Next Article in Journal
Exploring Micromonospora as Phocoenamicins Producers
Previous Article in Journal
Enhancing Bioproducts in Seaweeds via Sustainable Aquaculture: Antioxidant and Sun-Protection Compounds
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Infective Secondary Metabolites of the Marine Cyanobacterium Lyngbya Morphotype between 1979 and 2022

1
Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
2
Natural Products Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
3
Suez Canal University Hospital, Suez Canal University, Ismailia 41522, Egypt
*
Author to whom correspondence should be addressed.
Mar. Drugs 2022, 20(12), 768; https://doi.org/10.3390/md20120768
Submission received: 2 November 2022 / Revised: 1 December 2022 / Accepted: 5 December 2022 / Published: 7 December 2022
(This article belongs to the Section Marine Chemoecology for Drug Discovery)

Abstract

:
Cyanobacteria ascribed to the genus Lyngbya (Family Oscillatoriaceae) represent a potential therapeutic gold mine of chemically and biologically diverse natural products that exhibit a wide array of biological properties. Phylogenetic analyses have established the Lyngbya ‘morpho-type’ as a highly polyphyletic group and have resulted in taxonomic revision and description of an additional six new cyanobacterial genera in the same family to date. Among the most prolific marine cyanobacterial producers of biologically active compounds are the species Moorena producens (previously L. majuscula, then Moorea producens), M. bouillonii (previously L. bouillonii), and L. confervoides. Over the years, compounding evidence from in vitro and in vivo studies in support of the significant pharmaceutical potential of ‘Lyngbya’-derived natural products has made the Lyngbya morphotype a significant target for biomedical research and novel drug leads development. This comprehensive review covers compounds with reported anti-infective activities through 2022 from the Lyngbya morphotype, including new genera arising from recent phylogenetic re-classification. So far, 72 anti-infective secondary metabolites have been isolated from various Dapis, Lyngbya, Moorea, and Okeania species. These compounds showed significant antibacterial, antiparasitic, antifungal, antiviral and molluscicidal effects. Herein, a comprehensive literature review covering the natural source, chemical structure, and biological/pharmacological properties will be presented.

Graphical Abstract

1. Introduction

Infectious diseases, also known as transmissible diseases or communicable diseases, are illnesses caused by a harmful pathogen. Infections can be a result of a wide range of pathogens, such as bacteria, viruses, parasites, and fungi. The immune system of the host is always responsible for the fight against the cause of infection. Anti-infective drugs have improved the way for modern medicine and saved the life of millions of people. They are considered a vital group of drugs in this regard and have contributed significantly to the improvement of lifestyle and the rise in life expectation over the past decades [1,2]. Natural products contributed significantly to the major group of current anti-infective drugs [3]. However, for a long time, the pharmaceutical industry has ignored the search for natural product-derived and novel anti-infective drug discovery [4]. This fact resulted in a situation where illness with antibiotic resistant microbes regularly cannot be effectively treated [5]. Illnesses with Gram-negative bacteria and a developed treatment resistance, such as Enterobacter and Pseudomonas, are considered serious issues, while a few new antibiotics are under development [6].
The development of Antimicrobial Resistance (AMR) and multi-drug resistance to current therapeutics represent a serious issue for the patients, health care system and for the economy worldwide [7,8,9,10,11,12,13,14,15,16,17]. In addition, viral infections are spreading worldwide rapidly [18,19,20,21,22,23,24,25,26] and are considered a threat to the healthy community and represent additional economic burden to the concerned countries worldwide [18,19,20,21,22,23,24,25,26]. Therefore, it is of an urgent need to develop new drugs for the combat against AMR and viral infections.
Recently, the pandemic of coronavirus (COVID-19) resulted in hundreds of thousands of deaths in several countries around the world. SARS-CoV-2, a newly found coronavirus strain, causes COVID-19, an infectious respiratory illness. Remdesivir is being used in conjunction with the anti-inflammatory drugs dexamethasone or baricitinib as treatment for this coronavirus. Because of Remdesivir’s side effects, which include respiratory failure, hypokalemia, and headaches, new medications for effective COVID-19 therapy are urgently needed [27]. Therefore, more research is needed to develop powerful antivirals as alternative treatments that can improve the management of epidemics and eventually lead to their elimination.
Water occupies more than 70% of our Blue Planet. Distinct environments from oceans and seas to freshwater lakes, wetlands, ponds, rivers and streams, all of which exhibit diverse ecosystems, allowing the inhabitation of species with distinct characteristics, differing from terrestrial organisms. The advent of SCUBA and submersible technologies has enabled scientists to explore the plethora of marine natural products created by biologically diverse marine organisms and to investigate their biological activities. The lack of cures and treatments for many diseases compels the scientific community, academia and industry to search untapped avenues and focus on underexplored territories, which makes marine drug discovery a prime target for pharmaceutical research. Marine drug discovery is still considered in its infancy, and the full therapeutic potential of marine natural products is yet to be realized.
In this regard, scientists have investigated the biological activities of numerous natural products isolated from marine organisms in order to search for any promising pharmacological properties that may be utilized in the development of viable treatment modalities. Some of the marine organisms most widely studied for their natural products are benthic filamentous cyanobacteria collected pantropically.
For more than three billion years, cyanobacteria inhabited the earth, representing one of the eldest known organisms [28]. Cyanobacteria are diverse in terms of their physiology, metabolism and morphology. They inhabit all environments worldwide, including freshwater, marine environment and extreme habitats [29]. The production of highly potent/toxic secondary metabolites is considered as an evolutionary strategy for cyanobacteria to survive from planktivores or other ecological competitors [30,31]. It was noticed that a significant portion of the cyanobacterial secondary metabolites in the literature possessed cytotoxic activity [32]. Certainly, cytotoxicity is still today a noticeable activity of cyanobacteria-derived compounds.
Specimens characterized by relatively large filaments of discoid cells within a distinct sheath and lacking nitrogen-fixing heterocyst cells have routinely been assigned to the genus Lyngbya (Family Oscillatoriaceae, Order Oscillatoriales). This traditional morphology-based taxonomic identification has underestimated the biological diversity of filamentous marine cyanobacteria [33] leading to more than 500 marine-derived compounds being ascribed to the single genus Lyngbya. While this Lyngbya ‘morpho-type’ has evolved to additionally include the new genera Dapis [34], Limnoraphis [35], Moorena (previously Moorea) [36], Microseira [37] and Okeania [38], and further taxonomic revisions are in progress, it remains a group with tremendous therapeutic potential.
Currently, there are 17 marketed marine-derived compounds or analogs, or derivatives therefrom and an additional 34 drug candidates in different phases of clinical trials (I, II, and III) [39]. Astonishingly, amongst the 17 marketed compounds, there are 5 compounds (29%) of cyanobacterial/molluscs origin. Further, among the 34 drug candidates in different clinical phases (I, II, and II), there are 23 compounds (67%) of cyanobacterial/molluscs origin [39]. This fact makes cyanobacteria/molluscs the main source of marine derived marketed drugs and drug leads under development today. Since all marketed drugs and drug candidates in clinical phases are targeting different types of cancer, more research focus from the academia and industry should be directed to combat microbial resistance and viral infections worldwide.
A literature search was conducted using the keywords “Lyngbya”, “Moorea”, “Moorena”, “Neolyngbya”, “Okeania”, “Limnoraphis”, and “Dapis”, on SciFinder, resulting in 2956 hits, including 2423 for Lyngbya, 480 for Moorea, 35 for Okeania, 9 for Moorena, 3 for Neolyngbya, 3 for Limnoraphis, and 3 for Dapis, some of which were duplicated between the databases MEDLINE and CAplus. Results showed that, while mostly cytotoxic secondary metabolites have been described from Lyngbya morphotype, there are also a substantial number of these compounds with anti-infective properties.
This comprehensive review addresses all antibacterial, antifungal, antiviral, antiparasitic, molluscicidal and anti-diatom activities isolated from the marine cyanobacterium Lyngbya morpho-type. The review will focus on the source organisms, geographical locations of the studied species, chemical diversity of the isolated compounds and associated biological activities and their mode of action, if any. About 72 compounds belonging to a diverse group of chemical classes were reported from Lyngbya morpho-type over 43 years between 1979 and 2022.

2. Collected Species and Geographical Locations

The first report about an anti-infective compound from the genus Lyngbya was published in 1979 [40]. Between then and 2022, a total of about 72 secondary metabolites with anti-infective properties were reported from 11 morphotype species belonging to the genus Lyngbya (Table 1).
Interestingly, more than 50% of the reported compounds come from L. majuscula (Table 1). These species have been collected from locations around the world, focused on tropical regions (Figure 1).
A notable number of reported compounds (17%) comes from species that collected in Panama, making this place a diverse and rich location for collecting cyanobacterial strains. Japan comes in the second place with 9 compounds (13%) followed by Guam with 8 compounds (11%). In third place, Florida (USA), Red Sea and Japan, each comes with 6 compounds (8%). (Figure 2).
These data indicate that genus Lyngbya continues to be a rich source of secondary metabolites that are new to science and suggest potential locations for further discovery.

3. Compounds with Antibacterial Activities

Among the diverse bioactivities that Lyngbya secondary metabolites have displayed is the activity against bacteria. In 1979, malyngolide (1) (Figure 3), a δ-lactone was reported from Hawaiian Lyngbya majuscula in Kahala Beach, showed effective antibacterial activity against Mycobacterium smegmatis and Streptococcus pyogenes and was less active against Staphylococcus aureus and Bacillus subtilis, and inactive towards Enterobacter aerogenes, Escherichia coli, Pseudomonas aeruginosa, Salmonella enteritidis, and Staphylococcus marcescens [40].
In 1987, the fatty acid (-)-7(S)-methoxytetradec-4(E)-enoate (lyngbic acid) (2) (Figure 3), was purified from Moorea producens collected at the Red Sea, near Jeddah, Saudi Arabia [41], displayed antibacterial activity against S. aureus and B. subtilis [41].
The related amide of lyngbic acid, malyngamide D acetate (3) (Figure 3), which were isolated from Caribbean L. majuscula in Isla Guayacan, Puerto Rico in 1987, displayed slight activity against S. aureus [42].
In 2001, the cyclic depsipeptides pitipeptolides A (4) and B (5) (Figure 4) are reported from L. majuscula collected in Piti Bomb Holes, Guam [43]. The compounds displayed moderate activity against Mycobacterium tuberculosis strains (ATCC 25177 and ATCC 35818) in the antimycobacterial diffusion susceptibility assay. Pitipeptolide A (4) gave a diameter of growth inhibition zone for ATCC 25177 strain equivalent to 25 and 10 mm, and for ATCC 35818 strain equivalent to 15 and 9 mm upon treatment with 100 and 25 µg, respectively. Pitipeptolide B (5) gave a diameter of growth inhibition zone for ATCC 25177 strain equivalent to 30 and 15 mm, and for ATCC 35818 strain equivalent to 15 and 10 mm upon treatment with 100 and 25 µg, respectively. For comparison, treatments with 25, 5 and 1 µg of streptomycin resulted in superior activity, giving diameters of 50, 15 and 0 mm, respectively, for ATCC 25177 strain, and 55, 33 and 10 mm, respectively, for ATCC 35818 [43].
Ten years later, in 2011, pitipeptolides C-F (69) (Figure 4) are reported from L. majuscula in Piti Bomb Holes, Guam. The study showed that pitipeptolide F (9) was the most potent compound in the antimycobacterial disc diffusion assay (M. tuberculosis ATCC 25177 strain) [44]. Treatment with pitipeptolides A-F (49) resulted in inhibition zones of 28, 30, 26, 10, 21 and 40 mm, respectively, at 100 µg, 23, 24, 21, 0, 15 and 30 mm, respectively, at 50 µg, and 9, 14, 18, 0, 0 and 10 mm, respectively, at 10 µg. For comparison, streptomycin gave 40, 30 and 0 mm inhibition zone upon using 10, 5 and 1 µg treatment, respectively [44].
SAR studies revealed that the N-methylation in the Phe unit is essential for both cytotoxic and antibacterial activities, whereas the π system in the fatty acid unit was found to be one of the important structural features for the cytotoxic activity in mammalian cells, but it was not required for antibacterial activity. Furthermore, decreasing the hydrophobicity of certain units (2-Hydroxy 3-methyl pentanoic acid (Hmpa) → 2-Hydroxy isovaleric acid (Hiva) and Ile → Val) caused a reduction in the anticancer activity (as seen with pitipeptolides E and F), while on the other hand resulted in an increase in antimycobacterial potency (particularly pitipeptolide F) [44].
Pitiprolamide (10) (Figure 5), a dolastatin 16 analog and a proline rich cyclic depsipeptide was purified in 20111 from the same Guamanian cyanobacterium Lyngbya majuscula collected at Piti Bomb Holes, displayed weak antimycobacterial effect against M. tuberculosis (ATCC 25177 strain) starting at 50 μg in a disk diffusion assay. The compound displayed zone of inhibition of 23, 13 and 0 mm after 100, 50 and 10 µg treatment. Also, the compound exerted weak antibacterial activity against Bacillus cereus (ATCC 10987 strain) starting at 1 μM in a microtiter plate-based assay with an approximate IC50 value of 70 μM and lacked the activity against S. aureus and P. aeruginosa [45].
Table 2 that purified in 2013 from the cyanobacterium M. producens collected at the Red Sea, near Jeddah, Saudi Arabia, significantly inhibited the growth of M. tuberculosis H37Rv in vitro (65% inhibition) at a concentration of 12.5 μg/mL, while the chlorinated lipopetides malyngamides A (11), B (12) and 4 (13) (Figure 6) (obtained from the same cyanobacterial collection) displayed much weaker antimycobacterial activity at the same tested concentration, which was deemed as ineffective (18, 10 and 17% inhibition, respectively) [41]. This result suggests the importance of a terminal free carboxylic acid moiety for the antimycobacterial effect.
Another group of antimicrobial natural products is the cyclic undecapeptides lyngbyazothrins A and B (14 and 15) and C and D (16 and 17) (Figure 7), which were isolated as binary mixtures from Lyngbya sp. 36.91 SAG (Culture Collection of Algae, Gottingen, Germany) in 2009. The mixture of lyngbyazothrins A and B (14 and 15) showed minimal antibacterial activity against Micrococcus flavus SBUG 16 in the agar diffusion disk (100 μg/disk: 8 mm diameter of inhibition zone). The mixture of lyngbyazothrins C (16) and D (17) showed modest activity against B. subtilis SBUG 14 (25 μg/disk: 18 mm), E. coli ATCC 11229 (100 μg/disk: 18 mm), and E. coli SBUG 13 (100 μg/disk: 15 mm) and low activity against P. aeruginosa ATCC 27853 (100 μg/disk: 8 mm) and Serratia marcescens SBUG 9 (200 μg/disk: 8 mm). When used at the same concentrations, the lyngbyazothrins A and B (14 and 15) mixture lacked activity against the aforementioned strains, which suggests that the linkage of the acyl residue at C-5 of the 3-amino-2,5,7,8-tetrahydroxy-10-methylundecanoic acid (Aound) unit may be responsible for the antimicrobial activity [46].
The intriguing cyclic depsipeptides, tiahuramides A-C (1820) (Figure 8), are isolated in 2018 from L. majuscula collected at Tiahura sector, Moorea Island in French Polynesia, displayed growth inhibitory activities on opportunistic marine pathogenic bacteria (Aeromonas salmonicida (CIP 103209T strain), Vibrio anguillarum (CIP 63.36T), and Shewanella baltica (CIP 105850T)) and terrestrial bacteria (E. coli (CIP 54.8) and Micrococcus luteus (CIP A270)). The MIC values against A. salmonicida, V. anguillarum, S. baltica, E. coli and M. luteus were as follows: 27, 33, >50, 35 and 47 μM, respectively, for tiahuramide A; 9.4, 8.5, 22, 12 and 29 μM, respectively, for tiahuramide B; and 6.7, 7.4, 16, 14 and 17 μM, respectively, for tiahuramide C. As evidenced by the MIC values, tiahuramide C (20) exhibited the greatest antibacterial potency followed by tiahuramide B (19), whereas tiahuramide A (18) was the least active among this series of compounds [47].
Table 2 summarizes all compounds with reported antibacterial effects, their sources and collection sites as well as the targeted bacteria and observed effects.

4. Compounds with Anti-Swarming and Anti-Quorum Sensing Activities

Some compounds exert their antibacterial activities by inhibiting swarming, a mechanism used by bacteria to spread across surfaces supplied with nutrients through the use of rotating flagella in order to speed their growth [48,49].
Lagunamides A-C (2123) (Figure 9), cyclic depsipeptides purified in 2010 and 2011 from L. majuscula found in Pulau Hantu Besar, Singapore, exhibited moderate to weak anti-swarming activities against the Gram-negative bacterial strain P. aeruginosa PA01 (62, 56 and 49% compared to control, respectively) when tested at 100 ppm; P. aeruginosa PA01 (62, 56 and 49% compared to control, respectively) when tested at 100 ppm [50,51].
On the other hand, other compounds exert their antimicrobial activities by interfering with quorum sensing (QS), a mechanism that is responsible for the regulating of the bacterial gene expression in response to fluctuations in cell-population density [52,53].
In 2010, malyngamide C (24) and 8-epi-malyngamide C (25) (Figure 10) are reported from L. majuscula collected in Bush Key, Dry Tortugas, Florida, displayed activity against the QS reported pSB1075, which expresses LasR and responds to 3-oxo-C12-HSL (N-3-oxo-dodecanoyl-L-homoserine lactone). Using concentrations of both compounds that did not actually inhibit bacterial cell growth (10, 100 and 1000 µM) resulted in reducing 3-oxo-C12-HSL signalling in the QS reporter [54].
Malyngolide (1), an antibiotic isolated from L. majuscula in South Florida, inhibited violacein pigment production by Chromobacterium violaceum CV017 in the QS bioassay. Effective concentrations ranged from 0.07 to 0.22 mM, with an EC50 value of 0.11 mM, and the growth of the C. violaceum reporter strain was not inhibited even at the higher concentration used (0.22 mM). In the presence of 14 µM of 3-oxo-C12-HSL, malyngolide (1) inhibited responses of the lasR+PlasI-luxCDABE reporter pSB1075 when used at concentrations ranging from 3.57 to 57 µM (EC50 = 12.2 µM) without affecting bacterial growth. At these concentrations, malyngolide (1) also significantly reduced the production of elastase by P. aeruginosa PAO1, which is an extracellular enzyme regulated by 3-oxo-C12-HSL and LasR, with an EC50 value of 10.6 µM. At higher concentrations of malyngolide, elastase production was inhibited to the level observed in the QS mutant of P. aeruginosa JP2. It is worth mentioning that a decline in the activity of malyngolide was noticed upon storing it in plastic instead of glass vials [55].
Another disruptor of QS in P. aeruginosa is lyngbyoic acid (26) (Figure 11), a small cyclopropane-containing fatty acid isolated was reported in 2019 from L. majuscula collected at various sites in Florida. The compound was evaluated against four reporters based on different acylhomoserine lactone (AHL) receptors (LuxR, AhyR, TraR and LasR), and LasR turned out to be the most reported being affected by lyngbyoic acid (26). It also reduced the production of pyocyanin and elastase (LasB) both on the protein and transcript level in wild-type P. aeruginosa, and directly inhibited LasB enzymatic activity with a Ki of 5.4 mM, without affecting bacterial growth [56].
Finally, in 2019, doscadenamide A (27) (Figure 11), was isolated from M. bouillonii collected in Fingers Reef, Apra Harbor, Guam, displayed QS agonistic activities in a LasR-dependent manner. Doscadenamide A and the QS signaling molecule 3-oxo-C12-HSL share structural similarities as they both contain a five-membered ring core and long alkyl side chain. Doscadenamide A activated the 3-oxo-C12-HSL-responsive reporter plasmid pSB1075, which encodes LasR and contains a light-producing luxCDABE cassette expressed in E. coli; however, it was not able to activate the related reporter pTIM5319, which is identical to pSB1075, except for lacking the AHL-binding site LasR, thereby suggesting that doscadenamide A activates QS via the AHL-binding site. The effect of the compound was tested on wild-type P. aeruginosa, using effective doses of 10, 100 and 1000 µM, and maximal induction of the QS pigment pyocyanin production was observed upon usage of even the lowest concentration. Levels of pyocyanin increased after only 6 h of treatment with 10 µM of doscadenamide A, which was a comparable result with using 10 µM of the positive control 3-oxo-C12-HSL [57].
Table 3 summarizes all compounds with reported anti-swarming and anti-quorum sensing effects, their sources and collection sites as well as the targeted bacteria and observed effects.

5. Compounds with Antifungal Activities

Antifungal assays are among the widely used bioassays for testing the activities of natural compounds isolated from cyanobacteria. Majusculamide C (28) (Figure 12), a cyclic depsipeptide reported in 1984 from L. majuscula in Marshall Islands, inhibited the growth of a number of fungal plant pathogens such as Phytophthora infestans and Plasmopora viticola, the causative organisms of tomato late blight and grape downy mildew, respectively [58].
In 1988, 57-normajusculamide C (29) (Figure 12) was purified from the marine cyanobacterium L. majuscula collected in Marshall Islands. The compound displayed antimycotic activity against the indicator organism Saccharomyces pastorianus [59].
Microcolins A (30) and B (31) (Figure 13), lipopeptides isolated from Floridian L. polychroa, showed only little activity against two strains (SIO and EBGJ) of the marine fungus Dendryphiella salina, which has been linked to diseases among marine algae and seagrasses, where the LD50 values were above 200 μg/mL in the antifungal assay. The antifungal activities of microcolins A (30) and B (31), were significantly lower than the known antifungal compound amphotericin B, which resulted in 100% inhibition of marine fungus Dendryphiella salina in the same assay at concentrations as low as 3.13 μg/mL [60].
The majority of natural products have been tested for their antifungal activity against Candida albicans as reported herein. Laxaphycin B (32) (Figure 14), a cyclic lipopetide reported in 1997 from L. majuscula in Moorea Atoll, French Polynesia, exhibited antifungal activity against C. albicans. Interestingly, laxaphycin A (33) (Figure 14), inactive by itself, exerted a synergistic effect when combined with laxaphycin B (32) and potentialized its antifungal activity. This unique difference in activity might be attributed to the chemical structures of the compounds. Laxaphycin A (33) is an undecapeptide with segregated hydrophobic and hydrophilic residues, while laxaphycin B (32) is a dodecapeptide with alternating hydrophobic and hydrophilic residues [61].
Tanikolide (34) (Figure 15), a lipid lactone that was reported in 1999 from L. majuscula found in Tanikeli Island, Madagascar, showed antifungal activity towards C. albicans with 13 mm diameter zone of inhibition at 100 µg/disk using paper disk-agar plate methodology [62].
Lyngbyabellin B (35) (Figure 15), a cyclic depsipeptide that reported in 2000 from L. majuscula found in Dry Tortugas National Park in Florida, displayed antifungal effect towards C. albicans (ATCC 14053) in a disk diffusion assay with a 10.5 mm zone of inhibition at 100 µg/disk and a slight halo at 10 µg/disk [63].
In 2002, the lipopeptide hectochlorin (36) (Figure 15), was reported from L. majuscula found in both Hector Bay, Jamaica, and Boca del Drago Beach, Panama. The compound produced a 16 mm zone of inhibition at 100 µg/disk and an 11 mm zone of inhibition at 10 µg/disk against C. albicans (ATCC 14053) [64].
In 2002, the lipopeptides lobocyclamides A–C (3739) (Figure 16) are obtained from a cyanobacterial mat containing L. confervoides found in Cay Lobos, Southern Bahamas. The compounds exhibited moderate antifungal activities when tested in disk diffusion assay at 150 µg/disk against fluconazole-resistant fungus C. albicans 96–489 giving 7, 8 and 10 mm inhibition zones, respectively. When evaluated towards C. glabrata, lobocyclamide B (38) and C (39) produced 6 and 8 mm inhibition zone, respectively, at 150 µg/disk [65].
In the microbroth dilution assay against C. albicans 96–489, lobocyclamide A (37) displayed MIC value of 100 µg/mL, while lobocyclamide B (38) showed an MIC value of 30–100 µg/mL [65].
A mixture of lobocyclamides A (37) and B (38) exhibited significant synergism (e.g., 1:1 mixture of A and B produced a MIC of 10–30 µg/mL) with higher activity than either of the pure compounds used individually [65], a phenomenon also reported with laxaphycins A (33) and B (32) [61].
Table 4 summarizes all compounds with reported antifungal activities, their sources and collection sites as well as the targeted fungi and observed effects.

6. Compounds with Antiparasitic Activities

Tropical parasitic diseases can be life-threatening if not treated appropriately from an early stage. The most common tropical infectious parasite is Plasmodium falciparum, the causative organism of malaria. Several Lyngbya-derived compounds displayed inhibitory activities on this parasite.
Carmabin A (40), dragomabin (41) and dragonamide A (42) (Figure 17), are linear alkynoic lipopeptides are reported in 2007 from L. majuscula that was collected from Isla Bastimentos in Bocas del Toro, Panama, possessed good antimalarial activities against a chloroquine-resistant strain (Indochina W2) of P. falciparum with IC50 values of 4.3, 6.0 and 7.7 µM, respectively. It was also found that carmabin A (40) was more cytotoxic to mammalian Vero cells (IC50 = 9.8 µM) than dragomabin (41) (IC50 = 182.3 µM) or dragonamide A (42) (IC50 = 67.8 µM), indicating that dragomabin (41) exhibited the best differential toxicity between parasitic and mammalian cells among the tested compounds in this series. The presence of three extra carbons in the aliphatic chain of carmabin A (40) may have contributed to its increased cytotoxicity over that displayed by dragomabin (41) [66].
On the other hand, the nonaromatic analog, dragonamide B (43) (Figure 18), was reported from a L. majuscula collected in Panama in 2007, was found to be completely inactive suggesting the necessity of an aromatic amino acid moiety at the carboxy terminus for the antimalarial activity [66]. Interestingly, when dragonamide A (42) was subjected to the same antimalarial assay on a later date, no activity was shown against the parasite (maximum test concentration 10 μM) [67].
In 2010, the antimalarial malyngolide dimer (44) (Figure 18), a symmetric cyclodepside isolated from L. majuscula in Coiba National Park, Panama was reported. It showed an IC50 value of 19 μM when tested against the chloroquine-resistant P. falciparum strain (W2) [68].
The intriguing cyclic depsipeptides lagunamides A-C (2123) (Figure 9), purified from L. majuscula found in Pulau Hantu Besar, Singapore, also showed significant activity against the drug-sensitive NF54 strain of P. falciparum, with IC50 values of 0.19, 0.91 and 0.29 μM, respectively [50,51].
Ikoamide (45) (Figure 19), an antimalarial lipopeptide reported in 2020 from a marine cyanobacterium Okeania sp. collected in Okinawa, Japan. The compound displayed strong antimalarial activity against P. falciparum with an IC50 value of 0.14 μM without cytotoxicity against human cancer cell lines (HeLa and HL60) at 10 μM [69].
Mabuniamide (46) (Figure 20), a lipopeptide from of an Okinawan Okeania sp. in 2019 exhibited moderate antimalarial activity with IC50 of 1.4 μM against P. falciparum [70].
Bastimolide B (47) (Figure 21), a 24-membered polyhydroxy macrolide with a long aliphatic polyhydroxylated side chain and unique terminal tertbutyl group was purified from Okeania hirsuta collected in Panama [71]. It showed a strong antimalarial activity against chloroquine-sensitive P. falciparum strain HB3 with IC50 of 5.7 µM.
On the other hand, bastimolide A (48) (Figure 21), which was obtained from the cyanobacterium Okeania hirsuta collected at the Caribbean coast of Panama, showed IC50 with 2.6 µM against chloroquine-sensitive P. falciparum strain [72]. Interestingly, 2-(E)-bastimolide A (49) (Figure 21), a methanolysis product of bastimolide A, displayed the greatest antimalarial activity with IC50 of 1.4 µM. It was found that, the existence of the double bond (at C-2/C-3) as well as the 1,3-diol (at C-9 and C-11) and 1,3,5-triol (at C-19, C-21, and C-23) functionalities were found to be important for the antimalarial activity [72].
In 2020, lyngbyabellins A (50) (Figure 22) was reported from the Malaysian Moorea bouillonii, while lyngbyabellein G (51) (Figure 22) was isolated from the Saudi Red Sea Okeania sp. Both compounds inhibited P. falciparum with IC50 of 0.3 and 1.1 µM, respectively [73].
On the other hand, homohydroxydolabellin (52) (Figure 22), which was isolated from the Malaysian M. bouillonii displayed IC50 of 6.4 µM against P. falciparum [73].
Another tropical parasite, which is the causative organism of the disease leishmaniasis, is Leishmania donovani. Antileishmanial activity has been displayed by a number of compounds isolated from Lyngbya sp.
Dragonamides A (42) (Figure 17) and E (53) (Figure 23) and herbamide B (54) (Figure 23), modified linear lipopeptides isolated in 2010 from Panamanian L. majuscula found around mangrove roots in the Bastimentos National Park, Bocas del Toro, Panama, showed inhibitory activities against L. donovani (LD-1S/MHOM/SD/00-strain 1S) with IC50 values of 6.5, 5.1 and 5.9 μM, respectively [67].
Almiramides B (55) and C (56) (Figure 24), members of another class of linear lipopeptides isolated in 2010 from the Panamanian collection of the marine cyanobacterium Lyngbya majuscula, also exhibited antileishmanial activities, with IC50 values of 2.4 and 1.9 μM, respectively, whereas almiramide A (57) (Figure 24) was completely inactive up to 13.5 μM. This lack of activity might be attributed to the absence of an unsaturated terminus on the side chain, which was present in the active compounds, almiramides B (55) and C (56). Additionally, these compounds did not exert significant cytotoxicity to mammalian Vero cells and were selective for parasitic cells [74].
The cyclic depsipeptides dudawalamides A-D (5861) (Figure 25) are reported in 2017 from M. producens collected in Papua New Guinean M found in Dudawali Bay. The compounds exhibited broad and variable antiparasitic activities against malaria-, leishmaniasis- and Chagas disease-causing microorganisms (P. falciparum, L. donovani and Trypanosoma cruzi, respectively).
It was found that dudawalamides A (58) and D (61) were more potent against P. falciparum with IC50 values of 3.6 and 3.5 μM, respectively, compared to dudawalamides B (59) and C (60) (IC50 = 8.0 and 10 μM, respectively). Dudawalamides A (58) and B (59) possessed 12 and 7% growth inhibition at 10 μg/mL, respectively, against T. cruzi, and they both had an IC50 value > 10 μM against L. donovani. Dudawalamide D (61) was the most potent antiparasitic compound in this series since it exhibited an IC50 value of 2.6 μM against L. donovani, and inhibited T. cruzi by 60% when used at a concentration of 10 μg/mL [75].
It is interesting to note that cyclic depsipeptides with 2,2-dimethy-3-hydroxy-7-octynoic acid (Dhoya) moiety, which belong to the kulolide superfamily, possess only minor differences in structure and stereochemistry between each other; nevertheless, their potency was affected by such slight changes, indicating the significant role that configuration and residue sequence plays in the bioactivity of this class of compounds [75].
In 2020, the linear peptides iheyamides A-C (6264) (Figure 26) were reported from the cyanobacterium Dapis sp., collected in Okinawa, Japan [76]. Iheyamide A (62) showed moderate antitrypanosomal effect against Trypanosoma brucei rhodesiense and T. bhurstuerusei brucei with an IC50 value of 1.5 µM. It was found that the isopropyl-O-Me-pyrrolinone moiety is essential for the antitrypanosomal activity [76].
In 2016, janadolide (65) (Figure 27), a cyclic polyketide−peptide hybrid with a tert-butyl group was reported from an Okeania sp., collected in Okinawa, Japan. The compound showed potent antitrypanosomal activity against Trypanosoma brucei brucei GUTat 3.1 strain with an IC50 value of 47 nM without cytotoxicity against human cells at 10 μM [77].
Finally, the polyketide beru’amide (66) (Figure 27) with 4S,5R-configuration was purified in very small amount (68 µg) from a cyanobacterium Okeania sp. collected in Kagoshima, Japan. Two synthetic enantiomers of beru’amide, 4S,5R and 4R,5S, were prepared and evaluated for their growth inhibition effects the causative parasite of African sleeping sickness Trypanosoma brucei rhodesiensec strains IL-1501. Interestingly, the enantiomers 4S,5R and 4R,5S of beru’amide displayed a closely similar and strong antitrypanosomal activity against Trypanosoma brucei rhodesiense with IC50 values of 1.2 and 1.0 μM, respectively. Accordingly, the absence of any noteworthy variance in the antitrypanosomal activities between the synthetic enantiomers, 4S,5R and 4R,5S, suggests that the absolute configurations are insignificant for the antitrypanosomal effect [78].
Table 5 displays all compounds with reported antiparasitic activities, their sources and collection sites as well as the targeted parasites and observed effects.

7. Compounds with Antiviral Activities

Purification of the culture of the marine cyanobacterium L. lagerheimii that was collected in Hawaii resulted in the purification of two sulfoglycolipids (compounds 67 and 68) (Figure 28). The compounds displayed activity against HIV-1 in cultured lymphoblastoid CEM, LDV-7, MT-2 and C3–44 cell lines in the tetrazolium assay and inp24 viral protein and syncytium formation assay [79]. The degree of inhibition HIV-1 by the compounds was generally comparable within a given cell line, but the degree of protection varied substantially among the different cell lines. The protective effects of the compounds were studied over a wide range of concentration range (about l–l00 µg/mL), depending on the target cell line and the mode of infection. Both compounds displayed similar levels of activity, suggesting that the length of the aliphatic side chain length and degree of unsaturation have no critical effect on the potency. Interestingly, sulfoglycolipids represent the first cyanobacterial derived compounds with antiviral activity [79].
In another studies, sulfoglycolipids inhibited the DNA polymerase function of the HIV-1 RT with IC50 values in the range 24–2950 nM without any significant effect on the ribonuclease H [80,81]. It was described that, the existence of a sulfate moiety in the sugar part as well as the aliphatic side chain are crucial for sulfoglycolipid’s effect on HIV RT [81].
Table 6 displays the compounds with reported antiviral activities, their sources and collection sites as well as the targeted viruses and observed effects.

8. Compounds with Molluscicidal Anti-Diatoms Activities (Table 7)

Snails and slugs can damage crops by feeding on them; therefore, farmers and gardeners depend on molluscicides to protect their plants. There are some chemical compounds isolated from Lyngbya that possess molluscicidal activities.
Tanikolide (34), a lipid lactone was reported in 1999 from L. majuscula found in Tanikeli Island, Madagascar. The compound exhibited molluscicidal activity against the same snail (LD50 = 9.0 µg/mL) [62].
In addition, in 1996, a chlorinated lipopeptide, barbamide (69) (Figure 29), was reported from L. majuscula collected from Barbara Beach in Curaçao. It showed toxic effect on the mollusc Biomphalaria glabrata with LC100 of 10 µg/mL [82].
Finally, in 2010, the greatest potency of molluscicidal activity against B. glabrata was observed with cyanolide A (70) (Figure 30), a glycosidic macrolide isolated from Papua New Guinean L. bouillonii in Pigeon Island. The compound displayed molluscicidal effect with LC50 value against B. glabrata of 1.2 μM [83].
In 2021, debromooscillatoxin G (71) and I (72) (Figure 31) were purified from an Okinawan cyanobacterium Moorea prducens. Both compounds moderately inhibited the growth of the marine diatom Nitzschia amabilis at a concentration of 10 µg/mL by 30% and 50%, respectively [84].
Table 7 displays the compounds with reported molluscicidal and anti-diatom activities, their sources and collection sites as well as the targeted organism and observed effects
Table 7. Compounds with reported molluscicidal and anti-diatom activities.
Table 7. Compounds with reported molluscicidal and anti-diatom activities.
CompoundSource OrganismCollection SiteTargeted OrganismLC50/LC100/LD50/% of InhibitionReference
Tanikolide (34)L. majusculaMadagascarB. glabrataLD50 = 9.0 µg/mL[62]
Barbamide (69)L. majusculaCuraçaoB. glabrataLC100 = 10 µg/mL[82]
Cyanolide A (70)L. bouilloniiPapua New GuineaB. glabrataLC50 = 1.2 μM[83]
Debromooscillatoxin G (71)M. producensOkinawa, JapanN. amabilis30% at 10 μg/mL[84]
Debromooscillatoxin I (72)M. producensOkinawa, JapanN. amabilis30% at 10 μg/mL[84]

9. Summary

Secondary metabolites originating from the marine Lyngbya morphotype showed a huge chemical diversity and important biological activities, providing an unexploited potential for biodiscovery and therapeutics’ candidates. This marine-inspired genus Lyngbya has been a vital example since its first discovery back in 1979 as an untapped resource of marine-derived drug candidates. The existence of 72 compounds with anti-infective properties of marine derived Lyngbya morphotype worldwide (Figure 1), together with more than 40 years (Figure 32) of research efforts fashioned a resource empowering the biosynthetic capabilities of this genus. In aquatic environments, members of the marine derived Lyngbya morphotype have typically been obtained from different locations worldwide. Accordingly, the interest in marine derived Lyngbya species was growing, and became an essential source of chemical diversity with anti-infective effects.
Since the first report of the antibacterial malyngolide (1) in 1979, additional 71 compounds with anti-infective properties have been reported until now from 10 marine Lyngbya morphotype. The field was most active in the years 2002 (4 compounds from one species), 2007 (4 compound from one species), 2009 (6 compounds from 2 species), 2010 (12 compounds from 7 species), 2011 (7 compounds from 3 species), 2013 (4 compounds from one species), 2017 (4 compounds from one species), 2018 (4 compounds from 2 species), 2020 (7 compounds from 3 species), 2021 (2 compounds from one species) and finally in 2022 (one compound from one species) (Figure 32). Between 1979 and 2001 and in the years 2015, 2016, 2019, 2021 and 2022 there are reports about only one or two compounds from one or two species (Figure 32).
With regards to the source of the reported anti-infective compounds and as shown in Figure 33, it is clear that the morphotype Lyngbya is the main source of the compounds with 48 records (66%), followed by the morphotypes Moorea with 15 compounds (20%), Okeania with 9 compounds (10%) and Dapis with 3 compounds (4%) (Figure 33). Detailed contribution of the individual cyanobacterial morphotype is as follows: Dapis sp. (3 compounds), Lyngbya sp. (5 compounds), Lyngbya confervoides (3 compounds), Lyngbya lagerheimii (one compound), Lyngbya majuscula (37 compounds), Lygnbya polychora (2 compounds), Moorea bouilloni (4 compounds), Moorea producens (8 compounds), Okeania sp. (4 compounds and finally Okeania hirsuta (3 compounds) (Figure 33).
As per the chemical diversity of the genus Lyngbya, it could be noticed that nitrogenous compounds represented as a predominant class of reported secondary metabolites with 59 nitrogenous compounds (83%) and 12 non-nitrogenous compounds (17%). This existence of these enormous nitrogenous secondary metabolites could be attributed to the capability of the members of cyanobacteria of fixing atmospheric nitrogen. Peptides are represented by 71% (51 compounds) from the nitrogen-containing secondary metabolites, while regular nitrogenous compounds, including alkaloids and others are represented by 9 compounds (12%) (Figure 34). Interestingly, there are 14 halogenated compounds among the reported anti-infective secondary metabolites.
Most Lyngbya-derived compounds have demonstrated excellent antibacterial and antiprotozoal activities against different pathogens and parasites. Out of the 72 reported secondary metabolites from Lygnbya morphotype, 31 compounds (about 40%) have been reported to possess antiparasitic activities. In addition, 28 compounds (36%) of the reported compounds displayed antibacterial effects. With antifungal effects, the number was much less with only 12 compounds (15%). Finally, 3 compounds contributed to molluscicidal activity, 2 compounds for each of the antiviral and anti-diatom effects (Figure 35).

10. Conclusions

Herein, 72 compounds, mostly peptides, derived from different Lyngbya morphotype are described. To the best of our knowledge, the anti-infective compounds in this review showed significant activities, including antibacterial, anti-swarming, ant-quorum sensing, antifungal, antiparasitic, antiviral and molluscicidal activities. Therefore, members of the genus Lyngbya morphotype represent a therapeutic gold mine of chemically and biologically diverse natural products that exhibit a wide array of anti-infective effects. The isolation of these chemical compounds over the span of more than forty years and the compounding evidence collected from biological and pharmacological investigations in support of the compounds’ pharmaceutical potential makes this intriguing cyanobacterium a significant target for biomedical research and novel drug leads development. Therefore, special attention should be given to the original source of such compounds when searching for medically or environmentally useful natural products. Therefore, a potential way to drug development from the marine cyanobacterium Lyngbya would be the optimization of its cultivation in the laboratory under the condition which would optimize the production of the desired biologically active metabolites. Due to the special supplies, which are required not only for cyanobacterial growth but also for the optimization of the production of cyanobacterial secondary metabolites, broad efforts are worried with this approach.
In summary, members of the Lyngbya morphotype have been exceptional sources of biosynthetic and biochemical novelty applied to drug discovery. Even facing significant headwinds, new discoveries from Lyngbya morphotype continue apace.

Author Contributions

Conceptualization, D.T.A.Y. and L.A.S.; formal analysis, L.A.S. and D.T.A.Y.; investigation, L.A.S. and D.T.A.Y.; resources, L.A.S. and D.T.A.Y.; data curation, D.T.A.Y. and L.A.S.; writing—original draft preparation, D.T.A.Y. and S.J.M.; Drawing chemical structures, D.T.A.Y., S.J.M. and A.A.B., writing—review and editing, D.T.A.Y. and L.A.S.; supervision, D.T.A.Y.; project administration, D.T.A.Y.; funding acquisition, D.T.A.Y. and L.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research work was funded by Institutional Fund Projects under grant no. (IFPRP: 174-166-1442).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This research work was funded by Institutional Fund Projects under grant no. (IFPRP: 174-166-1442). Therefore, authors gratefully acknowledge technical and financial support from the Ministry of Education and King Abdulaziz University, DSR, Jeddah, Saudi Arabia. We thank Mostafa Rateb and Kerry McPhail for providing some recent SciFinder search.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hayes, G.W.; Keating, C.L.; Newman, J.S. The golden anniversary of the silver bullet. JAMA 1993, 270, 1610–1611. [Google Scholar] [CrossRef]
  2. Kardos, N.; Demain, A.L. Penicillin: The medicine with the greatest impact on therapeutic outcomes. Appl. Microbiol. Biotechnol. 2011, 92, 677–687. [Google Scholar] [CrossRef]
  3. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef] [Green Version]
  4. Cooper, M.A.; Shlaes, D. Fix the antibiotics pipeline. Nature 2011, 472, 32. [Google Scholar] [CrossRef]
  5. Boucher, H.W.; Talbot, G.H.; Bradley, J.S.; Edwards, J.E.; Gilbert, D.; Rice, L.B.; Scheld, M.; Spellberg, B.; Bartlett, J. Bad bugs, no drugs: No ESKAPE! An update from the Infectious Diseases Society of America. Clin. Infect. Dis. 2009, 48, 1334. [Google Scholar] [CrossRef] [Green Version]
  6. Freire-Moran, L.; Aronsson, B.; Manz, C.; Gyssens, I.C.; So, A.D.; Monnet, D.L.; Cars, O.; ECDC-EMA Working Group. Critical shortage of new antibiotics in development against multidrug-resistant bacteria-time to react is now. Drug Resist. Updat. 2011, 14, 118–124. [Google Scholar] [CrossRef]
  7. Founou, R.C.; Founou, L.L.; Essack, S.Y. Clinical and economic impact of antibiotic resistance in developing countries: A systematic review and meta-analysis. PLoS ONE 2017, 12, e0189621. [Google Scholar] [CrossRef] [Green Version]
  8. Prestinaci, F.; Pezzotti, P.; Pantosti, A. Antimicrobial resistance: A global multifaceted phenomenon. Pathog. Glob. Health 2015, 109, 309–318. [Google Scholar] [CrossRef] [Green Version]
  9. Chokshi, A.; Sifri, Z.; Cennimo, D.; Horng, H. Global contributors to antibiotic resistance. J. Glob. Infect. Dis. 2019, 1, 36–42. [Google Scholar]
  10. Shrestha, P.; Cooper, B.S.; Coast, J.; Oppong, R.; Do Thi Thuy, N.; Phodha, T.; Celhay, O.; Guerin, P.J.; Wertheim, H.; Lubell, Y. Enumerating the economic cost of antimicrobial resistance per antibiotic consumed to inform the evaluation of interventions affecting their use. Antimicrob. Resist. Infect. Control 2018, 7, 98. [Google Scholar] [CrossRef] [Green Version]
  11. Llor, C.; Bjerrum, L. Antimicrobial resistance: Risk associated with antibiotic overuse and initiatives to reduce the problem. Ther. Adv. Drug Saf. 2014, 5, 229–241. [Google Scholar] [CrossRef] [PubMed]
  12. Antibiotic Resistance Threats in the United States; 2013. Available online: https://www.cdc.gov/drugresistance/pdf/ar-threats-2013-508.pdf (accessed on 25 October 2022).
  13. Bassetti, M.; Peghin, M.; Vena, A.; Giacobbe, D.R. Treatment of infections due to MDR gram-negative bacteria. Front Med. 2019, 6, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ramírez-Castillo, F.Y.; Moreno-Flores, A.C.; Avelar-González, F.J.; Márquez-Díaz, F.; Harel, J.; Guerrero-Barrera, A.L. An evaluation of multidrug-resistant Escherichia coli isolates in urinary tract infections from Aguascalientes, Mexico: Cross-sectional study. Ann. Clin. Microbiol. Antimicrob. 2018, 17, 34. [Google Scholar] [CrossRef] [PubMed]
  15. Annavajhala, M.K.; Gomez-Simmonds, A.; Uhlemann, A.-C. Multidrug-resistant Enterobacter cloacae complex emerging as a global, diversifying threat. Front. Microbiol. 2019, 10, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Chatterjee, S.; Poonawala, H.; Jain, Y. Drug-resistant tuberculosis: Is India ready for the challenge? BMJ Glob. Health 2018, 3, e000971. [Google Scholar] [CrossRef]
  17. Friedrich, M.J. Drug-resistant tuberculosis predicted to increase in high-burden countries. JAMA 2017, 318, 231. [Google Scholar] [CrossRef]
  18. Bloom, D.E.; Cadarette, D. Infectious disease threats in the twenty-first century: Strengthening the global response. Front. Immunol. 2019, 10, 549. [Google Scholar] [CrossRef] [Green Version]
  19. Luo, G.D.; Gao, S.-J. Global health concerns stirred by emerging viral infections. Med. Virol. 2020, 92, 399–400. [Google Scholar] [CrossRef]
  20. Piret, J.; Boivin, G. Pandemics throughout history. Front. Microbiol. 2021, 11, 3594. [Google Scholar] [CrossRef]
  21. Muñoz, L.S.; Garcia, M.A.; Gordon-Lipkin, E.; Parra, B.; Pardo, C.A. Emerging viral infections and their impact on the global burden of neurological disease. Semin. Neurol. 2018, 38, 163–175. [Google Scholar] [CrossRef]
  22. Bleibtreu, A.; Bertine, M.; Bertin, C.; Houhou-Fidouh, N.; Visseaux, B. Focus on Middle East respiratory syndrome coronavirus (MERS-CoV). Med. Mal. Infect. 2020, 50, 243–251. [Google Scholar] [CrossRef]
  23. Roychoudhury, S.; Das, A.; Sengupta, P.; Dutta, S.; Roychoudhury, S.; Choudhury, A.P.; Ahmed, A.B.F.; Bhattacharjee, S.; Slama, P. Viral pandemics of the last four decades: Pathophysiology, health impacts and perspectives. Int. J. Environ. Res. Public Health 2020, 17, 9411. [Google Scholar] [CrossRef]
  24. Schaefer, T.J.; Panda, P.K.; Wolford, R.W. Dengue Fever; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  25. Bhatt, S.; Gething, P.W.; Brady, O.J.; Messina, J.P.; Farlow, A.W.; Moyes, C.L.; Drake, J.M.; Brownstein, J.S.; Hoen, A.G.; Sankoh, O.; et al. The global distribution and burden of dengue. Nature 2013, 496, 504–507. [Google Scholar] [CrossRef] [Green Version]
  26. Powell, J.R.; Human, M.-B. Viral diseases: Why Aedes aegypti? Am. J. Trop. Med. Hyg. 2018, 8, 1563–1565. [Google Scholar] [CrossRef]
  27. Singh, R.; Chauhan, N.; Kuddus, M. Exploring the therapeutic potential of marine-derived bioactive compounds against COVID-19. Environ. Sci. Pollut. Res. Int. 2021, 28, 52798–52809. [Google Scholar] [CrossRef]
  28. Schopf, J.W.; Packer, B.M. Early Archean (3.3-billion to 3.5-billion-yearold) microfossils from Warrawoona Group, Australia. Science 1987, 237, 70–73. [Google Scholar] [CrossRef]
  29. Whitton, B.A.; Potts, M. The Ecology of Cyanobacteria: Their Diversity in Time and Space; Springer: Dordrecht, The Netherlands, 2000. [Google Scholar]
  30. Nagle, D.G.; Paul, V.J. Production of secondary metabolites by filamentous tropical marine cyanobacteria: Ecological functions of the compounds. J. Phycol. 1999, 35, 1412–1421. [Google Scholar]
  31. Berry, J.P.; Gantar, M.; Perez, M.H.; Berry, G.; Noriega, F.G. Cyanobacterial toxins as allelochemicals with potential applications as algaecides, herbicides and insecticides. Mar. Drugs 2008, 6, 117–146. [Google Scholar] [CrossRef]
  32. Burja, A.M.; Banaigs, B.; Abou-Mansour, E.; Burgess, J.G.; Wright, P.C. Marine cyanobacteria—A prolific source of natural products. Tetrahedron 2001, 57, 9347–9377. [Google Scholar] [CrossRef]
  33. Engene, N.; Rottacker, E.C.; Kas, J.; Byrum, T.; Gerwick, W.H.; Choi, H.; Ellisman, M.H. Moorea producens Gen. Nov., Sp. Nov. and Moorea bouillonii Comb. Nov., Tropical marine cyanobacteria rich in bioactive secondary metabolites. Int. J. Syst. Evol. Microbiol. 2012, 62 Pt 5, 1171–1178. [Google Scholar]
  34. Engene, N.; Tronholm, A.; Paul, V.J. Uncovering cryptic diversity of Lyngbya: The new tropical marine cyanobacterial genus Dapis (Oscillatoriales). J. Phycol. 2018, 54, 435–446. [Google Scholar] [CrossRef] [PubMed]
  35. Jiří, K.; Eliška, Z.; Jan, Š.; Jiří, K.; Jason, W.; Brett, A.N.; Jaroslava, K. Polyphasic evaluation of Limnoraphis robusta, a water—Bloom forming cyanobacterium from Lake Atitlán, Guatemala, with a description of Limnoraphis gen. nov. J. Czech Phycol. Soc. 2013, 13, 39–52. [Google Scholar]
  36. Tronholm, A.; Engene, N. Moorena Gen. Nov., a valid name for “Moorea Engene & Al.” Nom. Inval. (Oscillatoriaceae, Cyanobacteria). Not. Algarum 2019, 122, 1–2. [Google Scholar]
  37. Mcgregor, G.B.; Sendall, B.C. Phylogeny and toxicology of Lyngbya wollei (Cyanobacteria, Oscillatoriales) from North-Eastern Australia, with description of Microseira gen. nov. J. Phycol. 2015, 51, 109–119. [Google Scholar] [CrossRef] [PubMed]
  38. Engene, N.; Paul, V.J.; Byrum, T.; Gerwick, W.H.; Thor, A.; Ellisman, M.H. Five chemically rich species of tropical marine cyanobacteria of the genus Okeania gen. nov. (Oscillatoriales, Cyanoprokaryota). J. Phycol. 2013, 49, 1095–1106. [Google Scholar] [CrossRef]
  39. Marine Pharmacology. Available online: https://www.marinepharmacology.org/ (accessed on 1 November 2022).
  40. Cardllina, J.H.C.; Moore, R.E.; Arnold, E.V.; Clardy, J. Structure and absolute configuration of malyngolide, an antibiotic from the marine blue-green alga Lyngbya majuscula Gomont. J. Org. Chem. 1979, 44, 4039–4042. [Google Scholar] [CrossRef]
  41. Shaala, L.A.; Youssef, D.T.A.; McPhail, K.L.; Elbandy, M. Malyngamide 4, a new lipopeptide from the Red Sea marine cyanobacterium Moorea producens (Formerly Lyngbya majuscula). Phytochem. Lett. 2013, 6, 183–188. [Google Scholar] [CrossRef]
  42. Gekwick, W.H.; Reyes, S.; Alvarado, B. Two malyngamides from the Caribbean cyanobacterium Lyngbya majuscula. Phytochemistry 1987, 26, 1701–1704. [Google Scholar] [CrossRef]
  43. Luesch, H.; Pangilinan, R.; Yoshida, W.Y.; Moore, R.E.; Paul, V.J. Pitipeptolides A and B, new cyclodepsipeptides from the marine cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2001, 64, 304–307. [Google Scholar] [CrossRef]
  44. Montaser, R.; Paul, V.J.; Luesch, H. Pitipeptolides C-F, antimycobacterial cyclodepsipeptides from the marine cyanobacterium Lyngbya majuscula from Guam. Phytochemistry 2011, 72, 2068–2074. [Google Scholar] [CrossRef] [Green Version]
  45. Montaser, R.; Abboud, K.A.; Paul, V.J.; Luesch, H. Pitiprolamide, a proline-rich dolastatin 16 analogue from the marine cyanobacterium Lyngbya majuscula from Guam. J. Nat. Prod. 2011, 74, 109–112. [Google Scholar] [CrossRef] [Green Version]
  46. Zainuddin, E.N.; Jansen, R.; Nimtz, M.; Wray, V.; Preisitsch, M.; Lalk, M.; Mundt, S. Lyngbyazothrins A-D, antimicrobial cyclic undecapeptides from the cultured cyanobacterium Lyngbya sp. J. Nat. Prod. 2009, 72, 2080. [Google Scholar] [CrossRef] [Green Version]
  47. Levert, A.; Alvarin, R.; Bornancin, L.; Mansour, E.A.; Burja, A.M.; Genevie, A.; Bonnard, I.; Alonso, E.; Botana, L.; Banaigs, B. Structures and activities of tiahuramides A−C, cyclic depsipeptides from a Tahitian collection of the marine cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2018, 81, 1301–1310. [Google Scholar] [CrossRef]
  48. Kearns, D.B. A field guide to bacterial swarming motility. Nat. Rev. Microbiol. 2010, 8, 634–644. [Google Scholar] [CrossRef] [Green Version]
  49. Guttenplan, S.B.; Kearns, D.B. Regulation of flagellar motility during biofilm formation Sarah. FEMS Microbiol. Rev. 2013, 37, 849–871. [Google Scholar] [CrossRef]
  50. Tripathi, A.; Puddick, J.; Prinsep, M.R.; Rottmann, M.; Tan, L.T. Lagunamides A and B: Cytotoxic and antimalarial cyclodepsipeptides from the marine Cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2010, 73, 1810–1814. [Google Scholar] [CrossRef]
  51. Tripathi, A.; Puddick, J.; Prinsep, M.R.; Rottmann, M.; Chan, K.P.; Chen, D.Y.K.; Tan, L.T. Lagunamide C, a cytotoxic cyclodepsipeptide from the marine cyanobacterium Lyngbya majuscula. Phytochemistry 2011, 72, 2369–2375. [Google Scholar] [CrossRef]
  52. Rémy, B.; Mion, S.; Plener, L.; Elias, M.; Chabrière, E.; Daudé, D. Interference in bacterial quorum sensing: A biopharmaceutical perspective. Front. Pharmacol. 2018, 9, 203. [Google Scholar] [CrossRef]
  53. Zhao, X.; Yu, Z.; Ding, T. Quorum-sensing regulation of antimicrobial resistance in bacteria. Microorganisms 2020, 8, 425. [Google Scholar] [CrossRef] [Green Version]
  54. Kwan, J.C.; Teplitski, M.; Gunasekera, S.P.; Paul, V.J.; Luesch, H. Isolation and biological evaluation of 8-epi-malyngamide C from the Floridian marine cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2010, 73, 463–466. [Google Scholar] [CrossRef] [Green Version]
  55. Dobretsov, S.; Teplitski, M.; Alagely, A.; Gunasekera, S.P.; Paul, V.J. Malyngolide from the cyanobacterium Lyngbya majuscula interferes with quorum sensing circuitry. Environ. Microbiol. Rep. 2010, 2, 739–744. [Google Scholar] [CrossRef] [PubMed]
  56. Kwan, J.C.; Meickle, T.; Ladwa, D.; Teplitski, M.; Paul, V.; Luesch, H. Lyngbyoic acid, a “Tagged” fatty acid from a marine cyanobacterium, disrupts quorum sensing in Pseudomonas aeruginosa. Mol. Biosyst. 2011, 7, 1205–1216. [Google Scholar] [CrossRef] [PubMed]
  57. Liang, X.; Matthew, S.; Chen, Q.Y.; Kwan, J.C.; Paul, V.J.; Luesch, H. Discovery and total synthesis of doscadenamide A: A quorum sensing signaling molecule from a marine cyanobacterium. Org. Lett. 2019, 21, 7274–7278. [Google Scholar] [CrossRef] [PubMed]
  58. Carter, D.C.; Moore, R.E.; Mynderse, J.S.; Niemczura, W.P.; Todd, J.S. Structure of majusculamide C, a cyclic depsipeptide from Lyngbya majuscula. J. Org. Chem. 1984, 49, 236–241. [Google Scholar] [CrossRef]
  59. Mynderse, J.S.; Hunt, A.H.; Moore, R.E. 57-Normajusculamide C, a minor cyclic depsipeptide isolated from Lyngbya majuscula. J. Nat. Prod. 1988, 51, 1299–1301. [Google Scholar] [CrossRef]
  60. Meickle, T.; Matthew, S.; Ross, C.; Luesch, H.; Paul, V. Bioassay-guided isolation and identification of desacetylmicrocolin B from Lyngbya Cf. Polychroa. Planta Med. 2009, 75, 1427–1430. [Google Scholar] [CrossRef] [Green Version]
  61. Bonnard, I.; Rolland, M.; Francisco, C.; Banaigs, B. Total Structure and biological properties of laxaphycins A and B, cyclic lipopeptides from the marine cyanobacterium Lyngbya majuscula. Int. J. Pept. Res. Ther. 1997, 4, 289–292. [Google Scholar] [CrossRef]
  62. Singh, I.P.; Milligan, K.E.; Gerwick, W.H. Tanikolide, a toxic and antifungal lactone from the marine cyanobacterium Lyngbya majuscula. J. Nat. Prod. 1999, 62, 1333–1335. [Google Scholar] [CrossRef]
  63. Milligan, K.E.; Marquez, B.L.; Williamson, R.T.; Gerwick, W.H. Lyngbyabellin B, a toxic and antifungal secondary metabolite from the marine cyanobacterium Lyngbya mojuscula. J. Nat. Prod. 2000, 63, 1440–1443. [Google Scholar] [CrossRef]
  64. Marquez, B.L.; Watts, K.S.; Yokochi, A.; Roberts, M.A.; Verdier-Pinard, P.; Jimenez, J.I.; Hamel, E.; Scheuer, P.J.; Gerwick, W.H. Structure and absolute stereochemistry of hectochlorin, a potent stimulator of actin assembly. J. Nat. Prod. 2002, 65, 866–871. [Google Scholar] [CrossRef]
  65. MacMillan, J.B.; Ernst-Russell, M.A.; De Ropp, J.S.; Molinski, T.F. Lobocyclamides A-C, lipopeptides from a cryptic cyanobacterial mat containing Lyngbya confervoides. J. Org. Chem. 2002, 67, 8210–8215. [Google Scholar] [CrossRef]
  66. McPhail, K.L.; Correa, J.; Linington, R.G.; González, J.; Ortega-Barría, E.; Capson, T.L.; Gerwick, W.H. Antimalarial linear lipopeptides from a Panamanian strain of the marine cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2007, 70, 984–988. [Google Scholar] [CrossRef] [Green Version]
  67. Balunas, M.J.; Linington, R.G.; Tidgewell, K.; Fenner, A.M.; Ureña, L.D.; Togna, G.D.; Kyle, D.E.; Gerwick, W.H. Dragonamide E, a modified linear lipopeptide from Lyngbya majuscula with antileishmanial activity. J. Nat. Prod. 2010, 73, 60–66. [Google Scholar] [CrossRef] [Green Version]
  68. Gutiérrez, M.; Tidgewell, K.; Capson, T.L.; Engene, N.; Almanza, A.; Schemies, J.; Jung, M.; Gerwick, W.H. Malyngolide dimer, a bioactive symmetric cyclodepside from the Panamanian marine cyanobacterium Lyngbya majuscula. J. Nat. Prod. 2010, 73, 709–711. [Google Scholar] [CrossRef] [Green Version]
  69. Nozaki, T.; Saito-nakano, Y.; Suenaga, K. Ikoamide, an antimalarial lipopeptide from an Okeania sp. marine cyanobacterium. J. Nat. Prod. 2020, 83, 481–488. [Google Scholar]
  70. Ozaki, K.; Iwasaki, A.; Sezawa, D.; Fujimura, H.; Nozaki, T.; Saito-nakano, Y.; Suenaga, K.; Teruya, T. Isolation and total synthesis of mabuniamide, a lipopeptide from an Okeania sp. marine cyanobacterium. J. Nat. Prod. 2019, 82, 2907–2915. [Google Scholar] [CrossRef]
  71. Shao, C.; Mou, X.; Cao, F.; Spadafora, C.; Glukhov, E.; Gerwick, L.; Wang, C.; Gerwick, W.H. Bastimolide B, an antimalarial 24-membered marine macrolide possessing a tert-butyl group. J. Nat. Prod. 2018, 81, 211–215. [Google Scholar] [CrossRef]
  72. Shao, C.L.; Linington, R.G.; Balunas, M.J.; Centeno, A.; Boudreau, P.; Zhang, C.; Engene, N.; Spadafora, C.; Mutka, T.S.; Kyle, D.E.; et al. Bastimolide A, a potent antimalarial polyhydroxy macrolide from the marine cyanobacterium Okeania hirsuta. J. Org. Chem. 2015, 80, 7849–7855. [Google Scholar] [CrossRef]
  73. Fathoni, I.; Petitbois, J.G.; Alarif, W.M.; Abdel-Lateff, A.; Al-lihaibi, S.S.; Yoshimura, E.; Nogata, Y.; Vairappan, C.S.; Sholikhah, E.N.; Okino, T. Bioactivities of lyngbyabellins from cyanobacteria of Moorea and Okeania genera. Molecules 2020, 5, 3986. [Google Scholar] [CrossRef]
  74. Sanchez, L.M.; Lopez, D.; Vesely, B.A.; Della Togna, G.; Gerwick, W.H.; Kyle, D.E.; Linington, R.G. Almiramides A-C: Discovery and development of a new class of leishmaniasis lead compounds. J. Med. Chem. 2010, 53, 4187–4197. [Google Scholar] [CrossRef] [Green Version]
  75. Almaliti, J.; Malloy, K.L.; Glukhov, E.; Spadafora, C.; Gutiérrez, M.; Gerwick, W.H. Dudawalamides A-D, antiparasitic cyclic depsipeptides from the marine cyanobacterium Moorea producens. J. Nat. Prod. 2017, 80, 1827–1836. [Google Scholar] [CrossRef] [PubMed]
  76. Kurisawa, N.; Iwasaki, A.; Jeelani, G.; Nozaki, T.; Suenaga, K. Iheyamides A-C, antitrypanosomal linear peptides isolated from a marine Dapis sp. cyanobacterium. J. Nat. Prod. 2020, 83, 1684–1690. [Google Scholar] [CrossRef] [PubMed]
  77. Ogawa, H.; Iwasaki, A.; Sumimoto, S.; Kanamori, Y.; Ohno, O.; Iwatsuki, M.; Ishiyama, A.; Hokari, R.; Otoguro, K.; Omura, S.; et al. Janadolide, a cyclic polyketide-peptide hybrid possessing a tert-butyl group from an Okeania sp. marine cyanobacterium. J. Nat. Prod. 2016, 79, 1862–1866. [Google Scholar] [CrossRef] [PubMed]
  78. Taguchi, R.; Iwasaki, A.; Ebihara, A.; Jeelani, G.; Nozaki, T.; Suenaga, K. Isolation and total synthesis of beru’amide, an antitrypanosomal polyketide from a marine cyanobacterium Okeania sp. Org. Lett. 2022, 24, 4710–4714. [Google Scholar] [CrossRef] [PubMed]
  79. Gustafson, K.R.; Cardellina, J.H., 2nd; Fuller, R.W.; Weislow, O.S.; Kiser, R.F.; Snader, K.M.; Patterson, G.M.; Boyd, M.R. AIDS-antiviral sulfolipids from cyanobacteria (blue-green algae). J. Natl. Cancer Inst. 1989, 81, 1254–1258. [Google Scholar] [CrossRef]
  80. Reshef, V.; Mizrachi, E.; Maretzki, T.; Silberstein, C.; Loya, S.; Hizi, A.; Carmeli, S. New acylated sulfoglycolipids and digalactolipids and related known glycolipids from cyanobacteria with a potential to inhibit the reverse transcriptase of HIV-1. J. Nat. Prod. 1997, 60, 1251–1260. [Google Scholar] [CrossRef]
  81. Loya, S.; Reshef, V.; Mizrachi, E.; Silberstein, C.; Rachamim, Y.; Carmeli, S.; Hizi, A. The inhibition of the reverse transcriptase of HIV-1 by the natural sulfoglycolipids from cyanobacteria: Contribution of different moieties to their high potency. J. Nat. Prod. 1998, 61, 891–895. [Google Scholar] [CrossRef]
  82. Orjala, J.; Gerwick, W.H. Barbamide, a chlorinated metabolite with molluscicidal activity from the Caribbean cyanobacterium Lyngbya majuscula. J. Nat. Prod. 1996, 59, 427–430. [Google Scholar] [CrossRef]
  83. Pereira, A.R.; McCue, C.F.; Gerwick, W.H. Cyanolide A, a glycosidic macrolide with potent molluscicidal activity from the Papua New Guinea cyanobacterium Lyngbya bouillonii. J. Nat. Prod. 2010, 73, 217–220. [Google Scholar] [CrossRef]
  84. Iguchi, K.; Satake, M.; Nishio, Y.; Zhang, B.; Kawashima, K.; Uchida, H.; Nagai, H. Debromooscillatoxins G and I from the cyanobacterium Moorea Producens. Heterocycles 2021, 102, 1287–1293. [Google Scholar]
Figure 1. A map with red dots indicating collection locations for Lyngbya-morphotype described in this review.
Figure 1. A map with red dots indicating collection locations for Lyngbya-morphotype described in this review.
Marinedrugs 20 00768 g001
Figure 2. Number of reported anti-infective compounds related to collection site.
Figure 2. Number of reported anti-infective compounds related to collection site.
Marinedrugs 20 00768 g002
Figure 3. Chemical structures of compounds 13.
Figure 3. Chemical structures of compounds 13.
Marinedrugs 20 00768 g003
Figure 4. Chemical structures of compounds 49.
Figure 4. Chemical structures of compounds 49.
Marinedrugs 20 00768 g004
Figure 5. Chemical structure of compound 10.
Figure 5. Chemical structure of compound 10.
Marinedrugs 20 00768 g005
Figure 6. Chemical structures of compounds 1113.
Figure 6. Chemical structures of compounds 1113.
Marinedrugs 20 00768 g006
Figure 7. Chemical structures of compounds 1417.
Figure 7. Chemical structures of compounds 1417.
Marinedrugs 20 00768 g007
Figure 8. Chemical structures of compounds 1820.
Figure 8. Chemical structures of compounds 1820.
Marinedrugs 20 00768 g008
Figure 9. Chemical structures of compounds 2123.
Figure 9. Chemical structures of compounds 2123.
Marinedrugs 20 00768 g009
Figure 10. Chemical structures of compounds 24 and 25.
Figure 10. Chemical structures of compounds 24 and 25.
Marinedrugs 20 00768 g010
Figure 11. Chemical structures of compounds 26 and 27.
Figure 11. Chemical structures of compounds 26 and 27.
Marinedrugs 20 00768 g011
Figure 12. Chemical structures of compounds 28 and 29.
Figure 12. Chemical structures of compounds 28 and 29.
Marinedrugs 20 00768 g012
Figure 13. Chemical structures of compounds 30 and 31.
Figure 13. Chemical structures of compounds 30 and 31.
Marinedrugs 20 00768 g013
Figure 14. Chemical structures of compounds 32 and 33.
Figure 14. Chemical structures of compounds 32 and 33.
Marinedrugs 20 00768 g014
Figure 15. Chemical structures of compounds 3436.
Figure 15. Chemical structures of compounds 3436.
Marinedrugs 20 00768 g015
Figure 16. Chemical structures of compounds 3739.
Figure 16. Chemical structures of compounds 3739.
Marinedrugs 20 00768 g016
Figure 17. Chemical structures of compounds 4042.
Figure 17. Chemical structures of compounds 4042.
Marinedrugs 20 00768 g017
Figure 18. Chemical structures of compounds 33 and 34.
Figure 18. Chemical structures of compounds 33 and 34.
Marinedrugs 20 00768 g018
Figure 19. Chemical structure of compound 45.
Figure 19. Chemical structure of compound 45.
Marinedrugs 20 00768 g019
Figure 20. Chemical structure of compound 46.
Figure 20. Chemical structure of compound 46.
Marinedrugs 20 00768 g020
Figure 21. Chemical structures of compounds 4749.
Figure 21. Chemical structures of compounds 4749.
Marinedrugs 20 00768 g021
Figure 22. Chemical structures of compounds 5052.
Figure 22. Chemical structures of compounds 5052.
Marinedrugs 20 00768 g022
Figure 23. Chemical structures of compounds 53 and 54.
Figure 23. Chemical structures of compounds 53 and 54.
Marinedrugs 20 00768 g023
Figure 24. Chemical structures of compounds 5557.
Figure 24. Chemical structures of compounds 5557.
Marinedrugs 20 00768 g024
Figure 25. Chemical structures of compounds 5861.
Figure 25. Chemical structures of compounds 5861.
Marinedrugs 20 00768 g025
Figure 26. Chemical structures of compounds 6264.
Figure 26. Chemical structures of compounds 6264.
Marinedrugs 20 00768 g026
Figure 27. Chemical structures of compounds 65 and 66.
Figure 27. Chemical structures of compounds 65 and 66.
Marinedrugs 20 00768 g027
Figure 28. Chemical structures of compounds 67 and 68.
Figure 28. Chemical structures of compounds 67 and 68.
Marinedrugs 20 00768 g028
Figure 29. Chemical structure of compound 69.
Figure 29. Chemical structure of compound 69.
Marinedrugs 20 00768 g029
Figure 30. Chemical structure of compound 70.
Figure 30. Chemical structure of compound 70.
Marinedrugs 20 00768 g030
Figure 31. Chemical structures of compounds 71 and 72.
Figure 31. Chemical structures of compounds 71 and 72.
Marinedrugs 20 00768 g031
Figure 32. Number of investigated Lyngbya-morphotype and reported anti-infective compounds over time.
Figure 32. Number of investigated Lyngbya-morphotype and reported anti-infective compounds over time.
Marinedrugs 20 00768 g032
Figure 33. Number of reported anti-infective compounds per Lyngbya morphotype.
Figure 33. Number of reported anti-infective compounds per Lyngbya morphotype.
Marinedrugs 20 00768 g033
Figure 34. Distribution of the nitrogenous and non-nitrogenous compounds in Lyngbya morpho-type.
Figure 34. Distribution of the nitrogenous and non-nitrogenous compounds in Lyngbya morpho-type.
Marinedrugs 20 00768 g034
Figure 35. Number of reported compounds associated with biological activities.
Figure 35. Number of reported compounds associated with biological activities.
Marinedrugs 20 00768 g035
Table 1. Number of reported anti-infective compounds from Lyngbya morphotype.
Table 1. Number of reported anti-infective compounds from Lyngbya morphotype.
Species Name aNumber of Reported Compounds
Dapis sp.3
Lyngbya sp.4
Lyngbya bouillonii1
Lyngbya confervoides3
Lyngbya lagerheimii2
Lyngbya majuscula35
Lygnbya polychora2
Moorea bouillonii4
Moorea producens11
Okeania sp.5
Okeania hirsuta2
a Names are given as reported in the original manuscripts.
Table 2. Compounds with reported antibacterial activities.
Table 2. Compounds with reported antibacterial activities.
CompoundSource OrganismCollection SiteTargeted BacteriaMIC/Inhibition Zone/IC50Reference
Malyngolide (1)L. majusculaHawaii, USAM. smegmatis, S. pyogenes, S. aureus and B. subtilisMore active against M. smegmatis and S. pyogenes than S. aureus and B. subtilis[40]
Lyngbic acid (2)M. producensRed SeaM. tuberculosis H37Rv65% inhibition at 12.5 μg/mL[41]
Lyngbic acid (2)L. majusculaCaribbean regionS. aureus and B. subtilisAntibacterial activity[42]
Malyngamide D acetate (3)L. majusculaCaribbean regionS. aureusSlight activity[42]
Pitipeptolide A (4)L. majusculaGuamM. tuberculosis ATCC 2517725 mm at 100 µg
10 mm at 25 µg
[43]
Pitipeptolide A (4)L. majusculaGuamM. tuberculosis ATCC 3581815 mm at 100 µg
9 mm at 25 µg
[43]
Pitipeptolide B (5)L. majusculaGuamM. tuberculosis ATCC 2517730 mm at 100 µg
15 mm at 25 µg
[43]
Pitipeptolide B (5)L. majusculaGuamM. tuberculosis ATCC 3581815 mm at 100 µg
10 mm at 25 µg
[43]
Pitipeptolide A (4)L. majusculaGuamM. tuberculosis ATCC 2517728 mm at 100 µg
23 mm at 50 µg
9 mm at 10 µg
[44]
Pitipeptolide B (5)L. majusculaGuamM. tuberculosis ATCC 2517730 mm at 100 µg
24 mm at 50 µg
14 mm at 10 µg
[44]
Pitipeptolide C (6)L. majusculaGuamM. tuberculosis ATCC 2517726 mm at 100 µg
21 mm at 50 µg
18 mm at 10 µg
[44]
Pitipeptolide D (7)L. majusculaGuamM. tuberculosis ATCC 2517710 mm at 100 µg
0 mm at 50 µg
0 mm at 10 µg
[44]
Pitipeptolide E (8)L. majusculaGuamM. tuberculosis ATCC 2517721 mm at 100 µg
15 mm at 50 µg
0 mm at 10 µg
[44]
Pitipeptolide F (9)L. majusculaGuamM. tuberculosis ATCC 2517740 mm at 100 µg
30 mm at 50 µg
10 mm at 10 µg
[44]
Pitiprolamide (10)L. majusculaGuamM. tuberculosis ATCC 2517723 mm at 100 µg
13 mm at 50 µg
0 mm at 10 µg
[45]
Pitiprolamide (10)L. majusculaGuamB. cereus ATCC 10987IC50 = 70 μM at 1 μM[45]
Mixture of lyngbyazothrins A and B (14 and 15)Lyngbya sp.Germany (Culture)M. flaVus SBUG 168 mm at 100 μg/disk[46]
Mixture of lyngbyazothrins C (16) and D (17)Lyngbya sp.Germany (Culture)B. subtilis SBUG 14
E. coli ATCC 11229
E. coli SBUG 13
P. aeruginosa ATCC 27853
S. marcescens SBUG 9
18 mm at 25 μg/disk
18 mm at 100 μg/disk
15 mm at 100 μg/disk
8 mm at 100 μg/disk
8 mm at 200 μg/disk
[46]
Tiahuramide A (18)L. majusculaFrench PolynesiaA. salmonicida (CIP 103209T strain), V. anguillarum (CIP 63.36T), S. baltica (CIP 105850T), E. coli (CIP 54.8) and M. luteus (CIP A270)MIC = 27, 33, >50, 35 and 47 μM, respectively[47]
Tiahuramide B (19)L. majusculaFrench PolynesiaA. salmonicida (CIP 103209T strain), V. anguillarum (CIP 63.36T), S. baltica (CIP 105850T), E. coli (CIP 54.8) and M. luteus (CIP A270)MIC = 9.4, 8.5, 22, 12 and 29 μM, respectively[47]
Tiahuramide C (20)L. majusculaFrench PolynesiaA. salmonicida (CIP 103209T strain), V. anguillarum (CIP 63.36T), S. baltica (CIP 105850T), E. coli (CIP 54.8) and M. luteus (CIP A270)MIC = 6.7, 7.4, 16, 14 and 17 μM, respectively[47]
Table 3. Compounds with reported anti-swarming and anti-quorum sensing activities.
Table 3. Compounds with reported anti-swarming and anti-quorum sensing activities.
CompoundSource
Organism
Collection SiteTargeted Bacteria/ReceptorAnti-Swarming/Anti-Quorum SensingReference
Lagunamide A (21)L. majusculaSingaporeP. aeruginosa PA01Anti-swarming effect: 62% at 100 ppm[50,51]
Lagunamide B (22)L. majusculaSingaporeP. aeruginosa PA01Anti-swarming effect: 56% at 100 ppm[50,51]
Lagunamide C (23)L. majusculaSingaporeP. aeruginosa PA01Anti-swarming effect: 49%, at 100 ppm[50,51]
Malyngamide C (24)L. majusculaFlorida, USA3-oxo-C12-HSL (N-3-oxo-dodecanoyl-L-homoserine lactone) signaling in a LasR-based quorum sensing (QS) reporter pSB1075QS inhibitor reduction in 3-oxo-C12-HSL signaling at 10, 100 and 1000 µM[54]
8-epi-Malyngamide C (25)L. majusculaFlorida, USA3-oxo-C12-HSL (N-3-oxo-dodecanoyl-L-homoserine lactone) signaling in a LasR-based quorum sensing (QS) reporter pSB1075QS inhibitor reduction in 3-oxo-C12-HSL signaling at 10, 100 and 1000 µM[54]
Malyngolide (1)L. majusculaFlorida, USAProduction of violacein pigment by C. violaceum CV017 in the QS bioassayQS inhibitor inhibition of violacein production with effective concentrations ranged from 0.07 to 0.22 mM; EC50 = 0.11 mM[55]
Responses of lasR+PlasI-luxCDABE reporter pSB1075 in the presence of 14 µM of 3-oxo-C12-HSLInhibition of responses of the lasR+PlasI-luxCDABE reporter pSB1075 with concentrations ranging from 3.57 to 57; EC50 = 12.2 µM[55]
Production of elastase by P. aeruginosa PAO1 (an extracellular enzyme regulated by 3-oxo-C12-HSL and LasR)Significant reduction in elastase production; EC50 = 10.6 µM, at higher concentrations of MAL, elastase production was inhibited to the level observed in the QS mutant of P. aeruginosa JP2[55]
Lyngbyoic acid (26)L. majusculaFlorida, USAFour reporters based on different acylhomoserine lactone (AHL) receptors acylhomoserine lactone (AHL) receptors (LuxR, AhyR, TraR and LasR)QS inhibitor, most effective inhibition against LasR reporter[56]
Production of pyocyanin and elastase (LasB) both on the protein and transcript level in wild-type P. aeruginosa.Reduction in the production of pyocyanin and elastase (LasB) and direct inhibition of LasB enzymatic activity; Ki = 5.4 mM
Doscadenamide A (27)L. bouilloniiGuam3-Oxo-C12-HSL-responsive reporter plasmid pSB1075, which encodes LasR and contains a light-producing luxCDABE cassette expressed in E. coliQS agonist in a LasR-dependent manner and activation of 3-oxo-C12-HSL-responsive reporter plasmid pSB1075[57]
Production of QS pigment pyocyanin in wild-type P. aeruginosaIncrease pyocyanin production at 10 µM
Table 4. Compounds with reported antifungal activity.
Table 4. Compounds with reported antifungal activity.
CompoundSource
Organism
Collection SiteTargeted FungiMIC/Inhibition Zone/LD50Reference
Majusculamide C (28)L. majusculaMarshall IslandsP. infestans and P. viticolaGrowth inhibition[58]
57-Normajusculamide C (29)L. majusculaMarshall IslandsS. pastorianusAntimycotic activity[59]
Microcolin A (30)L. polychroaMarshall IslandsD. salina (SIO and EBGJ strains)LD50 = >200 μg/mL[60]
Microcolin B (31)L. polychroaMarshall IslandsD. salina (SIO and EBGJ strains)LD50 = >200 μg/mL[60]
Laxaphycin B (32)L. majusculaFrench PolynesiaC. albicansAntifungal activity[61]
Mixture of laxaphycins A (33) and B (32)L. majusculaFrench PolynesiaC. albicansLaxaphycin B produces synergetic effect to the inactive laxaphycin A
Tanikolide (34)L. majusculaMadagascarC. albicans13 mm at 100 µg/disk[62]
Lyngbyabellin B (35)L. majusculaFlorida, USAC. albicans (ATCC 14053)10.5 mm at 100 µg/disk[63]
Hectochlorin (36)L. majusculaJamaicaC. albicans (ATCC 14053)16 mm at 100 µg/disk
11 mm at 10 µg/disk
[64]
Lobocyclamide A (37)L. confervoidesSouthern BahamasC. albicans 96–489 (Fluconazole-resistant)7 mm at 150 µg/disk and MIC = 100 µg/mL[65]
Lobocyclamide B (38)L. confervoidesSouthern BahamasC. albicans 96–489 (Fluconazole-resistant)8 mm at 150 µg/disk and MIC = 30–100 µg/mL[65]
Lobocyclamide B (38)L. confervoidesSouthern BahamasC. glabrata6 mm at 150 µg/disk[65]
Mixture of lobocyclamides A and B (37 and 38)L. confervoidesSouthern Bahamas-MIC = 10–30 µg/mL[65]
Lobocyclamide C (39)L. confervoidesSouthern BahamasC. albicans 96–489 (Fluconazole-resistant)10 mm at 150 µg/disk[65]
Lobocyclamides C (39)L. confervoidesSouthern BahamasC. glabrata8 mm at 150 µg/disk[65]
Table 5. Compounds with reported antiparasitic activities.
Table 5. Compounds with reported antiparasitic activities.
CompoundSource OrganismCollection SiteTargeted Microbe/ParasiteIC50/% of InhibitionReference
Lagunamide A (21)L. majusculaSingaporeP. falciparum (NF54 strain)IC50 = 0.19 μM[50,51]
Lagunamide B (22)L. majusculaSingaporeP. falciparum (NF54 strain)IC50 = 0.91 μM[50,51]
Lagunamide C (23)L. majusculaSingaporeP. falciparum (NF54 strain)IC50 = 0.29 μM[50,51]
Carmabin A (40)L. majusculaPanamaP. falciparum (Indochina W2 strain)IC50 = 4.3 µM[66,67]
Dragomabin (41)L. majusculaPanamaP. falciparum (Indochina W2 strain)IC50 = 6.0 µM[66,67]
Dragonamide A (42)L. majusculaPanamaP. falciparum (Indochina W2 strain)IC50 = 7.7 µM[66,67]
Dragonamide A (42)L. majusculaPanamaL. donovani (LD-1S/MHOM/SD/00-strain 1S)IC50 = 6.5 μM[67]
Malyngolide dimer (44)L. majusculaPanamaP. falciparum (W2 strain)IC50 = 19 μM[68]
Dragonamide E (53)L. majusculaPanamaL. donovani (LD-1S/MHOM/SD/00-strain 1S)IC50 = 5.1 μM[67]
Herbamide B (54)L. majusculaPanamaL. donovani (LD-1S/MHOM/SD/00-strain 1S)IC50 = 5.9 μM[67]
Almiramide B (55)L. majusculaPanamaL. donovani (LD-1S/MHOM/SD/00-strain 1S)IC50 = 2.4 μM[74]
Almiramide C (56)L. majusculaPanamaL. donovani (LD-1S/MHOM/SD/00-strain 1S)IC50 = 1.9 μM[74]
Dudawalamide A (58)M. producensPapua New GuineaP. falciparumIC50 = 3.6 μM[75]
Dudawalamide A (58)M. producensPapua New GuineaT. cruzi12% inhibition at 10 μg/mL[75]
Dudawalamide A (58)M. producensPapua New GuineaL. donovaniIC50 = >10 μM[75]
Dudawalamide B (59)M. producensPapua New GuineaP. falciparumIC50 = 10 μM[75]
Dudawalamide B (59)M. producensPapua New GuineaT. cruzi7% inhibition at 10 μg/mL[75]
Dudawalamide B (59)M. producensPapua New GuineaL. donovaniIC50 >10 μM[75]
Dudawalamide C (60)M. producensPapua New GuineaP. falciparumIC50 = 3.5 μM[75]
Dudawalamide D (61)M. producensPapua New GuineaP. falciparumIC50 = 8.0 μM[75]
Dudawalamide D (61)M. producensPapua New GuineaT. cruzi60% inhibition at 10 μg/mL[75]
Dudawalamide D (61)M. producensPapua New GuineaL. donovaniIC50 = 2.6 μM[75]
Iheyamide A (62)Dapis sp.Okinawa, JapanT. brucei rhodesiense
T. bhurstuerusei brucei
IC50 = 1.5 μM
IC50 = 1.5 μM
[76]
Janadolide (65)Okeania sp.Okinawa, JapanT. brucei bruceiIC50 = 47 nM[77]
Beru’amide (66)Okeania sp.Kagoshima, JapanT. brucei rhodesienseIC5 = 1.2 μM[78]
Table 6. Compounds with reported antiviral activities.
Table 6. Compounds with reported antiviral activities.
CompoundSource OrganismCollection SiteTargeted VirusIC50/% of InhibitionReference
67L. lagerheimiiHawaii, USAHIV-1HIV-1 inhibition at l-l00 µg/mL[79]
68L. lagerheimiiHawaii, USAHIV-1HIV-1 inhibition at l-l00 µg/mL[79]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Youssef, D.T.A.; Mufti, S.J.; Badiab, A.A.; Shaala, L.A. Anti-Infective Secondary Metabolites of the Marine Cyanobacterium Lyngbya Morphotype between 1979 and 2022. Mar. Drugs 2022, 20, 768. https://doi.org/10.3390/md20120768

AMA Style

Youssef DTA, Mufti SJ, Badiab AA, Shaala LA. Anti-Infective Secondary Metabolites of the Marine Cyanobacterium Lyngbya Morphotype between 1979 and 2022. Marine Drugs. 2022; 20(12):768. https://doi.org/10.3390/md20120768

Chicago/Turabian Style

Youssef, Diaa T. A., Shatha J. Mufti, Abeer A. Badiab, and Lamiaa A. Shaala. 2022. "Anti-Infective Secondary Metabolites of the Marine Cyanobacterium Lyngbya Morphotype between 1979 and 2022" Marine Drugs 20, no. 12: 768. https://doi.org/10.3390/md20120768

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

Article Metrics

Back to TopTop