldiacylglycerols, which inhibit nitric ox‑
ide production [178181]. The haptophyte Tisochrysis lutea (formerly known as Isochrysis
anis 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 [182184]. 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 [187190].
At present, China is the major world producer of microalgal biomass for human con‑
sumption [191], while the US and several European countries are at the forefront of mi‑
croalgal biotechnology research [192,193]. Bioprospecting for industrially relevant microal‑
gae, instead, is conducted worldwide, mainly in inhospitable habitats, since extremophiles
hyper‑accumulate bioactive compounds [194] and display robust growth phenotypes un‑
der co‑cultivation [195197].
Mar. Drugs 2024,22, 304 8 of 32
The full biotechnological exploitation of non‑conventional microalgae, however, re‑
quires 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‑inammatory and immunomodulatory properties, including
polysaccharides, phenols, avonoids, and phycobiliproteins [201204]. Notably, the light‑
harvesting pigment C‑phycocyanin is a selective inhibitor of the enzyme cyclooxigenase‑2
producing the pro‑inammatory mediator prostaglandin E2[205208].
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 mys‑
terious 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 At‑
lantic 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 bi‑
ology 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 eld 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 labora‑
tory [219]. In 1979, the manned submersible vessel Alvin enabled the discovery of hy‑
drothermal vents in the Galápagos Rift of the East Pacic 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 identied 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) [223226]. Deep‑sea biodi‑
versity 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 [227229].
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 inves‑
tigation of their secondary metabolites and, recently, genetic engineering [230,231]. Bio‑
prospecting for anti‑inammatory compounds of deep‑sea bacteria resulted in the identi‑
cation 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‑inammatory gene ex‑
pression and prevented the production of the inammatory mediators interleukin‑1βand
IL‑6 [232]. Another example of a bacterium‑derived compound with immunomodulatory
properties is the exopolysaccharide from Planococcus rietoensis (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 heteroge‑
neous, as revealed by recent investigations into the sponge‑associated microbiome [234].
Mar. Drugs 2024,22, 304 9 of 32
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 reected by the increasing number of
newly identied 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‑inammatory com‑
pounds 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 hepi‑
alide (Figure 2.4) from Samsoniella hepiali. The laer also produces uridine, ergosterol, wal‑
terolactone A, (4R, 5S)‑5‑hydroxyhexan‑4‑olide, and myrothecol (Figure 2.5–10). Further‑
more, Acremonium sp. and Eutypella sp. accumulate eremophilane‑like sesquiterpenoids
(Figure 2.11) [237241], while the Ascomycetes Penicillium oxalicum and chrysogenum pro‑
duce alkaloids and chrysamides (Figure 2.12–15), respectively, capable of suppressing the
synthesis of pro‑inammatory 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‑neuroinammatory properties
(Figure 2.16) [244].
Table 1. Recently discovered anti‑inammatory and immunomodulatory compounds from deep‑
sea microorganisms.
Molecule Source Organism(s) Biological Activity—Half Maximal Inhibitory
Concentration (IC50)
Development
Stage Ref.
7,13‑epoxyl‑macrolactin A Bacillus 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 Exopolysaccharide
Planococcus rietoensis
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‑1βand 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 hepialide Samsoniella 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 A Samsoniella hepiali W7
(Ascomycota) Suppression of nitric oxide release by cultured
activated murine microglia (IC50 1 µM). Preclinical trial [238]
(4R,5S)‑5‑hydroxyhexan‑4‑olide Samsoniella 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 µgmL1). 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]
Mar. Drugs 2024,22, 304 10 of 32
Table 1. Cont.
Molecule Source Organism(s) Biological Activity—Half Maximal Inhibitory
Concentration (IC50)
Development
Stage Ref.
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‑inammatory 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]
Mar. Drugs 2024, 22, 304 11 of 34
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 Ba-
cillus subtilis B5; (2) cyclopenol from Aspergillus sp. [241]; (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. [243]; (11) eremophilane from
Acremonium sp. and Eutypella sp. [244,245] (1214) oxaline, isorhodoptilometrin, and 5-hydroxy-7-
(2’-hydroxypropyl)-2-methyl-chromone from Penicillium oxalicum [246]; (15) chrysamide from Pen-
icillium chrysogenum [247]; and (16) phenazostatin from Cystobasidium laryngis [248].
7. Emerging Marine Immunomodulatory Lead Compounds
7.1. Seaweeds (Macroalgae)
Seaweeds, or macroalgae, are plant-like multicellular phototrophs found in inter-
tidal waters attached to rocks or floating free in the open sea [198]. The edible red sea-
weed Porphyra dentata (Rhodophyta, Bangiales) is widely used worldwide in TM and
contains phenolic compounds which inhibit the synthesis of the pro-inflammatory sig-
naling molecule nitric oxide (NO) and suppress the NF-kB pathway in vitro [249].
Several species are farmed in the ocean due to their highly nutritious composition
and content of bioactive compounds, mainly sulfated polysaccharides [250–252]. For in-
stance, porphyran, a sulfated galactan from red seaweeds (Porphyra), stimulates the
immune function and apoptotic/autophagic processes, and is considered a candidate an-
ti-cancer drug (IC
50
20 µg/mL) [253]. Similarly, the polysaccharide fraction from
Lithothamnion muelleri (Hapalidiaceae) displayed immunomodulatory activity by inhibit-
ing the synthesis of pro-inflammatory chemokines in an animal model of arthritis [254].
Moreover, fucoidan and ulvan extracted from the brown macroalga (kelp) Undaria pin-
natifida and the green alga Ulva lactuca (IC
50
623.58–785.48 µg/mL), respectively, are po-
tent immunostimulators and antioxidants, and thus, have potential applications to re-
Figure 2. Chemical structures of recently identied immunomodulatory and anti‑inammatory
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,
Mar. Drugs 2024,22, 304 11 of 32
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 Peni‑
cillium chrysogenum [243]; and (16) phenazostatin from Cystobasidium laryngis [244].
7. Emerging Marine Immunomodulatory Lead Compounds
7.1. Seaweeds (Macroalgae)
Seaweeds, or macroalgae, are plant‑like multicellular phototrophs found in intertidal
waters aached to rocks or oating free in the open sea [194]. The edible red seaweed Por‑
phyra dentata (Rhodophyta, Bangiales) is widely used worldwide in TM and contains phe‑
nolic compounds which inhibit the synthesis of the pro‑inammatory 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 [246248]. For in‑
stance, porphyran, a sulfated galactan from red seaweeds (Porphyra), stimulates the im‑
mune function and apoptotic/autophagic processes, and is considered a candidate anti‑
cancer drug (IC50 20 µg/mL) [249]. Similarly, the polysaccharide fraction from Lithotham‑
nion muelleri (Hapalidiaceae) displayed immunomodulatory activity by inhibiting the syn‑
thesis of pro‑inammatory chemokines in an animal model of arthritis [250]. Moreover,
fucoidan and ulvan extracted from the brown macroalga (kelp) Undaria pinnatida and the
green alga Ulva lactuca (IC50 623.58–785.48 µg/mL), respectively, are potent immunostim‑
ulators and antioxidants, and thus, have potential applications to reduce the side eects
of immunosuppressive therapies [251,252]. Anti‑inammatory eects were reported for
polysaccharide extracts from Halimeda tuna (Ulvophyceae) [10] and Posidonia oceanica (Al‑
ismatales) [253], and for carrageenans derived from dierent red seaweeds (Chondrus cris‑
pus,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‑inammatory mediators in animal models of
arthritis [255,256].
7.2. Invertebrates
Several marine invertebrate phyla are known producers of anti‑inammatory and im‑
munomodulatory compounds [257,258]. Corals are colonial organisms of the class An‑
thozoa (Cnidaria), typically found in reef ecosystems in tropical and sub‑tropical water.
Coral bioprospecting is mainly focussed on the octocorallia order Alcyonacea (Gorgoni‑
ans or soft corals), which is known to produce a vast repertoire of secondary metabo‑
lites 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‑inammatory 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 oxy‑
genated metabolites of C20 fay acids involved in the modulation and resolution of inam‑
mation [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) inhib‑
ited the synthesis of several pro‑inammatory mediators in arthritis animal
models [266268]. Lastly, peptides isolated from the venom of the jellysh 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 bioac‑
tive compounds, mainly derived from their innate immune system [270]. Recently, a syn‑
thetic peptide derived from the sea squirt Styela clava was shown to exert both antimicrobial
Mar. Drugs 2024,22, 304 12 of 32
and immunomodulatory activities in animal models. In particular, the clavanin‑MO pep‑
tide (used at 2 µM) promoted the synthesis of the anti‑inammatory cytokine IL‑10 while
suppressing the release of the pro‑inammatory factors IL‑12 and tumor necrosis factor‑
α(TNF‑α) upon bacterial infection [271]. Another study described the in vitro inhibitory
eects of chemical inhibitors based on the structure of metabolites from Herdmania mo‑
mus against multiple pro‑inammatory enzymes and the release of pro‑inammatory cy‑
tokines 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 sulde donors in vitro, sup‑
pressing the pro‑inammatory 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 eects
of the mucus of Bolinus brandaris, suggesting that a still‑uncharacterized compound could
trigger the immune system against cancerous cells by inducing monocyte dierentiation
(IC50 ranging between 1 and ~10 µg/mL) [274]. Moreover, lipids extracted from the mus‑
sel Mytilus coruscus exerted a strong anti‑inammatory eect in a murine arthritis model,
reducing the levels of the pro‑inammatory mediators leukotriene B₄, prostaglandin E₂,
and thromboxane B₂, but also of the cytokines IL‑1β, IL‑6, interferon‑γ, and tumor necro‑
sis 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‑inammatory enzymes and nitric oxide pro‑
duction 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‑inammatory lead compound identi‑
ed in Scaphechinus mirabilis (described in detail in Section 8.1) [279]. Another example in‑
cludes Isostichopus badionotus, whose extracts suppress the expression of pro‑inammatory
genes in vivo [280]. Moreover, a recent study described the anti‑inammatory activity
of the protein cargo of extracellular vesicles of the sea cucumber (Holothuroidea) Sticho‑
pus japonicus, showing a strong inhibition of the release of pro‑inammatory 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 ter‑
penoids [282]. Early studies have reported the in vitro and in vivo anti‑inammatory activ‑
ity 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 chem‑
ical nature of their bioactive compounds could not be identied [284]. Recently, stular‑
ins (bromotyrosine acids) isolated from Ecionemia acervus (Ancorinidae) strongly inhibited
the activity of pro‑inammatory enzymes and the release of inammatory cytokines from
cultured macrophages (tested range between 5 and µM) [285]. Similarly, the brominated
alkaloid aeroplysinin derived from Aplysina aerophoba displayed inhibitory eects on cul‑
tured vascular endothelial cells by suppressing the NF‑kB pathway (IC50 3µM) [286]. Fur‑
thermore, 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‑inammatory cytokine
IL‑6 (tested range between 0.3 and 10 µM) [287]. Finally, lipids extracted from Halichondria
sitiens exerted immunomodulatory eects on cultured dendritic cells (used at 10 µg/mL)
by suppressing the secretion on the pro‑inammatory cytokines IL‑12 and Il‑6, but also
prevented the production of IFN‑γby CD4+ T lymphocytes, thus blocking the so‑called
Th1‑type immune response (explained in detail in Section 8.2) [288].
Mar. Drugs 2024,22, 304 13 of 32
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 [290293], and recent ethnopharmacological stud‑
ies have isolated several phytochemicals with anti‑inammatory 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‑
inammatory cytokines (TNF‑α, IL‑6, and IL‑12) by in vitro cultured immune cells [295],
while the extracts from Lumniera racemosa displayed anti‑angiogenic properties (IC50 rang‑
ing between 2,57 and 4,95 µM) [297]. Moreover, agalloide terpenoids from Ceriops decan‑
dra (tested at 100 µM), and, mainly, Excoecaria agallocha, suppressed NF‑kB pathway ac‑
tivation [298300]. 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
identied in the fungus Roussoella sp. after isolation from a mangrove sediment, which
inhibited the synthesis of pro‑inammatory cytokines by cultured immune cells [302].
8. Marine Pharmacology, Quo Vadis?
Several marine lead compounds are currently being assessed in pre‑clinical and clin‑
ical studies, while seven marine drugs have already received “rst‑in‑class” status, i.e.,
are endowed with “new and unique mechanism(s) of action” [303306]. Most marine
drugs in clinical use nd application in cancer immunotherapy, as antibody–drug con‑
jugates and in the management of chronic inammatory conditions (reviewed in detail
in [307,308]). Two examples of recently identied 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 charac‑
terized by microvascular damage followed by aberrant autoimmune responses of the skin
and internal organs, including the gastrointestinal tract, kidneys, lungs, and heart [309]. Fi‑
brosis is a hallmark of SSc pathogenesis. This process is driven by activated pro‑brotic my‑
obroblasts, highly dierentiated cells which produce contractile proteins such as alpha‑
smooth muscle actin, resulting in excessive extracellular matrix deposition [310]. Although
myobroblasts contribute to the physiological process of wound healing in damaged tis‑
sues, their aberrant activation contributes to diuse brosis and chronic
inammation [311,312]. Innate immune cells—particularly monocytes and macro‑
phages—are established mediators of the brotic process in SSc [313]. Therefore, the dis‑
covery of novel immunomodulatory and anti‑brotic molecules is of great clinical rele‑
vance for slowing SSc progression.
The marine compound Echinochrome A (6‑ethyl‑2,3,5,7,8‑pentahydroxy‑1,4‑naphtho
quinone, EchA) is a natural pigment from the echinoderm Scaphechinus mirabilis [314] en‑
dowed with antioxidant anti‑brotic properties [315318]. 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‑inammatory immune cells. The administra‑
tion of EchA suppressed dierent mechanisms involved in the brotic process, includ‑
ing broblast activation and myobroblast maturation; tumor growth factor (TGF)‑β1‑
mediated expression of smooth muscle actin; phosphorylation of pro‑brotic transcription
factors in skin broblasts; and, nally, dierentiation of macrophages into both M1 and M2
cells (Figure 3). These eects resulted in lower serum concentrations of pro‑inammatory
cytokines TNF‑αand IFN‑γ. Overall, EchA is a promising marine lead compound with
anti‑brotic properties and, thus, potential application in the clinical management of SSc.
Recently, a novel administration system based on polymeric nanobers was developed to
Mar. Drugs 2024,22, 304 14 of 32
improve the water solubility of EchA. This pharmacological advance is expected to pro‑
mote the controlled release of the drug, enhancing its bioavailability [319].
Mar. Drugs 2024, 22, 304 15 of 34
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 Bio-
render (accessed on 12 June 2024), based on [283].
8.2. Deep-Sea Bacteria Exopolysaccharides and Their Applications in Cancer Immunotherapy
The interplay between immunity and tumorigenesis is a cornerstone of cancer biol-
ogy, since the immune system exerts a multifaceted influence in terms of thwarting tu-
mor initiation, progression, and metastasis [324]. 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 [325]. It is
now well established that tumor recognition and rejection by the immune system in-
volve 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), regulato-
ry T cells (Tregs), and myeloid-derived suppressor cells [326]. Moreover, macrophages
highly adaptable phagocytic immune cells [327]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
[328]. M1 and M2, however, are the extremes of a broader cell type spectrum [329–331],
since macrophages are known to interfere with tumorigenesis by influencing angiogene-
sis, fibrosis, and tumor cell phagocytosis. Moreover, macrophages orchestrate immuno-
surveillance by expressing costimulatory molecules like CD86 (B7-2) or T cell inhibitory
molecules and promote the recruitment of immunosuppressive T-reg cells [332].
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 [333]. Notably, upon cytokine signaling, APCs mediate the “im-
Figure 3. Antibrotic activity of EchA in SSc model. EchA reduces skin cell inltration (M1 and
M2 macrophages) and myobroblast activation, ameliorating skin thickness. Cytokines (IL), cluster
of dierentiation (CD), reactive oxygen species (ROS), protein‑coupled receptor signaling pathway
(CCL), transforming growth factor (TGF‑β), platelet‑derived growth factor receptors (PDGF‑Rs), ‑
broblast growth factor receptor (FGFRs), and Echinochrome A (EchA). Figure created with Biorender
(accessed on 12 June 2024), based on [279].
8.2. Deep‑Sea Bacteria Exopolysaccharides and Their Applications in Cancer Immunotherapy
The interplay between immunity and tumorigenesis is a cornerstone of cancer biol‑
ogy, since the immune system exerts a multifaceted inuence 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 sarco‑
mas 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 com‑
plex 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 dierenti‑
ate into classically activated (M1) and alternatively activated (M2) types, with the former
promoting inammation and the laer fostering tissue repair [324]. M1 and M2, how‑
ever, are the extremes of a broader cell type spectrum [325327], since macrophages are
known to interfere with tumorigenesis by inuencing angiogenesis, brosis, and tumor
cell phagocytosis. Moreover, macrophages orchestrate immunosurveillance by expressing
Mar. Drugs 2024,22, 304 15 of 32
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 activa‑
tion, 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, neoplas‑
tic cells can still escape the immune system defense mechanisms [331]. Immunotherapy
aims at overcoming this phenomenon by unleashing the host immune system against ma‑
lignant 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 regula‑
tory 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 autoim‑
munity represents a continuously advancing area of study. Therefore, the identication of
novel immunoactive molecules is crucial to developing new therapeutic strategies.
A recent study described the immunostimulating eect of a marine exopolysaccha‑
ride (EPS) produced by the deep‑sea psychrotolerant Gram‑positive bacterium Planococcus
rietoensis AP‑5 [233]. This compound was tested on in vitro‑cultured THP‑1 monocytes
dierentiated into macrophage‑like cells and treated with dierent 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) produc‑
tion, 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).
Mar. Drugs 2024,22, 304 16 of 32
Mar. Drugs 2024, 22, 304 17 of 34
Figure 4. Immune surveillance and immunotherapy with immune checkpoint inhibitors. Illustra-
tion 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), clus-
ter of differentiation (CD), human monocytic cell line (THP-1), tumor necrosis factor (TNF), phor-
bol 12-myristate13-acetate (PMA), and extracellular polysaccharides s(EPS). Figure created with
Biorender (accessed on 12 June 2024), based on [237].
8.3. Synthetic Biology and Molecular Pharming in Microalgae
The genetic manipulation of microalgal genomes represents a booming field in bio-
technology, projecting photosynthetic microbes as viable alternatives to conventional
heterotrophic hosts (bacteria and yeasts) for the production of high-value recombinant
therapeutics [341,342]. Advanced genetic tools are constantly being developed to: (i)
achieve high-expression of foreign DNA sequences coupled to synthetic cis-acting regu-
latory elements [343,344]; (ii) introduce multigene expression constructs [345]; (iii) con-
duct iterative editing interventions in the nuclear genome [346]; and (iv) exploit the chlo-
roplast genome for metabolic engineering and production of recombinant proteins
[347,348]. 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 [349]. Different
classes of recombinant therapeutics can be produced in microalgae, including full-length
antibodies [350], anti-cancer cytokines [351], and immune receptors [352].
Furthermore, genetic egineering can be employed to enhance the yield of functional
metabolites. For instance, endogenous metabolic circuits can be rewired to hyperaccu-
mulate specific pathway intermediates or modified to accumulate non-native metabo-
lites. Alternatively, entirely new biosynthetic pathways can be introduced to produce ex-
Figure 4. Immune surveillance and immunotherapy with immune checkpoint inhibitors. Illus‑
tration of in vitro EPS immunostimulant eect 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
dierentiation (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].
8.3. Synthetic Biology and Molecular Pharming in Microalgae
The genetic manipulation of microalgal genomes represents a booming eld in biotech‑
nology, 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 pro‑
teins [343,344]. Microalgae are suitable platforms for producing recombinant protein‑based
therapeutics since they perform eukaryotic post‑translational modications and can be en‑
gineered to secrete heterologous products in the cultivation media [345]. Dierent classes
of recombinant therapeutics can be produced in microalgae, including full‑length antibod‑
ies [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 hyperaccu‑
mulate specic pathway intermediates or modied to accumulate non‑native metabolites.
Alternatively, entirely new biosynthetic pathways can be introduced to produce exotic
Mar. Drugs 2024,22, 304 17 of 32
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 facili‑
tate 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 rein‑
hardtii, engineering strategies are currently being tested in non‑conventional strains, in‑
cluding the marine rhodophyte Porphyridium purpureum, in which the expression of a gly‑
cosylated viral antigen was recently reported [354]. This is expected to signicantly expand
the range of therapeutic uses of microalgae, with respect to both recombinant protein ex‑
pression [355] and hyper‑accumulation of high‑value pigments and lipids [356359]. Fi‑
nally, current developments of synthetic biology in marine cyanobacteria are expected to
introduce signicant novelties into the biomanufacturing of therapeutics in these highly
productive photosynthetic microbes [360,361].
8.4. The Marine Viral Dark Maer and Its Potential for Medical Biotechnology
Despite not nding a place in the tree of life, viruses are the undisputed engines of evo‑
lution in the marine biosphere and are major drivers of its biogeochemical
cycles [362,363]. The genetic complexity of the “marine viral dark maer” is just begin‑
ning 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, re‑
spectively. Moreover, certain structural features of viral proteins have broader relevance
for biotechnology. One example is inteins, self‑cleavable protein splicing elements en‑
riched in marine viral genomes, which have been engineered into valuable biotechnolog‑
ical tools [369,370]. However, due to the scarcity of reports describing the use of marine
viruses in biotechnology, at present, it is dicult to predict their impact on future devel‑
opments 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 bio‑
prospecting of marine environments has identied hundreds of lead compounds with po‑
tential applications in the clinical management of chronic inammatory diseases and can‑
cer. 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 threaten‑
ing marine biodiversity due to the intense activities of shipping, transportation, sheries,
tourism, and renewable energy production. Among these, seabed mining has been pro‑
posed as a severe cause of biodiversity erosion [371374], 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 ecophysiol‑
ogy. 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,
Mar. Drugs 2024,22, 304 18 of 32
puing at risk these under‑explored fragile ecosystems and, thus, precluding future bio‑
prospecting endeavours [377]. On the other hand, ocean acidication, a direct consequence
of rising atmospheric CO2levels, is suggested to interfere with the metabolism of marine
ora, particularly of seaweeds, aecting their polysaccharide, fay acid, and secondary
metabolite composition and prole and potentially altering their reported pharmacologi‑
cal uses [378]. Therefore, the ultimate frontier of marine pharmacology lies in the devel‑
opment 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: Wewish to thank Arianna (Cai Hang) for assistance with the revision of Chinese
writing in the text.
Conicts of Interest: The authors declare no conict 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.
2. Deacon, M. Scientists and the Sea, 1650–1900: A Study of Marine Science; Academic Press: Cambridge, MA, USA, 1971.
3. Mora, C.; Tiensor, 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. [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. [CrossRef] [PubMed]
5. Baldrian, P.; Větrovský, T.; Lepinay, C.; Kohout, P. High‑throughput sequencing view on the magnitude of global fungal diver‑
sity. Fungal Divers. 2022,114, 539–547. [CrossRef]
6. Bouchet, P.; Decock, W.; Lonneville, B.; Vanhoorne, B.; Vandepie, L. Marine biodiversity discovery: The metrics of new species
descriptions. Front. Mar. Sci. 2023,10, 929989. [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. [CrossRef] [PubMed]
8. Moles, J.; Torrent, A.; Alcaraz, M.J.; Ruhí, R.; Avila, C. Anti‑inammatory activity in selected Antarctic benthic organisms. Front.
Mar. Sci. 2014,1, 24. [CrossRef]
9. Lindequist, U. Marine‑Derived Pharmaceuticals—Challenges and Opportunities. Biomol. Ther. 2016,24, 561–571. [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‑Inammatory and Immunomodulatory Properties of a Crude Polysaccharide Derived from Green Seaweed Halimeda
tuna: Computational and Experimental Evidences. Mar. Drugs 2024,22, 85. [CrossRef] [PubMed]
11. Haque, N.; Parveen, S.; Tang, T.; Wei, J.; Huang, Z. Marine Natural Products in Clinical Use. Mar. Drugs 2022,20, 528. [CrossRef]
[PubMed]
12. Mayer, A.M.; Pierce, M.; Reji, M.; Wu, A.C.; Jekielek, K.K.; Le, H.Q.; Howe, K.; Bu, M.; Seo, S.; Newman, D.J.; et al. Marine‑
Derived Pharmaceuticals in Clinical Trials in 2022. J. Pharmacol. Exp. Ther. 2023,385, 299.
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.
[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 inammatory diseases:
Findings from the global burden of disease study 2019. eClinicalMedicine 2023,64, 102193. [CrossRef] [PubMed]
15. Miller, F.W. The increasing prevalence of autoimmunity and autoimmune diseases: An urgent call to action for improved un‑
derstanding, diagnosis, treatment, and prevention. Curr. Opin. Immunol. 2023,80, 102266. [CrossRef] [PubMed]
16. Michels, N.; Aart, C.v.; Morisse, J.; Huybrechts, I. Chronic Inammation Toward Cancer Incidence: An Epidemiological System‑
atic Review. J. Glob. Oncol. 2018,4, 24s. [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. [CrossRef] [PubMed]
18. Wraith, D.C. The Future of Immunotherapy: A 20‑Year Perspective. Front. Immunol. 2017,8, 1668. [CrossRef] [PubMed]
Mar. Drugs 2024,22, 304 19 of 32
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. Le. 2024,59, 124–135.
[CrossRef]
20. Karthikeyan, A.; Joseph, A.; Nair, B.G. Promising bioactive compounds from the marine environment and their potential eects
on various diseases. J. Genet. Eng. Biotechnol. 2022,20, 14. [CrossRef] [PubMed]
21. Malve, H. Exploring the ocean for new drug developments: Marine pharmacology. J. Pharm. Bioallied Sci. 2016,8,
83–91. [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.
23. Li, C.‑Q.; Ma, Q.‑Y.; Gao, X.‑Z.; Wang, X.; Zhang, B.‑L. Research Progress in Anti‑Inammatory Bioactive Substances Derived
from Marine Microorganisms, Sponges, Algae, and Corals. Mar. Drugs 2021,19, 572. [CrossRef] [PubMed]
24. Mayer, A.M.S.; Pierce, M.L.; Howe, K.; Rodríguez, A.D.; Taglialatela‑Scafati, O.; Nakamura, F.; Fusetani, N. Marine pharmacol‑
ogy in 2018: Marine compounds with antibacterial, antidiabetic, antifungal, anti‑inammatory, antiprotozoal, antituberculosis
and antiviral activities; aecting the immune and nervous systems, and other miscellaneous mechanisms of action. Pharmacol.
Res. 2022,183, 106391. [CrossRef]
25. Ahmad, B.; Shah, M.; Choi, S. Oceans as a Source of Immunotherapy. Mar. Drugs 2019,17, 282. [CrossRef] [PubMed]
26. Montuori, E.; de Pascale, D.; Lauritano, C. Recent Discoveries on Marine Organism Immunomodulatory Activities. Mar. Drugs
2022,20, 422. [CrossRef] [PubMed]
27. Khursheed, M.; Ghelani, H.; Jan, R.K.; Adrian, T.E. Anti‑Inammatory Eects of Bioactive Compounds from Seaweeds, Bry‑
ozoans, Jellysh, Shellsh and Peanut Worms. Mar. Drugs 2023,21, 524. [CrossRef] [PubMed]
28. Bekiari, M. Marine Bioprospecting: Understanding the Activity and Some Challenges Related to Environmental Protection,
Scientic 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, Swierland, 2023; pp. 237–252.
29. Reverter, M.; Rohde, S.; Parchemin, C.; Tapissier‑Bontemps, N.; Schupp, P.J. Metabolomics and Marine Biotechnology: Coupling
Metabolite Proling and Organism Biology for the Discovery of New Compounds. Front. Mar. Sci. 2020,7, 613471. [CrossRef]
30. Bayona, L.M.; de Voogd, N.J.; Choi, Y.H. Metabolomics on the study of marine organisms. Metabolomics 2022,18, 17. [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. [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. [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. [CrossRef]
34. World Health Organization. WHO Global Report on Traditional and Complementary Medicine 2019; World Health Organization:
Geneva, Swierland, 2019.
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. [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. [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. [CrossRef]
38. Moseley, M.E. The Maritime Foundations of Andean Civilization; Springer: Berlin/Heidelberg, Germany, 1975.
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. [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. [CrossRef]
41. Ruiz Vigo, W.M. Importancia de las Cyanophytas. Rev. Perspect. 2019,20, 240–244. [CrossRef]
42. Godínez‑Ortega, J.L.; Ortega, M.M.; Garduño‑Solórzano, G.; Oliva, M.G.; Vilaclara, G. Traditional knowledge of Mexican conti‑
nental algae. J. Ethnobiol. 2001,21, 57–88.
43. Sowunmi, A.A. Fin‑shes in Yorùbánatural healing practices from southwest Nigeria. J. Ethnopharmacol. 2007,113,
72–78. [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.
Mar. Drugs 2024,22, 304 20 of 32
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 Albu‑
querque, U.P.; Moura, G.J.B.d. Ichthyofauna used in traditional medicine in Brazil. Evid.‑Based Complement. Altern. Med. 2012,
2012, 474716. [CrossRef] [PubMed]
46. Alves, R.R.; Rosa, I.L. From cnidarians to mammals: The use of animals as remedies in shing communities in NE Brazil.
J. Ethnopharmacol. 2006,107, 259–276. [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. [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.
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. [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. [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. [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 tracking networks for marine wildlife. Sci. Adv. 2019,5, eaav5948. [CrossRef] [PubMed]
53. Siddiqui, S.A.; Baruah, S.; Wu, Y.S.; Yuansah, S.C.; Castro‑Muñoz, R.; Szymkowiak, A.; Kulawik, P. Investigating the sustainabil‑
ity, utilisation, consumption and conservation of sea mammals—A systematic review. Sustain. Prod. Consum. 2024,46, 400–417.
[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. [CrossRef]
55. Huxtable, R.J. The pharmacology of extinction. J. Ethnopharmacol. 1992,37, 1–11. [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. [CrossRef]
57. Nurse, A. Preventing marine wildlife crime: An evaluation of legal protection and enforcement perspectives. Front. Conserv. Sci.
2023,3, 1102823. [CrossRef]
58. Müller, W.E.; Batel, R.; Schröder, H.C.; Müller, I.M. Traditional and Modern Biomedical Prospecting: Part I‑the History: Sus‑
tainable Exploitation of Biodiversity (Sponges and Invertebrates) in the Adriatic Sea in Rovinj (Croatia). Evid. Based Complement.
Altern. Med. 2004,1, 71–82. [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. [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.
61. Papadopoulos, D. Medical Knowledge and the Aquatic Animals in Claudius Aelianus’s on the Characteristics of Animals. Avail‑
able 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 reme‑
dies, medicine, surgery, and pharmacopoeia. J. Appl. Phycol. 2023,35, 2049–2068. [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. [CrossRef]
64. Cavaillon, J.M. Once upon a time, inammation. J. Venom. Anim. Toxins Incl. Trop. Dis. 2021,27, e20200147. [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. [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. [CrossRef]
67. Peters, M.A. The ancient Silk Road and the birth of merchant capitalism. Educ. Philos. Theory 2021,53, 955–961. [CrossRef]
68. Bradley, S. The Silk Road and Sources of Chinese Medicine Expansion: Part 2—Formularies. Chin. Med. Cult. 2018,1, 68–70.
[CrossRef]
69. Bradley, S. The Silk Road and Sources of Chinese Medicine Expansion: Part 1—Materia Medica. Chin. Med. Cult. 2018,1, 29–31.
[CrossRef]
70. Díaz, J.L. Ethnopharmacology of sacred psychoactive plants used by the Indians of Mexico. Annu. Rev. Pharmacol. Toxicol. 1977,
17, 647–675. [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, Swierland, 2020; pp. 1–11.
72. World Health Organization. The International Pharmacopoeia; World Health Organization: Geneva, Swierland, 2022;
Online Version.
73. Silverstein, A.M. Autoimmunity versus horror autotoxicus: The struggle for recognition. Nat. Immunol. 2001,2,
279–281. [CrossRef] [PubMed]
Mar. Drugs 2024,22, 304 21 of 32
74. Mackay, I.R. Travels and travails of autoimmunity: A historical journey from discovery to rediscovery. Autoimmun. Rev. 2010,
9, A251–A258. [CrossRef] [PubMed]
75. Kaufmann, S.H.E. Immunology’s Coming of Age. Front. Immunol. 2019,10, 684. [CrossRef] [PubMed]
76. Plytycz, B.; Seljelid, R. From inammation to sickness: Historical perspective. Arch. Immunol. Ther. Exp. 2003,51, 105–109.
77. Tilden, J.E. The marine and fresh water algae of China. Lingnan Sci. J. 1929,7, 349–398.
78. Gao, K. Chinese studies on the edible blue‑green alga, Nostoc agelliforme: A review. J. Appl. Phycol. 1998,10, 37–49. [CrossRef]
79. Chase, F.M. Useful Algae; Government Printing Oce: Washington, DC, USA, 1942.
80. Tseng, C.K. The Past, Present and Future of Phycology in China; Springer: Dordrecht, The Netherlands, 2004; pp. 11–20.
81. Zhao, Z.; Guo, P.; Brand, E. A concise classication of bencao (materia medica). Chin. Med. 2018,13, 18. [CrossRef]
82. Pan, M.H.; Chiou, Y.S.; Tsai, M.L.; Ho, C.T. Anti‑inammatory activity of traditional Chinese medicinal herbs. J. Tradit. Comple‑
ment. Med. 2011,1, 8–24. [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. [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. [CrossRef]
85. Chen, X.; Ni, L.; Fu, X.; Wang, L.; Duan, D.; Huang, L.; Xu, J.; Gao, X. Molecular Mechanism of Anti‑Inammatory Activities of
a Novel Sulfated Galactofucan from Saccharina japonica. Mar. Drugs 2021,19, 430. [CrossRef] [PubMed]
86. Pan, L.; Fu, T.; Cheng, H.; Mi, J.; Shang, Q.; Yu, G. Polysaccharide from edible alga Gloiopeltis furcata aenuates intestinal
mucosal damage by therapeutically remodeling the interactions between gut microbiota and mucin O‑glycans. Carbohydr. Polym.
2022,278, 118921. [CrossRef] [PubMed]
87. Peng, H.S.; Huang, L.Q. Foreign medicinal materials in Bencao Tujing. Zhonghua Yi Shi Za Zhi 2021,51, 15–23. [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 (Illus‑
trated Classic of Materia Medica): A Preliminary Study. Chin. Med. Cult. 2022,5, 84–90. [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. [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; Schoenhammer, A., Ed.; Springer International Publishing: Cham,
Swierland, 2019; pp. 261–293.
91. Buell, P.D.; Anderson, E.N. Arabic Medicine in China: Tradition, Innovation, and Change; Brill: Leiden, The Netherlands, 2021.
[CrossRef]
92. Shi, J.; Yang, Y.; Zhou, X.; Zhao, L.; Li, X.; Yusuf, A.; Hosseini, M.; Sedkon, F.; Hu, X. The current status of old traditional
medicine introduced from Persia to China. Front. Pharmacol. 2022,13, 953352. [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. [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. [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.
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. [CrossRef]
97. Ryu, B.; Qian, Z.J.; Kim, S.K. Purication of a peptide from seahorse, that inhibits TPA‑induced MMP, iNOS and COX‑2 expres‑
sion through MAPK and NF‑kappaB activation, and induces human osteoblastic and chondrocytic dierentiation. Chem. Biol.
Interact. 2010,184, 413–422. [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. [CrossRef]
99. Malemud, C.J. The role of the JAK/STAT signal pathway in rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis. 2018,10, 117–127.
[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. [CrossRef]
101. Li, Y.; Qian, Z.J.; Kim, S.K. Cathepsin B inhibitory activities of three new phthalate derivatives isolated from seahorse, Hippocam‑
pus Kuda Bleeler. Bioorg Med. Chem. Le. 2008,18, 6130–6134. [CrossRef] [PubMed]
102. Sche, G.; Zwerina, J.; Firestein, G. The p38 mitogen‑activated protein kinase (MAPK) pathway in rheumatoid arthritis. Ann.
Rheum. Dis. 2008,67, 909–916. [CrossRef] [PubMed]
103. Law, S. Dried seahorse in traditional medicine: A narrative review. Infect. Dis. Herb. Med. 2021,2, 158. [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.
Mar. Drugs 2024,22, 304 22 of 32
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.
106. IUCN. The IUCN Red List of Threatened Species; IUCN: Gland, Swierland, 2023.
107. Walravens, H. Father Verbiest’s Chinese World Map (1674). Imago Mundi 1991,43, 31–47. [CrossRef]
108. Wang, Z.; Wu, Q. Restructuring Trade: Circulation of Medicinal Materials in East Asia in the 18th Century. Uisahak 2023,32,
279–319. [CrossRef]
109. Elshakry, M. When Science Became Western: Historiographical Reections. Isis 2010,101, 98–109. [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. [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 ora: Recent research advances and perspective. Acta Pharmacol. Sin. 2022,43, 3062–3079. [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. [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. [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. [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, Dierences, and Trends Directions. Evid.‑Based Complement. Altern. Med. 2022,
2022, 3971967. [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. [CrossRef]
117. Wang, Y. The scientic nature of traditional Chinese medicine in the post‑modern era. J. Tradit. Chin. Med. Sci. 2019,6, 195–200.
[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. [CrossRef]
119. Jaeger, S. A Geomedical Approach to Chinese Medicine: The Origin of the Yin‑Yang Symbol; IntechOpen: London, UK, 2012.
120. Verbeke, G. L’evolution de la Doctrine du Pneuma: Du Stoicisme a S. Augustin; Desclee de Brouwer: Bilbao, Spain, 1945.
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. [CrossRef]
122. Boaccioli, F.; Boaccioli, A.G. The suggestions of ancient Chinese philosophy and medicine for contemporary scientic research,
and integrative care. Brain Behav. Immun. Integr. 2024,5, 100024. [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. [CrossRef]
[PubMed]
124. Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018,154, 3–20. [CrossRef] [PubMed]
125. Lee, G.R. The Balance of Th17 versus Treg Cells in Autoimmunity. Int. J. Mol. Sci. 2018,19, 730. [CrossRef] [PubMed]
126. Hirano, T. IL‑6 in inammation, autoimmunity and cancer. Int. Immunol. 2021,33, 127–148. [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. [CrossRef] [PubMed]
128. Yang, C.; Li, D.; Ko, C.‑N.; Wang, K.; Wang, H. Active ingredients of traditional Chinese medicine for enhancing the eect of
tumor immunotherapy. Front. Immunol. 2023,14, 1133050. [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. [CrossRef] [PubMed]
130. Leeuwenhoek, A.V. Observations, communicated to the publisher by Mr. Antony van Leewenhoeck, in a dutch leer of the
9th Octob. 1676. here English’d: Concerning lile 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. [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.
133. ZoBell, C.E. Marine Microbiology: A Monograph on Hydrobacteriology; Chronica Botanica Company: Waltham, MA, USA, 1946.
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. [CrossRef]
135. Chisholm, S.W.; Olson, R.J.; Zeler, 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. [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
Mar. Drugs 2024,22, 304 23 of 32
MArine RESearcher in Oldenburg, Germany; Jungblut, S., Liebich, V., Bode‑Dalby, M., Eds.; Springer International Publishing:
Cham, Swierland, 2020; pp. 159–180.
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. [CrossRef] [PubMed]
138. Nicholls, H. Sorcerer II: The search for microbial diversity roils the waters. PLoS Biol. 2007,5, e74. [CrossRef] [PubMed]
139. Rusch, D.B.; Halpern, A.L.; Suon, G.; Heidelberg, K.B.; Williamson, S.; Yooseph, S.; Wu, D.; Eisen, J.A.; Homan, J.M.; Reming‑
ton, K.; et al. The Sorcerer II Global Ocean Sampling expedition: Northwest Atlantic through eastern tropical Pacic. PLoS Biol.
2007,5, e77. [CrossRef] [PubMed]
140. Duarte, C.M. Seafaring in the 21St Century: The Malaspina 2010 Circumnavigation Expedition. Limnol. Oceanogr. Bull. 2015,24,
11–14. [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. [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 com‑
munities. Commun. Biol. 2021,4, 604. [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. [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. [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.
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. [CrossRef] [PubMed]
147. Rodrigues, C.J.C.; de Carvalho, C.C.C.R. Cultivating marine bacteria under laboratory conditions: Overcoming the “uncultur‑
able” dogma. Front. Bioeng. Biotechnol. 2022,10, 964589. [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. [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. [CrossRef]
150. Liu, L.; Zheng, Y.‑Y.; Shao, C.‑L.; Wang, C.‑Y. Metabolites from marine invertebrates and their symbiotic microorganisms: Molec‑
ular diversity discovery, mining, and application. Mar. Life Sci. Technol. 2019,1, 60–94. [CrossRef]
151. Wilson, M.C.; Mori, T.; Rückert, C.; Uria, A.R.; Helf, M.J.; Takada, K.; Gernert, C.; Steens, U.A.E.; Heycke, N.; Schmi, S.; et al.
An environmental bacterial taxon with a large and distinct metabolic repertoire. Nature 2014,506, 58–62. [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. [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. [CrossRef] [PubMed]
154. Rath, C.M.; Janto, B.; Earl, J.; Ahmed, A.; Hu, F.Z.; Hiller, L.; Dahlgren, M.; Kreft, R.; Yu, F.; Wol, 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. [CrossRef] [PubMed]
155. Knobloch, S.; Jóhannsson, R.; Marteinsson, V. Co‑cultivation of the marine sponge Halichondria panicea and its associated mi‑
croorganisms. Sci. Rep. 2019,9, 10403. [CrossRef]
156. Kamat, S.; Kumar, S.; Philip, S.; Kumari, M. Chapter 10—Secondary metabolites from marine fungi: Current status and applica‑
tion. In Microbial Biomolecules; Kumar, A., Bilal, M., Ferreira, L.F.R., Kumari, M., Eds.; Academic Press: Cambridge, MA, USA,
2023; pp. 181–209. [CrossRef]
157. Xu, J.; Yi, M.; Ding, L.; He, S. A Review of Anti‑Inammatory Compounds from Marine Fungi, 2000‑2018. Mar. Drugs 2019,
17, 636. [CrossRef] [PubMed]
158. Nicolei, R.; Bellavita, R.; Falanga, A. The Outstanding Chemodiversity of Marine‑Derived Talaromyces. Biomolecules 2023,
13, 1021. [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. [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.; Lei, 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.
[CrossRef]
Mar. Drugs 2024,22, 304 24 of 32
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. [CrossRef]
162. Dawczynski, C.; Dirich, M.; Neumann, T.; Goee, 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. sunower oil. Clin. Nutr. 2018,37, 494–504. [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. [CrossRef] [PubMed]
164. Levasseur, W.; Perré, P.; Pozzobon, V. A review of high value‑added molecules production by microalgae in light of the classi‑
cation. Biotechnol. Adv. 2020,41, 107545. [CrossRef]
165. Riccio, G.; Lauritano, C. Microalgae with Immunomodulatory Activities. Mar. Drugs 2019,18, 2. [CrossRef] [PubMed]
166. Ghallab, D.S.; Ibrahim, R.S.; Mohyeldin, M.M.; Shawky, E. Marine algae: A treasure trove of bioactive anti‑inammatory com‑
pounds. Mar. Pollut. Bull. 2024,199, 116023. [CrossRef] [PubMed]
167. Dullius, A.; Buon, G.; Junior, M.F.; Giuliai, S. Articial Intelligence in Phycochemicals Recognition. In Value‑Added Products
from Algae: Phycochemical Production and Applications; Abomohra, A., Ende, S., Eds.; Springer International Publishing: Cham,
Swierland, 2024; pp. 97–122.
168. Galasso, C.; Corinaldesi, C.; Sansone, C. Carotenoids from Marine Organisms: Biological Functions and Industrial Applications.
Antioxidants 2017,6, 96. [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. [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. [CrossRef] [PubMed]
171. Razzak, S.A. Comprehensive overview of microalgae‑derived carotenoids and their applications in diverse industries. Algal Res.
2024,78, 103422. [CrossRef]
172. Chekanov, K.; Lukyanov, A.; Boussiba, S.; Aalo, 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.
[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. [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. [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. [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. Biofu‑
els Bioprod. 2023,16, 166. [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. [CrossRef]
178. Riccio, G.; De Luca, D.; Lauritano, C. Monogalactosyldiacylglycerol and Sulfolipid Synthesis in Microalgae. Mar. Drugs 2020,
18, 237. [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: Struc‑
tural 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. [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 Inammatory Response: A Critical Review. Int. J. Mol. Sci. 2021,22, 9825.
[CrossRef] [PubMed]
181. Banskota, A.H.; Gallant, P.; Stefanova, R.; Melanson, R.; O’Leary, S.J. Monogalactosyldiacylglycerols, potent nitric oxide in‑
hibitors from the marine microalga Tetraselmis chui. Nat. Prod. Res. 2013,27, 1084–1090. [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 fay acids for human consumption. Biotechnol. Adv. 2024,74, 108381. [CrossRef]
183. Thurn, A.‑L.; Schobel, J.; Weuster‑Bo, D. Photoautotrophic Production of Docosahexaenoic Acid‑ and Eicosapentaenoic Acid‑
Enriched Biomass by Co‑Culturing Golden‑Brown and Green Microalgae. Fermentation 2024,10, 220. [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. [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. Paerns and drivers of diatom diversity and abundance in the global ocean. bioRxiv 2024. [CrossRef]
186. Peng, K.; Amenorfenyo, D.K.; Rui, X.; Huang, X.; Li, C.; Li, F. Eect of Iron Concentration on the Co‑Production of Fucoxanthin
and Fay Acids in Conticribra weissogii. Mar. Drugs 2024,22, 106. [CrossRef] [PubMed]
Mar. Drugs 2024,22, 304 25 of 32
187. Mogal, A.; Kukreja, S.; Gautam, S. Sustainable Production of Diatom‑Based Omega‑3 Fay 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, Swierland, 2024; pp. 131–137.
188. Di Dato, V.; Orece, 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. [CrossRef] [PubMed]
189. Di Costanzo, F.; Di Dato, V.; Ianora, A.; Romano, G. Prostaglandins in Marine Organisms: A Review. Mar. Drugs 2019,17, 428.
[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. [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. [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. [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. [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. [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. [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. [CrossRef]
197. Badary, A.; Hidasi, N.; Ferrari, S.; Mayeld, S.P. Isolation and characterization of microalgae strains able to grow on complex
biomass hydrolysate for industrial application. Algal Res. 2024,78, 103381. [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. [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. [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.
201. Tan, L.T.; Phyo, M.Y. Marine Cyanobacteria: A Source of Lead Compounds and their Clinically‑Relevant Molecular Targets.
Molecules 2020,25, 2197. [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‑inammatory properties of ten cyanobacteria strains. Algal Res. 2023,
74, 103182. [CrossRef]
203. Cruz, J.D.; Delare, 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. [CrossRef] [PubMed]
204. Jha, S.; Singh, V.K.; Singh, A.P.; Gupta, A.; Rana, P.; Sinha, R.P. The Radiant World of Cyanobacterial Phycobiliproteins: Exam‑
ining Their Structure, Functions, and Biomedical Potentials. Targets 2024,2, 32–51. [CrossRef]
205. Ismail, M.M.; El‑Fakharany, E.M.; Hegazy, G.E. Purication and fractionation of phycobiliproteins from Arthrospira platensis
and Corallina ocinalis with evaluating their biological activities. Sci. Rep. 2023,13, 14270. [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. [CrossRef]
207. Chen, H.; Qi, H.; Xiong, P. Phycobiliproteins—A Family of Algae‑Derived Biliproteins: Productions, Characterization and Phar‑
maceutical Potentials. Mar. Drugs 2022,20, 450. [CrossRef] [PubMed]
208. Khazi, M.I.; Li, C.; Liaqat, F.; Malec, P.; Li, J.; Fu, P. Acclimation and Characterization of Marine Cyanobacterial Strains Euryha‑
linema and Desertilum for C‑Phycocyanin Production. Front. Bioeng. Biotechnol. 2021,9, 752024. [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.
210. Carpenter, W.B.; Jereys, J.G.; Thomson, C.T.W. Preliminary report of the scientic exploration of the deep sea in H. M.
surveying‑vessel ‘Porcupine’, during the summerof 1869. Proc. R. Soc. Lond. 1870,18, 397–492. [CrossRef]
Mar. Drugs 2024,22, 304 26 of 32
211. Thomson, C.W.S.; Carpenter, W.B.; Jereys, 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 Scientic Direction of Dr. Carpenter, F.R.S., J.
Gwyn Jereys, F.R.S., and Dr. Wyville Thomson, F.R.S.; Macmillan and Co: London, UK, 1873.
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.
213. Certes, A.‑A. Sur la culture, àl’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.
214. Certes, A.‑A. Note relative àl’action des hautes pressions sur la vitalitédes microorganismes d’eau douce et d’eau de mer. Comptes
Rendue Hebd. Des Séances De L’académie Des Sci. 1884,36, 385–388.
215. Dohrn, A. The Foundation of Zoological Stations. Nature 1872,5, 277–280. [CrossRef]
216. Groeben, C. The Stazione Zoologica Anton Dohrn as a place for the circulation of scientic ideas: Vision and management. In
Proceedings of the 31st Annual Conference of IAMSLIC, Rome, Italy, 10–14 October 2005.
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.
218. Zobell, C.E. Bacterial Life at the Boom of the Philippine Trench. Science 1952,115, 507–508. [CrossRef] [PubMed]
219. Yayanos, A.A.; Die, A.S.; Van Boxtel, R. Obligately barophilic bacterium from the Mariana trench. Proc. Natl. Acad. Sci. USA
1981,78, 5212–5215. [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. [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. [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. [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. [CrossRef] [PubMed]
224. Saide, A.; Lauritano, C.; Ianora, A. A Treasure of Bioactive Compounds from the Deep Sea. Biomedicines 2021,9, 1556. [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. [CrossRef] [PubMed]
226. Wang, Y.N.; Meng, L.H.; Wang, B.G. Progress in Research on Bioactive Secondary Metabolites from Deep‑Sea Derived Microor‑
ganisms. Mar. Drugs 2020,18, 614. [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. [CrossRef]
[PubMed]
228. Zhivkoplias, E.; Pranindita, A.; Dunshirn, P.; Jouray, J.‑B.; Blasiak, R. Novel Database Reveals Growing Prominence Of Deep‑Sea
Life for Marine Bioprospecting; Research Square: Durham, NC, USA, 2023.
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. [CrossRef]
230. Peit, 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. [CrossRef] [PubMed]
231. Zhang, Y.; Li, X.; Bartle, 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. [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.
[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 rietoensis AP‑5 from deep‑sea sediments of the Northwest Pacic. Int. J. Biol.
Macromol. 2023,245, 125452. [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.
[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. [CrossRef]
236. Kamjam, M.; Sivalingam, P.; Deng, Z.; Hong, K. Deep Sea Actinomycetes and Their Secondary Metabolites. Front. Microbiol.
2017,8, 760. [CrossRef]
Mar. Drugs 2024,22, 304 27 of 32
237. Wang, L.; Li, M.; Lin, Y.; Du, S.; Liu, Z.; Ju, J.; Suzuki, H.; Sawada, M.; Umezawa, K. Inhibition of cellular inammatory mediator
production and amelioration of learning decit in ies by deep sea Aspergillus‑derived cyclopenin. J. Antibiot. 2020,73, 622–629.
[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. Hep‑
ialiamides A–C: Aminated Fusaric Acid Derivatives and Related Metabolites with Anti‑Inammatory Activity from the Deep‑
Sea‑Derived Fungus Samsoniella hepiali W7. Mar. Drugs 2023,21, 596. [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‑inammatory
and antioxidant components from deep marine‑derived fungus Myrothecium sp. Bzo‑l062. Mar. Drugs 2020,18, 597. [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. [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. [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‑
neuroinammatory eect 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. [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.
Le. 2016,18, 3650–3653. [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. [CrossRef] [PubMed]
245. Kazłowska, K.; Hsu, T.; Hou, C.C.; Yang, W.C.; Tsai, G.J. Anti‑inammatory properties of phenolic compounds and crude extract
from Porphyra dentata. J. Ethnopharmacol. 2010,128, 123–130. [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. [CrossRef]
247. Souza, C.R.M.; Bezerra, W.P.; Souto, J.T. Marine Alkaloids with Anti‑Inammatory Activity: Current Knowledge and Future
Perspectives. Mar. Drugs 2020,18, 147. [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. [CrossRef]
249. Pradhan, B.; Rout, L.; Ki, J.‑S. Immunomodulatory and anti‑inammatory and anticancer activities of porphyran, a sulfated
galactan. Carbohydr. Polym. 2023,301, 120326. [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 inammatory and hypernociceptive responses in antigen‑
induced arthritis in mice. PLoS ONE 2015,10, e0118356. [CrossRef] [PubMed]
251. Yoo, H.J.; You, D.‑J.; Lee, K.‑W. Characterization and Immunomodulatory Eects of High Molecular Weight Fucoidan Fraction
from the Sporophyll of Undaria pinnatida in Cyclophosphamide‑Induced Immunosuppressed Mice. Mar. Drugs 2019,17, 447.
[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. [CrossRef] [PubMed]
253. Vasarri, M.; Leri, M.; Barlea, E.; Ramazzoi, M.; Marzocchini, R.; Degl’Innocenti, D. Anti‑inammatory properties of the marine
plant Posidonia oceanica (L.) Delile. J. Ethnopharmacol. 2020,247, 112252. [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‑inammatory, wound healing, cytoprotective, and
anticoagulant activities. Int. J. Biol. Macromol. 2024,260, 129433. [CrossRef] [PubMed]
255. Sarithakumari, C.H.; Renju, G.L.; Kurup, G.M. Anti‑inammatory and antioxidant potential of alginic acid isolated from the
marine algae, Sargassum wightii on adjuvant‑induced arthritic rats. Inammopharmacology 2013,21, 261–268. [CrossRef]
256. Jung, H.A.; Jin, S.E.; Ahn, B.R.; Lee, C.M.; Choi, J.S. Anti‑inammatory 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.
[CrossRef] [PubMed]
257. Pereira, D.M.; Valentão, P.; Andrade, P.B. Chapter 7—Lessons from the Sea: Distribution, SAR, and Molecular Mechanisms of
Anti‑inammatory Drugs from Marine Organisms. In Studies in Natural Products Chemistry; Aa ur, R., Ed.; Elsevier: Amsterdam,
The Netherlands, 2013; Volume 40, pp. 205–228.
258. Senthilkumar, K.; Kim, S.‑K. Marine Invertebrate Natural Products for Anti‑Inammatory and Chronic Diseases. Evid.‑Based
Complement. Altern. Med. Ecam 2013,2013, 572859. [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,
Swierland, 2019; pp. 55–85.
260. Imbs, A.B.; Dembitsky, V.M. Coral Lipids. Mar. Drugs 2023,21, 539. [CrossRef] [PubMed]
Mar. Drugs 2024,22, 304 28 of 32
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. [CrossRef]
262. Lai, K.‑H.; Fan, Y.‑C.; Peng, B.‑R.; Wen, Z.‑H.; Chung, H.‑M. Capnellenes from Capnella imbricata: Deciphering Their Anti‑
Inammatory‑Associated Chemical Features. Pharmaceuticals 2023,16, 916. [CrossRef]
263. González, Y.; Torres‑Mendoza, D.; Jones, G.E.; Fernandez, P.L. Marine Diterpenoids as Potential Anti‑Inammatory Agents.
Mediat. Inamm. 2015,2015, 263543. [CrossRef]
264. Mustonen, A.‑M.; Nieminen, P. Fay Acids and Oxylipins in Osteoarthritis and Rheumatoid Arthritis—A Complex Field with
Signicant Potential for Future Treatments. Curr. Rheumatol. Rep. 2021,23, 41. [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. [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‑Inammatory
and Analgesic Eects of the Marine‑Derived Compound Excavatolide B Isolated from the Culture‑Type Formosan Gorgonian
Briareum excavatum. Mar. Drugs 2015,13, 2559–2579. [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. [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 inammatory response and bone destruction in adjuvant‑induced
arthritis. PLoS ONE 2013,8, e62926. [CrossRef] [PubMed]
269. Ayed, Y.; Sghaier, R.M.; Laouini, D.; Bacha, H. Evaluation of anti‑proliferative and anti‑inammatory activities of Pelagia noc‑
tiluca venom in Lipopolysaccharide/Interferon‑γstimulated RAW264.7 macrophages. Biomed. Pharmacother. 2016,84, 1986–1991.
[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 Articial Intelligence. Mar. Drugs 2024,22, 6.
[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 activ‑
ities. Sci. Rep. 2016,6, 35465. [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‑inammatory and/or COX‑2 inhibitory ascidian metabolites. Eur. J. Med. Chem. 2019,180, 86–98. [CrossRef]
273. Casertano, M.; Esposito, E.; Bello, I.; Indol, C.; Putra, M.Y.; Di Cesare Mannelli, L.; Ghelardini, C.; Menna, M.; Sorrentino, R.;
Cirino, G.; et al. Searching for Novel Sources of Hydrogen Sulde Donors: Chemical Proling of Polycarpa aurata Extract and
Evaluation of the Anti‑Inammatory Eects. Mar. Drugs 2023,21, 641. [CrossRef]
274. Sansone, C.; Balestra, C.; Pistelli, L.; Del Mondo, A.; Osca, D.; Brunet, C.; Crocea, F. A Comparative Analysis of Mucus
Immunomodulatory Properties from Seven Marine Gastropods from the Mediterranean Sea. Cells 2022,11, 2340. [CrossRef]
[PubMed]
275. Li, G.; Fu, Y.; Zheng, J.; Li, D. Anti‑inammatory activity and mechanism of a lipid extract from hard‑shelled mussel (Mytilus
coruscus) on chronic arthritis in rats. Mar. Drugs 2014,12, 568–588. [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. [CrossRef]
[PubMed]
277. Pereira, R.B.; Taveira, M.; Valentão, P.; Sousa, C.; Andrade, P.B. Fay acids from edible sea hares: Anti‑inammatory capacity
in LPS‑stimulated RAW 264.7 cells involves iNOS modulation. RSC Adv. 2015,5, 8981–8987. [CrossRef]
278. Ghelani, H.; Khursheed, M.; Adrian, T.E.; Jan, R.K. Anti‑Inammatory Eects of Compounds from Echinoderms. Mar. Drugs
2022,20, 693. [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 Inammation in Bleomycin‑Induced Scleroderma. Mar. Drugs 2021,19, 237.
[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‑inammatory activity in vivo. PharmaNutrition 2018,6, 74–80. [CrossRef]
281. Jo, S.‑H.; Kim, C.; Park, S.‑H. Novel Marine Organism‑Derived Extracellular Vesicles for Control of Anti‑Inammation. Tissue
Eng. Regen. Med. 2021,18, 71–79. [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. [CrossRef] [PubMed]
283. Youssef, D.T.; Ibrahim, A.K.; Khalifa, S.I.; Mesbah, M.K.; Mayer, A.M.; van Soest, R.W. New anti‑inammatory sterols from the
Red Sea sponges Scalarispongia aqabaensis and Callyspongia siphonella. Nat. Prod. Commun. 2010,5, 27–31. [PubMed]
Mar. Drugs 2024,22, 304 29 of 32
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‑inammatory Activities of Marine Sponges Aplysina Caissara, Haliclona sp. and Dragmacidon Reticulatum. Braz. Arch.
Biol. Technol. 2018,61, e18180104. [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 Eect on Intestinal Inammation. Mar. Drugs 2021,
19, 170. [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 Inammatory Response through Inhibition of
the NF‑κB Pathway. Mar. Drugs 2022,20, 605. [CrossRef] [PubMed]
287. Ciaglia, E.; Maltano, A.M.; Laezza, C.; Fontana, A.; Nuzzo, G.; Cutignano, A.; Abate, M.; Pelin, M.; Sosa, S.; Bifulco, M. Immuno‑
modulatory and anti‑inammatory eects of dihydrogracilin A, a terpene derived from the marine sponge Dendrilla membra‑
nosa. Int. J. Mol. Sci. 2017,18, 1643. [CrossRef] [PubMed]
288. Di, X.; Oskarsson, J.T.; Omarsdoir, S.; Freysdoir, J.; Hardardoir, I. Lipophilic fractions from the marine sponge Halichon‑
dria sitiens decrease secretion of pro‑inammatory 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. [CrossRef] [PubMed]
289. Palit, K.; Rath, S.; Chaerjee, 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. [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. [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, Swierland, 2020; pp. 1–29.
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. [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. [CrossRef]
294. Roome, T.; Dar, A.; Ali, S.; Naqvi, S.; Choudhary, M.I. A study on antioxidant, free radical scavenging, anti‑inammatory and
hepatoprotective actions of Aegiceras corniculatum (stem) extracts. J. Ethnopharmacol. 2008,118, 514–521. [CrossRef] [PubMed]
295. Kumari, C.S.; Yasmin, N.; Hussain, M.R.; Babuselvam, M. Invitro anti‑inammatory and anti‑artheritic property of Rhizopora
mucronata leaves. Int. J. Pharma Sci. Res. 2015,6, 482–485.
296. Islam, M.T. Chemical prole and biological activities of Sonneratia apetala (Buch.‑Ham.). Adv. Tradit. Med. 2020,20, 123–132.
[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 Lumniera racemosa with anti‑angiogenic and anti‑inammatory eects. Mar. Drugs 2018,16, 404. [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. [CrossRef] [PubMed]
299. Deepa, M.; Darsan, M.B.; Ramalingam, C. In Vitro Evaluation of the antioxidant, anti‑inammatory and antiproliferative activi‑
ties of the leaf extracts of Excoecaria agallocha L. Int. J. Pharm. Sci. 2015,7, 346–352.
300. Babuselvam, M.; Ravikumar, S.; Farook, K.M.; Abideen, S.; Mohamed, M.P.; Uthiraselvam, M. Evaluation of anti‑inammatory
and analgesic eects on the extracts of dierent parts of Excoecaria agallocha L. J. Appl. Pharm. Sci. 2012,2, 108–112. [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. [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.
[CrossRef] [PubMed]
303. Danioi, 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 Scientic Literature and Business Models. Mar.
Drugs 2021,19, 61. [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. [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. [CrossRef] [PubMed]
306. Malaweera, D.B.O.; Wijesundara, N.M. Trends in Development of Marine‑based Anti‑inammatory Pharmaceuticals. In Ency‑
clopedia of Marine Biotechnology; Wiley: Hoboken, NJ, USA, 2020; pp. 2471–2485. [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.
[CrossRef] [PubMed]
Mar. Drugs 2024,22, 304 30 of 32
308. Kapoor, S.; Nailwal, N.; Kumar, M.; Barve, K. Recent Patents and Discovery of Anti‑inammatory Agents from Marine Source.
Recent Pat. Inamm. Allergy Drug Discov. 2019,13, 105–114. [CrossRef] [PubMed]
309. Volkmann, E.R.; Andréasson, K.; Smith, V. Systemic sclerosis. Lancet 2023,401, 304–318. [CrossRef] [PubMed]
310. Denton, C.P.; Khanna, D. Systemic sclerosis. Lancet 2017,390, 1685–1699. [CrossRef] [PubMed]
311. Micallef, L.; Vedrenne, N.; Billet, F.; Coulomb, B.; Darby, I.A.; Desmoulière, A. The myobroblast, multiple origins for major
roles in normal and pathological tissue repair. Fibrogenesis Tissue Repair 2012,5, S5. [CrossRef] [PubMed]
312. Corallo, C.; Cutolo, M.; Volpi, N.; Franci, D.; Aglianò, M.; Montella, A.; Chirico, C.; Gonnelli, S.; Nuti, R.; Giordano, N.
Histopathological ndings in systemic sclerosis‑related myopathy: Fibrosis and microangiopathy with lack of cellular inam‑
mation. Ther. Adv. Musculoskelet. Dis. 2017,9, 3–10. [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. [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. [CrossRef] [PubMed]
315. Sedova, K.; Bernikova, O.; Azarov, J.; Shmakov, D.; Vityazev, V.; Kharin, S. Eects of echinochrome on ventricular repolarization
in acute ischemia. J. Electrocardiol. 2015,48, 181–186. [CrossRef]
316. Mohamed, A.S.; Sadek, S.A.; Hassanein, S.S.; Soliman, A.M. Hepatoprotective Eect of Echinochrome Pigment in Septic Rats.
J. Surg. Res. 2019,234, 317–324. [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.
318. Lebed’ko, O.A.; Ryzhavskii, B.Y.; Demidova, O.V. Eect of Antioxidant Echinochrome A on Bleomycin‑Induced Pulmonary
Fibrosis. Bull. Exp. Biol. Med. 2015,159, 351–354. [CrossRef] [PubMed]
319. Kikionis, S.; Papakyriakopoulou, P.; Mavrogiorgis, P.; Vasileva, E.A.; Mishchenko, N.P.; Fedoreyev, S.A.; Valsami, G.; Ioan‑
nou, 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. [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. [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.
322. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor‑inltrating
immune cells and their therapeutic implications. Cell Mol. Immunol. 2020,17, 807–821. [CrossRef] [PubMed]
323. Gordon, S. Elie Metchniko: Father of natural immunity. Eur. J. Immunol. 2008,38, 3257–3264. [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. [CrossRef]
325. Toledo, D.M.; Pioli, P.A. Macrophages in Systemic Sclerosis: Novel Insights and Therapeutic Implications. Curr. Rheumatol. Rep.
2019,21, 31. [CrossRef] [PubMed]
326. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011,11, 723–737.
[CrossRef] [PubMed]
327. Long, K.B.; Beay, G.L. Harnessing the antitumor potential of macrophages for cancer immunotherapy. Oncoimmunology 2013,
2, e26860. [CrossRef] [PubMed]
328. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012,12, 252–264. [CrossRef]
[PubMed]
329. Brzostek, J.; Gascoigne, N.R.; Rybakin, V. Cell Type‑Specic Regulation of Immunological Synapse Dynamics by B7 Ligand
Recognition. Front. Immunol. 2016,7, 24. [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. [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. [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.
[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. [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. [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. [CrossRef] [PubMed]
Mar. Drugs 2024,22, 304 31 of 32
336. Cutolo, M.; Campitiello, R.; Gotelli, E.; Soldano, S. The Role of M1/M2 Macrophage Polarization in Rheumatoid Arthritis Syn‑
ovitis. Front. Immunol. 2022,13, 867260. [CrossRef]
337. Goold, H.D.; Moseley, J.L.; Lauersen, K.J. The synthetic future of algal genomes. Cell Genom. 2024,4, 100505. [CrossRef]
338. Li, J.; Agathos, S.N.; Gao, Z. Editorial: Emerging trends in genetic engineering of microalgae. Front. Bioeng. Biotechnol. 2024,
12, 1403711. [CrossRef]
339. Torres‑Tiji, Y.; Sethuram, H.; Gupta, A.; McCauley, J.; Dutra‑Molino, J.V.; Pathania, R.; Saxton, L.; Kang, K.; Hillson, N.J.; May‑
eld, S.P. Bioinformatic prediction and high throughput in vivo screening to identify cis‑regulatory elements for the develop‑
ment of algal synthetic promoters. bioRxiv 2024. [CrossRef]
340. McQuillan, J.L.; Berndt, A.J.; Sproles, A.E.; Mayeld, 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.
[CrossRef] [PubMed]
341. Jacobebbinghaus, N.; Lauersen, K.J.; Kruse, O.; Baier, T. Bicistronic expression of nuclear transgenes in Chlamydomonas rein‑
hardtii. Plant J. 2024,118, 1400–1412. [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. [CrossRef]
343. Cutolo, E.A.; Mandalà, G.; Dall’Osto, L.; Bassi, R. Harnessing the Algal Chloroplast for Heterologous Protein Production. Mi‑
croorganisms 2022,10, 743. [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. [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. [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. [CrossRef] [PubMed]
347. Akram, M.; Khan, M.A.; Ahmed, N.; Bhai, R.; Pervaiz, R.; Malik, K.; Tahir, S.; Abbas, R.; Ashraf, F.; Ali, Q. Cloning and expres‑
sion of an anti‑cancerous cytokine: Human IL‑29 gene in Chlamydomonas reinhardtii. AMB Express 2023,13, 23. [CrossRef]
348. Torres‑Tiji, Y.; Fields, F.J.; Yang, Y.; Heredia, V.; Horn, S.J.; Keremane, S.R.; Jin, M.M.; Mayeld, 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. [CrossRef]
349. Stavridou, E.; Karapetsi, L.; Nteve, G.M.; Tsinou, G.; Chaikonstantinou, 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. [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. [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. [CrossRef]
352. Kneip, J.S.; Kniepkamp, N.; Jang, J.; Mortaro, M.G.; Jin, E.; Kruse, O.; Baier, T. CRISPR/Cas9‑Mediated Knockout of the Ly‑
copene ε‑Cyclase for Ecient Astaxanthin Production in the Green Microalga Chlamydomonas reinhardtii. Plants 2024,13,
1393. [CrossRef] [PubMed]
353. Marcolungo, L.; Bellamoli, F.; Cecchin, M.; Lopatriello, G.; Rossato, M.; Cosentino, E.; Rombauts, S.; Delledonne, M.; Balloari,
M. Haematococcus lacustris genome assembly and annotation reveal diploid genetic traits and stress‑induced gene expression
paerns. Algal Res. 2024,80, 103567. [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: Ecient production of immunologically active hepatitis C virus
glycoprotein. Proc. Natl. Acad. Sci. USA 2024,121, e2400145121. [CrossRef] [PubMed]
355. Hammel, A.; Neupert, J.; Bock, R. Optimized transgene expression in the red alga Porphyridium purpureum and ecient re‑
combinant protein secretion into the culture medium. Plant Mol. Biol. 2024,114, 18. [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. [CrossRef] [PubMed]
357. Mariam, I.; Beiga, M.; Rova, U.; Christakopoulos, P.; Matsakas, L.; Patel, A. Ameliorating microalgal OMEGA productionusing
omics platforms. Trends Plant Sci. 2024. [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. [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. [CrossRef] [PubMed]
360. Melis, A.; Hidalgo Martinez, D.A.; Beerle, N. Perspectives of cyanobacterial cell factories. Photosynth. Res. 2023. [CrossRef]
[PubMed]
Mar. Drugs 2024,22, 304 32 of 32
361. Bourgade, B.; Stensjö, K. Synthetic biology in marine cyanobacteria: Advances and challenges. Front. Microbiol. 2022,13, 994365.
[CrossRef] [PubMed]
362. Middelboe, M.; Brussaard, C.P.D. Marine Viruses: Key Players in Marine Ecosystems. Viruses 2017,9, 302. [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. [CrossRef] [PubMed]
364. Santiago‑Rodriguez, T.M.; Hollister, E.B. Unraveling the viral dark maer through viral metagenomics. Front. Immunol. 2022,
13, 1005107. [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.
[CrossRef]
366. D’Adamo, S.; Kormelink, R.; Martens, D.; Barbosa, M.J.; Wijels, R.H. Prospects for viruses infecting eukaryotic microalgae in
biotechnology. Biotechnol. Adv. 2021,54, 107790. [CrossRef] [PubMed]
367. Wilson, W.H.; Van Een, J.L.; Allen, M.J. The Phycodnaviridae: The story of how tiny giants rule the world. Curr. Top. Microbiol.
Immunol. 2009,328, 1–42. [CrossRef] [PubMed]
368. Shitrit, D.; Hackl, T.; Laurenceau, R.; Raho, N.; Carlson, M.C.G.; Sabehi, G.; Schwar, 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.
[CrossRef] [PubMed]
369. Novikova, O.; Topilina, N.; Belfort, M. Enigmatic distribution, evolution, and function of inteins. J. Biol. Chem. 2014,289,
14490–14497. [CrossRef]
370. Allen, M.J.; Lanzén, A.; Bratbak, G. Characterisation of the coccolithovirus intein. Mar. Genom. 2011,4, 1–7. [CrossRef] [PubMed]
371. Roer, 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. [CrossRef]
372. Evans, L.S.; Buchan, P.M.; Fortnam, M.; Honig, M.; Heaps, L. Puing coastal communities at the center of a sustainable blue
economy: A review of risks, opportunities, and strategies. Front. Political Sci. 2023,4, 1032204. [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. [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. [CrossRef]
375. Ferguson, D.K.; Li, C.; Chakraborty, A.; Giins, D.A.; Fowler, M.; Webb, J.; Campbell, C.; Morrison, N.; MacDonald, A.; Hubert,
C.R.J. Multi‑year seabed environmental baseline in deep‑sea oshore oil prospective areas established using microbial biodiver‑
sity. Mar. Pollut. Bull. 2023,194, 115308. [CrossRef] [PubMed]
376. Zhang, H.; Chen, S. Overview of research on marine resources and economic development. Mar. Econ. Manag. 2022,5, 69–83.
[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. [CrossRef] [PubMed]
378. Kumar, A.; AbdElgawad, H.; Castellano, I.; Selim, S.; Beemster, G.T.S.; Asard, H.; Buia, M.C.; Palumbo, A. Eects of ocean
acidication on the levels of primary and secondary metabolites in the brown macroalga Sargassum vulgare at dierent time
scales. Sci. Total Environ. 2018,643, 946–956. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual au‑
thor(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.
... The vastness and unique environmental conditions of the ocean have fostered the evolution of these fungi, endowing them with the capacity to produce an array of secondary metabolites. To date, over 35,000 marine natural products have been characterized, and 13 marine-derived drugs have been approved for clinical use [1][2][3][4][5][6]. Among them, the genus Talaromyces has shown great potential in agriculture, food, cosmetics, medicine, and environmental protection, emerging as a promising source for bioactive compound discovery [7][8][9]. ...
... PSU-MF07. Talaroterpenoids D-F (6)(7)(8) and fusariumin A (9) are orthoester groups containing 3,4-seco-meroterpenoid featuring highly oxygenated seven/six-membered rings, which bear three quaternary spirocarbons. Up to now, only 10 natural products containing an orthoester group have been reported from fungi. ...
... The refinement process was carried out using a full-matrix least-squares approach based on F2. Crystallographic data (Table S1, Supporting Information) have been deposited at the Cambridge Crystallographic Data Center under deposition numbers CCDC 2264255 (5) and 2385763 (6). These data can be obtained free of charge via the internet at www.ccdc.cam.ac.uk (accessed on 20 May 2023 for 5 and 23 September 2024 for 6). ...
Article
Full-text available
Six new highly oxidized seco-terpenoids, including three 3-nor-labdane type diterpenes, talaroterpenoids A–C (1–3), and three meroterpenoids containing an orthoester group, talaroterpenoids D–F (6–8), together with five known compounds (4–5 and 9–11), were isolated from the marine-derived fungus Talaromyces aurantiacus. Their chemical structures were elucidated through 1D, 2D NMR, HRESIMS, J-based configuration analysis (JBCA), computational ECD calculations, and single-crystal X-ray diffraction analysis. Compounds 1 and 2 contain an unusual 6,20-γ-lactone-bridged scaffold. Compounds 10 and 11 presented inhibitory effects on NO release in lipopolysaccharide (LPS)-induced BV-2 cells with IC50 values of 11.47 and 11.32 μM, respectively. Talaroterpenoid C (3) showed moderate antifungal activity against A. alternata and P. theae Steyaert.
... Seaweeds, plant-like multicellular algae that inhabit the tidewater either clung to stone or hanging in the open ocean, have been historically employed for food choices and therapeutic uses since antiquity [45]. Despite the availability of numerous treatment options for anxiety, depression, sleep disturbances, and diarrhea, finding a way to completely alleviate these conditions without side effects remains elusive. ...
Article
Full-text available
Najas gracillima, a marine seaweed found in North America and Asia, was investigated for its neuropharmacological and antidiarrheal properties. Acetone extracts of N. gracillima (ANG) were analyzed using both in vivo and in silico methods. GC‐MS analysis was conducted to identify bioactive compounds present in the extract. In vivo assessments, including the elevated plus maze, light‐dark box, and hole board tests, showed that ANG at doses of 200 and 400 mg/kg exhibited significant (P < 0.001) anxiolytic effects. Both doses also demonstrated antidepressant effects in the forced swimming and tail suspension tests by significantly (P < 0.001) reducing immobility time, with the 200 mg/kg dose showing more pronounced effects. Sedative activity was confirmed through open field and hole cross tests, where both doses exhibited significant (P < 0.001) sedative effects. ANG also demonstrated significant antidiarrheal effects at 400 mg/kg in castor oil‐induced diarrhea (P < 0.05) and gastrointestinal motility tests (P < 0.01). Molecular docking simulations revealed that compounds from ANG had strong binding affinities to critical drug targets involved in anxiety, depression, sleep disorders, and diarrhea. These findings suggest that N. gracillima holds potential for therapeutic use in treating neuropharmacological disorders and diarrhea, warranting further investigation.
... Compared to terrestrial environments, marine conditionssuch as high salinity, pressure, oligotrophy, darkness, and extreme temperatures-have driven marine organisms to develop unique metabolic pathways and defense mechanisms. These adaptations enable the production of biologically active substances with complex and diverse structures, offering valuable opportunities to discover novel marine-derived compounds with bioactive potential [3,4]. In recent years, numerous marine compounds with antitumor, anti-inflammatory, antioxidant, and cardiovascular protective properties have been identified. ...
Article
Full-text available
Marine natural products are increasingly utilized in nutrition, cosmetics, and medicine, garnering significant attention from researchers globally. With the expansion of marine resource exploration in recent years, the demand for marine natural products has risen, necessitating rapid and cost-effective activity evaluations using model organisms. Zebrafish, a valuable vertebrate model, has become an efficient tool for screening and identifying safe, active molecules from marine natural products. This review, based on nearly 10 years of literature, summarizes the current status and progress of zebrafish models in evaluating marine natural product bioactivity. It also highlights their potential in exploring marine resources with health benefits, offering a reference for the future development and utilization of marine biological resources.
Article
Full-text available
Marine phytoplankton is an emerging source of immunomodulatory bioactive lipids (BLs). Under physiological growth conditions and upon stress challenges, several eukaryotic microalgal species accumulate lipid metabolites that resemble the precursors of animal mediators of inflammation: eicosanoids and prostaglandins. Therefore, marine phytoplankton could serve as a biotechnological platform to produce functional BLs with therapeutic applications in the management of chronic inflammatory diseases and other clinical conditions. However, to be commercially competitive, the lipidic precursor yields should be enhanced. Beside tailoring the cultivation of native producers, genetic engineering is a feasible strategy to accrue the production of lipid metabolites and to introduce heterologous biosynthetic pathways in microalgal hosts. Here, we present the state-of-the-art clinical research on immunomodulatory lipids from eukaryotic marine phytoplankton and discuss synthetic biology approaches to boost their light-driven biosynthesis.
Article
Full-text available
Algae biotechnology holds immense promise for revolutionizing the bioeconomy through the sustainable and scalable production of various bioproducts. However, their development has been hindered by the lack of advanced genetic tools. This study introduces a synthetic biology approach to develop such tools, focusing on the construction and testing of synthetic promoters. By analyzing conserved DNA motifs within the promoter regions of highly expressed genes across six different algal species, we identified cis-regulatory elements (CREs) associated with high transcriptional activity. Combining the algorithms POWRS, STREME, and PhyloGibbs, we predicted 1511 CREs and inserted them into a minimal synthetic promoter sequence in 1, 2, or 3 copies, resulting in 4533 distinct synthetic promoters. These promoters were evaluated in vivo for their capacity to drive the expression of a transgene in a high-throughput manner through next-generation sequencing post antibiotic selection and fluorescence-activated cell sorting. To validate our approach, we sequenced hundreds of transgenic lines showing high levels of GFP expression. Further, we individually tested 14 identified promoters, revealing substantial increases in GFP expression—up to nine times higher than the baseline synthetic promoter, with five matching or even surpassing the performance of the native AR1 promoter. As a result of this study, we identified a catalog of CREs that can now be used to build superior synthetic algal promoters. More importantly, here we present a validated pipeline to generate building blocks for innovative synthetic genetic tools applicable to any algal species with a sequenced genome and transcriptome data set.
Article
Full-text available
The increasing applications for eicosapentaenoic acid (EPA) and the potential shortfall in supply due to sustainability and contamination issues related with its conventional sources (i.e., fish oils; seafood) led to an extensive search for alternative and sustainable sources, as well as production processes. The present mini-review covers all the steps involved in the production of EPA from microorganisms, with a deeper focus on microalgae. From production systems to downstream processing, the most important achievements within each area are briefly highlighted. Comparative tables of methodologies are also provided, as well as additional references of recent reviews, so that readers may deepen their knowledge in the different issues addressed. Key points • Microorganisms are more sustainable alternative sources of EPA than fish. • Due to the costly separation from DHA, species that produce only EPA are preferable. • EPA production can be optimised using non-genetic and genetic tailoring engineering.
Preprint
Full-text available
Diatoms constitute one of the most diverse and ecologically important phytoplankton groups, yet their large-scale diversity patterns and drivers of abundance are unclear due to limited observations. Here, we utilize Tara Oceans molecular and morphological data, spanning pole to pole, to describe marine diatom diversity, abundance, and environmental adaptation and acclimation strategies. The dominance of diatoms among phytoplankton in terms of relative abundance and diversity was confirmed, and the most prevalent genera were Chaetoceros, Thalassiosira, Actinocyclus and Pseudo-nitzschia. We defined 25 distinct diatom communities with varying environmental preferences illustrative of different life strategies. The Arctic Ocean stands out as a diatom hotspot with 6 of the diatom communities being exclusive to it. Light harvesting and photoprotection were among the cellular functions in which natural diatom populations invested the bulk of their transcriptional efforts. The biogeography of diatom lineages was consistent with their copy number for genes related to photoacclimation. This comprehensive study sheds light on marine diatom distributions, offering insights amid global change and oceanic anthropogenic impacts.
Article
Full-text available
Microalgae are promising production platforms for the cost-effective production of recombinant proteins. We have recently established that the red alga Porphyridium purpureum provides superior transgene expression properties, due to the episomal maintenance of transformation vectors as multicopy plasmids in the nucleus. Here, we have explored the potential of Porphyridium to synthesize complex pharmaceutical proteins to high levels. Testing expression constructs for a candidate subunit vaccine against the hepatitis C virus (HCV), we show that the soluble HCV E2 glycoprotein can be produced in transgenic algal cultures to high levels. The antigen undergoes faithful posttranslational modification by N-glycosylation and is recognized by conformationally selective antibodies, suggesting that it adopts a proper antigenic conformation in the endoplasmic reticulum of red algal cells. We also report the experimental determination of the structure of the N-glycan moiety that is attached to glycosylated proteins in Porphyridium . Finally, we demonstrate the immunogenicity of the HCV antigen produced in red algae when administered by injection as pure protein or by feeding of algal biomass.
Article
Full-text available
The green alga Haematococcus lacustris (formerly Haematococcus pluvialis) is a primary source of astaxanthin, a ketocarotenoid with high antioxidant activity and several industrial applications. Here, the Haematococcus lacustris highly repetitive genome was reconstructed by exploiting next-generation sequencing integrated with Hi-C scaffolding, obtaining a 151 Mb genome assembly in 32 scaffolds at a near-chromosome level with high continuity. Surprisingly, the distribution of the single-nucleotide-polymorphisms identified demonstrates a diploid configuration for the Haematococcus genome, further validated by Sanger sequencing of heterozygous regions. Functional annotation and RNA-seq data enabled the identification of 13,946 nuclear genes, with >5000 genes not previously identified in this species, providing insights into the molecular basis for metabolic rearrangement in stressing conditions such as high light and/or nitrogen starvation, where astaxanthin biosynthesis is triggered. These data constitute a rich genetic resource for biotechnological manipulation of Haematococcus lacustris highlighting potential targets to improve astaxanthin and carotenoid productivity.
Article
Full-text available
Carotenoids are valuable pigments naturally occurring in all photosynthetic plants and microalgae as well as in selected fungi, bacteria, and archaea. Green microalgae developed a complex carotenoid profile suitable for efficient light harvesting and light protection and harbor great capacity for carotenoid production through the substantial power of the endogenous 2-C-methyl-D-erythritol 4-phosphate (MEP) pathway. Previous works established successful genome editing and induced significant changes in the cellular carotenoid content in Chlamydomonas reinhardtii. This study employs a tailored carotenoid pathway for engineered bioproduction of the valuable ketocarotenoid astaxanthin. Functional knockout of lycopene ε-cyclase (LCYE) and non-homologous end joining (NHEJ)-based integration of donor DNA at the target site inhibit the accumulation of α-carotene and consequently lutein and loroxanthin, abundant carotenoids in C. reinhardtii without changes in cellular fitness. PCR-based screening indicated that 4 of 96 regenerated candidate lines carried (partial) integrations of donor DNA and increased ß-carotene as well as derived carotenoid contents. Iterative overexpression of CrBKT, PacrtB, and CrCHYB resulted in a 2.3-fold increase in astaxanthin accumulation in mutant ΔLCYE#3 (1.8 mg/L) compared to the parental strain UVM4, which demonstrates the potential of genome editing for the design of a green cell factory for astaxanthin bioproduction.
Preprint
Chloroplast synthetic biology holds promise for developing improved crops through improving the function of plastids. However, chloroplast engineering efforts face limitations due to the scarcity of genetic tools and the low throughput of plant-based systems. To address these challenges, we here established Chlamydomonas reinhardtii as a prototyping chassis for chloroplast synthetic biology. We developed an automation workflow that enables the generation, handling, and analysis of thousands of transplastomic strains in parallel, expanded the repertoire of selection markers for chloroplast transformation, established new reporter genes, and characterized over 140 regulatory parts, including native and synthetic promoters, UTRs, and intercistronic expression elements. We integrated the system within the Phytobrick cloning standard and demonstrate several applications, including a library-based approach to develop synthetic promoter designs in plastids. Finally, we provide a proof-of-concept for prototyping novel traits in plastids by introducing a chloroplast-based synthetic photorespiration pathway and demonstrating a twofold increase in biomass production. Overall, our study advances chloroplast engineering, and provides a promising platform to rapidly prototype chloroplast manipulations before their transfer into higher plants and crops.