ArticlePDF AvailableLiterature Review

The Clinical Promise of Microalgae in Rheumatoid Arthritis: From Natural Compounds to Recombinant Therapeutics

MDPI
Marine Drugs
Authors:

Abstract and Figures

Rheumatoid arthritis (RA) is an invalidating chronic autoimmune disorder characterized by joint inflammation and progressive bone damage. Dietary intervention is an important component in the treatment of RA to mitigate oxidative stress, a major pathogenic driver of the disease. Alongside traditional sources of antioxidants, microalgae—a diverse group of photosynthetic prokaryotes and eukaryotes—are emerging as anti-inflammatory and immunomodulatory food supplements. Several species accumulate therapeutic metabolites—mainly lipids and pigments—which interfere in the pro-inflammatory pathways involved in RA and other chronic inflammatory conditions. The advancement of the clinical uses of microalgae requires the continuous exploration of phytoplankton biodiversity and chemodiversity, followed by the domestication of wild strains into reliable producers of said metabolites. In addition, the tractability of microalgal genomes offers unprecedented possibilities to establish photosynthetic microbes as light-driven biofactories of heterologous immunotherapeutics. Here, we review the evidence-based anti-inflammatory mechanisms of microalgal metabolites and provide a detailed coverage of the genetic engineering strategies to enhance the yields of endogenous compounds and to develop innovative bioproducts.
Cellular composition of the synovial membrane, its interaction with bone tissue and immune cell types, and related mediators involved in inflamed RA joints. (A) In healthy conditions, the synovium consist of a thin lining layer of lining fibroblasts in association with lining M1 directly exposed to the bone tissue. The underlying sub-lining layer is a connective tissue enriched in blood vessels, adipocytes, fibroblasts, and both innate and adaptive immune cells. The inflamed RA synovium is characterized by a hyperplastic lining layer surrounded by proinflammatory sub-lining fibroblasts and a massive infiltration of B-cells, monocyte-derived macrophages, autoantibody-secreting plasma cells, and differentiated cytotoxic effector memory CD4⁺ T-cells in the sub-lining layer. The secretion of pro-inflammatory interleukins (IL-1: interleukin 1; IL-6: interleukin 6; TNF- α: tumor necrosis factor-alpha) by activated immune cells stimulates the production of the soluble cytokine Mediator Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL), which binds to its receptor RANK on monocytes and macrophages causing their differentiation into bone-resorbing osteoclasts. Red arrows indicate pro-inflammatory processes. (B) Interleukin (IL) isoforms produced by different types of synovial innate immune cells and macrophages (IL-6: interleukin 6; IL-1: interleukin 1; TNF-α: tumor necrosis factor-alpha; TNF-β: tumor necrosis factor-beta; VEGF: vascular endothelial growth factor). (C) Differentiation and interconversion of adaptive immune cells (Treg: regulatory T cells; TH1: T helper 1 cells; TH 17: T helper 17 cells; TH2: T helper 2 cells; B cells; APCs: antigen-presenting cells). (D) Effects of secreted ILs on bone-remodeling processes depending on the osteoprotegerin (OPG)–RANKL axis, which regulates the differentiation of osteoclasts in bone-resorbing osteoclasts. Figures created with BioRender.com, accessed on 15 November 2023.
… 
Structures of microalgal carotenoids and ROS detoxification mechanisms of astaxanthin. Lycopene (A), beta-carotene (B), lutein (C), astaxanthin (ASTX, (D)), fucoxanthin (E), and diatoxanthin (F). Pink and purple bars indicate the hydrophobic and hydrophilic regions of the molecules, respectively; in red, the oxygen of the keto groups, and in blue, the oxygen of the carboxylic groups; in green, the R and R’ functional groups of astaxanthin. Panel (G) outlines the two main routes of ASTX-mediated singlet molecule oxygen (¹O2) detoxification. The top pathway is based on an electron transfer process involving: (i) the formation of a weakly bound ASTX-¹O2 complex followed by direct electron transfer from the highest occupied molecular orbital (HOMO) of ASTX to the lowest unoccupied molecular orbital (LUMO) of singlet oxygen (¹O2), and the formation of radicals; (ii) a reverse reaction restoring the electron distribution between the two molecules. The overall process converts ¹O2 to its triplet unreactive form (³O2) upon spin inversion, while ASTX is restored from ³ASTX via internal conversion [55]. The bottom pathway shows the free radical scavenging activity based on a two-step transfer involving both an electron and proton (H⁺) from ASTX to ¹O2. The formed hydrogen peroxide is readily removed by peroxidase enzymes while ASTX is spontaneously restored by ascorbate. These mechanisms of action are iterative, meaning that a single ASTX molecule can perform multiple ROS detoxification cycles. Figures created with BioRender.com, accessed on 15 November 2023.
… 
of evidence-based interference of selected microalgal metabolites with the intracellular pro-inflammatory signaling pathways involved in the pathogenesis of RA. Microalgal carotenoids and lipids exert inhibitory effects on major intracellular pro-inflammatory signaling pathways involved in the onset and progression of rheumatoid arthritis. Blunt-ended solid lines indicate an inhibitory pharmacological effect, while dashed lines suggest proposed interference. Astaxanthin (ASTX), fucoxanthin, diatoxanthin, and β-carotene target different subunits of the Inhibitor of κB (IkB) kinase complex, preventing the phosphorylation-dependent release of the pro-inflammatory transcriptional activator NF-kB and its nuclear translocation. ASTX further acts upstream of this pathway through inhibiting the NF-κB-Inducing Kinase (NIK), preventing the autophosphorylation of the Mitogen- and Stress-activated protein Kinase-1 (MSK1), a nucleus-localized effector which activates NF-κB and the cAMP-responsive Element-Binding Protein (CREB) pro-inflammatory transcription factor. ASTX and β-carotene also inhibit the JNK/p38 MAPK signaling cascade, blocking the nuclear translocation of the JNK/p38 MAPK complex, and thus phosphorylation of downstream targets: the pro-inflammatory transcription factors ELK1, MEF2, ATF2, and STAT1, and the MAPK-activated kinase 2 (MK2) responsible for stabilizing IL mRNAs. β-carotene inhibits the JAK2/STAT3 pathway through blocking phosphorylation of the pro-inflammatory transcriptional activator STAT3 by JAK2 and its nuclear translocation. ASTX positively regulates the nuclear factor-erythroid 2-Related factor 2 (Nrf2)-mediated pathway involved in antioxidant and anti-inflammatory gene expression. ASTX and β-carotene are proposed to directly bind interleukin 6 (IL-6) and tumor necrosis factor-alpha (TNF-α), blocking their receptor interaction and activation of the downstream pathways. Lutein is suggested to inhibit both NF-kB and JNK/p38 MAPK pathways. ASTX detoxifies free radicals on both sides of the lipid bilayer and appears to suppress mitochondrial ROS production. The betaine lipid DGTS interferes with the activity of the IKKβ subunit of the IkB kinase complex, while the PUFAs docosahexaenoic (DHA), eicosapentaenoic (EPA), and dihomo-γ-linolenic (DGLA) acids modulate the NF-kB signaling cascade through targeting unknown pathway components. Figures created with BioRender.com, accessed on 15 November 2023.
… 
This content is subject to copyright.
Citation: 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. https://doi.org/10.3390/
md21120630
Academic Editor: Alberto Amato
Received: 17 October 2023
Revised: 4 December 2023
Accepted: 5 December 2023
Published: 7 December 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
marine drugs
Review
The Clinical Promise of Microalgae in Rheumatoid Arthritis:
From Natural Compounds to Recombinant Therapeutics
Edoardo Andrea Cutolo 1, *, Roberto Caferri 1, Rosanna Campitiello 2and Maurizio Cutolo 2
1Laboratory of Photosynthesis and Bioenergy, Department of Biotechnology, University of Verona,
Strada le Grazie 15, 37134 Verona, Italy; roberto.caferri@univr.it
2Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine,
IRCCS San Martino Polyclinic Hospital, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy;
rosannacampitiello@hotmail.it (R.C.)
*Correspondence: edoardoandrea.cutolo@univr.it
Abstract:
Rheumatoid arthritis (RA) is an invalidating chronic autoimmune disorder characterized
by joint inflammation and progressive bone damage. Dietary intervention is an important component
in the treatment of RA to mitigate oxidative stress, a major pathogenic driver of the disease. Along-
side traditional sources of antioxidants, microalgae—a diverse group of photosynthetic prokaryotes
and eukaryotes—are emerging as anti-inflammatory and immunomodulatory food supplements.
Several species accumulate therapeutic metabolites—mainly lipids and pigments—which interfere
in the pro-inflammatory pathways involved in RA and other chronic inflammatory conditions. The
advancement of the clinical uses of microalgae requires the continuous exploration of phytoplankton
biodiversity and chemodiversity, followed by the domestication of wild strains into reliable produc-
ers of said metabolites. In addition, the tractability of microalgal genomes offers unprecedented
possibilities to establish photosynthetic microbes as light-driven biofactories of heterologous im-
munotherapeutics. Here, we review the evidence-based anti-inflammatory mechanisms of microalgal
metabolites and provide a detailed coverage of the genetic engineering strategies to enhance the
yields of endogenous compounds and to develop innovative bioproducts.
Keywords:
photosynthesis; polyunsaturated fatty acids; carotenoids; oxylipins; xanthophylls;
antioxidants; functional foods; synthetic biology; genetic engineering; inflammation; rheumatoid
arthritis; autoimmunity; docosahexaenoic acid (DHA); eicosapenteanoic acid (EPA); astaxanthin;
rheumatology; interleukins; chloroplast; molecular pharming; novel foods; bioeconomy
1. Introduction
Chronic inflammation is a defining feature of autoimmune diseases, a group of condi-
tions in which immunological self-tolerance is disturbed due to the recognition of autoanti-
gens by immune cells. Rheumatoid arthritis (RA), the most common chronic inflammatory
arthropathy [
1
,
2
], is a systemic autoimmune disorder affecting the synovial joints, with a
higher incidence in women [
3
]. RA displays a complex pathophysiology involving the up-
regulation of pro-inflammatory mediators (interleukins, ILs) and enhanced production of
reactive oxygen species (ROS) [
4
,
5
]. Both genetic and modifiable lifestyle factors contribute
to the risk of RA predisposition [
6
,
7
], with diet highly influencing disease activity [
8
,
9
]. In
particular, a high antioxidant intake is known to reduce onset risk and to ameliorate the
clinical course of the disease [
10
], therefore, the identification of new sources of antioxidant
and anti-inflammatory molecules is of high clinical relevance.
Microalgae are photosynthetic prokaryotes and eukaryotes adapted to diverse envi-
ronments, including extreme habitats [
11
,
12
], which are consumed in human nutrition as
sources of proteins and other bioactive compounds [
13
17
]. Several species are non-toxic
producers of essential vitamins, lipids, and pigments of therapeutic value [
18
22
], which
could be employed as complementary agents in the management of chronic inflammatory
Mar. Drugs 2023,21, 630. https://doi.org/10.3390/md21120630 https://www.mdpi.com/journal/marinedrugs
Mar. Drugs 2023,21, 630 2 of 27
diseases. Moreover, the fast life cycle and light-powered autotrophic metabolism of mi-
croalgae allows for large-scale cultivation with lower inputs compared with heterotrophic
microorganisms [23].
In this review, we summarize the evidence-based putative interference of microalgal
compounds in pro-inflammatory pathways involved in the pathogenesis of RA and discuss
how bioprospecting for novel pharmacologically relevant strains, and their domestication,
can advance the clinical use of photosynthetic microorganisms. Lastly, we provide an
update on the available genetic engineering strategies to enhance the production of endoge-
nous microalgal metabolites and introduce emerging approaches to achieve light-driven
conversion of CO2into high-value recombinant biopharmaceuticals.
Pathogenesis and Mediators of Rheumatoid Arthritis
Although the exact etiology of RA remains unknown, the balance between immune
cells and the production of inflammatory ILs in the connective tissue that lines the joint
capsule (synovium) is altered in the disease onset and progression [
4
,
5
]. The healthy syn-
ovium consists of a thin lining layer of fibroblasts covering a connective tissue surrounded
by blood vessels and enriched in fibroblasts, and innate and adaptive immune cells: the
sub-lining layer [
24
] (Figure 1A). In RA, the lining layer is hyperplastic while the sub-lining
layer is infiltrated with B-cells, monocyte-derived macrophages, autoantibody-secreting
plasma cells, and differentiated cytotoxic CD4
+
T-cells involved in the breakdown of tissue
tolerance [
25
27
]. The release of pro-inflammatory ILs by monocyte-derived M1 macrophages,
and osteoclast activation cause progressive bone resorption [
28
], and autoantibodies pro-
duced by differentiated plasma cells further contribute to joint damage [29].
Mar. Drugs 2023, 21, x 3 of 28
Figure 1. Cellular composition of the synovial membrane, its interaction with bone tissue and
immune cell types, and related mediators involved in inflamed RA joints. (A) In healthy conditions,
the synovium consist of a thin lining layer of lining fibroblasts in association with lining M1 directly
exposed to the bone tissue. The underlying sub-lining layer is a connective tissue enriched in blood
vessels, adipocytes, fibroblasts, and both innate and adaptive immune cells. The inflamed RA
synovium is characterized by a hyperplastic lining layer surrounded by proinflammatory sub-lining
fibroblasts and a massive infiltration of B-cells, monocyte-derived macrophages, autoantibody-
secreting plasma cells, and differentiated cytotoxic effector memory CD4+ T-cells in the sub-lining
layer. The secretion of pro-inflammatory interleukins (IL-1: interleukin 1; IL-6: interleukin 6; TNF-
α: tumor necrosis factor-alpha) by activated immune cells stimulates the production of the soluble
cytokine Mediator Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL), which binds to
its receptor RANK on monocytes and macrophages causing their differentiation into bone-resorbing
osteoclasts. Red arrows indicate pro-inflammatory processes. (B) Interleukin (IL) isoforms produced
by different types of synovial innate immune cells and macrophages (IL-6: interleukin 6; IL-1:
interleukin 1; TNF-α: tumor necrosis factor-alpha; TNF-β: tumor necrosis factor-beta; VEGF:
vascular endothelial growth factor). (C) Differentiation and interconversion of adaptive immune
cells (Treg: regulatory T cells; TH1: T helper 1 cells; TH 17: T helper 17 cells; TH2: T helper 2 cells; B
cells; APCs: antigen-presenting cells). (D) Effects of secreted ILs on bone-remodeling processes
depending on the osteoprotegerin (OPG)RANKL axis, which regulates the differentiation of
osteoclasts in bone-resorbing osteoclasts. Figures created with BioRender.com, accessed on 15
November 2023.
Figure 1.
Cellular composition of the synovial membrane, its interaction with bone tissue and
immune cell types, and related mediators involved in inflamed RA joints. (
A
) In healthy conditions,
Mar. Drugs 2023,21, 630 3 of 27
the synovium consist of a thin lining layer of lining fibroblasts in association with lining M1 directly
exposed to the bone tissue. The underlying sub-lining layer is a connective tissue enriched in blood
vessels, adipocytes, fibroblasts, and both innate and adaptive immune cells. The inflamed RA syn-
ovium is characterized by a hyperplastic lining layer surrounded by proinflammatory sub-lining
fibroblasts and a massive infiltration of B-cells, monocyte-derived macrophages, autoantibody-
secreting plasma cells, and differentiated cytotoxic effector memory CD4
+
T-cells in the sub-lining
layer. The secretion of pro-inflammatory interleukins (IL-1: interleukin 1; IL-6: interleukin 6; TNF-
α
: tumor necrosis factor-alpha) by activated immune cells stimulates the production of the soluble
cytokine Mediator Receptor Activator of Nuclear Factor Kappa-B Ligand (RANKL), which binds to
its receptor RANK on monocytes and macrophages causing their differentiation into bone-resorbing
osteoclasts. Red arrows indicate pro-inflammatory processes. (
B
) Interleukin (IL) isoforms pro-
duced by different types of synovial innate immune cells and macrophages (IL-6: interleukin 6;
IL-1: interleukin 1; TNF-
α
: tumor necrosis factor-alpha; TNF-
β
: tumor necrosis factor-beta; VEGF:
vascular endothelial growth factor). (
C
) Differentiation and interconversion of adaptive immune cells
(Treg: regulatory T cells; TH1: T helper 1 cells; TH 17: T helper 17 cells; TH2: T helper 2 cells; B cells;
APCs: antigen-presenting cells). (
D
) Effects of secreted ILs on bone-remodeling processes depending
on the osteoprotegerin (OPG)–RANKL axis, which regulates the differentiation of osteoclasts in
bone-resorbing osteoclasts. Figures created with BioRender.com, accessed on 15 November 2023.
At the intracellular level, three main pro-inflammatory signaling kinase cascades
responding to soluble mediators are involved in RA, all being influenced by microalgal
metabolites: the Nuclear Factor Kappa-B (NF-kB)-mediated pathway [
30
], the Janus kinase
2/Signal Transducers and Activators of Transcription 3 (JAK2/STAT3) pathway [
31
], and
the Jun N-terminal kinase (JNK)/p38 Mitogen-Activated Protein Kinase (p38 MAPK)
pathway [32], the latter being predominant in the lining layer and endothelial cells.
Pro-inflammatory ILs (Figure 1D) [
33
] stimulate the production of the soluble cy-
tokine Mediator Receptor Activator of Nuclear Factor Kappa-B Ligand (RANKL) by im-
mune cells. RANKL binds its receptor RANK on monocytes and macrophages causing
their differentiation into bone-resorbing osteoclasts [
34
]. Other ILs produced by anti-
inflammatory M2 macrophages positively influence the osteoprotegerin (OPG)–RANKL
ratio, promoting bone homeostasis [
35
] (Figure 1D), while the IL-10 family (Figure 1D)
appears to play dual roles. Some members stimulate osteogenesis and suppress the syn-
thesis of pro-inflammatory mediators like IL-6, Tumor Necrosis Factor-
α
(TNF-
α
), and
Vascular endothelial growth factor (VEGF) [
36
], while others activate the NF-kB and MAPK
pathways, stimulating TNF-
α
, IL-1
β
, and RANKL production by synovial fibroblasts,
promoting osteoclastogenesis.
In conclusion, given the diversified nature of factors regulating the balance between
pro- and anti-inflammatory mediators in RA, there is a strong interest in discovering novel
molecules to be used as complementary tools alongside conventional therapies.
2. Anti-Inflammatory and Immunomodulatory Metabolites from Microalgae
2.1. Carotenes and Xanthophylls
Like bacteria, fungi, and plants, microalgae synthetize C40 lipophilic pigments con-
sisting of a polyene chain of conjugated double bonds (Figure 2) with terminally linked
ionone rings known as carotenoids [
37
,
38
].
β
-carotene (Figure 2B)—a structural element
of photosystems [
39
]—and the antioxidant lycopene (Figure 2A) are anti-inflammatory
carotenes [
40
,
41
] usually introduced in the diet with carrots (Daucus carota) and tomatoes
(Solanum lycopersicum), respectively, although present also in microalgae [42].
Xanthophylls are oxygenated carotenoids containing hydroxyl and ketone groups
in the ionone rings, which serve different functions in phototrophs. The non-ketolated
xanthophyll lutein (Figure 2C) participates in light-harvesting and photoprotection, while
the ketocarotenoid astaxanthin (ASTX, Figure 2D) scavenges harmful ROS generated by
photosynthetic electron transport under excess light [
43
]. Lutein is an anti-inflammatory
Mar. Drugs 2023,21, 630 4 of 27
carotenoid [
44
] abundantly found in green leafy vegetables and egg yolk, while ASTX is a
potent antioxidant uniquely synthetized by a few microalgal species.
Abiotic stresses induce a hypercarotenogenic response in several chlorophytes, in-
cluding the halophile Dunaliella salina (Chlorophyceae), which overaccumulates
β
-carotene
in lipid bodies (plastoglobules) inside the chloroplast [
45
], and in the freshwater species
Haematococcus pluvialis (Haematococcaceae), which forms haematocysts filled with ASTX-
rich cytoplasmic lipid droplets [
46
]. Other microalgal xanthophylls with anti-inflammatory
and immunomodulatory properties are fucoxanthin and diatoxanthin produced by sev-
eral diatoms (stramenopiles) and by the haptophyte Tisochrysis lutea (Coccolithophyceae)
(Figure 2E,F) [
47
,
48
]. As discussed in the following paragraphs, carotenoids appear to
interfere in all major pro-inflammatory pathways implicated in the onset and progression
of RA.
Astaxanthin: The Red Gold of Algae
With recognized safety for human consumption [
49
], approved Novel Food sta-
tus [
50
], and an established role in promoting bone homeostasis in degenerative skeletal
diseases [51], ASTX is the microalgal pigment of highest biopharmaceutical value.
Clinical studies have shown that ASTX intake reduces the levels of systemic inflam-
matory biomarkers [
52
,
53
] and potentiates the pain-relieving effect of anti-inflammatory
therapies [
54
]. The pharmacological effects of ASTX derive from its strong antioxidant-
activity mediated via ROS quenching [
55
] (Figure 2G, top panel) and direct free radical
scavenging [
56
,
57
] (Figure 2G, bottom panel). This amphipathic molecule is symmetrically
arranged within the lipid bilayer [
58
], thus exerting antioxidant activity on both intra- and
extracellular environments. Notably, the higher number of hydroxyl groups compared with
other carotenoids confers to ASTX superior ROS-detoxifying capacity [
59
]. Early studies
showed that ASTX suppressed ROS production [
60
63
] and secretion of pro-inflammatory
ILs by cultured human-activated monocytes [
64
]. Moreover ASTX stimulated the expres-
sion of ROS-scavenging enzymes in chondrocytes challenged with IL-1
β
[
65
], and inhibited
pro-inflammatory and osteoclastogenic gene expression in macrophages challenged with
RANKL [
66
]. Lastly, the administration of ASTX promoted cartilage health in animal
models of arthritis and osteoasthritis [6769].
Notably, the esterified biological form of ASTX displays higher bioavailability com-
pared with synthetic ester-free derivatives [
70
73
], suggesting the need to improve its
production from natural sources. However, a major limitation to the clinical use of ASTX
lies in its low solubility in gastrointestinal fluids [
74
], requiring encapsulation in polysac-
charide, lipid, and protein nanoparticles to enhance its delivery and release [7580].
2.2. Anti-Inflammatory Mechanisms of Action of Astaxanthin and Other Carotenoids
2.2.1. NF-κB Pathway
ASTX and
β
-carotene interfere with the NF-
κ
B pathway blocking the transloca-
tion of the NF-
κ
B transcription factor to the nucleus, thereby suppressing ROS and pro-
inflammatory gene expression. This effect is likely mediated through targeting the Inhibitor
of the NF-
κ
B
γ
subunit (IKK-
γ
) of the IkB kinase complex [
81
84
]. This prevents the phos-
phorylation and subsequent proteasome-mediated degradation of the IkB
α
binding factor,
which abolishes the release of NF-
κ
B [
30
]. A similar inhibitory effect has been proposed for
fucoxanthin and diatoxanthin [85,86].
The Mitogen- and Stress-activated protein Kinase-1 (MSK1) is a nucleus-localized
factor, which activates the NF-
κ
B pathway [
87
] and the transcriptional regulator cAMP-
responsive Element-Binding Protein (CREB) [
88
]. Phosphorylated CREB binds CREB-
Responsive Elements (CRE) promoting pro-inflammatory gene expression [
89
]. These
events are suppressed by ASTX, which inhibits MSK1 autophosphorylation [
90
]. Lastly, in
silico simulations suggested that ASTX and
β
-carotene extracellularly interact with IL-6
and TNF-
α
, preventing their binding to membrane receptors [
91
]. ASTX may also interact
Mar. Drugs 2023,21, 630 5 of 27
with the NF-
κ
B-Inducing Kinase (NIK) and block the phosphorylation of the IKK-
α
subunit
of the IkB αkinase complex, suppressing the NF-κB pathway [92].
Mar. Drugs 2023, 21, x 5 of 28
Clinical studies have shown that ASTX intake reduces the levels of systemic
inflammatory biomarkers [52,53] and potentiates the pain-relieving effect of anti-
inflammatory therapies [54]. The pharmacological effects of ASTX derive from its strong
antioxidant-activity mediated via ROS quenching [55] (Figure 2G, top panel) and direct
free radical scavenging [56,57] (Figure 2G, boom panel). This amphipathic molecule is
symmetrically arranged within the lipid bilayer [58], thus exerting antioxidant activity on
both intra- and extracellular environments. Notably, the higher number of hydroxyl
groups compared with other carotenoids confers to ASTX superior ROS-detoxifying
capacity [59]. Early studies showed that ASTX suppressed ROS production [6063] and
secretion of pro-inflammatory ILs by cultured human-activated monocytes [64]. Moreover
ASTX stimulated the expression of ROS-scavenging enzymes in chondrocytes challenged
with IL-1β [65], and inhibited pro-inflammatory and osteoclastogenic gene expression in
macrophages challenged with RANKL [66]. Lastly, the administration of ASTX promoted
cartilage health in animal models of arthritis and osteoasthritis [6769].
Figure 2. Structures of microalgal carotenoids and ROS detoxification mechanisms of astaxanthin.
Lycopene (A), beta-carotene (B), lutein (C), astaxanthin (ASTX, (D)), fucoxanthin (E), and
diatoxanthin (F). Pink and purple bars indicate the hydrophobic and hydrophilic regions of the
molecules, respectively; in red, the oxygen of the keto groups, and in blue, the oxygen of the
carboxylic groups; in green, the R and R’ functional groups of astaxanthin. Panel (G) outlines the
two main routes of ASTX-mediated singlet molecule oxygen (1O2) detoxification. The top pathway
Figure 2.
Structures of microalgal carotenoids and ROS detoxification mechanisms of astaxanthin.
Lycopene (
A
), beta-carotene (
B
), lutein (
C
), astaxanthin (ASTX, (
D
)), fucoxanthin (
E
), and diatoxan-
thin (
F
). Pink and purple bars indicate the hydrophobic and hydrophilic regions of the molecules,
respectively; in red, the oxygen of the keto groups, and in blue, the oxygen of the carboxylic groups;
in green, the R and R’ functional groups of astaxanthin. Panel (
G
) outlines the two main routes
of ASTX-mediated singlet molecule oxygen (
1
O
2
) detoxification. The top pathway is based on an
electron transfer process involving: (i) the formation of a weakly bound ASTX-
1
O
2
complex followed
by direct electron transfer from the highest occupied molecular orbital (HOMO) of ASTX to the lowest
unoccupied molecular orbital (LUMO) of singlet oxygen (
1
O
2
), and the formation of radicals; (ii) a
reverse reaction restoring the electron distribution between the two molecules. The overall process
converts
1
O
2
to its triplet unreactive form (
3
O
2
) upon spin inversion, while ASTX is restored from
3
ASTX via internal conversion [
55
]. The bottom pathway shows the free radical scavenging activity
based on a two-step transfer involving both an electron and proton (H
+
) from ASTX to
1
O
2
. The
formed hydrogen peroxide is readily removed by peroxidase enzymes while ASTX is spontaneously
restored by ascorbate. These mechanisms of action are iterative, meaning that a single ASTX molecule
can perform multiple ROS detoxification cycles. Figures created with BioRender.com, accessed on
15 November 2023.
Mar. Drugs 2023,21, 630 6 of 27
2.2.2. JAK2/STAT3 and JNK/p38 MAPK Pathways
β
-carotene and ASTX further modulate the pro-inflammatory pathways mediated by
the JNK/p38 MAPK [
93
] and JAK2/STAT3 kinases [
84
,
94
], the latter responding to IL-6
in the pathogenesis of RA and osteoarthritis [
31
,
95
]. Phosphorylation of STAT3 dimers
by JAK2 induces nuclear translocation and the differentiation of CD4
+
T cells into the
highly reactive T helper 17 (Th17) cell type [
96
]. TNFs and IL-1 activate the JNK/p38
MAPK pathway starting a phosphorylation cascade ending with JNK/p38 MAPK nuclear
translocation [
97
], and phosphorylation of pro-inflammatory transcription factors (ELK1,
MEF2, ATF2, and STAT1) [
98
] and of the MAPK-activated kinase 2 (MK2), which, in turn,
targets the tristetrapolin (TTP) factor, promoting stabilization of IL mRNAs [
99
]. These
oxidant-sensitive inflammatory pathways are also modulated by lutein [
100
], as reported
using extracts enriched in this xanthophyll from different species of the chlorophyte genus
Tetraselmis (Chlorodendrophyceae) [101].
2.2.3. Other Pro-Inflammatory Pathways Targeted by Microalgal Carotenoids
Mitochondrial disfunction is a key pathogenic driver in RA [
102
], and ASTX was
reported to attenuate organellar ROS production in human chondrocytes treated with
IL-1
β
[
69
]. In addition to suppressing pro-inflammatory pathways, ASTX is also suggested
to promote cartilage homeostasis via the transcriptional regulator nuclear factor-erythroid
2-related factor 2 (Nrf2) [
68
,
93
]. ASTX is suggested to stabilize and promote the nuclear
translocation of Nfr2, which binds so-called antioxidant response elements (AREs), enhanc-
ing the expression of anti-inflammatory and ROS-detoxifying genes [103,104].
2.3. Lipids and Their Derivatives
A hallmark of RA is the altered fatty acid profile of the synovium [
105
,
106
], while the
intake of polyunsaturated fatty acids (PUFAs) correlates with joint health and mitigates
the risk of RA onset [
107
]. Microalgal mass cultivation is a more sustainable way to derive
functional lipids compared with cold water fish [
108
112
]. Phytoplankton occupies the
lowest trophic level in oceans and freshwater basins, representing the primary PUFAs pro-
ducer in aquatic food webs [
113
]. Global warning and ocean acidification are predicted to
affect phytoplankton ecology [
114
,
115
], thus reducing PUFAs availability to higher trophic
levels and, eventually, putting at risk the supply for human nutrition [
116
]. Moreover,
upon stress acclimation, microalgae synthetise a wider range of anti-inflammatory and
immunomodulatory lipids compared to animals [117121].
2.3.1. Long-Chain Polyunsaturated Fatty Acids
Several microalgae accumulate very long-chain PUFAs [
122
,
123
], including the
omega-3
(
ω
-3, n-3) PUFAs [
124
]
α
-linolenic (18:3), docosahexaenoic (DHA, 22:6, n-3, Figure 3A)
,
do-
cosapentaenoic (DPA, n-3, 22:5, Figure 3B), and eicosapentaenoic (EPA, 20:5, n-3,
Figure 3C) acids but also
ω
-6 PUFAs like arachidonic (ARA, 20:4, n-6, Figure 3H),
γ
-linolenic (18:3), linoleic (18:2), and dihomo-
γ
-linolenic (DGLA, 20:3, n-6, Figure 3D) acids.
These molecules are biosynthetic precursors of anti-inflammatory signaling molecules and
interfere with pro-inflammatory pathways [125,126].
The administration of lipids from the DGLA-hyperproducing freshwater chloro-
phyte Lobosphaera incisa (Trebouxiophyceae) suppressed the expression of the NF-
κ
B
pathway-related genes in an animal model of chronic inflammation [
127
]. Similarly, lipid
extracts from the haptophyte Pavlova lutheri (Prymnesiophyceae), an EPA- and DHA-
hyperaccumulating strain, inhibited IL-6 and TNF-
α
production in activated human
macrophages, possibly through suppressing the NF-κB pathway [128].
Several heterotrophic marine microorganisms are strong DHA producers, such as the
dinoflagellate Crypthecodinium cohnii (Dinophyceae) and, above all, the thraustochytrids pro-
tists Thraustochytrium spp., Aurantiochytrium (formerly Schizochytrium) limacinum (Labyrinthu-
lomycetes) [
129
133
], and related genera [
134
]. Although these microorganisms can-
not exploit light energy to drive their metabolism, they are capable of fermenting plant
Mar. Drugs 2023,21, 630 7 of 27
biomass hydrolysates, affording cost-effective heterotrophic cultivation using renewable
feedstocks [
135
141
]. Arguably, the most valuable heterotrophic sources of DHA are
Schizochytrium spp., which recently obtained Novel Food status [
142
,
143
]. Notably, a recent
human clinical trial investigated the supplementation of an EPA- and DHA-enriched oil
from a Schizochytrium sp. in RA patients, reporting beneficial effects on joint health and the
blood levels of inflammatory markers [144].
Mar. Drugs 2023, 21, x 8 of 28
Figure 3. (A) Docosahexaenoic (DHA, 22:6, n-3); (B) docosapentaenoic (DPA, 22:5, n-3); (C)
eicosapentaenoic (EPA, 20:5, n-3); (D) dihomo-γ-linolenic (DGLA, 20:3, n-6); (E) 1,2-diacylglyceryl-
3-O-4-(N,N,N-trimethyl)-homoserine (DGTS); (F) 1,2-diacylglyceryl-3-O-carboxy-
(hydroxymethyl)-choline (DGCC); (G) 1,2-diacylglyceryl-3-O-2-(hydroxymethyl)-(N,N,N-
trimethyl)-β-alanine (DGTA); (H) arachidonic (ARA, 20:4, n-6). Figures created with BioRender.com,
accessed on 15 November 2023.
The administration of lipids from the DGLA-hyperproducing freshwater
chlorophyte Lobosphaera incisa (Trebouxiophyceae) suppressed the expression of the NF-
κB pathway-related genes in an animal model of chronic inflammation [127]. Similarly,
lipid extracts from the haptophyte Pavlova lutheri (Prymnesiophyceae), an EPA- and DHA-
hyperaccumulating strain, inhibited IL-6 and TNF-α production in activated human
macrophages, possibly through suppressing the NF-κB pathway [128].
Figure 3.
(
A
) Docosahexaenoic (DHA, 22:6, n-3); (
B
) docosapentaenoic (DPA, 22:5, n-3); (
C
) eicos-
apentaenoic (EPA, 20:5, n-3); (
D
) dihomo-
γ
-linolenic (DGLA, 20:3, n-6); (
E
) 1,2-diacylglyceryl-3-
O-4’-(N,N,N-trimethyl)-homoserine (DGTS); (
F
) 1,2-diacylglyceryl-3-O-carboxy-(hydroxymethyl)-
choline (DGCC); (
G
) 1,2-diacylglyceryl-3-O-2’-(hydroxymethyl)-(N,N,N-trimethyl)-
β
-alanine (DGTA);
(
H
) arachidonic (ARA, 20:4, n-6). Figures created with BioRender.com, accessed on 15 November 2023.
2.3.2. Betaine Lipids
Betaine lipids are anti-inflammatory and immunomodulatory glycerolipids in which
the phosphate and carbohydrate moieties attached to the glycerol backbone are replaced
with positively charged ether-bond betaine groups. Betaine lipids are widely distributed in
all clades of eukaryotic microalgae, where they act as acyl group donors upon membrane
lipid remodelling and during the accumulation of storage neutral lipids [145,146].
Mar. Drugs 2023,21, 630 8 of 27
Betaine lipids derive from the turnover of membrane phospholipids under abiotic
stresses, mainly temperature and nutrient starvation [
147
149
]. The betaine lipid 1,2-
diacylglyceryl-3-O-4’-(N,N,N-trimethyl)-homoserine (DGTS, Figure 3E) is the most abundant
betaine lipid in microalgae, followed by 1,2-diacylglyceryl-3-O-carboxy-(hydroxymethyl)-
choline (DGCC, Figure 3F), and 1,2-diacylglyceryl-3-O-2’-(hydroxymethyl)-(N,N,N-trimethyl)-
β
-alanine (DGTA, Figure 3G), with evidence of DGTS-mediated inhibition of the IKK-
β
kinase causing suppression of the NF-
κ
B pathway and secretion of pro-inflammatory ILs
by Th1 and Th2 cells, as recently reported with extracts from the oleaginous chlorophyte
Chromochloris zofingiensis (Chlorophyceae) [150].
2.3.3. Oxylipins
The eicosanoids, prostaglandins and leukotrienes, and the thromboxanes, are hormone-
like oxygenated metabolites of C20 fatty acids involved in the modulation and resolution
of inflammation in the RA synovium [
106
]. In mammals, oxylipins are produced by
the enzyme phospholipase A2 via release of sn-2 PUFAs from membrane phospholipids.
Typical oxylipin precursors are linoleic,
α
-linolenic, and ARA, which are substrates of
cyclooxygenases, lipoxygenases, and cytochrome P450 enzymes, respectively [151].
Several microalgal species are known to accumulate prostaglandin-like
oxylipins [
152
154
]. Their synthesis can occur either enzymatically via animal-like biosyn-
thetic pathways [
155
], as in diatoms Skeletonema marinoi and Thalassiosira rotula (Bacillario-
phyceae) [
156
158
], or via spontaneous oxidation of ARA, EPA, and DHA in T. lutea [
159
].
The latter, known as isoprostanoids, are functional lipids which regulate bone health
through preventing osteoclast differentiation [160].
The oxylipins isolated from the chlorophytes Chlamydomonas debaryana (Chlorophyceae)
and Nannochloropsis gaditana inhibited TNF-
α
production in cultured macrophages [
161
],
while the oral administration of biomass of the former suppressed the production of pro-
inflammatory ILs in an animal model of chronic inflammation, possibly through inhibiting
the NF-κB pathway [162,163]
Figure 4summarizes the evidence-based interference of the above-mentioned microal-
gal carotenoids and lipids with the main intracellular pro-inflammatory pathways involved
in the pathogenesis of RA.
Mar. Drugs 2023, 21, x 10 of 28
Figure 4. Summary of evidence-based interference of selected microalgal metabolites with the in-
tracellular pro-inflammatory signaling pathways involved in the pathogenesis of RA. Microalgal
carotenoids and lipids exert inhibitory effects on major intracellular pro-inflammatory signaling
pathways involved in the onset and progression of rheumatoid arthritis. Blunt-ended solid lines
indicate an inhibitory pharmacological effect, while dashed lines suggest proposed interference.
Astaxanthin (ASTX), fucoxanthin, diatoxanthin, and β-carotene target different subunits of the In-
hibitor of κB (IkB) kinase complex, preventing the phosphorylation-dependent release of the pro-
inflammatory transcriptional activator NF-kB and its nuclear translocation. ASTX further acts up-
stream of this pathway through inhibiting the NF-κB-Inducing Kinase (NIK), preventing the auto-
phosphorylation of the Mitogen- and Stress-activated protein Kinase-1 (MSK1), a nucleus-localized
effector which activates NF-κB and the cAMP-responsive Element-Binding Protein (CREB) pro-in-
flammatory transcription factor. ASTX and β-carotene also inhibit the JNK/p38 MAPK signaling
cascade, blocking the nuclear translocation of the JNK/p38 MAPK complex, and thus phosphoryla-
tion of downstream targets: the pro-inflammatory transcription factors ELK1, MEF2, ATF2, and
STAT1, and the MAPK-activated kinase 2 (MK2) responsible for stabilizing IL mRNAs. β-carotene
inhibits the JAK2/STAT3 pathway through blocking phosphorylation of the pro-inflammatory tran-
scriptional activator STAT3 by JAK2 and its nuclear translocation. ASTX positively regulates the
nuclear factor-erythroid 2-Related factor 2 (Nrf2)-mediated pathway involved in antioxidant and
anti-inammatory gene expression. ASTX and β-carotene are proposed to directly bind interleukin
6 (IL-6) and tumor necrosis factor-alpha (TNF-α), blocking their receptor interaction and activation
of the downstream pathways. Lutein is suggested to inhibit both NF-kB and JNK/p38 MAPK path-
ways. ASTX detoxifies free radicals on both sides of the lipid bilayer and appears to suppress mito-
chondrial ROS production. The betaine lipid DGTS interferes with the activity of the IKKβ subunit
of the IkB kinase complex, while the PUFAs docosahexaenoic (DHA), eicosapentaenoic (EPA), and
dihomo-γ-linolenic (DGLA) acids modulate the NF-kB signaling cascade through targeting un-
known pathway components. Figures created with BioRender.com, accessed on 15 November 2023.
3. Bioprospecting and Domestication of Pharmacologically Relevant Microalgae
Of the over 70,000 estimated existing algal strains [164], only a few species are used
in human nutrition and health. Bioprospecting for novel pharmacologically relevant spe-
cies [165,166] requires scrupulous large-scale screening of phytoplankton biodiversity and
chemodiversity [167], and the establishment of optimal cultivation strategies [168]. Inhos-
pitable environments are excellent ecosystems to discover species with industrial applica-
tions since extremophiles are physiologically adapted to harsh conditions and hyperaccu-
mulate pigments and lipids [11,12,169171].
Figure 4.
Summary of evidence-based interference of selected microalgal metabolites with the
intracellular pro-inflammatory signaling pathways involved in the pathogenesis of RA. Microalgal
Mar. Drugs 2023,21, 630 9 of 27
carotenoids and lipids exert inhibitory effects on major intracellular pro-inflammatory signaling path-
ways involved in the onset and progression of rheumatoid arthritis. Blunt-ended solid lines indicate
an inhibitory pharmacological effect, while dashed lines suggest proposed interference. Astaxanthin
(ASTX), fucoxanthin, diatoxanthin, and
β
-carotene target different subunits of the Inhibitor of
κ
B
(IkB) kinase complex, preventing the phosphorylation-dependent release of the pro-inflammatory
transcriptional activator NF-kB and its nuclear translocation. ASTX further acts upstream of this path-
way through inhibiting the NF-
κ
B-Inducing Kinase (NIK), preventing the autophosphorylation of the
Mitogen- and Stress-activated protein Kinase-1 (MSK1), a nucleus-localized effector which activates
NF-
κ
B and the cAMP-responsive Element-Binding Protein (CREB) pro-inflammatory transcription
factor. ASTX and
β
-carotene also inhibit the JNK/p38 MAPK signaling cascade, blocking the nuclear
translocation of the JNK/p38 MAPK complex, and thus phosphorylation of downstream targets: the
pro-inflammatory transcription factors ELK1, MEF2, ATF2, and STAT1, and the MAPK-activated
kinase 2 (MK2) responsible for stabilizing IL mRNAs.
β
-carotene inhibits the JAK2/STAT3 pathway
through blocking phosphorylation of the pro-inflammatory transcriptional activator STAT3 by JAK2
and its nuclear translocation. ASTX positively regulates the nuclear factor-erythroid 2-Related factor 2
(Nrf2)-mediated pathway involved in antioxidant and anti-inflammatory gene expression. ASTX
and
β
-carotene are proposed to directly bind interleukin 6 (IL-6) and tumor necrosis factor-alpha
(TNF-
α
), blocking their receptor interaction and activation of the downstream pathways. Lutein is
suggested to inhibit both NF-kB and JNK/p38 MAPK pathways. ASTX detoxifies free radicals on
both sides of the lipid bilayer and appears to suppress mitochondrial ROS production. The betaine
lipid DGTS interferes with the activity of the IKK
β
subunit of the IkB kinase complex, while the
PUFAs docosahexaenoic (DHA), eicosapentaenoic (EPA), and dihomo-
γ
-linolenic (DGLA) acids
modulate the NF-kB signaling cascade through targeting unknown pathway components. Figures
created with BioRender.com, accessed on 15 November 2023.
3. Bioprospecting and Domestication of Pharmacologically Relevant Microalgae
Of the over 70,000 estimated existing algal strains [
164
], only a few species are used
in human nutrition and health. Bioprospecting for novel pharmacologically relevant
species [
165
,
166
] requires scrupulous large-scale screening of phytoplankton biodiversity
and chemodiversity [
167
], and the establishment of optimal cultivation strategies [
168
].
Inhospitable environments are excellent ecosystems to discover species with industrial
applications since extremophiles are physiologically adapted to harsh conditions and
hyperaccumulate pigments and lipids [11,12,169171].
Among cryophilic species, the extracts of two Antarctic chlorophytes, Chloromonas retic-
ulata (Chlorophyceae) and Micractinium simplicissimus (Trebouxiophyceae), were recently
reported to exert an anti-inflammatory effect on activated macrophages [
172
,
173
]. At
the other extreme, the rhodophyte Cyanidioschyzon merolae (Cyanidiophyceae) thrives at
40
C and low pH, producing heat-stable carotenoids [
174
], while a stress-resilient strain
of the marine chlorophyte species Tetraselmis striata [
175
] accumulates anti-inflammatory
carotenoids and lipids [
176
]. A PUFAs-hyperproducing rhodophyte strain of the Galdieria
genus (Cyanidiophyceae) was identified in acid thermal springs, and its lipid content could
be enhanced via cultivation at temperatures below its optimal range [
177
]. Finally, the
extracts of a chlorophyte Mucidosphaerium sp. (Trebouxiophyceae) isolated from a similar
environment suppressed pro-inflammatory gene expression in human fibroblasts, as well
as mitochondrial ROS production and inflammation in cultured synoviocytes [178,179].
Turning Wild Species into “Unicellular Medicinal Crops”
Although the accumulation of therapeutic compounds in microalgae can be accrued
through abiotic stress challenges [
180
185
], wild organisms are usually not suited for
industrial applications. For instance, ASTX production via mass cultivation H. pluvialis is
restrained by its slow growth and elevated risk of pest contamination [186,187].
Adaptive laboratory evolution and random mutagenesis are powerful strategies for strain
improvement based, respectively, on spontaneous and enhanced mutation rates [
188
,
189
]
(Figure 5A). This approach generated lipid- and carotenoid-hyperproducing strains of
Mar. Drugs 2023,21, 630 10 of 27
chlorophyte Chlorella vulgaris (Trebouxiophyceae) [
190
,
191
], enabling the discovery of new
genetic targets to enhance lutein content [
192
]. Similarly, a simultaneous enhancement
of ASTX and EPA was reported in the Nannochloropsis species gaditiana [
193
] and in the
Tetraselmis striata [
194
], while DHA accrual was achieved in Schizochytrium sp. [
195
197
],
C. cohnii [
198
], and P. lutheri [
199
]. It should be noted, however, that evolved strains are
susceptible to the risk of retromutation and trait drift [200].
Mar. Drugs 2023, 21, x 12 of 28
Figure 5. Nuclear and chloroplast engineering to produce immunotherapeutics in microalgae. (A)
Foreign DNA sequences can be introduced in the nuclear genome of microalgae resulting in the
stable integration of transgene(s). Trait evolution can be achieved through random mutagenesis us-
ing physical (e.g., UV radiation, violet ash) or chemical agents followed by the identification of
phenotypes of interest. The recombinant protein products synthetized on cytoplasmic ribosomes
can be either accumulated in the cell or secreted in the cultivation medium to facilitate their recov-
ery. Alternatively, recombinant metabolic enzymes can be targeted to the chloroplast to enhance the
biosynthesis of native long-chain polyunsaturated fay acids (PUFAs), or even exotic metabolites
with immunomodulatory/anti-inflammatory properties. (B) Transgenes are introduced in the chlo-
roplast via biolistic delivery and targeted to defined chromosomal loci exploiting homologous re-
combination-enabled homology arm sequences (HA, purple) flanking the gene of interest (GOI, blue
segment). The high copy number of plastid chromosomes ensures a significantly greater synthesis
of different classes of recombinant immunotherapeutics compared with the engineering of the nu-
clear haploid genome. Figures created with BioRender.com, accessed on 15 November 2023.
Figure 5.
Nuclear and chloroplast engineering to produce immunotherapeutics in microalgae.
(
A
) Foreign DNA sequences can be introduced in the nuclear genome of microalgae resulting in the
stable integration of transgene(s). Trait evolution can be achieved through random mutagenesis
using physical (e.g., UV radiation, violet flash) or chemical agents followed by the identification of
phenotypes of interest. The recombinant protein products synthetized on cytoplasmic ribosomes can
Mar. Drugs 2023,21, 630 11 of 27
be either accumulated in the cell or secreted in the cultivation medium to facilitate their recovery.
Alternatively, recombinant metabolic enzymes can be targeted to the chloroplast to enhance the
biosynthesis of native long-chain polyunsaturated fatty acids (PUFAs), or even exotic metabolites with
immunomodulatory/anti-inflammatory properties. (
B
) Transgenes are introduced in the chloroplast
via biolistic delivery and targeted to defined chromosomal loci exploiting homologous recombination-
enabled homology arm sequences (HA, purple) flanking the gene of interest (GOI, blue segment).
The high copy number of plastid chromosomes ensures a significantly greater synthesis of different
classes of recombinant immunotherapeutics compared with the engineering of the nuclear haploid
genome. Figures created with BioRender.com, accessed on 15 November 2023.
4. Biomanufacturing of Immunomodulatory Metabolites
4.1. Engineering Carotenoid Metabolism
Although microalgae can potentially substitute plants in the production of carotenoids like
lutein [
201
,
202
], their yields are still lagging behind heterotrophic microorganisms [
203
206
].
Genetic engineering can greatly enhance pigment productivity in microalgae [
207
] but
requires a detailed knowledge of algal genomes and of the transcriptional networks regu-
lating carotenoid biosynthesis to manipulate key metabolic genes [
208
211
] (Figure 5A). In
this respect, a model organism for ketocarotenoid biosynthesis is C. zofingiensis [
212
,
213
],
from which the
β
-carotene ketolase (BKT) enzyme was identified as a rate-limiting factor
for ASTX production [
214
], while the chlorophytes D. salina and Desmodesmus spp. (Chloro-
phyceae) are useful resources for
β
-carotene and lutein metabolism, respectively [
215
,
216
].
Carotenoid enhancement and the production of non-native ASTX isomers can be
achieved through different engineering strategies [
217
], such as: (i) heterologous expression
of chaperones stabilizing biosynthetic enzymes [
218
,
219
] and cyanobacterial proteins to im-
prove pigment storage capacity [
220
], (ii) revival of endogenous silent BKT genes [
221
,
222
],
and (iii) multigene overexpression to circumvent several biosynthetic bottlenecks [
223
].
Simultaneous enhancement of fucoxanthin and
β
-carotene was achieved with a similar ap-
proach in the diatom Phaeodactylum tricornutum (Bacillariophyceae) through overexpressing
endogenous biosynthetic genes [
224
,
225
] and a plastoglobule protein to augment pigment
sequestration [
226
]. Ketocarotenoids engineering was recently reported in the industrially
relevant species C. merolae via heterologous expression of two biosynthetic genes [
227
],
and in the thraustochytrid A. limacinum via overexpression of an endogenous
β
-carotene
hydroxylase gene [228].
4.2. Synthetic Long-Chain Carotenoids
Algal chloroplasts provide excellent metabolic chassis to engineer the synthesis of
non-native pigments with superior therapeutic properties [
229
], such as carotenoids with
extended polyene chains and extra hydroxyl groups [
230
,
231
]. In this respect, extremophilic
microorganisms are invaluable genetic resources to implement novel metabolic pathways
in microalgae [
232
234
]. Archaea produce long-chain (C50) carotenoids through adding iso-
prene (C5) units to lycopene (C40) [
235
]. This biosynthetic pathway was successfully intro-
duced in bacteria to produce C50 ASTX [
236
,
237
] and non-natural C60 ketocarotenoids [
238
],
and it would be of extreme interest to verify its feasibility in microalgae to accumulate
extremely valuable synthetic metabolites.
4.3. Enhancing PUFAs Accumulation
Microalgae can be engineered to maximize the yields of the endogenous anti-inflammatory
lipids [
239
]. Overexpression of endogenous or heterologous fatty acid biosynthesis genes is
a standard approach to enhance PUFAs productivity [
240
], and the availability of transcrip-
tomes investigating stress adaptation is crucial to identify new factors to modulate lipid
profiles [
241
]. For instance, P. lutheri, different Prasinophyte species of the genus Ostreococcus
(Mamiellophyceae), and the diatom Fragilariopsis cylindrus (Bacillariophyceae) are model
species for studying DHA and EPA biosynthesis [
242
244
]. Transcriptomics analysis of
Aurantiochytrium revealed fatty acid synthase isoforms involved in DHA production under
Mar. Drugs 2023,21, 630 12 of 27
nitrogen starvation [
245
], while an acyl-CoA binding protein related to lipid droplet remod-
eling and EPA synthesis in P. tricornutum was recently suggested as a target to enhance the
yields of therapeutic PUFAs [246].
Lipid enhancement was reported in the oleaginous chlorophyte Neochloris oleoabundans
(Chlorophyceae) via heterologous expression of Kennedy pathway genes from the chloro-
phyte Acutodesmus obliquus (Chlorophyceae) [
247
], while DHA was increased in P. tricornu-
tum through overexpressing the
-6 desaturase [
248
], and the heterologous
-5 elongase and
acyl-CoA-dependent 6-desaturase from Ostreococcus tauri [249].
DHA enhancement was achieved in Aurantiochytrium sp. via overexpression of glucose-
6-phosphate dehydrogenase to increase NADPH regeneration for fatty acid biosynthesis [
250
],
and in a Schizochytrium sp. via overexpression of ATP-citrate lyase and acetyl-CoA carboxy-
lase [
251
]. Lastly, EPA was increased in D. salina [
252
] with a heterologous
-6 desaturase
gene from T. pseudonana [253].
Prostaglandin biosynthesis was engineered in the oleaginous diatom Fistulifera solaris
(Bacillariophyceae), a natural producer of C20 PUFAs, via expression of a cyclooxygenase
gene from the red macroalga Agarophyton vermiculophyllum (Gracilariaceae), resulting in the
highest reported heterologous prostaglandin production in a photosynthetic host.
5. Production of Heterologous Immunotherapeutics in Microalgae
Microalgae can be genetically engineered [
254
] to produce heterologous biopharma-
ceuticals [255,256], including human immune-related proteins [257260].
Nuclear transgenesis affords eukaryotic-like post-translational modifications of recom-
binant proteins (mainly N-terminal glycosylation) [
261
,
262
] and extracellular secretion to
facilitate recovery [
258
,
263
] (Figure 5A). Although nuclear transgenesis results in low ex-
pression titers and requires intensive screening efforts to identify high-yielding strains [
264
],
combinatorial transgene assembly and synthetic regulatory elements can greatly improve
translation rates [265,266].
As in plants [
267
], the algal chloroplast genome affords high-level accumulation of
recombinant therapeutics [
268
271
] (Figure 5B). The prokaryotic features of the polyploid
chloroplast genome (plastome) enable efficient homologous recombination-based transgene
insertion and multigene expression with synthetic operons [
272
]. A rich genetic toolkit
allows plastome manipulation in non-model species [
273
], including marker-free and
metabolism-dependent selection strategies [
274
276
]. Among these, the type D phosphite
dehydrogenase (PtxD) gene, which enables the oxidization of non-assimilable phosphite
ions in phosphate, is a biosafe solution to maintain axenic cultures [
277
,
278
], especially in
mixotrophic conditions where pest contamination risk is high [279].
Fast Tracking Microalgal Immunotherapeutics: The Time Is Now
Among newly proposed therapeutic agents for autoimmune diseases, fragment crys-
tallisable (Fc) multimers against autoantibodies and peptides targeting Fc/Fc
γ
receptors
are highly promising candidates [
280
]. Since microalgae can assemble full-length human
monoclonal antibodies binding Fc
γ
receptors [
281
283
], it should be possible to produce
antibodies targeting RA mediators [
284
]. Notably, polycistronic nuclear gene expression
was reported in microalgae [
285
], potentially affording simultaneous production of multiple
immunotherapeutic variants.
Viral nanoparticles are emerging tools with diagnostic and therapeutic applications in
autoimmune diseases [
286
,
287
]. This strategy was pioneered in the plantNicotiana benthamiana
via self-assembled nanoparticles exposing RA autoantigens, which induced immunotol-
erance in animal RA models [
288
]. Although few studies have explored the use of viral
vectors to produce recombinant therapeutics in microalgae so far [
289
,
290
], the ongoing
characterization of microalgae-infecting viruses [
291
294
] should facilitate this approach in
photosynthetic microbes [295,296].
Mar. Drugs 2023,21, 630 13 of 27
6. Final Observations, Comments, and Outlook
Microalgae are key players in the transition towards a bioeconomy [
297
], and are
the focus of intense academic and private research [
298
,
299
]. In Europe, the number of
companies engaging in microalgae production is growing [
300
], and so is the list of patents
describing their therapeutic uses in inflammatory diseases [
301
]. Microalgal biotechnology,
however, is still restrained by low product yields and recovery [
302
306
], high operating
costs, and barriers to market entry [307,308], especially for engineered strains [309].
6.1. Building a Good Reputation
Arguably, the biggest obstacle to the nutritional uses of photosynthetic microbes
is their recognition as safe [
310
]. Currently, only five eukaryotic microalgae and three
cyanoprokaryotes hold Novel Food status [
311
]. The occasional finding of hazardous
contaminants such as heavy metals, toxins, pathogens, and pesticides in the harvested
algal biomass [
312
,
313
] calls for rigorous adherence to good manufacturing practice in the
microalgal food industry [
314
,
315
], especially in the case of emerging strains [
316
]. An
even greater barrier is the fear of horizontal transfer of antibiotic resistance genes from
engineered strains to the gut microbiome and human pathogens [
317
], thus the adoption of
marker-free selection [
274
] and metabolic markers [
275
] are expected to promote a more
favourable attitude towards engineered microalgae and their derived products.
6.2. A Road Map for Clinical Uses of Microalgae in Chronic Inflammation
To advance the applications of microalgae in autoimmune diseases, the therapeu-
tic potential of novel classes of bioactive compounds should be explored. For example,
some microalgal strains are sustainable sources of the lipid-based prohormone vitamin
D
3
, the biosynthetic precursor of the biologically active 1,25-dihydroxyvitamin D (cal-
citriol) [
318
,
319
] whose deficiency is a major risk factor for RA [
320
,
321
]. Several microalgae
accumulate bioactive ergosterol and
β
-sitosterol [
322
,
323
], precursors of vitamin D
2
(ergo-
calciferol) and D
3
, respectively [
324
,
325
]. Moreover, the chlorophyte Botryococcus braunii
(Trebouxiophyceae) and Schizochytrium mangrovei can be engineered to hyperaccumulate
squalene, an anti-inflammatory triterpene and precursor of ergosterol [326331].
The photosynthetic protist Euglena gracilis (Euglenoidea), a recently authorised Novel
Food species [
332
], also deserves attention, being a natural producer of anti-inflammatory
carotenoids, DHA [
333
335
], and paramylon, an immunomodulatory (1,3)-
β
-glucan [
336
]
with reported inhibitory activity towards Th17 cells [337].
Finally, photosynthetic prokaryotes are still largely unexplored biomanufacturing plat-
forms, despite producing a vast repertoire of unique compounds [
338
], including molecules
with antioxidant and anti-inflammatory properties [
339
,
340
] like the pigment–protein com-
plex phycocyanin, which is a selective inhibitor of pro-inflammatory oxylipin synthe-
sis [
341
,
342
], and various polysaccharides [
343
345
] and peptides [
346
348
]. Last but not
least, an increasing number of genetic engineering and synthetic biology tools [
349
] can
be employed to produce recombinant therapeutics in cyanoprokaryotes [
350
], as already
reported for immunomodulatory proteins [351].
7. Conclusions
The exploration of microalgal biodiversity and chemodiversity is a promising ap-
proach to discover new complementary therapeutic approaches for the management of
RA and related chronic inflammatory conditions. Indeed, several preclinical studies have
highlighted multiple mechanisms of action of microalgal compounds and their interfer-
ence with pro-inflammatory pathways. Moreover, the availability of advanced genetic
engineering tools holds tremendous potential to develop innovative biopharmaceuticals in
photosynthetic microbes and expand their clinical applications.
Funding: This research received no external funding.
Data Availability Statement: No new data were created or analyzed in this study.
Mar. Drugs 2023,21, 630 14 of 27
Acknowledgments:
E.A.C. acknowledges the support of the post-doctoral research fellowship “Borsa
Valeria e Vincenzo Landi per Ricerche nel Campo della Genetica Agraria” from the Accademia
Nazionale dei Lincei. R.C. acknowledges the financial support of the European Research Council
(ERC) Advanced Grant 101053983-GrInSun (Harvesting Light for Life: Green Proteins at the Interface
between Sun Energy and Biosphere, Scientific Coordinator: Roberto Bassi).
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Finckh, A.; Gilbert, B.; Hodkinson, B.; Bae, S.-C.; Thomas, R.; Deane, K.D.; Alpizar-Rodriguez, D.; Lauper, K. Global epidemiology
of rheumatoid arthritis. Nat. Rev. Rheumatol. 2022,18, 591–602. [CrossRef]
2.
Meyer, A.; Cirpus, P.; Ott, C.; Schlecker, R.; Zähringer, U.; Heinz, E. Biosynthesis of Docosahexaenoic Acid in Euglena gracilis:
Biochemical and Molecular Evidence for the Involvement of a
4-Fatty Acyl Group Desaturase. Biochemistry
2003
,42, 9779–9788.
[CrossRef]
3.
Kvien, T.K.; Uhlig, T.; Ødegård, S.; Heiberg, M.S. Epidemiological aspects of rheumatoid arthritis: The sex ratio. Ann. N. Y Acad.
Sci. 2006,1069, 212–222. [CrossRef] [PubMed]
4.
Jang, S.; Kwon, E.J.; Lee, J.J. Rheumatoid Arthritis: Pathogenic Roles of Diverse Immune Cells. Int. J. Mol. Sci.
2022
,23, 905.
[CrossRef] [PubMed]
5.
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] [PubMed]
6. Dedmon, L.E. The genetics of rheumatoid arthritis. Rheumatology 2020,59, 2661–2670. [CrossRef] [PubMed]
7.
Schäfer, C.; Keyßer, G. Lifestyle Factors and Their Influence on Rheumatoid Arthritis: A Narrative Review. J. Clin. Med.
2022
,
11, 7179. [CrossRef] [PubMed]
8.
Philippou, E.; Petersson, S.D.; Rodomar, C.; Nikiphorou, E. Rheumatoid arthritis and dietary interventions: Systematic review of
clinical trials. Nutr. Rev. 2021,79, 410–428. [CrossRef] [PubMed]
9.
Nelson, J.; Sjöblom, H.; Gjertsson, I.; Ulven, S.M.; Lindqvist, H.M.; Bärebring, L. Do Interventions with Diet or Dietary
Supplements Reduce the Disease Activity Score in Rheumatoid Arthritis? A Systematic Review of Randomized Controlled Trials.
Nutrients 2020,12, 2991. [CrossRef] [PubMed]
10. Cutolo, M.; Nikiphorou, E. Nutrition and Diet in Rheumatoid Arthritis. Nutrients 2022,14, 888. [CrossRef]
11.
Malavasi, V.; Soru, S.; Cao, G. Extremophile Microalgae: The potential for biotechnological application. J. Phycol.
2020
,56, 559–573.
[CrossRef] [PubMed]
12.
Varshney, P.; Mikulic, P.; Vonshak, A.; Beardall, J.; Wangikar, P.P. Extremophilic micro-algae and their potential contribution in
biotechnology. Bioresour. Technol. 2015,184, 363–372. [CrossRef] [PubMed]
13.
Williamson, E.; Ross, I.L.; Wall, B.T.; Hankamer, B. Microalgae: Potential novel protein for sustainable human nutrition. Trends
Plant Sci. 2021,49, D1004–D1011. [CrossRef] [PubMed]
14.
Niccolai, A.; Chini Zittelli, G.; Rodolfi, L.; Biondi, N.; Tredici, M.R. Microalgae of interest as food source: Biochemical composition
and digestibility. Algal Res. 2019,42, 101617. [CrossRef]
15.
Ampofo, J.; Abbey, L. Microalgae: Bioactive Composition, Health Benefits, Safety and Prospects as Potential High-Value
Ingredients for the Functional Food Industry. Foods 2022,11, 1744. [CrossRef]
16.
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]
17.
Levasseur, W.; Perré, P.; Pozzobon, V. A review of high value-added molecules production by microalgae in light of the
classification. Biotechnol. Adv. 2020,41, 107545. [CrossRef]
18. Borowitzka, M.A. Chapter 9—Microalgae in Medicine and Human Health: A Historical Perspective. In Microalgae in Health and
Disease Prevention; Levine, I.A., Fleurence, J., Eds.; Academic Press: Cambridge, MA, USA, 2018; pp. 195–210.
19. Riccio, G.; Lauritano, C. Microalgae with Immunomodulatory Activities. Mar. Drugs 2019,18, 2. [CrossRef]
20.
Abu-Ghosh, S.; Dubinsky, Z.; Verdelho, V.; Iluz, D. Unconventional high-value products from microalgae: A review. Bioresour.
Technol. 2021,329, 124895. [CrossRef]
21.
Saide, A.; Martínez, K.A.; Ianora, A.; Lauritano, C. Unlocking the Health Potential of Microalgae as Sustainable Sources of
Bioactive Compounds. Int. J. Mol. Sci. 2021,22, 4383. [CrossRef]
22.
Del Mondo, A.; Smerilli, A.; Sané, E.; Sansone, C.; Brunet, C. Challenging microalgal vitamins for human health. Microb. Cell
Factories 2020,19, 201. [CrossRef] [PubMed]
23.
Novoveská, L.; Nielsen, S.L.; Eroldo˘gan, O.T.; Haznedaroglu, B.Z.; Rinkevich, B.; Fazi, S.; Robbens, J.; Vasquez, M.; Einarsson, H.
Overview and Challenges of Large-Scale Cultivation of Photosynthetic Microalgae and Cyanobacteria. Mar. Drugs
2023
,21, 445.
[CrossRef] [PubMed]
24.
Singh, J.A.; Arayssi, T.; Duray, P.; Schumacher, H.R. Immunohistochemistry of normal human knee synovium: A quantitative
study. Ann. Rheum. Dis. 2004,63, 785–790. [CrossRef] [PubMed]
Mar. Drugs 2023,21, 630 15 of 27
25.
Zhang, F.; Wei, K.; Slowikowski, K.; Fonseka, C.Y.; Rao, D.A.; Kelly, S.; Goodman, S.M.; Tabechian, D.; Hughes, L.B.; Salomon-
Escoto, K.; et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell
transcriptomics and mass cytometry. Nat. Immunol. 2019,20, 928–942. [CrossRef] [PubMed]
26.
Cutolo, M.; Campitiello, R.; Gotelli, E.; Soldano, S. The Role of M1/M2 Macrophage Polarization in Rheumatoid Arthritis
Synovitis. Front. Immunol. 2022,13, 867260. [CrossRef] [PubMed]
27.
Makowski, L.; Chaib, M.; Rathmell, J.C. Immunometabolism: From basic mechanisms to translation. Immunol. Rev.
2020
,
295, 5–14. [CrossRef]
28. McInnes, I.B.; Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011,365, 2205–2219. [CrossRef]
29.
Fukui, S.; Iwamoto, N.; Takatani, A.; Igawa, T.; Shimizu, T.; Umeda, M.; Nishino, A.; Horai, Y.; Hirai, Y.; Koga, T.; et al. M1
and M2 Monocytes in Rheumatoid Arthritis: A Contribution of Imbalance of M1/M2 Monocytes to Osteoclastogenesis. Front.
Immunol. 2017,8, 1958. [CrossRef]
30.
Brown, K.D.; Claudio, E.; Siebenlist, U. The roles of the classical and alternative nuclear factor-
κ
B pathways: Potential implications
for autoimmunity and rheumatoid arthritis. Arthritis Res. Ther. 2008,10, 212. [CrossRef]
31.
Malemud, C.J. The role of the JAK/STAT signal pathway in rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis.
2018
,10, 117–127.
[CrossRef]
32.
Schett, 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]
33.
Komatsu, N.; Takayanagi, H. Mechanisms of joint destruction in rheumatoid arthritis—Immune cell-fibroblast-bone interactions.
Nat. Rev. Rheumatol. 2022,18, 415–429. [CrossRef] [PubMed]
34.
Ono, T.; Hayashi, M.; Sasaki, F.; Nakashima, T. RANKL biology: Bone metabolism, the immune system, and beyond. Inflamm.
Regen. 2020,40, 2. [CrossRef] [PubMed]
35.
Ni, S.; Shan, F.; Geng, J. Interleukin-10 family members: Biology and role in the bone and joint diseases. Int. Immunopharmacol.
2022,108, 108881. [CrossRef] [PubMed]
36.
Müller, R.D.; John, T.; Kohl, B.; Oberholzer, A.; Gust, T.; Hostmann, A.; Hellmuth, M.; Laface, D.; Hutchins, B.; Laube, G.; et al. IL-10
overexpression differentially affects cartilage matrix gene expression in response to TNF-alpha in human articular chondrocytes
in vitro. Cytokine 2008,44, 377–385. [CrossRef] [PubMed]
37.
Sandmann, G. Diversity and origin of carotenoid biosynthesis: Its history of coevolution towards plant photosynthesis. New
Phytol. 2021,232, 479–493. [CrossRef]
38. Takaichi, S. Carotenoids in algae: Distributions, biosyntheses and functions. Mar. Drugs 2011,9, 1101–1118. [CrossRef]
39.
Gao, J.; Wang, H.; Yuan, Q.; Feng, Y. Structure and Function of the Photosystem Supercomplexes. Front. Plant Sci.
2018
,9, 357.
[CrossRef]
40.
Anjani, G.; Ayustaningwarno, F.; Eviana, R. Critical review on the immunomodulatory activities of carrot’s
β
-carotene and other
bioactive compounds. J. Funct. Foods 2022,99, 105303. [CrossRef]
41.
Moia, V.M.; Leal Portilho, F.; Almeida Pádua, T.; Barbosa Corrêa, L.; Ricci-Junior, E.; Cruz Rosas, E.; Magalhaes Rebelo Alencar,
L.; Savio Mendes Sinfronio, F.; Sampson, A.; Hussain Iram, S.; et al. Lycopene used as Anti-inflammatory Nanodrug for the
Treatment of Rheumathoid Arthritis: Animal assay, Pharmacokinetics, ABC Transporter and Tissue Deposition. Colloids Surf. B
Biointerfaces 2020,188, 110814. [CrossRef]
42.
Renju, G.L.; Muraleedhara Kurup, G.; Saritha Kumari, C.H. Anti-inflammatory activity of lycopene isolated from Chlorella
marina on Type II Collagen induced arthritis in Sprague Dawley rats. Immunopharmacol. Immunotoxicol.
2013
,35, 282–291.
[CrossRef] [PubMed]
43.
Caferri, R.; Guardini, Z.; Bassi, R.; Dall’Osto, L. Chapter Two—Assessing photoprotective functions of carotenoids in photosyn-
thetic systems of plants and green algae. In Methods in Enzymology; Wurtzel, E.T., Ed.; Academic Press: Cambridge, MA, USA,
2022; Volume 674, pp. 53–84.
44.
Zhao, K.; Zhou, T.; Yang, J.; Li, Y.; Qin, J.; Wang, S.; Li, D.; Chen, J.; Zheng, W.V. Lutein shows a protective effect against the aging
of mesenchymal stem cells by downregulating inflammation. Int. Immunopharmacol. 2023,116, 109749. [CrossRef]
45.
Pick, U.; Zarka, A.; Boussiba, S.; Davidi, L. A hypothesis about the origin of carotenoid lipid droplets in the green algae Dunaliella
and Haematococcus. Planta 2019,249, 31–47. [CrossRef] [PubMed]
46.
Chekanov, K.; Lukyanov, A.; Boussiba, S.; Aflalo, C.; Solovchenko, A. Modulation of photosynthetic activity and photoprotection
in Haematococcus pluvialis cells during their conversion into haematocysts and back. Photosynth. Res.
2016
,128, 313–323.
[CrossRef] [PubMed]
47.
Bigagli, E.; D’Ambrosio, M.; Cinci, L.; Niccolai, A.; Biondi, N.; Rodolfi, L.; Dos Santos Nascimiento, L.B.; Tredici, M.R.; Luceri, C.
A Comparative In Vitro Evaluation of the Anti-Inflammatory Effects of a Tisochrysis lutea Extract and Fucoxanthin. Mar. Drugs
2021,19, 334. [CrossRef] [PubMed]
48.
Sansone, C.; Pistelli, L.; Del Mondo, A.; Calabrone, L.; Fontana, A.; Noonan, D.M.; Albini, A.; Brunet, C. The Microalgal
Diatoxanthin Inflects the Cytokine Storm in SARS-CoV-2 Stimulated ACE2 Overexpressing Lung Cells. Antioxidants
2022
,
11, 1515. [CrossRef] [PubMed]
49.
Satoh, A.; Tsuji, S.; Okada, Y.; Murakami, N.; Urami, M.; Nakagawa, K.; Ishikura, M.; Katagiri, M.; Koga, Y.; Shirasawa, T.
Preliminary Clinical Evaluation of Toxicity and Efficacy of A New Astaxanthin-rich Haematococcus pluvialis Extract. J. Clin.
Biochem. Nutr. 2009,44, 280–284. [CrossRef]
Mar. Drugs 2023,21, 630 16 of 27
50.
Turck, D.; Castenmiller, J.; de Henauw, S.; Hirsch-Ernst, K.I.; Kearney, J.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.;
Pelaez, C.; et al. Safety of astaxanthin for its use as a novel food in food supplements. EFSA J. 2020,18, e05993.
51.
Valenti, M.T.; Perduca, M.; Romanelli, M.G.; Mottes, M.; Dalle Carbonare, L. A potential role for astaxanthin in the treatment of
bone diseases (Review). Mol. Med. Rep. 2020,22, 1695–1701. [CrossRef]
52.
Donoso, A.; González-Durán, J.; Muñoz, A.A.; González, P.A.; Agurto-Muñoz, C. Therapeutic uses of natural astaxanthin: An
evidence-based review focused on human clinical trials. Pharmacol. Res. 2021,166, 105479. [CrossRef]
53.
Hajizadeh-Sharafabad, F.; Zahabi, E.S.; Malekahmadi, M.; Zarrin, R.; Alizadeh, M. Carotenoids supplementation and inflamma-
tion: A systematic review and meta-analysis of randomized clinical trials. Crit. Rev. Food Sci. Nutr.
2022
,62, 8161–8177. [CrossRef]
[PubMed]
54.
Zhang, L.; Chen, H.; Ding, K.; He, S.; Li, G.; Qu, J.; Qiao, Y.; Zhang, L.; Sui, X.; Fan, C.; et al. Astaxanthin intake alleviates
gouty arthritis in patients and rats by modulating the levels of various inflammatory markers. J. Funct. Foods
2021
,87, 104823.
[CrossRef]
55.
Tamura, H.; Ishikita, H. Quenching of Singlet Oxygen by Carotenoids via Ultrafast Superexchange Dynamics. J. Phys. Chem. A
2020,124, 5081–5088. [CrossRef]
56.
Dose, J.; Matsugo, S.; Yokokawa, H.; Koshida, Y.; Okazaki, S.; Seidel, U.; Eggersdorfer, M.; Rimbach, G.; Esatbeyoglu, T. Free
Radical Scavenging and Cellular Antioxidant Properties of Astaxanthin. Int. J. Mol. Sci. 2016,17, 103. [CrossRef] [PubMed]
57.
Zhang, J.; Sun, Z.; Sun, P.; Chen, T.; Chen, F. Microalgal carotenoids: Beneficial effects and potential in human health. Food Funct.
2014,5, 413–425. [CrossRef] [PubMed]
58.
Yang, Y.; Kim, B.; Lee, J.-Y. Astaxanthin Structure, Metabolism, and Health Benefits. 2014. Available online: https://www.
jscimedcentral.com/public/assets/articles/nutrition-1-1003.pdf (accessed on 15 November 2023).
59.
Fukuzawa, K.; Inokami, Y.; Tokumura, A.; Terao, J.; Suzuki, A. Rate constants for quenching singlet oxygen and activities for
inhibiting lipid peroxidation of carotenoids and α-tocopherol in liposomes. Lipids 1998,33, 751–756. [CrossRef] [PubMed]
60.
Bolin, A.P.; Macedo, R.C.; Marin, D.P.; Barros, M.P.; Otton, R. Astaxanthin prevents
in vitro
auto-oxidative injury in human
lymphocytes. Cell Biol. Toxicol. 2010,26, 457–467. [CrossRef]
61.
Macedo, R.C.; Bolin, A.P.; Marin, D.P.; Otton, R. Astaxanthin addition improves human neutrophils function:
In vitro
study. Eur.
J. Nutr. 2010,49, 447–457. [CrossRef]
62.
Guerra, B.A.; Otton, R. Impact of the carotenoid astaxanthin on phagocytic capacity and ROS/RNS production of human
neutrophils treated with free fatty acids and high glucose. Int. Immunopharmacol. 2011,11, 2220–2226. [CrossRef]
63.
Guerra, B.A.; Bolin, A.P.; Otton, R. Carbonyl stress and a combination of astaxanthin/vitamin C induce biochemical changes in
human neutrophils. Toxicol Vitr. 2012,26, 1181–1190. [CrossRef]
64.
Speranza, L.; Pesce, M.; Patruno, A.; Franceschelli, S.; Lutiis, M.A.d.; Grilli, A.; Felaco, M. Astaxanthin Treatment Reduced
Oxidative Induced Pro-Inflammatory Cytokines Secretion in U937: SHP-1 as a Novel Biological Target. Mar. Drugs
2012
,
10, 890–899. [CrossRef] [PubMed]
65.
Kimble, L.; Mathison, B.; Chew, B.P. Astaxanthin mediates inflammatory biomarkers associated with arthritis in human chon-
drosarcoma cells induced with interleukin-1β.FASEB J. 2013,27, 638.6. [CrossRef]
66.
Mamun-Or-Rashid, A.N.M.; Lucy, T.T.; Yagi, M.; Yonei, Y. Inhibitory Effects of Astaxanthin on CML-HSA-Induced Inflammatory
and RANKL-Induced Osteoclastogenic Gene Expression in RAW 264.7 Cells. Biomedicines 2022,10, 54. [CrossRef] [PubMed]
67.
Kumar, A.; Dhaliwal, N.; Dhaliwal, J.; Dharavath, R.N.; Chopra, K. Astaxanthin attenuates oxidative stress and inflammatory
responses in complete Freund-adjuvant-induced arthritis in rats. Pharmacol. Rep. 2020,72, 104–114. [CrossRef]
68.
Sun, K.; Luo, J.; Jing, X.; Guo, J.; Yao, X.; Hao, X.; Ye, Y.; Liang, S.; Lin, J.; Wang, G.; et al. Astaxanthin protects against osteoarthritis
via Nrf2: A guardian of cartilage homeostasis. Aging 2019,11, 10513–10531. [CrossRef] [PubMed]
69.
Wang, X.; Liu, Z.; Peng, P.; Gong, Z.; Huang, J.; Peng, H. Astaxanthin attenuates osteoarthritis progression via inhibiting
ferroptosis and regulating mitochondrial function in chondrocytes. Chem.-Biol. Interact. 2022,366, 110148. [CrossRef]
70.
Budriesi, R.; Micucci, M.; Daglia, M.; Corazza, I.; Biotti, G.; Mattioli, L.B. Chemical Features and Biological Effects of Astaxanthin
Extracted from Haematococcus pluvialis Flotow: Focus on Gastrointestinal System. Biol. Life Sci. Forum 2022,12, 31.
71.
Régnier, P.; Bastias, J.; Rodriguez-Ruiz, V.; Caballero-Casero, N.; Caballo, C.; Sicilia, D.; Fuentes, A.; Maire, M.; Crepin, M.;
Letourneur, D.; et al. Astaxanthin from Haematococcus pluvialis Prevents Oxidative Stress on Human Endothelial Cells without
Toxicity. Mar. Drugs 2015,13, 2857–2874. [CrossRef]
72.
Capelli, B.; Bagchi, D.; Cysewski, G.R. Synthetic astaxanthin is significantly inferior to algal-based astaxanthin as an antioxidant
and may not be suitable as a human nutraceutical supplement. Nutrafoods 2013,12, 145–152. [CrossRef]
73.
Yang, L.; Qiao, X.; Gu, J.; Li, X.; Cao, Y.; Xu, J.; Xue, C. Influence of molecular structure of astaxanthin esters on their stability and
bioavailability. Food Chem. 2021,343, 128497. [CrossRef]
74.
Madhavi, D.; Kagan, D.; Seshadri, S. A Study on the Bioavailability of a Proprietary, Sustained-release Formulation of Astaxanthin.
Integr. Med. 2018,17, 38–42.
75.
Liu, X.; Xie, J.; Zhou, L.; Zhang, J.; Chen, Z.; Xiao, J.; Cao, Y.; Xiao, H. Recent advances in health benefits and bioavailability of
dietary astaxanthin and its isomers. Food Chem. 2023,404, 134605. [CrossRef] [PubMed]
76.
Jafari, Z.; Bigham, A.; Sadeghi, S.; Dehdashti, S.M.; Rabiee, N.; Abedivash, A.; Bagherzadeh, M.; Nasseri, B.; Karimi-Maleh, H.;
Sharifi, E.; et al. Nanotechnology-Abetted Astaxanthin Formulations in Multimodel Therapeutic and Biomedical Applications. J.
Med. Chem. 2022,65, 2–36. [CrossRef] [PubMed]
Mar. Drugs 2023,21, 630 17 of 27
77.
Li, B.; Lee, J.-Y.; Luo, Y. Health benefits of astaxanthin and its encapsulation for improving bioavailability: A review. J. Agric. Food
Res. 2023,14, 100685. [CrossRef]
78.
Abdol Wahab, N.R.; Meor Mohd Affandi, M.M.R.; Fakurazi, S.; Alias, E.; Hassan, H. Nanocarrier System: State-of-the-Art in Oral
Delivery of Astaxanthin. Antioxidants 2022,11, 1676. [CrossRef] [PubMed]
79.
Chen, S.; Wang, J.; Feng, J.; Xuan, R. Research progress of Astaxanthin nano-based drug delivery system: Applications, prospects
and challenges? Front. Pharmacol. 2023,14, 1102888. [CrossRef] [PubMed]
80.
Hien, H.T.M.; Oanh, H.T.; Quynh, Q.T.; Thu, N.T.H.; Van Hanh, N.; Hong, D.D.; Hoang, M.H. Astaxanthin-loaded nanoparticles
enhance its cell uptake, antioxidant and hypolipidemic activities in multiple cell lines. J. Drug Deliv. Sci. Technol.
2023
,80, 104133.
[CrossRef]
81.
Bai, S.-K.; Lee, S.-J.; Na, H.-J.; Ha, K.-S.; Han, J.-A.; Lee, H.; Kwon, Y.-G.; Chung, C.-K.; Kim, Y.-M.
β
-Carotene inhibits
inflammatory gene expression in lipopolysaccharide-stimulated macrophages by suppressing redox-based NF-
κ
B activation. Exp.
Mol. Med. 2005,37, 323–334. [CrossRef]
82.
Lee, S.J.; Bai, S.K.; Lee, K.S.; Namkoong, S.; Na, H.J.; Ha, K.S.; Han, J.A.; Yim, S.V.; Chang, K.; Kwon, Y.G.; et al. Astaxanthin
inhibits nitric oxide production and inflammatory gene expression by suppressing I(kappa)B kinase-dependent NF-
κ
B activation.
Mol. Cells 2003,16, 97–105.
83.
Priyadarshini, L.; Aggarwal, A. Astaxanthin inhibits cytokines production and inflammatory gene expression by suppressing I
κ
B
kinase-dependent nuclear factor
κ
B activation in pre and postpartum Murrah buffaloes during different seasons. Vet. World
2018
,
11, 782–788. [CrossRef]
84.
Li, R.; Hong, P.; Zheng, X.
β
-carotene attenuates lipopolysaccharide-induced inflammation via inhibition of the NF-
κ
B,
JAK2/STAT3 and JNK/p38 MAPK signaling pathways in macrophages. Anim. Sci. J. 2019,90, 140–148. [CrossRef] [PubMed]
85.
Lee, A.H.; Shin, H.-Y.; Park, J.-H.; Koo, S.Y.; Kim, S.M.; Yang, S.-H. Fucoxanthin from microalgae Phaeodactylum tricornutum
inhibits pro-inflammatory cytokines by regulating both NF-
κ
B and NLRP3 inflammasome activation. Sci. Rep.
2021
,11, 543.
[CrossRef] [PubMed]
86.
Pistelli, L.; Sansone, C.; Smerilli, A.; Festa, M.; Noonan, D.M.; Albini, A.; Brunet, C. MMP-9 and IL-1
β
as Targets for Diatoxanthin
and Related Microalgal Pigments: Potential Chemopreventive and Photoprotective Agents. Mar. Drugs
2021
,19, 354. [CrossRef]
[PubMed]
87.
Vermeulen, L.; De Wilde, G.; Van Damme, P.; Vanden Berghe, W.; Haegeman, G. Transcriptional activation of the NF-
κ
B p65
subunit by mitogen- and stress-activated protein kinase-1 (MSK1). EMBO J. 2003,22, 1313–1324. [CrossRef] [PubMed]
88.
Takeba, Y.; Suzuki, N.; Wakisaka, S.; Takeno, M.; Kaneko, A.; Asai, T.; Sakane, T. Involvement of cAMP responsive element
binding protein (CREB) in the synovial cell hyperfunction in patients with rheumatoid arthritis. Clin. Exp. Rheumatol.
2000
,
18, 47–55. [PubMed]
89.
Wen, A.Y.; Sakamoto, K.M.; Miller, L.S. The role of the transcription factor CREB in immune function. J. Immunol.
2010
,
185, 6413–6419. [CrossRef] [PubMed]
90.
Terazawa, S.; Nakajima, H.; Shingo, M.; Niwano, T.; Imokawa, G. Astaxanthin attenuates the UVB-induced secretion of
prostaglandin E2 and interleukin-8 in human keratinocytes by interrupting MSK1 phosphorylation in a ROS depletion–
independent manner. Exp. Dermatol. 2012,21, 11–17. [CrossRef] [PubMed]
91.
Widyaningrum, D.; Oktafika, R.A.; Cecilia, D. Microalgae pigments as a promising immunomodulating food ingredient: In silico
study. IOP Conf. Ser. Earth Environ. Sci. 2022,998, 012056. [CrossRef]
92.
Pflug, K.M.; Sitcheran, R. Targeting NF-
κ
B-Inducing Kinase (NIK) in Immunity, Inflammation, and Cancer. Int. J. Mol. Sci.
2020
,
21, 8470. [CrossRef]
93.
Zhao, L.; Tao, X.; Wan, C.; Dong, D.; Wang, C.; Xi, Q.; Liu, Y.; Song, T. Astaxanthin alleviates inflammatory pain by regulating the
p38 mitogen-activated protein kinase and nuclear factor-erythroid factor 2-related factor/heme oxygenase-1 pathways in mice.
Food Funct. 2021,12, 12381–12394. [CrossRef]
94.
Yang, Y.; Li, R.; Hui, J.; Li, L.; Zheng, X.
β
-Carotene attenuates LPS-induced rat intestinal inflammation via modulating autophagy
and regulating the JAK2/STAT3 and JNK/p38 MAPK signaling pathways. J. Food Biochem.
2021
,45, e13544. [CrossRef] [PubMed]
95.
Chen, B.; Ning, K.; Sun, M.-l.; Zhang, X.-A. Regulation and therapy, the role of JAK2/STAT3 signaling pathway in OA: A
systematic review. Cell Commun. Signal. 2023,21, 67. [CrossRef] [PubMed]
96.
Tripathi, S.K.; Chen, Z.; Larjo, A.; Kanduri, K.; Nousiainen, K.; Äijo, T.; Ricaño-Ponce, I.; Hrdlickova, B.; Tuomela, S.;
Laajala, E.; et al. Genome-wide Analysis of STAT3-Mediated Transcription during Early Human Th17 Cell Differentiation.
Cell Rep. 2017,19, 1888–1901. [CrossRef] [PubMed]
97.
Maik-Rachline, G.; Zehorai, E.; Hanoch, T.; Blenis, J.; Seger, R. The nuclear translocation of the kinases p38 and JNK promotes
inflammation-induced cancer. Sci. Signal 2018,11, eaao3428. [CrossRef] [PubMed]
98.
Yang, Y.; Kim, S.C.; Yu, T.; Yi, Y.-S.; Rhee, M.H.; Sung, G.-H.; Yoo, B.C.; Cho, J.Y. Functional Roles of p38 Mitogen-Activated
Protein Kinase in Macrophage-Mediated Inflammatory Responses. Mediat. Inflamm. 2014,2014, 352371. [CrossRef] [PubMed]
99.
O’Neil, J.D.; Ammit, A.J.; Clark, A.R. MAPK p38 regulates inflammatory gene expression via tristetraprolin: Doing good by
stealth. Int. J. Biochem. Cell Biol. 2018,94, 6–9. [CrossRef] [PubMed]
100.
Ahn, Y.J.; Kim, H. Lutein as a Modulator of Oxidative Stress-Mediated Inflammatory Diseases. Antioxidants
2021
,10, 1448.
[CrossRef]
Mar. Drugs 2023,21, 630 18 of 27
101.
Kim, E.A.; Kang, N.; Heo, S.Y.; Oh, J.Y.; Lee, S.H.; Cha, S.H.; Kim, W.K.; Heo, S.J. Antioxidant, Antiviral, and Anti-Inflammatory
Activities of Lutein-Enriched Extract of Tetraselmis Species. Mar. Drugs 2023,21, 369. [CrossRef]
102.
Clayton, S.A.; MacDonald, L.; Kurowska-Stolarska, M.; Clark, A.R. Mitochondria as Key Players in the Pathogenesis and
Treatment of Rheumatoid Arthritis. Front. Immunol. 2021,12, 673916. [CrossRef]
103.
Kim, J.; Cha, Y.N.; Surh, Y.J. A protective role of nuclear factor-erythroid 2-related factor-2 (Nrf2) in inflammatory disorders.
Mutat. Res. 2010,690, 12–23. [CrossRef]
104.
Ferrándiz, M.L.; Nacher-Juan, J.; Alcaraz, M.J. Nrf2 as a therapeutic target for rheumatic diseases. Biochem. Pharmacol.
2018
,
152, 338–346. [CrossRef] [PubMed]
105.
Sigaux, J.; Bellicha, A.; Buscail, C.; Julia, C.; Flipo, R.M.; Cantagrel, A.; Laporte, F.; Beal, C.; Boissier, M.C.; Semerano, L. Serum
Fatty Acid Profiles Are Associated with Disease Activity in Early Rheumatoid Arthritis: Results from the ESPOIR Cohort.
Nutrients 2022,14, 2947. [CrossRef] [PubMed]
106.
Mustonen, A.-M.; Nieminen, P. Fatty Acids and Oxylipins in Osteoarthritis and Rheumatoid Arthritis—A Complex Field with
Significant Potential for Future Treatments. Curr. Rheumatol. Rep. 2021,23, 41. [CrossRef] [PubMed]
107.
Hahn, J.; Cook, N.R.; Alexander, E.K.; Friedman, S.; Walter, J.; Bubes, V.; Kotler, G.; Lee, I.-M.; Manson, J.E.; Costenbader, K.H.
Vitamin D and marine omega 3 fatty acid supplementation and incident autoimmune disease: VITAL randomized controlled
trial. BMJ 2022,376, e066452. [CrossRef] [PubMed]
108.
Chen, W.; Li, T.; Du, S.; Chen, H.; Wang, Q. Microalgal polyunsaturated fatty acids: Hotspots and production techniques. Front.
Bioeng. Biotechnol. 2023,11, 1146881. [CrossRef] [PubMed]
109.
Demets, R.; Foubert, I. Chapter 1—Traditional and novel sources of long-chain omega-3 fatty acids. In Omega-3 Delivery Systems;
García-Moreno, P.J., Jacobsen, C., Moltke Sørensen, A.-D., Yesiltas, B., Eds.; Academic Press: Cambridge, MA, USA, 2021;
pp. 3–23.
110.
Fernandes, T.; Cordeiro, N. Microalgae as Sustainable Biofactories to Produce High-Value Lipids: Biodiversity, Exploitation, and
Biotechnological Applications. Mar. Drugs 2021,19, 573. [CrossRef]
111.
Remize, M.; Brunel, Y.; Silva, J.L.; Berthon, J.Y.; Filaire, E. Microalgae n-3 PUFAs Production and Use in Food and Feed Industries.
Mar. Drugs 2021,19, 113. [CrossRef]
112.
Razali, W.A.W.; Pandhal, J. Outdoor pilot-scale cultivation and techno economic assessment of a novel omega-3 eicosapentaenoic
acid over-producing Nannochloropsis oculata strain. Bioresour. Technol. Rep. 2023,24, 10168. [CrossRef]
113.
Brett, M.; Müller-Navarra, D. The role of highly unsaturated fatty acids in aquatic foodweb processes. Freshw. Biol.
1997
,
38, 483–499. [CrossRef]
114.
Hixson, S.M.; Arts, M.T. Climate warming is predicted to reduce omega-3, long-chain, polyunsaturated fatty acid production in
phytoplankton. Glob. Chang. Biol. 2016,22, 2744–2755. [CrossRef]
115.
Wang, T.; Tong, S.; Liu, N.; Li, F.; Wells, M.L.; Gao, K. The fatty acid content of plankton is changing in subtropical coastal waters
as a result of OA: Results from a mesocosm study. Mar. Environ. Res. 2017,132, 51–62. [CrossRef] [PubMed]
116.
Puccinelli, E.; Sardenne, F.; Pecquerie, L.; Fawcett, S.E.; Machu, E.; Soudant, P. Omega-3 Pathways in Upwelling Systems: The
Link to Nitrogen Supply. Front. Mar. Sci. 2021,8, 664601. [CrossRef]
117.
Li-Beisson, Y.; Thelen, J.J.; Fedosejevs, E.; Harwood, J.L. The lipid biochemistry of eukaryotic algae. Progress. Lipid Res.
2019
,
74, 31–68. [CrossRef] [PubMed]
118.
Lopes, D.; Aveiro, S.S.; Conde, T.; Rey, F.; Couto, D.; Melo, T.; Moreira, A.S.P.; Domingues, M.R. Chapter 6—Algal lipids: Structural
diversity, analysis and applications. In Functional Ingredients from Algae for Foods and Nutraceuticals, 2nd ed.; Dominguez, H.,
Pereira, L., Kraan, S., Eds.; Woodhead Publishing: Sawston, UK, 2023; pp. 335–396.
119.
Kugler, A.; Zorin, B.; Didi-Cohen, S.; Sibiryak, M.; Gorelova, O.; Ismagulova, T.; Kokabi, K.; Kumari, P.; Lukyanov, A.; Boussiba, S.; et al.
Long-Chain Polyunsaturated Fatty Acids in the Green Microalga Lobosphaera incisa Contribute to Tolerance to Abiotic Stresses.
Plant Cell Physiol. 2019,60, 1205–1223. [CrossRef] [PubMed]
120.
Rousch, J.M.; Bingham, S.E.; Sommerfeld, M.R. Changes in fatty acid profiles of thermo-intolerant and thermo-tolerant marine
diatoms during temperature stress. J. Exp. Mar. Biol. Ecol. 2003,295, 145–156. [CrossRef]
121.
Conde, T.A.; Zabetakis, I.; Tsoupras, A.; Medina, I.; Costa, M.; Silva, J.; Neves, B.; Domingues, P.; Domingues, M.R. Microalgal
Lipid Extracts Have Potential to Modulate the Inflammatory Response: A Critical Review. Int. J. Mol. Sci.
2021
,22, 9825.
[CrossRef]
122. Khozin-Goldberg, I.; Leu, S.; Boussiba, S. Microalgae as a Source for VLC-PUFA Production. Subcell. Biochem. 2016,86, 471–510.
123.
Taipale, S.; Peltomaa, E.; Salmi, P. Variation in
ω
-3 and
ω
-6 Polyunsaturated Fatty Acids Produced by Different Phytoplankton
Taxa at Early and Late Growth Phase. Biomolecules 2020,10, 559. [CrossRef]
124.
Barta, D.G.; Coman, V.; Vodnar, D.C. Microalgae as sources of omega-3 polyunsaturated fatty acids: Biotechnological aspects.
Algal Res. 2021,58, 102410. [CrossRef]
125.
Lupette, J.; Benning, C. Human health benefits of very-long-chain polyunsaturated fatty acids from microalgae. Biochimie
2020
,
178, 15–25. [CrossRef]
126.
Nauroth, J.M.; Liu, Y.C.; Van Elswyk, M.; Bell, R.; Hall, E.B.; Chung, G.; Arterburn, L.M. Docosahexaenoic acid (DHA) and
docosapentaenoic acid (DPAn-6) algal oils reduce inflammatory mediators in human peripheral mononuclear cells
in vitro
and
paw edema in vivo. Lipids 2010,45, 375–384. [CrossRef] [PubMed]
Mar. Drugs 2023,21, 630 19 of 27
127.
Novichkova, E.; Nayak, S.; Boussiba, S.; Gopas, J.; Zilberg, D.; Khozin-Goldberg, I. Dietary Application of the Microalga Lo-
bosphaera incisa P127 Reduces Severity of Intestinal Inflammation, Modulates Gut-Associated Gene Expression, and Microbiome
in the Zebrafish Model of IBD. Mol. Nutr. Food Res. 2023,67, 2200253. [CrossRef] [PubMed]
128.
Robertson, R.C.; Guihéneuf, F.; Bahar, B.; Schmid, M.; Stengel, D.B.; Fitzgerald, G.F.; Ross, R.P.; Stanton, C. The Anti-Inflammatory
Effect of Algae-Derived Lipid Extracts on Lipopolysaccharide (LPS)-Stimulated Human THP-1 Macrophages. Mar. Drugs
2015
,
13, 5402–5424. [CrossRef] [PubMed]
129.
Morabito, C.; Bournaud, C.; Maës, C.; Schuler, M.; Aiese Cigliano, R.; Dellero, Y.; Maréchal, E.; Amato, A.; Rébeillé, F. The lipid
metabolism in thraustochytrids. Progress Lipid Res. 2019,76, 101007. [CrossRef] [PubMed]
130.
Ngoc Mai, D.T.; Ha, N.C.; Thom, L.T.; Hong, D.D. Initial studies on squalene from some marine microalgae isolated in Vietnam.
Acad. J. Biol. 2013,35, 333–341. [CrossRef]
131.
Chen, X.; He, Y.; Ye, H.; Xie, Y.; Sen, B.; Jiao, N.; Wang, G. Different carbon and nitrogen sources regulated docosahexaenoic acid
(DHA) production of Thraustochytriidae sp. PKU#SW8 through a fully functional polyunsaturated fatty acid (PUFA) synthase
gene (pfaB). Bioresour. Technol. 2020,318, 124273.
132.
Leyton, A.; Shene, C.; Chisti, Y.; Asenjo, J.A. Production of Carotenoids and Phospholipids by Thraustochytrium sp. in Batch and
Repeated-Batch Culture. Mar. Drugs 2022,20, 416. [CrossRef] [PubMed]
133.
Jaritkhuan, S.; Suanjit, S. Species diversity and polyunsaturated fatty acid content of thraustochytrids from fallen mangrove
leaves in Chon Buri province, Thailand. Agric. Nat. Resour. 2018,52, 24–32. [CrossRef]
134.
Dellero, Y.; Cagnac, O.; Rose, S.; Seddiki, K.; Cussac, M.; Morabito, C.; Lupette, J.; Aiese Cigliano, R.; Sanseverino, W.; Kuntz, M.; et al.
Proposal of a new thraustochytrid genus Hondaea gen. nov. and comparison of its lipid dynamics with the closely related
pseudo-cryptic genus Aurantiochytrium. Algal Res. 2018,35, 125–141. [CrossRef]
135.
Olsen, P.M.; Kósa, G.; Klüver, M.; Kohler, A.; Shapaval, V.; Horn, S.J. Production of docosahexaenoic acid from spruce sugars
using Aurantiochytrium limacinum.Bioresour. Technol. 2023,376, 128827. [CrossRef]
136.
Aini, U.N.; Lunprom, S.; Reungsang, A.; Salakkam, A. Docosahexaenoic acid (DHA) production by Aurantiochytrium limacinum
using cassava pulp hydrolysate as an alternative low-cost carbon source. Front. Mar. Sci. 2022,9, 985119. [CrossRef]
137.
Didrihsone, E.; Dubencovs, K.; Grube, M.; Shvirksts, K.; Suleiko, A.; Suleiko, A.; Vanags, J. Crypthecodinium cohnii Growth
and Omega Fatty Acid Production in Mediums Supplemented with Extract from Recycled Biomass. Mar. Drugs
2022
,20, 68.
[CrossRef] [PubMed]
138.
Ding, J.; Fu, Z.; Zhu, Y.; He, J.; Ma, L.; Bu, D. Enhancing docosahexaenoic acid production of Schizochytrium sp. by optimizing
fermentation using central composite design. BMC Biotechnol. 2022,22, 39. [CrossRef] [PubMed]
139.
Patel, A.; Liefeldt, S.; Rova, U.; Christakopoulos, P.; Matsakas, L. Co-production of DHA and squalene by thraustochytrid from
forest biomass. Sci. Rep. 2020,10, 1992. [CrossRef]
140.
Leong, H.Y.; Su, C.-A.; Lee, B.-S.; Lan, J.C.-W.; Law, C.L.; Chang, J.-S.; Show, P.L. Development of Aurantiochytrium limacinum
SR21 cultivation using salt-rich waste feedstock for docosahexaenoic acid production and application of natural colourant in food
product. Bioresour. Technol. 2019,271, 30–36. [CrossRef] [PubMed]
141.
Patel, A.; Rova, U.; Christakopoulos, P.; Matsakas, L. Simultaneous production of DHA and squalene from Aurantiochytrium sp.
grown on forest biomass hydrolysates. Biotechnol. Biofuels 2019,12, 255. [CrossRef]
142.
Allen, K.M.; Habte-Tsion, H.-M.; Thompson, K.R.; Filer, K.; Tidwell, J.H.; Kumar, V. Freshwater microalgae (Schizochytrium sp.) as
a substitute to fish oil for shrimp feed. Sci. Rep. 2019,9, 6178. [CrossRef]
143.
EFSA Panel on Nutrition; Novel Foods and Food Allergens (NDA); Turck, D.; Bohn, T.; Castenmiller, J.; De Henauw, S.; Hirsch-
Ernst, K.I.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; et al. Safety of oil from Schizochytrium sp. (strain ATCC 20889) for use in
infant and follow-on formula as a novel food pursuant to Regulation (EU) 2015/2283. EFSA J. 2022,20, e07083.
144.
Dawczynski, C.; Dittrich, M.; Neumann, T.; Goetze, K.; Welzel, A.; Oelzner, P.; Völker, S.; Schaible, A.M.; Troisi, F.;
Thomas, L.; et al. Docosahexaenoic acid in the treatment of rheumatoid arthritis: A double-blind, placebo-controlled, randomized
cross-over study with microalgae vs. sunflower oil. Clin. Nutr. 2018,37, 494–504. [CrossRef]
145.
Cañavate, J.P.; Armada, I.; Ríos, J.L.; Hachero-Cruzado, I. Exploring occurrence and molecular diversity of betaine lipids across
taxonomy of marine microalgae. Phytochemistry 2016,124, 68–78. [CrossRef]
146.
Hoffmann, D.Y.; Shachar-Hill, Y. Do betaine lipids replace phosphatidylcholine as fatty acid editing hubs in microalgae? Front.
Plant Sci. 2023,14, 1077347. [CrossRef] [PubMed]
147.
Martin, P.; Van Mooy, B.A.S.; Heithoff, A.; Dyhrman, S.T. Phosphorus supply drives rapid turnover of membrane phospholipids
in the diatom Thalassiosira pseudonana. ISME J. 2011,5, 1057–1060. [CrossRef] [PubMed]
148.
Murakami, H.; Nobusawa, T.; Hori, K.; Shimojima, M.; Ohta, H. Betaine Lipid Is Crucial for Adapting to Low Temperature and
Phosphate Deficiency in Nannochloropsis. Plant Physiol. 2018,177, 181–193. [CrossRef] [PubMed]
149.
Oishi, Y.; Otaki, R.; Iijima, Y.; Kumagai, E.; Aoki, M.; Tsuzuki, M.; Fujiwara, S.; Sato, N. Diacylglyceryl-N,N,N-trimethylhomoserine-
dependent lipid remodeling in a green alga, Chlorella kessleri. Commun. Biol. 2022,5, 19. [CrossRef]
150.
Leitner, P.D.; Jakschitz, T.; Gstir, R.; Stuppner, S.; Perkams, S.; Kruus, M.; Trockenbacher, A.; Griesbeck, C.; Bonn, G.K.;
Huber, L.A.; et al. Anti-Inflammatory Extract from Soil Algae Chromochloris zofingiensis Targeting TNFR/NF-
κ
B Signaling at
Different Levels. Cells 2022,11, 1407. [CrossRef]
151. Biringer, R.G. The enzymology of the human prostanoid pathway. Mol. Biol. Rep. 2020,47, 4569–4586. [CrossRef]
Mar. Drugs 2023,21, 630 20 of 27
152.
Di Costanzo, F.; Di Dato, V.; Ianora, A.; Romano, G. Prostaglandins in Marine Organisms: A Review. Mar. Drugs
2019
,17, 428.
[CrossRef]
153.
Blasio, M.; Balzano, S. Fatty Acids Derivatives From Eukaryotic Microalgae, Pathways and Potential Applications. Front. Microbiol.
2021,12, 718933. [CrossRef]
154.
Linares-Maurizi, A.; Reversat, G.; Awad, R.; Bultel-Poncé, V.; Oger, C.; Galano, J.-M.; Balas, L.; Durbec, A.; Bertrand-Michel, J.;
Durand, T.; et al. Bioactive Oxylipins Profile in Marine Microalgae. Mar. Drugs 2023,21, 136. [CrossRef]
155.
Orefice, I.; Romano, G.; Di Dato, V. Chapter Ten—The biosynthesis and metabolism of prostaglandins in microalgae. In Advances
in Botanical Research; Rébeillé, F., Maréchal, E., Eds.; Academic Press: Cambridge, MA, USA, 2022; Volume 101, pp. 375–436.
156.
Di Dato, V.; Orefice, I.; Amato, A.; Fontanarosa, C.; Amoresano, A.; Cutignano, A.; Ianora, A.; Romano, G. Animal-like
prostaglandins in marine microalgae. ISME J. 2017,11, 1722–1726. [CrossRef]
157.
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]
158.
Di Dato, V.; Di Costanzo, F.; Barbarinaldi, R.; Perna, A.; Ianora, A.; Romano, G. Unveiling the presence of biosynthetic pathways
for bioactive compounds in the Thalassiosira rotula transcriptome. Sci. Rep. 2019,9, 9893. [CrossRef] [PubMed]
159.
Vigor, C.; Oger, C.; Reversat, G.; Rocher, A.; Zhou, B.; Linares-Maurizi, A.; Guy, A.; Bultel-Poncé, V.; Galano, J.M.; Vercauteren, J.; et al.
Isoprostanoid Profiling of Marine Microalgae. Biomolecules 2020,10, 1073. [CrossRef] [PubMed]
160.
Lwin, S.; Brooks, J.; Jacobson, R.; Danks, L.; Lundberg, K.; Milne, G.; Morrow, J.; Edwards, J.R. Isoprostane levels are altered in
rheumatoid arthritis and suppress NFκB activity to inhibit osteoclast formation. Bone 2011,48, S136. [CrossRef]
161.
de Los Reyes, C.; Ávila-Román, J.; Ortega, M.J.; de la Jara, A.; García-Mauriño, S.; Motilva, V.; Zubía, E. Oxylipins from the
microalgae Chlamydomonas debaryana and Nannochloropsis gaditana and their activity as TNF-
α
inhibitors. Phytochemistry
2014
,
102, 152–161. [CrossRef]
162.
Ávila-Román, J.; Talero, E.; Rodríguez-Luna, A.; García-Mauriño, S.; Motilva, V. Anti-inflammatory effects of an oxylipin-
containing lyophilised biomass from a microalga in a murine recurrent colitis model. Br. J. Nutr.
2016
,116, 2044–2052. [CrossRef]
163.
Ávila-Román, J.; Talero, E.; de Los Reyes, C.; García-Mauriño, S.; Motilva, V. Microalgae-derived oxylipins decrease inflammatory
mediators by regulating the subcellular location of NFκB and PPAR-γ.Pharmacol. Res. 2018,128, 220–230. [CrossRef]
164. Guiry, M.D. How Many Species of Algae are There? J. Phycol. 2012,48, 1057–1063. [CrossRef]
165.
Serive, B.; Nicolau, E.; Bérard, J.-B.; Kaas, R.; Pasquet, V.; Picot, L.; Cadoret, J.-P. Community analysis of pigment patterns from 37
microalgae strains reveals new carotenoids and porphyrins characteristic of distinct strains and taxonomic groups. PLoS ONE
2017,12, e0171872. [CrossRef]
166.
Saeed, M.U.; Hussain, N.; Shahbaz, A.; Hameed, T.; Iqbal, H.M.N.; Bilal, M. Bioprospecting microalgae and cyanobacteria for
biopharmaceutical applications. J. Basic Microbiol. 2021,62, 1110–1124. [CrossRef]
167.
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.
168.
Wolf, J.; Ross, I.L.; Radzun, K.A.; Jakob, G.; Stephens, E.; Hankamer, B. High-throughput screen for high performance microalgae
strain selection and integrated media design. Algal Res. 2015,11, 313–325. [CrossRef]
169.
Lafarga, T.; Sánchez-Zurano, A.; Morillas-España, A.; Acién-Fernández, F.G. Extremophile microalgae as feedstock for high-value
carotenoids: A review. Int. J. Food Sci. Technol. 2021,56, 4934–4941. [CrossRef]
170.
Montuori, E.; Saggiomo, M.; Lauritano, C. Microalgae from Cold Environments and Their Possible Biotechnological Applications.
Mar. Drugs 2023,21, 292. [CrossRef] [PubMed]
171.
Steinrücken, P.; Erga, S.R.; Mjøs, S.A.; Kleivdal, H.; Prestegard, S.K. Bioprospecting North Atlantic microalgae with fast growth
and high polyunsaturated fatty acid (PUFA) content for microalgae-based technologies. Algal Res.
2017
,26, 392–401. [CrossRef]
[PubMed]
172.
Suh, S.-S.; Hong, J.-M.; Kim, E.J.; Jung, S.W.; Chae, H.; Kim, J.E.; Kim, J.H.; Kim, I.-C.; Kim, S. Antarctic freshwater microalga,
Chloromonas reticulata, suppresses inflammation and carcinogenesis. Int. J. Med. Sci. 2019,16, 189–197. [CrossRef] [PubMed]
173.
Chae, H.J.; Seo, J.B.; Kim, S.H.; Youn, E.J.; Kim, S.; Suh, S.S. Antarctic Freshwater Microalga, Micractinium simplicissimum,
Suppresses Inflammation. J. Nanosci. Nanotechnol. 2021,21, 4098–4103. [CrossRef]
174.
Villegas-Valencia, M.; González-Portela, R.E.; de Freitas, B.B.; Al Jahdali, A.; Romero-Villegas, G.I.; Malibari, R.; Kapoore, R.V.;
Fuentes-Grünewald, C.; Lauersen, K.J. Cultivation of the polyextremophile Cyanidioschyzon merolae 10D during summer
conditions on the coast of the Red Sea and its adaptation to hypersaline sea water. Front. Microbiol.
2023
,14, 1157151. [CrossRef]
175.
Schüler, L.M.; Gangadhar, K.N.; Duarte, P.; Placines, C.; Molina-Márquez, A.M.; Léon-Bañares, R.; Sousa, V.S.; Varela, J.; Barreira,
L. Improvement of carotenoid extraction from a recently isolated, robust microalga, Tetraselmis sp. CTP4 (chlorophyta). Bioprocess
Biosyst. Eng. 2020,43, 785–796. [CrossRef]
176.
Silva, M.; Kamberovic, F.; Uota, S.T.; Kovan, I.-M.; Viegas, C.S.B.; Simes, D.C.; Gangadhar, K.N.; Varela, J.; Barreira, L. Microalgae
as Potential Sources of Bioactive Compounds for Functional Foods and Pharmaceuticals. Appl. Sci. 2022,12, 5877. [CrossRef]
177.
López, G.; Yate, C.; Ramos, F.A.; Cala, M.P.; Restrepo, S.; Baena, S. Production of Polyunsaturated Fatty Acids and Lipids from
Autotrophic, Mixotrophic and Heterotrophic cultivation of Galdieria sp. strain USBA-GBX-832. Sci. Rep.
2019
,9, 10791. [CrossRef]
Mar. Drugs 2023,21, 630 21 of 27
178.
Miyata, M.; Iwata, S.; Mifude, C.K.; Tajima, M.; Kameyama, M.; Ihara, M.; Matsui, T.; Yamagishi, S.I.; Ishitobi, H.; Miyaki, S.; et al.
A Novel Mucidosphaerium sp. Downregulates Inflammatory Gene Expression in Skin and Articular Cells. Altern. Ther. Health Med.
2021,27, 28–34.
179. Kaseda, K.; Mifude, C.K.; Ishitobi, H.; Miyaki, S. Mitochondrial Regulation in the Pathogenic Process of Inflammatory Arthritis
by Microalgal Mucidosphaerium Species. Of 2017,6, 17–19.
180.
Ren, X.; Liu, Y.; Fan, C.; Hong, H.; Wu, W.; Zhang, W.; Wang, Y. Production, Processing, and Protection of Microalgal n-3
PUFA-Rich Oil. Foods 2022,11, 1215. [CrossRef] [PubMed]
181.
Ren, Y.; Sun, H.; Deng, J.; Huang, J.; Chen, F. Carotenoid Production from Microalgae: Biosynthesis, Salinity Responses and Novel
Biotechnologies. Mar. Drugs 2021,19, 713. [CrossRef] [PubMed]
182.
Shi, T.-Q.; Wang, L.-R.; Zhang, Z.-X.; Sun, X.-M.; Huang, H. Stresses as First-Line Tools for Enhancing Lipid and Carotenoid
Production in Microalgae. Front. Bioeng. Biotechnol. 2020,8, 610. [CrossRef]
183.
Sui, Y.; Muys, M.; Van de Waal, D.B.; D’Adamo, S.; Vermeir, P.; Fernandes, T.V.; Vlaeminck, S.E. Enhancement of co-production of
nutritional protein and carotenoids in Dunaliella salina using a two-phase cultivation assisted by nitrogen level and light intensity.
Bioresour. Technol. 2019,287, 121398. [CrossRef]
184.
Yin, F.-W.; Zhang, Y.-T.; Jiang, J.-Y.; Guo, D.-S.; Gao, S.; Gao, Z. Efficient docosahexaenoic acid production by Schizochytrium sp.
via a two-phase pH control strategy using ammonia and citric acid as pH regulators. Process Biochem. 2019,77, 1–7. [CrossRef]
185.
Montero-Lobato, Z.; Vázquez, M.; Navarro, F.; Fuentes, J.L.; Bermejo, E.; Garbayo, I.; Vílchez, C.; Cuaresma, M. Chemically-
Induced Production of Anti-Inflammatory Molecules in Microalgae. Mar. Drugs 2018,16, 478. [CrossRef]
186.
Dawidziuk, A.; Popiel, D.; Luboinska, M.; Grzebyk, M.; Wisniewski, M.; Koczyk, G. Assessing contamination of microalgal
astaxanthin producer Haematococcus cultures with high-resolution melting curve analysis. J. Appl. Genet.
2017
,58, 277–285.
[CrossRef]
187.
Jannel, S.; Caro, Y.; Bermudes, M.; Petit, T. Novel Insights into the Biotechnological Production of Haematococcus pluvialis-Derived
Astaxanthin: Advances and Key Challenges to Allow Its Industrial Use as Novel Food Ingredient. J. Mar. Sci. Eng.
2020
,8, 789.
[CrossRef]
188.
Arora, N.; Philippidis, G.P. Microalgae strain improvement strategies: Random mutagenesis and adaptive laboratory evolution.
Trends Plant Sci. 2021,26, 1199–1200. [CrossRef]
189.
Trovão, M.; Schüler, L.M.; Machado, A.; Bombo, G.; Navalho, S.; Barros, A.; Pereira, H.; Silva, J.; Freitas, F.; Varela, J. Random
Mutagenesis as a Promising Tool for Microalgal Strain Improvement towards Industrial Production. Mar. Drugs
2022
,20, 440.
[CrossRef] [PubMed]
190.
Varunraj, R.; Priyadharshini, U.; Vijay, K.; Balamurugan, S. Adaptive laboratory evolution empowers lipids and biomass
overproduction in Chlorella vulgaris for environmental applications. Environ. Res. 2023,238 Pt 1, 117125. [CrossRef] [PubMed]
191.
Guardini, Z.; Dall’Osto, L.; Barera, S.; Jaberi, M.; Cazzaniga, S.; Vitulo, N.; Bassi, R. High Carotenoid Mutants of Chlorella vulgaris
Show Enhanced Biomass Yield under High Irradiance. Plants 2021,10, 911. [CrossRef] [PubMed]
192.
McQuillan, J.L.; Cutolo, E.A.; Evans, C.; Pandhal, J. Proteomic characterization of a lutein-hyperaccumulating Chlamydomonas
reinhardtii mutant reveals photoprotection-related factors as targets for increasing cellular carotenoid content. Biotechnol. Biofuels
Bioprod. 2023,16, 166. [CrossRef]
193.
Cecchin, M.; Cazzaniga, S.; Martini, F.; Paltrinieri, S.; Bossi, S.; Maffei, M.E.; Ballottari, M. Astaxanthin and eicosapentaenoic acid
production by S4, a new mutant strain of Nannochloropsis gaditana.Microb. Cell Factories 2022,21, 117. [CrossRef]
194.
Schüler, L.M.; Bombo, G.; Duarte, P.; Santos, T.F.; Maia, I.B.; Pinheiro, F.; Marques, J.; Jacinto, R.; Schulze, P.S.C.; Pereira, H.; et al.
Carotenoid biosynthetic gene expression, pigment and n-3 fatty acid contents in carotenoid-rich Tetraselmis striata CTP4 strains
under heat stress combined with high light. Bioresour. Technol. 2021,337, 125385. [CrossRef]
195.
Sun, X.-M.; Ren, L.-J.; Bi, Z.-Q.; Ji, X.-J.; Zhao, Q.-Y.; Jiang, L.; Huang, H. Development of a cooperative two-factor adaptive-
evolution method to enhance lipid production and prevent lipid peroxidation in Schizochytrium sp. Biotechnol. Biofuels
2018
,
11, 65. [CrossRef]
196.
Ren, L.; Sun, X.; Zhang, L.; Huang, H.; Zhao, Q. Exergy analysis for docosahexaenoic acid production by fermentation and strain
improvement by adaptive laboratory evolution for Schizochytrium sp. Bioresour. Technol. 2020,298, 122562. [CrossRef]
197.
Sun, X.M.; Ren, L.J.; Ji, X.J.; Chen, S.L.; Guo, D.S.; Huang, H. Adaptive evolution of Schizochytrium sp. by continuous high oxygen
stimulations to enhance docosahexaenoic acid synthesis. Bioresour. Technol. 2016,211, 374–381. [CrossRef]
198.
Diao, J.; Song, X.; Cui, J.; Liu, L.; Shi, M.; Wang, F.; Zhang, W. Rewiring metabolic network by chemical modulator based
laboratory evolution doubles lipid production in Crypthecodinium cohnii.Metab. Eng. 2019,51, 88–98. [CrossRef] [PubMed]
199.
Meireles, L.A.; Guedes, A.C.; Malcata, F.X. Increase of the yields of eicosapentaenoic and docosahexaenoic acids by the microalga
Pavlova lutheri following random mutagenesis. Biotechnol. Bioeng. 2003,81, 50–55. [CrossRef] [PubMed]
200.
Jebali, A.; Sanchez, M.R.; Hanschen, E.R.; Starkenburg, S.R.; Corcoran, A.A. Trait drift in microalgae and applications for strain
improvement. Biotechnol. Adv. 2022,60, 108034. [CrossRef] [PubMed]
201.
Lin, J.H.; Lee, D.J.; Chang, J.S. Lutein production from biomass: Marigold flowers versus microalgae. Bioresour. Technol.
2015
,
184, 421–428. [CrossRef] [PubMed]
202.
Patel, A.; Rova, U.; Christakopoulos, P.; Matsakas, L. Microalgal lutein biosynthesis: Recent trends and challenges to enhance the
lutein content in microalgal cell factories. Front. Mar. Sci. 2022,9, 1015419. [CrossRef]
Mar. Drugs 2023,21, 630 22 of 27
203.
Dufossé, L. Back to nature, microbial production of pigments and colorants for food use. Adv. Food Nutr. Res.
2022
,102, 93–122.
[PubMed]
204.
Li, C.; Swofford, C.A.; Sinskey, A.J. Modular engineering for microbial production of carotenoids. Metab. Eng. Commun.
2020
,
10, e00118. [CrossRef] [PubMed]
205.
Wang, N.; Peng, H.; Yang, C.; Guo, W.; Wang, M.; Li, G.; Liu, D. Metabolic Engineering of Model Microorganisms for the
Production of Xanthophyll. Microorganisms 2023,11, 1252. [CrossRef]
206.
Seeger, J.; Wendisch, V.F.; Henke, N.A. Extraction and Purification of Highly Active Astaxanthin from Corynebacterium glutamicum
Fermentation Broth. Mar. Drugs 2023,21, 530. [CrossRef]
207.
Kato, Y.; Hasunuma, T. Metabolic Engineering for Carotenoid Production Using Eukaryotic Microalgae and Prokaryotic Cyanobac-
teria. In Carotenoids: Biosynthetic and Biofunctional Approaches; Misawa, N., Ed.; Springer Singapore: Singapore, 2021; pp. 121–135.
208.
Fachet, M.; Witte, C.; Flassig, R.J.; Rihko-Struckmann, L.K.; McKie-Krisberg, Z.; Polle, J.E.W.; Sundmacher, K. Reconstruction and
analysis of a carbon-core metabolic network for Dunaliella salina.BMC Bioinform. 2020,21, 1–9. [CrossRef]
209.
Narang, P.K.; Dey, J.; Mahapatra, S.R.; Roy, R.; Kushwaha, G.S.; Misra, N.; Suar, M.; Raina, V. Genome-based identification and
comparative analysis of enzymes for carotenoid biosynthesis in microalgae. World J. Microbiol. Biotechnol.
2021
,38, 8. [CrossRef]
[PubMed]
210.
Fang, H.; Liu, J.; Ma, R.; Zou, Y.; Ho, S.-H.; Chen, J.; Xie, Y. Functional Characterization of Lycopene
β
- and
ε
-Cyclases from a
Lutein-Enriched Green Microalga Chlorella sorokiniana FZU60. Mar. Drugs 2023,21, 418. [CrossRef] [PubMed]
211.
Grigoriev, I.V.; Hayes, R.D.; Calhoun, S.; Kamel, B.; Wang, A.; Ahrendt, S.; Dusheyko, S.; Nikitin, R.; Mondo, S.J.; Salamov, A.; et al.
PhycoCosm, a comparative algal genomics resource. Nucleic Acids Res. 2020,49, D1004–D1011. [CrossRef] [PubMed]
212.
Zhang, Y.; Ye, Y.; Bai, F.; Liu, J. The oleaginous astaxanthin-producing alga Chromochloris zofingiensis: Potential from production to
an emerging model for studying lipid metabolism and carotenogenesis. Biotechnol. Biofuels 2021,14, 119. [CrossRef] [PubMed]
213.
Roth, M.S.; Cokus, S.J.; Gallaher, S.D.; Walter, A.; Lopez, D.; Erickson, E.; Endelman, B.; Westcott, D.; Larabell, C.A.;
Merchant, S.S.; et al. Chromosome-level genome assembly and transcriptome of the green alga Chromochloris zofingiensis
illuminates astaxanthin production. Proc. Natl. Acad. Sci. USA 2017,114, E4296–E4305. [CrossRef] [PubMed]
214.
Ye, Y.; Huang, J.C. Defining the biosynthesis of ketocarotenoids in Chromochloris zofingiensis.Plant Divers.
2020
,42, 61–66.
[CrossRef] [PubMed]
215.
Kim, M.; Kim, J.; Lee, S.; Khanh, N.; Li, Z.; Polle, J.E.W.; Jin, E. Deciphering the
β
-carotene hyperaccumulation in Dunaliella by
the comprehensive analysis of Dunaliella salina and Dunaliella tertiolecta under high light conditions. Plant Cell Environ.
2023
,
47, 213–229. [CrossRef] [PubMed]
216.
Yeh, T.-J.; Tseng, Y.-F.; Chen, Y.-C.; Hsiao, Y.; Lee, P.-C.; Chen, T.-J.; Chen, C.-Y.; Kao, C.-Y.; Chang, J.-S.; Chen, J.-C.; et al.
Transcriptome and physiological analysis of a lutein-producing alga Desmodesmus sp. reveals the molecular mechanisms for high
lutein productivity. Algal Res. 2017,21, 103–119. [CrossRef]
217.
Velmurugan, A.; Kodiveri Muthukaliannan, G. Genetic manipulation for carotenoid production in microalgae an overview. Curr.
Res. Biotechnol. 2022,4, 221–228. [CrossRef]
218.
Yazdani, M.; Croen, M.G.; Fish, T.L.; Thannhauser, T.W.; Ahner, B.A. Overexpression of native ORANGE (OR) and OR mutant
protein in Chlamydomonas reinhardtii enhances carotenoid and ABA accumulation and increases resistance to abiotic stress. Metab.
Eng. 2021,68, 94–105. [CrossRef]
219.
Kumari, S.; Vira, C.; Lali, A.M.; Prakash, G. Heterologous expression of a mutant Orange gene from Brassica oleracea increases
carotenoids and induces phenotypic changes in the microalga Chlamydomonas reinhardtii.Algal Res. 2020,47, 101871. [CrossRef]
220.
Pivato, M.; Perozeni, F.; Licausi, F.; Cazzaniga, S.; Ballottari, M. Heterologous expression of cyanobacterial Orange Carotenoid
Protein (OCP2) as a soluble carrier of ketocarotenoids in Chlamydomonas reinhardtii.Algal Res.
2021
,55, 102255. [CrossRef]
[PubMed]
221.
Tran, N.T.; Kaldenhoff, R. Metabolic engineering of ketocarotenoids biosynthetic pathway in Chlamydomonas reinhardtii strain
CC-4102. Sci. Rep. 2020,10, 10688. [CrossRef] [PubMed]
222.
Perozeni, F.; Cazzaniga, S.; Baier, T.; Zanoni, F.; Zoccatelli, G.; Lauersen, K.J.; Wobbe, L.; Ballottari, M. Turning a green alga red:
Engineering astaxanthin biosynthesis by intragenic pseudogene revival in Chlamydomonas reinhardtii.Plant Biotechnol. J.
2020
,
18, 2053–2067. [CrossRef] [PubMed]
223.
Amendola, S.; Kneip, J.S.; Meyer, F.; Perozeni, F.; Cazzaniga, S.; Lauersen, K.J.; Ballottari, M.; Baier, T. Metabolic Engineering for
Efficient Ketocarotenoid Accumulation in the Green Microalga Chlamydomonas reinhardtii.ACS Synth. Biol.
2023
,12, 820–831.
[CrossRef] [PubMed]
224.
Hao, T.B.; Lu, Y.; Zhang, Z.H.; Liu, S.F.; Wang, X.; Yang, W.D.; Balamurugan, S.; Li, H.Y. Hyperaccumulation of fucoxanthin by
enhancing methylerythritol phosphate pathway in Phaeodactylum tricornutum.Appl. Microbiol. Biotechnol. 2021,105, 8783–8793.
225.
Cen, S.-Y.; Li, D.-W.; Huang, X.-L.; Huang, D.; Balamurugan, S.; Liu, W.-J.; Zheng, J.-W.; Yang, W.-D.; Li, H.-Y. Crucial
carotenogenic genes elevate hyperaccumulation of both fucoxanthin and
β
-carotene in Phaeodactylum tricornutum.Algal Res.
2022
,
64, 102691. [CrossRef]
226.
Jiang, E.-Y.; Fan, Y.; Phung, N.-V.; Xia, W.-Y.; Hu, G.-R.; Li, F.-L. Overexpression of plastid lipid-associated protein in marine
diatom enhances the xanthophyll synthesis and storage. Front. Microbiol. 2023,14, 1143017. [CrossRef]
Mar. Drugs 2023,21, 630 23 of 27
227.
Seger, M.; Mammadova, F.; Villegas-Valencia, M.; Bastos de Freitas, B.; Chang, C.; Isachsen, I.; Hemstreet, H.; Abualsaud, F.;
Boring, M.; Lammers, P.J.; et al. Engineered ketocarotenoid biosynthesis in the polyextremophilic red microalga Cyanidioschyzon
merolae 10D. Metab. Eng. Commun. 2023,17, e00226. [CrossRef]
228.
Sueishi, Y.; Ishikawa, M.; Yoshioka, D.; Endoh, N.; Oowada, S.; Shimmei, M.; Fujii, H.; Kotake, Y. Oxygen radical absorbance
capacity (ORAC) of cyclodextrin-solubilized flavonoids, resveratrol and astaxanthin as measured with the ORAC-EPR method. J.
Clin. Biochem. Nutr. 2012,50, 127–132. [CrossRef]
229.
Larrea-Alvarez, M.; Purton, S. Multigenic engineering of the chloroplast genome in the green alga Chlamydomonas reinhardtii.
Microbiology 2020,166, 510. [CrossRef] [PubMed]
230.
Albrecht, M.; Takaichi, S.; Steiger, S.; Wang, Z.-Y.; Sandmann, G. Novel hydroxycarotenoids with improved antioxidative
properties produced by gene combination in Escherichia coli.Nat. Biotechnol. 2000,18, 843–846. [CrossRef] [PubMed]
231.
Umeno, D.; Arnold, F.H. Evolution of a pathway to novel long-chain carotenoids. J. Bacteriol.
2004
,186, 1531–1536. [CrossRef]
[PubMed]
232.
Jehliˇcka, J.; Edwards, H.G.; Oren, A. Bacterioruberin and salinixanthin carotenoids of extremely halophilic Archaea and Bacteria:
A Raman spectroscopic study. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2013,106, 99–103. [CrossRef] [PubMed]
233.
Mandelli, F.; Miranda, V.S.; Rodrigues, E.; Mercadante, A.Z. Identification of carotenoids with high antioxidant capacity produced
by extremophile microorganisms. World J. Microbiol. Biotechnol. 2012,28, 1781–1790. [CrossRef] [PubMed]
234.
Grivard, A.; Goubet, I.; Duarte Filho, L.M.S.; Thiéry, V.; Chevalier, S.; de Oliveira-Junior, R.G.; El Aouad, N.; Guedes da Silva
Almeida, J.R.; Sitarek, P.; Quintans-Junior, L.J.; et al. Archaea Carotenoids: Natural Pigments with Unexplored Innovative
Potential. Mar. Drugs 2022,20, 524. [CrossRef] [PubMed]
235.
Yang, Y.; Yatsunami, R.; Ando, A.; Miyoko, N.; Fukui, T.; Takaichi, S.; Nakamura, S. Complete biosynthetic pathway of the
C50 carotenoid bacterioruberin from lycopene in the extremely halophilic archaeon Haloarcula japonica.J. Bacteriol.
2015
,
197, 1614–1623. [CrossRef]
236.
Heider, S.A.E.; Peters-Wendisch, P.; Wendisch, V.F.; Beekwilder, J.; Brautaset, T. Metabolic engineering for the microbial production
of carotenoids and related products with a focus on the rare C50 carotenoids. Appl. Microbiol. Biotechnol.
2014
,98, 4355–4368.
[CrossRef]
237.
Furubayashi, M.; Ikezumi, M.; Takaichi, S.; Maoka, T.; Hemmi, H.; Ogawa, T.; Saito, K.; Tobias, A.V.; Umeno, D. A highly selective
biosynthetic pathway to non-natural C50 carotenoids assembled from moderately selective enzymes. Nat. Commun.
2015
,6, 7534.
[CrossRef]
238.
Li, L.; Furubayashi, M.; Wang, S.; Maoka, T.; Kawai-Noma, S.; Saito, K.; Umeno, D. Genetically engineered biosynthetic pathways
for nonnatural C60 carotenoids using C5-elongases and C50-cyclases in Escherichia coli. Sci. Rep. 2019,9, 2982. [CrossRef]
239.
Jakhwal, P.; Kumar Biswas, J.; Tiwari, A.; Kwon, E.E.; Bhatnagar, A. Genetic and non-genetic tailoring of microalgae for the
enhanced production of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)—A review. Bioresour. Technol.
2022
,
344, 126250. [CrossRef] [PubMed]
240.
Muñoz, C.F.; Südfeld, C.; Naduthodi, M.I.S.; Weusthuis, R.A.; Barbosa, M.J.; Wijffels, R.H.; D’Adamo, S. Genetic engineering of
microalgae for enhanced lipid production. Biotechnol. Adv. 2021,52, 107836. [CrossRef] [PubMed]
241.
Qiu, X.; Xie, X.; Meesapyodsuk, D. Molecular mechanisms for biosynthesis and assembly of nutritionally important very long
chain polyunsaturated fatty acids in microorganisms. Progress Lipid Res. 2020,79, 101047. [CrossRef] [PubMed]
242.
Hulatt, C.J.; Wijffels, R.H.; Posewitz, M.C. The Genome of the Haptophyte Diacronema lutheri (Pavlova lutheri, Pavlovales): A
Model for Lipid Biosynthesis in Eukaryotic Algae. Genome Biol. Evol. 2021,13, evab178. [CrossRef] [PubMed]
243.
Zhou, X.-R.; Robert, S.S.; Petrie, J.R.; Frampton, D.M.F.; Mansour, M.P.; Blackburn, S.I.; Nichols, P.D.; Green, A.G.; Singh, S.P.
Isolation and characterization of genes from the marine microalga Pavlova salina encoding three front-end desaturases involved
in docosahexaenoic acid biosynthesis. Phytochemistry 2007,68, 785–796. [CrossRef] [PubMed]
244.
Vaezi, R.; Napier, J.A.; Sayanova, O. Identification and Functional Characterization of Genes Encoding Omega-3 Polyunsaturated
Fatty Acid Biosynthetic Activities from Unicellular Microalgae. Mar. Drugs 2013,11, 5116–5129. [CrossRef]
245.
Heggeset, T.M.B.; Ertesvåg, H.; Liu, B.; Ellingsen, T.E.; Vadstein, O.; Aasen, I.M. Lipid and DHA-production in Aurantiochytrium
sp.—Responses to nitrogen starvation and oxygen limitation revealed by analyses of production kinetics and global transcriptomes.
Sci. Rep. 2019,9, 19470. [CrossRef]
246.
Leyland, B.; Novichkova, E.; Dolui, A.K.; Jallet, D.; Daboussi, F.; Legeret, B.; Li, Z.; Li-Beisson, Y.; Boussiba, S.; Khozin-Goldberg, I.
Acyl-CoA binding protein is required for lipid droplet degradation in the diatom Phaeodactylum tricornutum.Plant Physiol.
2023
.
[CrossRef]
247.
Muñoz, C.F.; Weusthuis, R.A.; D’Adamo, S.; Wijffels, R.H. Effect of Single and Combined Expression of Lysophosphatidic
Acid Acyltransferase, Glycerol-3-Phosphate Acyltransferase, and Diacylglycerol Acyltransferase on Lipid Accumulation and
Composition in Neochloris oleoabundans.Front. Plant Sci. 2019,10, 1573. [CrossRef]
248.
Zhu, B.-H.; Tu, C.-C.; Shi, H.-P.; Yang, G.-P.; Pan, K.-H. Overexpression of endogenous delta-6 fatty acid desaturase gene enhances
eicosapentaenoic acid accumulation in Phaeodactylum tricornutum.Process Biochem. 2017,57, 43–49. [CrossRef]
249.
Hamilton, M.L.; Haslam, R.P.; Napier, J.A.; Sayanova, O. Metabolic engineering of Phaeodactylum tricornutum for the enhanced
accumulation of omega-3 long chain polyunsaturated fatty acids. Metab. Eng. 2014,22, 3–9. [CrossRef]
250.
Cui, G.-Z.; Ma, Z.; Liu, Y.-J.; Feng, Y.; Sun, Z.; Cheng, Y.; Song, X.; Cui, Q. Overexpression of glucose-6-phosphate dehydrogenase
enhanced the polyunsaturated fatty acid composition of Aurantiochytrium sp. SD116. Algal Res. 2016,19, 138–145. [CrossRef]
Mar. Drugs 2023,21, 630 24 of 27
251.
Han, X.; Zhao, Z.; Wen, Y.; Chen, Z. Enhancement of docosahexaenoic acid production by overexpression of ATP-citrate lyase and
acetyl-CoA carboxylase in Schizochytrium sp. Biotechnol. Biofuels 2020,13, 131. [CrossRef] [PubMed]
252.
Celente, G.d.S.; Rizzetti, T.M.; Sui, Y.; Schneider, R.d.C.d.S. Potential use of microalga Dunaliella salina for bioproducts with
industrial relevance. Biomass Bioenergy 2022,167, 106647.
253.
Shi, H.; Luo, X.; Wu, R.; Yue, X. Production of eicosapentaenoic acid by application of a delta-6 desaturase with the highest ALA
catalytic activity in algae. Microb. Cell Factories 2018,17, 7. [CrossRef]
254.
Sproles, A.E.; Fields, F.J.; Smalley, T.N.; Le, C.H.; Badary, A.; Mayfield, S.P. Recent advancements in the genetic engineering of
microalgae. Algal Res. 2021,53, 102158. [CrossRef]
255.
Banerjee, A.; Ward, V. Production of recombinant and therapeutic proteins in microalgae. Curr. Opin. Biotechnol.
2022
,78, 102784.
[CrossRef] [PubMed]
256.
Arias, C.A.D.; Oliveira, C.F.M.d.; Molino, J.V.D.; Ferreira-Camargo, L.S.; Matsudo, M.C.; Carvalho, J.C.M.d. Production of
Recombinant Biopharmaceuticals in Chlamydomonas reinhardtii.Int. J. Plant Biol. 2023,14, 39–52. [CrossRef]
257.
El-Ayouty, Y.; El-Manawy, I.; Nasih, S.; Hamdy, E.; Kebeish, R. Engineering Chlamydomonas reinhardtii for Expression of
Functionally Active Human Interferon-α.Mol. Biotechnol. 2019,61, 134–144. [CrossRef]
258.
Torres-Tiji, Y.; Fields, F.J.; Yang, Y.; Heredia, V.; Horn, S.J.; Keremane, S.R.; Jin, M.M.; Mayfield, S.P. Optimized production of
a bioactive human recombinant protein from the microalgae Chlamydomonas reinhardtii grown at high density in a fed-batch
bioreactor. Algal Res. 2022,66, 102786. [CrossRef]
259.
Dehghani, J.; Adibkia, K.; Movafeghi, A.; Pourseif, M.M.; Omidi, Y. Designing a new generation of expression toolkits for
engineering of green microalgae; robust production of human interleukin-2. Bioimpacts 2020,10, 259–268. [CrossRef] [PubMed]
260.
Smyth, D.J.; Ren, B.; White, M.P.J.; McManus, C.; Webster, H.; Shek, V.; Evans, C.; Pandhal, J.; Fields, F.; Maizels, R.M.; et al. Oral
delivery of a functional algal-expressed TGF-
β
mimic halts colitis in a murine DSS model. J. Biotechnol.
2021
,340, 1–12. [CrossRef]
[PubMed]
261.
Mathieu-Rivet, E.; Lerouge, P.; Bardor, M. Chlamydomonas reinhardtii: Protein Glycosylation and Production of Biopharmaceuti-
cals. In Chlamydomonas: Biotechnology and Biomedicine; Springer: Berlin/Heidelberg, Germany, 2017.
262.
Van Landuyt, L.; Lonigro, C.; Meuris, L.; Callewaert, N. Customized protein glycosylation to improve biopharmaceutical function
and targeting. Curr. Opin. Biotechnol. 2019,60, 17–28. [CrossRef]
263.
Ramos-Martinez, E.M.; Fimognari, L.; Sakuragi, Y. High-yield secretion of recombinant proteins from the microalga Chlamy-
domonas reinhardtii.Plant Biotechnol. J. 2017,15, 1214–1224. [CrossRef] [PubMed]
264.
Sproles, A.E.; Berndt, A.; Fields, F.J.; Mayfield, S.P. Improved high-throughput screening technique to rapidly isolate Chlamy-
domonas transformants expressing recombinant proteins. Appl. Microbiol. Biotechnol.
2022
,106, 1677–1689. [CrossRef] [PubMed]
265.
Perozeni, F.; Baier, T. Current Nuclear Engineering Strategies in the Green Microalga Chlamydomonas reinhardtii.Life
2023
,13, 1566.
[CrossRef] [PubMed]
266.
McQuillan, J.L.; Berndt, A.J.; Sproles, A.E.; Mayfield, S.P.; Pandhal, J. Novel cis-regulatory elements as synthetic promoters
to drive recombinant protein expression from the Chlamydomonas reinhardtii nuclear genome. New Biotechnol.
2022
,68, 9–18.
[CrossRef]
267.
LaManna, L.; Chou, C.-H.; Lei, H.; Barton, E.R.; Maliga, P. Chloroplast transformation for bioencapsulation and oral delivery
using the immunoglobulin G fragment crystallizable (Fc) domain. Sci. Rep. 2023,13, 18916. [CrossRef]
268.
Carrera-Pacheco, S.E.; Hankamer, B.; Oey, M. Environmental and nuclear influences on microalgal chloroplast gene expression.
Trends Plant Sci. 2023,28, 955–967. [CrossRef]
269.
Dyo, Y.M.; Purton, S. The algal chloroplast as a synthetic biology platform for production of therapeutic proteins. Microbiology
2018,164, 113–121. [CrossRef]
270.
Akram, M.; Khan, M.A.; Ahmed, N.; Bhatti, R.; Pervaiz, R.; Malik, K.; Tahir, S.; Abbas, R.; Ashraf, F.; Ali, Q. Cloning and
expression of an anti-cancerous cytokine: Human IL-29 gene in Chlamydomonas reinhardtii.AMB Express
2023
,13, 23. [CrossRef]
[PubMed]
271.
Taunt, H.N.; Stoffels, L.; Purton, S. Green biologics: The algal chloroplast as a platform for making biopharmaceuticals.
Bioengineered 2018,9, 48–54. [CrossRef] [PubMed]
272.
Yeon, J.; Miller, S.M.; Dejtisakdi, W. New Synthetic Operon Vectors for Expressing Multiple Proteins in the Chlamydomonas
reinhardtii Chloroplast. Genes 2023,14, 368. [CrossRef] [PubMed]
273.
Cutolo, E.A.; Mandalà, G.; Dall’Osto, L.; Bassi, R. Harnessing the Algal Chloroplast for Heterologous Protein Production.
Microorganisms 2022,10, 743. [CrossRef] [PubMed]
274.
Taunt, H.N.; Jackson, H.O.; Gunnarsson, Í.N.; Pervaiz, R.; Purton, S. Accelerating Chloroplast Engineering: A New System for
Rapid Generation of Marker-Free Transplastomic Lines of Chlamydomonas reinhardtii.Microorganisms 2023,11, 1967. [CrossRef]
275.
Cutolo, E.; Tosoni, M.; Barera, S.; Herrera-Estrella, L.; Dall’Osto, L.; Bassi, R. A Phosphite Dehydrogenase Variant with Promis-
cuous Access to Nicotinamide Cofactor Pools Sustains Fast Phosphite-Dependent Growth of Transplastomic Chlamydomonas
reinhardtii.Plants 2020,9, 473. [CrossRef]
276.
Dahlin, L.R.; Guarnieri, M.T. Heterologous expression of phosphite dehydrogenase in the chloroplast or nucleus enables phosphite
utilization and genetic selection in Picochlorum spp. Algal Res. 2022,62, 102604. [CrossRef]
Mar. Drugs 2023,21, 630 25 of 27
277.
Cutolo, E.; Tosoni, M.; Barera, S.; Herrera-Estrella, L.; Dall’Osto, L.; Bassi, R. A chimeric hydrolase-PTXD transgene enables
chloroplast-based heterologous protein expression and non-sterile cultivation of Chlamydomonas reinhardtii.Algal Res.
2021
,
59, 102429. [CrossRef]
278.
Changko, S.; Rajakumar, P.D.; Young, R.E.B.; Purton, S. The phosphite oxidoreductase gene, ptxD as a bio-contained chloroplast
marker and crop-protection tool for algal biotechnology using Chlamydomonas. Appl. Microbiol. Biotechnol.
2020
,104, 675–686.
[CrossRef]
279.
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]
280.
Zuercher, A.W.; Spirig, R.; Baz Morelli, A.; Rowe, T.; Käsermann, F. Next-generation Fc receptor–targeting biologics for autoim-
mune diseases. Autoimmun. Rev. 2019,18, 102366. [CrossRef] [PubMed]
281.
Vanier, G.; Stelter, S.; Vanier, J.; Hempel, F.; Maier, U.G.; Lerouge, P.; Ma, J.; Bardor, M. Alga-Made Anti-Hepatitis B Antibody
Binds to Human FcγReceptors. Biotechnol. J. 2018,13, 1700496. [CrossRef] [PubMed]
282.
Hempel, F.; Maurer, M.; Brockmann, B.; Mayer, C.; Biedenkopf, N.; Kelterbaum, A.; Becker, S.; Maier, U.G. From hybridomas to a
robust microalgal-based production platform: Molecular design of a diatom secreting monoclonal antibodies directed against the
Marburg virus nucleoprotein. Microb. Cell Factories 2017,16, 131. [CrossRef] [PubMed]
283.
Vanier, G.; Hempel, F.; Chan, P.; Rodamer, M.; Vaudry, D.; Maier, U.G.; Lerouge, P.; Bardor, M. Biochemical Characterization
of Human Anti-Hepatitis B Monoclonal Antibody Produced in the Microalgae Phaeodactylum tricornutum.PLoS ONE
2015
,
10, e0139282. [CrossRef] [PubMed]
284.
Lim, S.H.; Kim, K.; Choi, C.-I. Pharmacogenomics of Monoclonal Antibodies for the Treatment of Rheumatoid Arthritis. J. Pers.
Med. 2022,12, 1265. [CrossRef]
285.
Gallaher, S.D.; Craig, R.J.; Ganesan, I.; Purvine, S.O.; McCorkle, S.R.; Grimwood, J.; Strenkert, D.; Davidi, L.; Roth, M.S.;
Jeffers, T.L.; et al. Widespread polycistronic gene expression in green algae. Proc. Natl. Acad. Sci. USA
2021
,118, e2017714118.
[CrossRef]
286.
Tinazzi, E.; Merlin, M.; Bason, C.; Beri, R.; Zampieri, R.; Lico, C.; Bartoloni, E.; Puccetti, A.; Lunardi, C.; Pezzotti, M.; et al.
Plant-Derived Chimeric Virus Particles for the Diagnosis of Primary Sjögren Syndrome. Front. Plant Sci.
2015
,6, 1080. [CrossRef]
287. Clarke, J. Harnessing plant viruses to treat autoimmune diseases. Nat. Rev. Rheumatol. 2020,16, 352. [CrossRef]
288.
Zampieri, R.; Brozzetti, A.; Pericolini, E.; Bartoloni, E.; Gabrielli, E.; Roselletti, E.; Lomonosoff, G.; Meshcheriakova, Y.; Santi, L.;
Imperatori, F.; et al. Prevention and treatment of autoimmune diseases with plant virus nanoparticles. Sci. Adv.
2020
,6, eaaz0295.
[CrossRef]
289.
Malla, A.; Rosales-Mendoza, S.; Phoolcharoen, W.; Vimolmangkang, S. Efficient Transient Expression of Recombinant Proteins
Using DNA Viral Vectors in Freshwater Microalgal Species. Front. Plant Sci. 2021,12, 650820. [CrossRef]
290.
Bañuelos-Hernández, B.; Monreal-Escalante, E.; González-Ortega, O.; Angulo, C.; Rosales-Mendoza, S. Algevir: An Expression
System for Microalgae Based on Viral Vectors. Front. Microbiol. 2017,8, 1100. [CrossRef] [PubMed]
291.
Quispe, C.F.; Esmael, A.; Sonderman, O.; McQuinn, M.; Agarkova, I.; Battah, M.; Duncan, G.A.; Dunigan, D.D.; Smith, T.P.L.; De
Castro, C.; et al. Characterization of a new chlorovirus type with permissive and non-permissive features on phylogenetically
related algal strains. Virology 2017,500, 103–113. [CrossRef] [PubMed]
292.
Esmael, A.; Agarkova, I.V.; Dunigan, D.D.; Zhou, Y.; Van Etten, J.L. Viral DNA Accumulation Regulates Replication Efficiency of
Chlorovirus OSy-NE5 in Two Closely Related Chlorella variabilis Strains. Viruses 2023,15, 1341. [CrossRef] [PubMed]
293.
Nelson, D.R.; Hazzouri, K.M.; Lauersen, K.J.; Jaiswal, A.; Chaiboonchoe, A.; Mystikou, A.; Fu, W.; Daakour, S.; Dohai, B.;
Alzahmi, A.; et al. Large-scale genome sequencing reveals the driving forces of viruses in microalgal evolution. Cell Host Microbe
2021,29, 250–266. [CrossRef] [PubMed]
294.
Wilson, W.H.; Van Etten, J.L.; Allen, M.J. The Phycodnaviridae: The story of how tiny giants rule the world. Curr. Top. Microbiol.
Immunol. 2009,328, 1–42. [PubMed]
295.
D’Adamo, S.; Kormelink, R.; Martens, D.; Barbosa, M.J.; Wijffels, R.H. Prospects for viruses infecting eukaryotic microalgae in
biotechnology. Biotechnol. Adv. 2021,54, 107790. [CrossRef] [PubMed]
296.
Bolaños-Martínez, O.C.; Mahendran, G.; Rosales-Mendoza, S.; Vimolmangkang, S. Current Status and Perspective on the Use of
Viral-Based Vectors in Eukaryotic Microalgae. Mar. Drugs 2022,20, 434. [CrossRef]
297.
Fernández, F.G.A.; Reis, A.; Wijffels, R.H.; Barbosa, M.; Verdelho, V.; Llamas, B. The role of microalgae in the bioeconomy. New
Biotechnol. 2021,61, 99–107. [CrossRef]
298.
Fabris, M.; Abbriano, R.M.; Pernice, M.; Sutherland, D.L.; Commault, A.S.; Hall, C.C.; Labeeuw, L.; McCauley, J.I.; Kuzhiuparambil,
U.; Ray, P.; et al. Emerging Technologies in Algal Biotechnology: Toward the Establishment of a Sustainable, Algae-Based
Bioeconomy. Front. Plant Sci. 2020,11, 279. [CrossRef]
299.
Rumin, J.; Nicolau, E.; Junior, R.G.O.; Fuentes-Grünewald, C.; Picot, L. Analysis of Scientific Research Driving Microalgae Market
Opportunities in Europe. Mar. Drugs 2020,18, 264. [CrossRef]
300.
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]
301.
Kapoor, S.; Nailwal, N.; Kumar, M.; Barve, K. Recent Patents and Discovery of Anti-inflammatory Agents from Marine Source.
Recent. Pat. Inflamm. Allergy Drug Discov. 2019,13, 105–114. [CrossRef] [PubMed]
Mar. Drugs 2023,21, 630 26 of 27
302.
Tzima, S.; Georgiopoulou, I.; Louli, V.; Magoulas, K. Recent Advances in Supercritical CO
2
Extraction of Pigments, Lipids and
Bioactive Compounds from Microalgae. Molecules 2023,28, 1410. [CrossRef] [PubMed]
303.
Karan, H.; Roles, J.; Hankamer, B.; Ross, I.L. Targeting greens and yellows: A solar biorefinery analysis for the microalgae-based
co-production of pigments, proteins, and fuel. Algal Res. 2023,74, 103187. [CrossRef]
304.
Kholany, M.; Coutinho, J.A.P.; Ventura, S.P.M. Carotenoid Production from Microalgae: The Portuguese Scenario. Molecules
2022
,
27, 2540. [CrossRef]
305.
Zhou, J.; Wang, M.; Saraiva, J.A.; Martins, A.P.; Pinto, C.A.; Prieto, M.A.; Simal-Gandara, J.; Cao, H.; Xiao, J.; Barba, F.J. Extraction
of lipids from microalgae using classical and innovative approaches. Food Chem. 2022,384, 132236. [CrossRef]
306.
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]
307.
Rumin, J.; Junior, R.G.d.O.; Bérard, J.-B.; Picot, L. Improving Microalgae Research and Marketing in the European Atlantic Area:
Analysis of Major Gaps and Barriers Limiting Sector Development. Mar. Drugs 2021,19, 319. [CrossRef]
308.
Mendes, M.C.; Navalho, S.; Ferreira, A.; Paulino, C.; Figueiredo, D.; Silva, D.; Gao, F.; Gama, F.; Bombo, G.; Jacinto, R.; et al. Algae
as Food in Europe: An Overview of Species Diversity and Their Application. Foods 2022,11, 1871. [CrossRef]
309.
Spicer, A.; Molnar, A. Gene Editing of Microalgae: Scientific Progress and Regulatory Challenges in Europe. Biology
2018
,7, 21.
[CrossRef]
310.
Vilatte, A.; Spencer-Milnes, X.; Jackson, H.O.; Purton, S.; Parker, B. Spray Drying Is a Viable Technology for the Preservation of
Recombinant Proteins in Microalgae. Microorganisms 2023,11, 512. [CrossRef] [PubMed]
311.
Commission, E.; Centre, J.R.; Araújo, R.; Peteiro, C. Algae as Food and Food Supplements in Europe; Publications Office: Luxembourg, 2021.
312.
Rzymski, P.; Budzulak, J.; Niedzielski, P.; Klimaszyk, P.; Proch, J.; Kozak, L.; Poniedziałek, B. Essential and toxic elements in
commercial microalgal food supplements. J. Appl. Phycol. 2019,31, 3567–3579. [CrossRef]
313.
Muys, M.; Sui, Y.; Schwaiger, B.; Lesueur, C.; Vandenheuvel, D.; Vermeir, P.; Vlaeminck, S.E. High variability in nutritional value
and safety of commercially available Chlorella and Spirulina biomass indicates the need for smart production strategies. Bioresour.
Technol. 2019,275, 247–257. [CrossRef] [PubMed]
314.
Rzymski, P.; Niedzielski, P.; Kaczmarek, N.; Jurczak, T.; Klimaszyk, P. The multidisciplinary approach to safety and toxicity
assessment of microalgae-based food supplements following clinical cases of poisoning. Harmful Algae
2015
,46, 34–42. [CrossRef]
315.
Ferreira de Oliveira, A.P.; Bragotto, A.P.A. Microalgae-based products: Food and public health. Future Foods
2022
,6, 100157.
[CrossRef]
316.
Molino, A.; Iovine, A.; Casella, P.; Mehariya, S.; Chianese, S.; Cerbone, A.; Rimauro, J.; Musmarra, D. Microalgae Characterization
for Consolidated and New Application in Human Food, Animal Feed and Nutraceuticals. Int. J. Environ. Res. Public. Health
2018
,
15, 2436. [CrossRef]
317.
McInnes, R.S.; McCallum, G.E.; Lamberte, L.E.; van Schaik, W. Horizontal transfer of antibiotic resistance genes in the human gut
microbiome. Curr. Opin. Microbiol. 2020,53, 35–43. [CrossRef]
318.
Ljubic, A.; Jacobsen, C.; Holdt, S.L.; Jakobsen, J. Microalgae Nannochloropsis oceanica as a future new natural source of vitamin
D(3). Food Chem. 2020,320, 126627. [CrossRef]
319.
Ljubic, A.; Thulesen, E.T.; Jacobsen, C.; Jakobsen, J. UVB exposure stimulates production of vitamin D3 in selected microalgae.
Algal Res. 2021,59, 102472. [CrossRef]
320.
Mateen, S.; Moin, S.; Shahzad, S.; Khan, A.Q. Level of inflammatory cytokines in rheumatoid arthritis patients: Correlation with
25-hydroxy vitamin D and reactive oxygen species. PLoS ONE 2017,12, e0178879. [CrossRef]
321.
Athanassiou, L.; Kostoglou-Athanassiou, I.; Koutsilieris, M.; Shoenfeld, Y. Vitamin D and Autoimmune Rheumatic Diseases.
Biomolecules 2023,13, 709. [CrossRef] [PubMed]
322.
Kühn, J.; Brandsch, C.; Kiourtzidis, M.; Nier, A.; Bieler, S.; Matthäus, B.; Griehl, C.; Stangl, G.I. Microalgae-derived sterols do not
reduce the bioavailability of oral vitamin D(3) in mice. Int. J. Vitam. Nutr. Res. 2022. [CrossRef] [PubMed]
323.
Wang, L.; Jeon, Y.-J.; Kim, J.-I.
In vitro
and
in vivo
anti-inflammatory activities of a sterol-enriched fraction from freshwater green
alga, Spirogyra sp. Fish. Aquat. Sci. 2020,23, 27. [CrossRef]
324.
Fagundes, M.B.; Vendruscolo, R.G.; Wagner, R. Chapter 21—Sterols from microalgae. In Handbook of Microalgae-Based Processes and
Products; Jacob-Lopes, E., Maroneze, M.M., Queiroz, M.I., Zepka, L.Q., Eds.; Academic Press: Cambridge, MA, USA, 2020.
325.
Randhir, A.; Laird, D.W.; Maker, G.; Trengove, R.; Moheimani, N.R. Microalgae: A potential sustainable commercial source of
sterols. Algal Res. 2020,46, 101772. [CrossRef]
326.
Cárdeno, A.; Aparicio-Soto, M.; Montserrat-de la Paz, S.; Bermudez, B.; Muriana, F.J.G.; Alarcón-de-la-Lastra, C. Squalene targets
pro- and anti-inflammatory mediators and pathways to modulate over-activation of neutrophils, monocytes and macrophages. J.
Funct. Foods 2015,14, 779–790. [CrossRef]
327.
Fan, K.W.; Aki, T.; Chen, F.; Jiang, Y. Enhanced production of squalene in the thraustochytrid Aurantiochytrium mangrovei by
medium optimization and treatment with terbinafine. World J. Microbiol. Biotechnol. 2010,26, 1303–1309. [CrossRef]
328.
Hong, W.K.; Heo, S.Y.; Park, H.M.; Kim, C.H.; Sohn, J.H.; Kondo, A.; Seo, J.W. Characterization of a squalene synthase from the
thraustochytrid microalga Aurantiochytrium sp. KRS101. J. Microbiol. Biotechnol. 2013,23, 759–765. [CrossRef]
329.
Nakazawa, A.; Matsuura, H.; Kose, R.; Kato, S.; Honda, D.; Inouye, I.; Kaya, K.; Watanabe, M.M. Optimization of culture
conditions of the thraustochytrid Aurantiochytrium sp. strain 18W-13a for squalene production. Bioresour. Technol.
2012
,
109, 287–291. [CrossRef]
Mar. Drugs 2023,21, 630 27 of 27
330.
Okada, S.; Devarenne, T.P.; Chappell, J. Molecular Characterization of Squalene Synthase from the Green Microalga Botryococcus
braunii, Race B. Arch. Biochem. Biophys. 2000,373, 307–317. [CrossRef]
331.
Duan, R.; Pan, X.; Li, K.; Yang, Q.; Cui, X.; Zheng, Y.; Lu, Y.; Yao, C.; Ling, X. Metabolism balance regulation for squalene
production by disturbing triglyceride (TAG) synthesis in Schizochytrium sp. Algal Res. 2023,69, 102946. [CrossRef]
332.
EFSA Panel on Nutrition, Novel Foods; Food Allergens (NDA); Turck, D.; Castenmiller, J.; De Henauw, S.; Hirsch-Ernst, K.I.;
Kearney, J.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; et al. Safety of dried whole cell Euglena gracilis as a novel food pursuant to
Regulation (EU) 2015/2283. EFSA J. 2020,18, e06100. [PubMed]
333.
Yao, R.; Fu, W.; Du, M.; Chen, Z.-X.; Lei, A.-P.; Wang, J.-X. Carotenoids Biosynthesis, Accumulation, and Applications of a Model
Microalga Euglenagracilis.Mar. Drugs 2022,20, 496. [CrossRef] [PubMed]
334.
Piovan, A.; Filippini, R.; Corbioli, G.; Costa, V.D.; Giunco, E.M.V.; Burbello, G.; Pagetta, A.; Giusti, P.; Zusso, M. Carotenoid
Extract Derived from Euglena gracilis Overcomes Lipopolysaccharide-Induced Neuroinflammation in Microglia: Role of NF-
κ
B
and Nrf2 Signaling Pathways. Mol. Neurobiol. 2021,58, 3515–3528. [CrossRef] [PubMed]
335.
Brun, P.; Piovan, A.; Caniato, R.; Dalla Costa, V.; Pauletto, A.; Filippini, R. Anti-Inflammatory Activities of Euglena gracilis Extracts.
Microorganisms 2021,9, 2058. [CrossRef] [PubMed]
336.
Feuzing, F.; Mbakidi, J.P.; Marchal, L.; Bouquillon, S.; Leroy, E. A review of paramylon processing routes from microalga biomass
to non-derivatized and chemically modified products. Carbohydr. Polym. 2022,288, 119181. [CrossRef] [PubMed]
337.
Suzuki, K.; Nakashima, A.; Igarashi, M.; Saito, K.; Konno, M.; Yamazaki, N.; Takimoto, H. Euglena gracilis Z and its carbohydrate
storage substance relieve arthritis symptoms by modulating Th17 immunity. PLoS ONE
2018
,13, e0191462. [CrossRef] [PubMed]
338.
Dittmann, E.; Gugger, M.; Sivonen, K.; Fewer, D.P. Natural Product Biosynthetic Diversity and Comparative Genomics of the
Cyanobacteria. Trends Microbiol. 2015,23, 642–652. [CrossRef]
339.
Pagels, F.; Guedes, A.C.; Vasconcelos, V.; Lopes, G. Chapter 4—Anti-inflammatory compounds from cyanobacteria. In The
Pharmacological Potential of Cyanobacteria; Lopes, G., Silva, M., Vasconcelos, V., Eds.; Academic Press: Cambridge, MA, USA, 2022;
pp. 81–105.
340.
Wu, Q.; Liu, L.; Miron, A.; Klímová, B.; Wan, D.; Kuˇca, K. The antioxidant, immunomodulatory, and anti-inflammatory activities
of Spirulina: An overview. Arch. Toxicol. 2016,90, 1817–1840. [CrossRef]
341.
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]
342.
Jensen, G.S.; Attridge, V.L.; Beaman, J.L.; Guthrie, J.; Ehmann, A.; Benson, K.F. Antioxidant and anti-inflammatory properties of
an aqueous cyanophyta extract derived from Arthrospira platensis: Contribution to bioactivities by the non-phycocyanin aqueous
fraction. J. Med. Food 2015,18, 535–541. [CrossRef]
343.
Ngatu, N.R.; Okajima, M.K.; Yokogawa, M.; Hirota, R.; Eitoku, M.; Muzembo, B.A.; Dumavibhat, N.; Takaishi, M.; Sano, S.;
Kaneko, T.; et al. Anti-inflammatory effects of sacran, a novel polysaccharide from Aphanothece sacrum, on 2,4,6-
trinitrochlorobenzene–induced allergic dermatitis
in vivo
.Ann. Allergy Asthma Immunol.
2012
,108, 117–122. [CrossRef]
[PubMed]
344.
Motoyama, K.; Tanida, Y.; Hata, K.; Hayashi, T.; Hashim, I.I.A.; Higashi, T.; Ishitsuka, Y.; Kondo, Y.; Irie, T.; Kaneko, S.; et al.
Anti-inflammatory Effects of Novel Polysaccharide Sacran Extracted from Cyanobacterium Aphanothece sacrum in Various
Inflammatory Animal Models. Biol. Pharm. Bull. 2016,39, 1172–1178. [CrossRef] [PubMed]
345.
Zampieri, R.M.; Adessi, A.; Caldara, F.; Codato, A.; Furlan, M.; Rampazzo, C.; De Philippis, R.; La Rocca, N.; Dalla Valle, L.
Anti-Inflammatory Activity of Exopolysaccharides from Phormidium sp. ETS05, the Most Abundant Cyanobacterium of the
Therapeutic Euganean Thermal Muds, Using the Zebrafish Model. Biomolecules 2020,10, 582. [CrossRef] [PubMed]
346.
Vo, T.-S.; Ryu, B.; Kim, S.-K. Purification of novel anti-inflammatory peptides from enzymatic hydrolysate of the edible microalgal
Spirulina maxima. J. Funct. Foods 2013,5, 1336–1346. [CrossRef]
347.
Kirk, R.D.; He, H.; Wahome, P.G.; Wu, S.; Carter, G.T.; Bertin, M.J. New Micropeptins with Anti-Neuroinflammatory Activity
Isolated from a Cyanobacterial Bloom. ACS Omega 2021,6, 15472–15478. [CrossRef]
348.
Kirk, R.D.; Picard, K.; Christian, J.A.; Johnson, S.L.; DeBoef, B.; Bertin, M.J. Unnarmicin D, an Anti-inflammatory Cyanobacterial
Metabolite with
δ
and
µ
Opioid Binding Activity Discovered via a Pipeline Approach Designed to Target Neurotherapeutics.
ACS Chem. Neurosci. 2020,11, 4478–4488. [CrossRef] [PubMed]
349.
Santos-Merino, M.; Singh, A.K.; Ducat, D.C. New Applications of Synthetic Biology Tools for Cyanobacterial Metabolic Engineer-
ing. Front. Bioeng. Biotechnol. 2019,7, 33. [CrossRef]
350.
Jester, B.W.; Zhao, H.; Gewe, M.; Adame, T.; Perruzza, L.; Bolick, D.T.; Agosti, J.; Khuong, N.; Kuestner, R.; Gamble, C.; et al.
Development of spirulina for the manufacture and oral delivery of protein therapeutics. Nat. Biotechnol.
2022
,40, 956–964.
[CrossRef]
351.
Betterle, N.; Hidalgo Martinez, D.; Melis, A. Cyanobacterial Production of Biopharmaceutical and Biotherapeutic Proteins. Front.
Plant Sci. 2020,11, 237. [CrossRef]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... 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 [163,164]), which is amenable to fermentation at a large scale in seawater and wastewater [165]. Notably, the anti-inflammatory properties of DHA compounds were recently reported in a clinical trial involving patients with rheumatoid arthritis [166]. ...
... 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 [167]. Microalgae comprise a vast group of photosynthetic microbes producing a huge repertoire of anti-inflammatory and immunomodulatory pigments and lipids [163,[168][169][170][171]. ...
... Carotenoids are lipophilic pigments [172][173][174][175] designated as carotenes (lycopene and α-and β-carotene), which contribute to light-harvesting, and the oxygenated derivatives xanthophylls (or ketocarotenoids: astaxanthin, fucoxanthin and lutein), which are mainly involved in the detoxification of reactive oxidative species (ROS) generated by photosynthetic reactions. The ketocarotenoid astaxanthin is the microalgal pigment of greatest pharmacological value, being endowed with strong antioxidant capacity (extensively reviewed in [163]). The biological production of astaxanthin, however, is restrained by the slow growth of its native producer, the freshwater Chlorophyte Haematococcus lacustris (previously named Haematococcus pluvialis) [176]. ...
Article
Full-text available
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 can result in improved plant vigor and resilience to environmental stresses. 51 Paramylon has been reported to enhance water uptake and retention in plants, helping them maintain optimal hydration levels for growth and physiological functions. This can be particularly beneficial during periods of drought or water stress. ...
Article
Full-text available
Paramylon, a β-1,3-glucan polysaccharide found in microalgae like Euglena gracilis, has attracted significant interest in nutrition, health, and agriculture due to its diverse applications. It is highly adaptable, utilizing nutrients from wastewater, CO2 vaporization, or food waste, with waste-based cultivation yielding 1.77 g/L/day. Paramylon influences plant physiology, enhancing hormone content, photosynthetic efficiency, and dehydration tolerance. For example, during paramylon treatment, the photosynthetic efficiency of Solanum lycopersicum increased to 75%, compared to 50% in controls, while drought resistance rose to 80%, significantly higher than the 60% observed in untreated plants. It also improves water-use efficiency and regulates CO2 diffusion, offering protection against abiotic stresses such as drought, salinity, nutrient deficiencies, and extreme temperatures. As a sustainable alternative to traditional fertilizers, paramylon functions effectively as a biostimulant, enhancing crop yield and quality while increasing plant tolerance to environmental stress. Despite its promising potential, the precise molecular mechanisms and long-term effects on different crops and soil ecosystems remain underexplored. Future research should focus on optimizing paramylon formulations to enhance bioavailability, exploring its synergistic effects with other biostimulants, and assessing its economic feasibility and scalability for commercial adoption. Additionally, studying its impact on soil microbiota and plant-microbe interactions could reveal broader ecological benefits. This review highlights paramylon's potential as a sustainable fertilizer alternative to improve agricultural productivity and crop quality. While current findings are promising, further interdisciplinary research is essential to unlock its full potential in modern agricultural systems.
... Simultaneous biofixation of both carbon dioxide and nitrogen oxides has been shown by the microalgae species Chlorella vulgaris Beijerinck, Haematococcus pluvialis (Flotow) R. A. Saunders and Scenedesmus subspicatus Kützing [197]. Advances in medical treatments have been proposed for microalgae, including in patients with rheumatoid arthritis [198] and in hypoxic cancers [199], to rescue neuronal activity in the brain [200] and to improve the healing of diabetic ulcers [201]. These examples of current microalgae studies show their potential to provide technological support and cross-funding to support improved nutritional support and clinical interventions using selected microalgae. ...
Article
Full-text available
Undernutrition is an important global health problem, especially in children and older adults. Both reversal of maternal and child undernutrition and heathy ageing have become United Nations-supported global initiatives, leading to increased attention to nutritional interventions targeting undernutrition. One feasible option is microalgae, the precursor of all terrestrial plants. Most commercially farmed microalgae are photosynthetic single-celled organisms producing organic carbon compounds and oxygen. This review will discuss commercial opportunities to grow microalgae. Microalgae produce lipids (including omega-3 fatty acids), proteins, carbohydrates, pigments and micronutrients and so can provide a suitable and underutilised alternative for addressing undernutrition. The health benefits of nutrients derived from microalgae have been identified, and thus they are suitable candidates for addressing nutritional issues globally. This review will discuss the potential benefits of microalgae-derived nutrients and opportunities for microalgae to be converted into food products. The advantages of microalgae cultivation include that it does not need arable land or pesticides. Additionally, most species of microalgae are still unexplored, presenting options for further development. Further, the usefulness of microalgae for other purposes such as bioremediation and biofuels will increase the knowledge of these microorganisms, allowing the development of more efficient production of these microalgae as nutritional interventions.
... Microalgae are a broad microorganism category including nearly 2×10 6 species that comprise unicellular eukaryotic photosynthetic and prokaryotic cyanobacteria [8]. Photosynthetic microalgae consume atmospheric CO 2 and light energy to produce a wide range of biomolecules Abbreviations: G3P, Glyceraldehyde 3-phosphate; DOXP, 1-deoxyd-xylulose-5-phosphate; MEP, methylerythritol 4-phosphate; CDPME, 4-(cytidine 5-diphospho)-2-C-methyl-D-eriyhrotol; CDP-MEP, 2-phospho-4-(cytidine 5-diphospho)-2-C-methyl-D-erythritol; ME-cDP, 2-C-methyl-D-erythritol 2 4-cyclodiphosphate; HMBPP, 1-hydroxy-2-methyl-2-butenyl 4-diphosphate; IPP, isopentenyl pyrophosphate; DMAPP, dimethylallyl pyrophosphate; GGPP, geranylgeranyl pyrophosphate. ...
Article
Full-text available
Systems biology is an interdisciplinary field that aims to understand complex biological processes at the system level. The data, driven by high-throughput omics technologies, can be used to study the underpinning mechanisms of metabolite production under different conditions to harness this knowledge for the construction of regulatory networks, protein networks, metabolic models, and engineering of strains with enhanced target metabolite production in microalgae. In the current study, we comprehensively reviewed the recent progress in the application of these technologies for the characterization of carotenoid biosynthesis pathways in microalgae. Moreover, harnessing integrated approaches such as network analysis, meta-analysis, and machine learning models for deciphering the complexity of carotenoid biosynthesis pathways were comprehensively discussed.
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
Worldwide, osteoarthritis (OA) is regarded as the most widespread, distressing, and limiting chronic disease that affects degenerative joints. Currently, there is no treatment available to modify the progression of OA. The pathogenesis of OA is significantly linked with oxidative stress and pyroptosis. Astaxanthin (Ast) is a natural ketocarotenoid pigment with potent antioxidant activity and is shown to effectively alleviate cartilage damage in OA. However, its bioavailability is greatly limited due to poor water solubility, high sensitivity to light, temperature, and pH. In this study, Ast‐loaded tetrahedral framework nucleic acids (tFNAs) or tFNA/Ast complexes (TAC) for Ast delivery are developed. Compared with free Ast and tFNA alone, TAC exhibits improved drug stability and cellular uptake. Most importantly, TAC effectively protects chondrocytes against oxidative stress‐induced pyroptosis while promoting extracellular matrix anabolism by chondrocytes, and ultimately alleviates cartilage damage in a mouse destabilization of the medial meniscus (DMM) model. Thus, TAC holds great promise for the treatment of OA patients.
Article
Full-text available
Background Microalgae are emerging hosts for the sustainable production of lutein, a high-value carotenoid; however, to be commercially competitive with existing systems, their capacity for lutein sequestration must be augmented. Previous attempts to boost microalgal lutein production have focussed on upregulating carotenoid biosynthetic enzymes, in part due to a lack of metabolic engineering targets for expanding lutein storage. Results Here, we isolated a lutein hyper-producing mutant of the model green microalga Chlamydomonas reinhardtii and characterized the metabolic mechanisms driving its enhanced lutein accumulation using label-free quantitative proteomics. Norflurazon- and high light-resistant C. reinhardtii mutants were screened to yield four mutant lines that produced significantly more lutein per cell compared to the CC-125 parental strain. Mutant 5 (Mut-5) exhibited a 5.4-fold increase in lutein content per cell, which to our knowledge is the highest fold increase of lutein in C. reinhardtii resulting from mutagenesis or metabolic engineering so far. Comparative proteomics of Mut-5 against its parental strain CC-125 revealed an increased abundance of light-harvesting complex-like proteins involved in photoprotection, among differences in pigment biosynthesis, central carbon metabolism, and translation. Further characterization of Mut-5 under varying light conditions revealed constitutive overexpression of the photoprotective proteins light-harvesting complex stress-related 1 (LHCSR1) and LHCSR3 and PSII subunit S regardless of light intensity, and increased accrual of total chlorophyll and carotenoids as light intensity increased. Although the photosynthetic efficiency of Mut-5 was comparatively lower than CC-125, the amplitude of non-photochemical quenching responses of Mut-5 was 4.5-fold higher than in CC-125 at low irradiance. Conclusions We used C. reinhardtii as a model green alga and identified light-harvesting complex-like proteins (among others) as potential metabolic engineering targets to enhance lutein accumulation in microalgae. These have the added value of imparting resistance to high light, although partially compromising photosynthetic efficiency. Further genetic characterization and engineering of Mut-5 could lead to the discovery of unknown players in photoprotective mechanisms and the development of a potent microalgal lutein production system.
Article
Full-text available
Proinsulin Like Growth Factor I (prolGF-I) and myostatin (Mstn) regulate muscle regeneration and mass when intravenously delivered. We tested if chloroplast bioencapsulated forms of these proteins may serve as a non-invasive means of drug delivery through the digestive system. We created tobacco (Nicotiana tabacum) plants carrying GFP-Fc1, proIGF-I-Fc1, and Mstn-Fc1 fusion genes, in which fusion with the immunoglobulin G Fc domain improved both protein stability and absorption in the small intestine. No transplastomic plants were obtained with the Mstn-Fc1 gene, suggesting that the protein is toxic to plant cells. proIGF-I-Fc1 protein levels were too low to enable in vivo testing. However, GFP-Fc1 accumulated at a high level, enabling evaluation of chloroplast-made Fc fusion proteins for oral delivery. Tobacco leaves were lyophilized for testing in a mouse system. We report that the orally administered GFP-Fc1 fusion protein (5.45 µg/g GFP-Fc1) has been taken up by the intestinal epithelium cells, evidenced by confocal microscopy. GFP-Fc1 subsequently entered the circulation where it was detected by ELISA. Data reported here confirm that chloroplast expression and oral administration of lyophilized leaves is a potential delivery system of therapeutic proteins fused with Fc1, with the advantage that the proteins may be stored at room temperature.
Article
Full-text available
The marine carotenoid astaxanthin is one of the strongest natural antioxidants and therefore is used in a broad range of applications such as cosmetics or nutraceuticals. To meet the growing market demand, the natural carotenoid producer Corynebacterium glutamicum has been engineered to produce astaxanthin by heterologous expression of genes from the marine bacterium Fulvimarina pelagi. To exploit this promising source of fermentative and natural astaxanthin, an efficient extraction process using ethanol was established in this study. Appropriate parameters for ethanol extraction were identified by screening ethanol concentration (62.5–97.5% v/v), temperature (30–70 °C) and biomass-to-solvent ratio (3.8–19.0 mgCDW/mLsolvent). The results demonstrated that the optimal extraction conditions were: 90% ethanol, 60 °C, and a biomass-to-solvent ratio of 5.6 mgCDW/mLsolvent. In total, 94% of the cellular astaxanthin was recovered and the oleoresin obtained contained 9.4 mg/g astaxanthin. With respect to other carotenoids, further purification of the oleoresin by column chromatography resulted in pure astaxanthin (100%, HPLC). In addition, a 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging assay showed similar activities compared to esterified astaxanthin from microalgae and a nine-fold higher antioxidative activity than synthetic astaxanthin.
Article
Full-text available
Diatoms (Bacillariophyceae) accumulate neutral storage lipids in lipid droplets during stress conditions, which can be rapidly degraded and recycled when optimal conditions resume. Since nutrient and light availability fluctuate in marine environments, storage lipid turnover is essential for diatom dominance of marine ecosystems. Diatoms have garnered attention for their potential to provide a sustainable source of omega-3 fatty acids. Several independent proteomic studies of lipid droplets isolated from the model oleaginous pennate diatom Phaeodactylum tricornutum have identified a previously uncharacterized protein with an acyl-CoA binding (ACB) domain, Phatrdraft_48778, here referred to as Phaeodactylum tricornutum acyl-CoA binding protein (PtACBP). We report the phenotypic effects of CRISPR-Cas9 targeted genome editing of PtACBP. ptacbp mutants were defective in lipid droplet and triacylglycerol degradation, as well as lipid and eicosapentaenoic acid synthesis, during recovery from nitrogen starvation. Transcription of genes responsible for peroxisomal β-oxidation, triacylglycerol lipolysis, and eicosapentaenoic acid synthesis was inhibited. A lipid binding assay using a synthetic ACB domain from PtACBP indicated preferential binding specificity towards certain polar lipids. PtACBP fused to eGFP displayed an endomembrane-like pattern, which surrounded the periphery of lipid droplets. PtACBP is likely responsible for intracellular acyl transport, affecting cell division, development, photosynthesis, and stress response. A deeper understanding of the molecular mechanisms governing storage lipid turnover will be crucial for developing diatoms and other microalgae as biotechnological cell factories.
Article
Full-text available
Microalgae and cyanobacteria are diverse groups of organisms with great potential to benefit societies across the world. These organisms are currently used in food, feed, pharmaceutical and cosmetic industries. In addition, a variety of novel compounds are being isolated. Commercial production of photosynthetic microalgae and cyanobacteria requires cultivation on a large scale with high throughput. However, scaling up production from lab-based systems to large-scale systems is a complex and potentially costly endeavor. In this review, we summarise all aspects of large-scale cultivation, including aims of cultivation, species selection, types of cultivation (ponds, photobioreactors, and biofilms), water and nutrient sources, temperature, light and mixing, monitoring, contamination, harvesting strategies, and potential environmental risks. Importantly, we also present practical recommendations and discuss challenges of profitable large-scale systems associated with economical design, effective operation and maintenance, automation, and shortage of experienced phycologists.
Article
The green microalga Dunaliella salina hyperaccumulates β‐carotene in the chloroplast, which turns its cells orange. This does not occur in the sister species Dunaliella tertiolecta. However, the molecular mechanisms of β‐carotene hyperaccumulation were still unclear. Here, we discovered the reasons for β‐carotene hyperaccumulation by comparing the morphology, physiology, genome, and transcriptome between the carotenogenic D. salina and the noncarotenogenic D. tertiolecta after transfer to high light. The differences in photosynthetic capacity, cell growth, and the concentration of stored carbon suggest that these species regulate the supply and utilization of carbon differently. The number of β‐carotene‐containing plastid lipid globules increased in both species, but much faster and to a greater extent in D. salina than in D. tertiolecta. Consistent with the accumulation of plastid lipid globules, the expression of the methyl‐erythritol‐phosphate and carotenoid biosynthetic pathways increased only in D. salina, which explains the de novo synthesis of β‐carotene. In D. salina, the concomitantly upregulated expression of the carotene globule proteins suggests that hyperaccumulation of β‐carotene also requires a simultaneous increase in its sink capacity. Based on genomic analysis, we propose that D. salina has genetic advantages for routing carbon from growth to carotenoid metabolism.
Article
To support a global population of ~10 billion people in 2050, dietary protein demand is forecast to increase 32-78% compared to 2017, requiring significantly higher planetary resources. Microalgae are an attractive sustainable protein source compared with current plant and animal sources. Benefits include mass scalability, low CO2 emissions, and significantly reduced land and freshwater use per unit protein. Microalgae are already used as food products and numerous species exhibit high total protein contents and well-balanced essential amino acid (EAA) compositions for human dietary requirements. Microalgae proteins are also bioavailable for human digestion, and downstream processing steps are likely to further enhance protein digestibility. Species, cultivation, and process/product optimisation are actively being developed to enhance their nutritional, social, and environmental benefits.