Open access peer-reviewed chapter

Cyanobacteria Natural Products as Sources for Future Directions in Antibiotic Drug Discovery

Written By

Bahareh Nowruzi

Submitted: 13 April 2022 Reviewed: 06 July 2022 Published: 07 October 2022

DOI: 10.5772/intechopen.106364

From the Edited Volume

Cyanobacteria - Recent Advances and New Perspectives

Edited by Archana Tiwari

Chapter metrics overview

154 Chapter Downloads

View Full Metrics

Abstract

Cyanobacteria, an abundant source of natural products with a broad diversity of secondary metabolites, have emerged as a novel resource for the progression of synthetic analogs. Due to the rise of antibiotic resistance, there is a need for new medications and cyanobacteria-derived compounds have shown promising important alternatives for new therapeutics. These secondary metabolites are produced through nonribosomal peptide synthetase (NRPS), polyketide synthase (PKS), and mainly through mixed NRPS-PKS enzymatic systems. Current research is focused on the exploitation of cyanobacteria for the production of bioactive metabolites. Screening of cyanobacteria for pharmaceutically active compounds has received increasing attention; however, limited knowledge is available on biosynthetic mechanisms that would enhance the drug discovery process and culture-based production of desired metabolites. Overall, there is a promising outlook that cyanobacterial secondary metabolites will become alternatives for the development of new medications in a near future with enhanced pharmacological and pharmacokinetic properties.

Keywords

  • cyanobacteria
  • natural products
  • antibiotic
  • drug discovery
  • antibiotic resistance
  • polyketide synthase (PKS)
  • nonribosomal peptide synthetase (NRPS)
  • bioactive metabolites
  • synthetic analogs
  • biosynthetic mechanisms

1. Introduction

Antibiotics, the so-called “miracle drugs,” came into existence half a century ago; however, their current popularity swiftly leads to overuse. Over the last decade, it has become quite apparent that the efficiency of antibiotics is dropping due to the growth of pathogen resistance; a problem that increases as fewer new drugs become available in the market. Moreover, unraveling this resistance is not straightforward, since antibiotic resistance is actually produced in multiple ways. Considering the urgency of the issue, efforts to develop new antibiotics are being carried out by pharmaceutical companies. In this regard, natural products account for a thorough and important component of today’s pharmaceutical compendium as a fundamental source of chemical diversity. To date, several natural products have been studied, but many others still await investigation [1]. Cyanobacteria, being one of the eldest recognized creatures living on the earth with exclusive structural features, produce several bioactive compounds with varied biological activities. Moreover, cyanobacteria as photosynthetic microorganisms, which have been preserving the oxygen levels on the earth, structurally look like gram-negative bacteria. They include chlorophyll a and phycobiliproteins, as well as the photosystems II and I. The adaptation mechanisms shown by cyanobacteria allow them to survive in severe climate conditions and tolerate limiting factors, such as heat, drought, salinity, nitrogen starvation, cold, photo-oxidation, osmotic, and UV stress [2]. Additionally, cyanobacteria are able to produce biologically active natural products with known antifungal, antibacterial, anti-inflammatory, antiviral, and enzyme-inhibiting bioactivities mostly through either the nonribosomal polypeptide (NRP) or the mixed polyketide-NRP biosynthetic pathways [3]. An increasing number of cyanobacterial metabolites are found to target actin and tubulin filaments in eukaryotic cells, making them a noteworthy source of anticancer natural products. Definite bioactive compounds, for example, dolastatin-10 and curacin A, have gone through clinical trials as possible anticancer drugs [4]. Cyanobacterial bioactive products can be categorized consistently with diverse structural typologies comprising terpenes, polyketides, peptides, lipids, and alkaloids. Many structural modifications can be found in cyanobacterial compounds, especially polyketide-derived units [3]. Besides, each cyanobacterial strain produces a category of bioactive compounds, so that new drugs are being constantly discovered from these sources.

Along with all these advantageous features, cyanobacteria are also known to produce toxins, mainly neurotoxins and hepatotoxins [2, 5], which act also as activators (e.g., antillatoxin) or blockers (e.g., jamaicamide A and kalkitoxin) and in addition their possible neuroprotectant and analgesics properties, they are functional molecular to distinguish usefully channels [4, 6, 7, 8].

Patellamide and trunkamide have also clinical potential, showing moderate cytotoxicity but multi-drug resistance. Investigations about the cyanobacterial natural product and secondary metabolites have gradually adapted to the genomic revolution over the past 15 years, and the genetic characterization of these secondary metabolites has led to further investigations in the field of cyanobacterial natural product synthesis. Despite important achievements in this area, numerous pharmaceutical companies have decreased the use of natural bioactive products and drug discovery screening because of: a) difficulties associated with strain, b) troubles correlated to natural bioactive products, and c) problems with logical property rights [9, 10, 11, 12, 13, 14]. Finally, the use of compound collections prepared by combinatorial chemistry methods has been also influential.

Advertisement

2. Improving access to natural products

It is now evident that the chemical diversity of natural products is a better option than the variety of available synthetic compounds for drug discovery [15, 16]. Therefore, the use of natural chemical diversity in this regard is becoming increasingly frequent [11, 17]. Early publications showed that only a small number of cyanobacteria taxa were accessible for screening [9]. Now, extensive cyanobacteria collections, together with better cyanobacteria culture techniques, are providing new chemicals for use in drug discovery assays [11]. Progress is being made in the chemistry of natural products, leading to advances in synthetic methods seeking the production of compound analogs with enhanced pharmacological or pharmaceutical characteristics [18]. Another interesting feature that has made natural product “privileged” structures is their ability to be used as cores of compound (alkaloids, polyketides, terpenoids, and flavonoids) libraries produced through combinatorial techniques [19, 20].

Over the past 30 years, there has been a considerable reduction in the interest by the leading pharmaceutical companies in drug discovery from natural sources. Despite this, phycologists, associated with the manufacturing industry, are exploiting this niche so that there is now a renaissance related to new improvements in spectroscopy, analytical technologies, and high-throughput screening [21]. In addition, competing technologies, such as combinatorial chemistry, have not proved to be very successful in delivering the new drug in significant numbers [22]. With the use of alternative techniques to produce analogs and derivatives of natural products, new compounds can be patented, even if the primary structure had been previously disclosed [11].

Advertisement

3. New approaches to the value of natural products

Multitude reasons have been suggested in regards to why natural products are such appropriate sources for drug leads, but at least one study has endeavored to quantify a connection between the drug molecules and those typically found in natural products and combinatorial chemical libraries [22]. Combinatorial libraries are synthesized in large numbers, and structures have high randomness. A multivariate evaluation of the chemical space occupied by thousands of combinatorial drug compounds compared with that of natural products revealed a good correlation between clinically approved drug molecules with natural products. This means that the structure of drugs used nowadays can be simulated by that of natural products [15]. With the progress in analytical spectroscopy, numerous clarifications are currently accessible so that the discovery of new bioactive compounds needs only a few micrograms [22]. The improvement in fractionation methods intended for isolating and purifying natural bioactive products (counter-current chromatography [20], analytical structure determination [23], etc. has led to screening natural product mixtures with timescales suitable for those expected in high-throughput screening campaigns. Complex structures can be resolved now with much less than 1 mg of the compound using the recent NMR techniques [11]. According to Quinn (developing a drug-like natural product library, 2008), it is possible to prepare a screening a library of highly diverse plant-derived compounds by pre-selecting products from the dictionary of natural products to be drug-like in their physicochemical properties. Yet, many alternative approaches are also being tested in order to enhance the speed and efficiency of drug discovery from natural products [11]. For instance, bioinformatics has been used for predicting microbes, which are able to produce new chemicals on the basis of the gene sequences encoding polyketide synthesis; this method has led to the discovery of potential antifungal and anticancer activities in some compounds [24]. Furthermore, the Metagenomics approach, which has led to the discovery of antibiotic compounds, has been recently used to achieve a broader range of synthetic cyanobacterial capabilities. This involves the collection of the entire DNA from a field cyanobacteria sample and the cloning of this DNA in host organisms, such as E. coli. Recombinant bacteria are subsequently cultured and examined for the expression of bioactive metabolites [11]. Additionally, peptide synthetase genes and polyketide synthase genes have been explored, and manipulation of biosynthetic pathways in refractory microbes, such as uncultivable, is a promising line of research. Along with the most innovative tools of genetic engineering, new approaches to metagenomic mining of environmental DNA are being popularized, so that the genetic potential of many bacteria can be explored [25]. Even though more than 200 genome projects are either already completed or still undergoing publication, there are still some striking questions on what is actually being sequenced, considering the fact that these studies are limited to cultivable microbes. The Metagenomics approach, being culture-independent, can help to solve this problem and can also help with data mining with potential interest for a broad scientific community [25]. Different techniques unite enzymatic and synthetic methods to achieve multifaceted natural bioactive products, and refining the activity of obviously occurring antibiotics [11]. Mutasynthetic techniques are useful for making the antibiotic daptomycin-associated compounds [26, 27], vancomycin analogs and anticancer compound cryptophicin have been formed using the cytochrome P450 enzymes [12]. The biosynthesis of cyanobacterial compounds supports the creation of numerous functional groups, chiefly in the gene clusters related to cyanobacterial compounds, for instance, jamaicamide A, barbamide, or curacin A [28]. Hence, undescribed enzymatic mechanisms will be revealed thanks to biochemical studies in cyanobacterial secondary metabolic pathways. From the experience in the production of pharmaceuticals from invertebrate-derived microbes, it is evident that several obstacles must be overcome before this approach becomes a conventional technology. Still, there are good reasons to be optimistic about the future [22].

Advertisement

4. Activity profiling of extracts

An alternative technique to the time-consuming and expensive methods previously used for creating extensive collections of isolated and structurally characterized natural products [29] is screening the mixtures of compounds obtained from extracts of cyanobacteria strains [11]. Yet, obtaining extracts with potential biologically active novel compounds is not always simple from primary screenings. This probability can be predicted by comparing the ratio of the binding potencies at two receptor sites for a known selective ligand and for an extract by the “differential smart screens” method [30]. Furthermore, by means of a database of the usefulness of an extensive variety of identified bioactive compounds the analysis of drugs with the unknown process is imaginable. Therefore, information about previously unidentified compounds can be gained, which is precious for the antibiotic applications stated below [31]:

  1. Creation of original whole-cell assays for drug screening, such as multi-patch.

  2. Target identification with cDNA and quantitative real-time PCR (qRT-PCR) for confirmation of the results.

  3. Revisions on mechanisms-of-action (MOA) with antibiotic-induced expression profiling.

These techniques could lead to a novel understanding of the potential effects of untested compounds (or exposure to compounds not structurally analogous and, thus, not expected to act via the same biological target) [2].

Bioinformatics and proteomics experiments are used in studies at the mRNA (transcriptome) or protein (proteome) levels, which help with the identification of DNA binding sites of transcription factors [32] and the adjustment of biological functions, respectively, in order to characterize the complex organism responses to environmental stimulates [2]. Microarrays have been used for the identification of regulon members and stimulons by many groups in the transcriptome measurement level [33, 34].

Two-dimensional gel electrophoresis in which proteins are separated according to their molecular weight and isoelectric point, is useful in most cases, but intricate protein samples can also be analyzed using the liquid chromatography-tandem MS (LC-MS/MS) in which protein and peptide combinations are supplied to a mass spectrometer (MS) from a HPLC system. Isotopic dilution strategies on a MS instrument (e.g., isotope-coded affinity tags or ICAT) can be used for a comparative quantification of protein expression. ICAT approaches were advantageous when first released but are limited by their inability to analyze more than two conditions without a large amount of multiplexing [2, 35]. Currently, a developed version of the iTRAQ approach can analyze eight different conditions simultaneously. Despite all these tools, the most useful method would involve a concurrent quantification of the expression of all the genes and proteins of interest from a biological sample.

Advertisement

5. Natural products as pharmacological instruments

Aside from their curative activity, natural bioactive products can operate as pharmacological instruments demonstrating novel physiological features [14]. Cyanobacteria are stubbornly obstinate to genetic manipulation, which is accessible only for a small number of strains [3]. The modularity in cyanobacterial PKS-NRPS gene clusters authorizes the heterologous expression of natural bioactive products and, thus, genetic manipulation for combinatorial biosynthesis of innovative hybrid chemical bioactive products [4]. The prosperous production of nonribosomal and ribosomal peptides in heterologous hosts permits the usage of other cyanobacterial natural bioactive products [3]. Cyanobacteria usually synthesize multiple variants of the identical natural bioactive product; this can be ascribed to a deficiency of the inactivity of the NRPS tailoring enzymes or NRPS biosynthetic pathways. The genetic basis for this modification of secondary metabolite gene clusters is probably controlled by gene duplications, gene deletions, recombination, sequential mutation followed by natural selection, and loss and gain of tailoring enzymes [36]. However, the evolutionary and adaptive importance of these processes is deficiently understood.

Advertisement

6. Which cyanobacteria phyla produce therapeutics?

Throughout the prior decade, several natural bacterial compounds have been described, all of which originated from five bacterial phyla: Bacteroidetes (34 compounds), cyanobacteria (220), actinobacteria (256), proteobacteria (78), firmicutes (35), and four bioactive compounds from taxonomically unknown sources [37]. The variety of cyanobacterial natural bioactive products gathers > 1100 secondary compounds recognized with composite chemical structures, stated from different genera [3]. These metabolites represent a broad range of bioactivities including some that may be related to their natural environment (antibacterial, antifungal, antiviral, and cytotoxic) [29], but others demonstrate a clear pharmaceutical interest, for example, they can be used as anticancer agents, immunomodulators, or protease inhibitors [38]. Cyanobacteria exhibit different growth forms, from unicellular to filamentous or colonial forms, and depending on their environmental conditions they may be surrounded by a mucilaginous or gelatinous sheath [29]. The PKS and NRPS genes seem to be more widespread in undifferentiated filamentous and heterocystous cyanobacterial strains. Despite the current taxonomic instability within cyanobacteria, which makes assessing the actual occurrence of natural products difficult, cyanobacterial compounds are mainly obtained from the lyngbya, symploca, microcystis, nostac, and hapalosiphon (Table 1) [3, 37].

Cyanobacterial compounds (class)Cyanobacterial strainBiological targetPotential therapeutic usesReferences
Apratoxin ALyngbya bouilloniiSTAT3, KB, and LoVo cell lines
Cytotoxic against human tumor cell lines (0.36–0.52 nM)
Oncology, Early stage adenocarcinoma (induction of G-1 phase cell cycle arrest)[4, 29, 37, 39]
Apratoxin DLyngbya sp.AntiproliferativeOncology[40]
Coibamide ALeptolyngbya.AntiproliferativeOncology[41]
Curacin A-D
NRPS-PKS
Lyngbya majuscula 19 LColon, renal, and breast cancer cell lines. Involvement of HMG-CoA in formation of cyclopropyl ringOncology, Antimitotic, Inhibits microtubule assembly
Anti-inflammatory, Antiproliferative, Immunosuppressant, herbicidal
[4, 29, 37, 42, 43]

CryptophycinNostoc sp.Tubulin polymerization antiproliferative and antimitotic agents, Cytotoxicity against human tumor cell lines and human solid TumorsOncology, destabilization of microtubule dynamics and the induction of hyperphosphorylation of the anti-apoptotic protein B-cell leukemia/lymphoma 2 (BCl-2),triggering programed cell death[37, 44, 45]

LargazoleSymploca sp.Histone deacetylaseOncology, anti-epileptics, neurological disorders, mood stabilizer[46]
MicrocystinMicrocystis aeruginosa PCC 7806,M,aeruginosa K-139
Planktothrix agardhii CYA126
LymphocytesCytotoxic, inhibit membrane-bound leucine aminopeptidase
Enzyme inhibitor, cytotoxic, tumor promoter, anticancer
[47, 48, 49, 50, 51]



HassallidinsAnabaena sp. SYKE 748AAntifungal activity[52]
SulfoglycolipidScytonema sp.HIV-1Inhibit reverse transcriptase and DNA polymerases[29]
Dolastatin-10Symploca sp.Binds to tubulin on rhizoxin-binding SiteAffects microtubule assembly in P388 lymphocytic leukemia cell line (NCI)[53, 54]
Dolastatin-15Lyngbya sp.binds to vinca alkaloid site of tubulinBreast cancers treatment[55]
Jamaicamides (A-C)L. majusculaH-460 human lung cell carcinoma, neuro-2A-neuroblastoma cell lineNeurotoxic, cytotoxic against H-460 human lung and neuro-2a mouse neuroblastoma cell lines[56]
KalkitoxinL. majusculaBlock voltage sensitive Na+ channelNeurotoxic, Neural necrosis through N-methyl-D-aspartate Receptor mechanisms[57]
AstaxanthinHaematococcus pluvialisColon cancer cell linesExpression decrease of cyclin D1, increase of p53 and some cyclin kinase inhibitors (p21WAF-1/CIP-1, p27)[58]
PolysaccharideNavicula directaHSV1, 2, influenza A virusInhibition of hyaluronidase
[59, 60]
AllophycocyaninCryptomonadsEnterovirus 71Inhibition of cytopathic effect, delay in synthesis of viral RNA[61]
HectochlorinL. majusculaColon, melanoma, ovarianActin binding compounds,[62]
Diadinochrome A, B, Diatoxanthin, cynthiaxanthinPeridinium bipesHeLa cellsCytotoxic effect[63]
Pheophorbide a-, b-like compoundsDunaliella primolectaHSV1Inhibition of cytopathic effect[64]

Table 1.

Current status of potential cyanobacteria therapeutics.

Advertisement

7. Cyanobacterial drug discovery

Systems biology can help us with the acquisition consciousness of the ways living systems function using computational power [65]. So as to study some specific facts in a definite biosynthetic pathway, some information about both the proteins in charge and the responsible gene of that event is needed. The function of linking the chemical diversity of natural bioactive products and genomes in addition to modeling and prediction by incorporating such biological information could offer considerable information for the understanding of such a complex biological system [2].

According to the Comprehensive Microbial Resource Declaration, the genome sequences of human pathogenic bacteria and non-homologous in humans, have been documented. This could be an appropriate technique for the reporting of the new drug [66], and the improvements in synthetic biology now provide a solution to cyanobacteria being stubborn to genetic manipulation, opening up cyanobacteria as a valuable source of new enzymes and novel natural bioactive products.

Today pharmaceutical industry is concentrated on prominent output screening systems, genomics tools, and bioinformatics, containing combinatorial chemistry and logical design for the recognition of new bioactive compounds [29]. Recognizing groups of secondary bioactive compounds biosynthetic gene clusters with possible therapeutic competence involvement in an initial stage, which is conducted by the chemical structure of the identified bioactive compounds in cyanobacteria strains [3]. Cyanobacterial biologically active compounds are produced through NRPS, PKS, and mixed NRPS-PKS pathways [4]. Cyanobacteria strains presentation progressive screening outcomes are then designated for proteome mining and genomic characterization in order to classify biosynthetic gene clusters responsible for proteins connected to the making of these bioactive components [2]. This is imaginable because databases of biosynthetic gene clusters and cyanobacterial chemicals have been gathered through gene libraries (http://dtp.nci.nih.gov/docs/3d_database/dis3d.html, NCBI Pubchem http://pubchem.ncbi.nlm.nih.gov/, ChemIDPlus http://chem.sis.nlm.nih.gov/chemidplus, ANTIMIC [67], and Super Natural Database http://bioinformatics.charite.de/supernatural/) [68]. As a result of the increased antibiotic resistance, available drugs are effective against only one-third of the diseases, and the identification of new biologically active compounds is thus urgently necessary [29].

Advertisement

8. Web-based tools and databases for drug target identification

A variety of different silico tools and databases are available for drug target determination among the identified genes in pathogens for an initial screening. DrugBank(http://insilico.charite.de/supertarget/ main.html#Home), NCBI Entrez Gene(http://www.ebi.ac.uk/msd/), TarFisDock and MATADOR (http://matador.embl.de/) could be used either by a manual searching or by BLAST search of sequenced proteins. These facilities compensate the costs of screening through very large compound collections, minimizing the pace of drug discovery by both reducing the number of compounds used in real screens and the costs of screening [2].

Advertisement

9. Secondary metabolites derived from Cyanobacteria strains

Natural bioactive products have been isolated from a varied diversity of strains and verified for numerous biological activities. Among these strains, cyanobacteria strains signify such a source.

Secondary metabolites derived from cyanobacteria strains were identified as a rich source of bioactive compounds [69, 70, 71]. Several bioactive compounds isolated from different cyanobacterial strains showed a varied range of chemical structures and biological activities comprising new peptides, amides, terpenes, carbohydrates, polyketides, fatty acids, alkaloids, and other organic chemicals [41, 72, 73, 74]. These compounds are regarded as good candidates for drug discovery, with functions in the industry [75, 76, 77], agriculture [19], and in pharmacy [69, 77, 78].

The cyanobacterial bioactive compounds specify useful pharmaceuticals that are problematic to produce synthetically [79]. The variety of structures found in Lyngbya majuscula is just incredible. Compounds isolated from this strain are amino acids, fatty acids, depsipeptides, pyrroles, amides, alkaloids, lactones, lipopeptides, and many others [40, 72, 80, 81]. Totally, cyanobacterial bioactive compounds show an exciting range of biological activities ranging from insecticidal, immunosuppressant, antiviral, anticancer, antimicrobial, and anti-inflammatory to proteinase-inhibiting activities which are outstanding targets of biomedical research (Table 2) [ 2, 5, 6, 7, 8, 78, 113, 114, 115, 116, 117, 118].

Species of cyanobacteriaBioactive compoundsBiological activityClassReferences
Lyngbya Lagerheimii
Phormidium tenue
SulfolipidAnti-HIV-1 activityFatty Acid (sulfo)[82]
Lyngbya majuscula 19 LBarbamideAntimolluscicidalchlorinated lipopeptide[83, 84]
L. majuscule
L. majuscula
Antillatoxin
Antillatoxin B
Neurotoxic
Ichthyotoxic, activator of voltage-gated sodium channel
Cyclic lipopeptide[83, 85, 86]
Synechocystis trididemniDidemninAnticancer, antiviral, immunosuppressiveLipopeptide[87]
Cylindrospermum licheniformeCylindrocyclophaneAnticancer, cytotoxicAlkaloid
Macrocycle, chloro
[43, 88, 89]

Cylindrospermopsis raciborskiiCylindrospermopsinCytotoxicAlkaloid[90]
Prochloron didemniPatellamide A, B, C and DCytotoxic, biological activity against multi-drug resistant UO-31 renal cell linesCyclic lipopeptide[28, 91, 92, 93]

L. majusculalyngbyabellins A and B,Cytotoxic, Anticancer, Cytoskeleton disruptionLipopeptides[94]
L. majusculaAntillatoxin
Antillatoxin B
Neurotoxic
Ichthyotoxic, activator of voltage-gated sodium channel, with sodium channel-activating
Lipopeptide[4, 83, 85, 86]

Lyngbya semiplenaSemiplenamides A-GAll displayed weak to moderate toxicity in brine shrimp assay; and 38 showed weak affinity for the rat cannabinoid CB1 receptor; showed moderate inhibitor of anandamide membrane transporterLipopeptide[4]
Nostoc ellipsosporumCyanovirinAnti-HIV, antiviral HIV-1 (interacts with high mannose groups of envelope glycoproteins, gp120 and blocks its interaction with target cell receptors) HIV-2
HSV-6
Measles virus
SIV
FIV
Peptide and proteins[29, 95]
P. tenueMonogalactopyranosyl glycerol digalactopyranosyl glycerolAnti-HIV, anticancerSulfolipids[96]
Calothrix sp.CalothrixinAntimalarial, anticancer Against HeLa epithelial carcinomaIndoles[97]
Symploca hydnoides
Symploca sp VP453
Symplostatin 1
Symplostatin 3
Against Murine colon 38 and murine mammary 16/C cell lines Against Microtubule depolymerizationanalog of dolastatin-10[54, 98]
L. majusculaLyngbyatoxins A-CCytotoxic, Ichthyotoxic, Tumor promoter, Protein Kinase C activator, Skin irritant[2, 43]
L. majusculaHectochlorinAgainst Colon, melanoma, ovarian and renal and lung cancer cell lines, promote actin polymerizationLipopeptide[4, 62]
Lyngbya majusculeHermitamides A and BIchthyotoxic, Brine shrimp toxicity, Cytotoxic[43]
L. majusculaSomocystinamide ACytotoxic against neuro-2a neuroblastoma cells (IC50 = 1.4 lg/mL), Pluripotent inhibitor of angiogenesis and tumor cell proliferation, Induces apoptosis in endothelial cells.Lipopeptide[4, 99]
Oscillatoria NigroviridisOscillatoxinAnticancer, Toxic generalAromaic[43]
Microcystis aeruginosaMicroviridinAntibiotic, anticancertricyclic depsipeptides[100, 101]
Lyngbya sp.Kempopeptins A, BAgainst colon cancerCyclodepsipeptides[102]
L. majusculaHermitamides AeBAgainst lung cancer, Potent blockers of the hNav1.2 voltage-gated sodium channel., IchthyotoxicLipopeptide[43, 103]
L. majusculaLyngbyatoxin A-CIchthyotoxic, Cytotoxic, Tumor Promoter, Protein Kinase C activator, Skin irritant[43]
Caulerpa taxifolia
Green algae
CaulerpenyneCytotoxicity, anticancer, antitumor, and antiproliferative activities[104]
L. majusculaCarmabin A-BAnticancer, AntiproliferativeN-Methylated Peptide[43]
Cyanobacteria Nostoc linckia and Nostoc spongiaeforme var. tenueBoromycinCytotoxicity against human epidermoid carcinoma (LoVo), human colorectal adenocarcinoma activity, potent cytotoxicity against drug-resistant murine and human solid tumorsPolyether-macrolide antibiotic[97, 105]
L. majusculaMajuscolamid A-DAntifungal, Antimycotic activity[106]
L. majusculaMicrocolin A-CAntiproliferative, Anticancer, Cytotoxic, Immunosuppressive[107]
L. majusculaMalyngamide A-UAntimicrobial, Antifeedant, Cytotoxic, ImmunosuppressiveLipopeptide[43]
L. majusculaPitiamide A-BAntifeedantFatty Acid Amides[107, 108]
L. majusculaYanucamides A and BBrine shrimp toxicityDepsipeptides[43]
Nodularia spumigenaNodularia toxinEnzyme inhibitionLipopeptide[2, 109, 110]
Aulosira fertilissimaAulosirazoleAnticancerAromatic[111]
Oscillatoria acutissimaAcutiphycin and 20,21-didehydroacutiphycinAntineoplastic agentLipopeptide[112]

Table 2.

Bioactive compounds from cyanobacteria.

Advertisement

10. Antiviral activity

The extension of fatal, virus-related diseases, such as HIV, has resulted in several considerable consequences. Since the only accredited therapy (HAART, highly active antiretroviral therapy) has shown toxic effects, severe induction to viral resistance, and disability to eliminate viral agents, thus the need for new and safe antiviral therapies is an urgent issue [119, 120]. Some potential antiviral compounds are described below:

10.1 Polysaccharides

Spirulan and Ca-spirulan derived from Spirulina sp. are regarded as the most notable antiviral polysaccharide compounds provided their broad-spectrum activity against HIV-1, HIV-2, H, influenza and other enveloped viruses. These compounds disable the reverse transcriptase activity of HIV-1 and prevent the attachment and fusion of virus cells with host cells. Additionally, the fusion between HIV-infected and uninfected CD4+ lymphocytes, which boosts the viral infectivity, is inhibited [29]. Their reduced anticoagulant properties make them more advantageous antiviral agents over other sulfated polysaccharides. Another interesting compound is nostoflan from Nostoc flagelliforme, an acidic polysaccharide showing potent virucidal activity against herpes simplex virus-1 [121, 122].

10.2 Carbohydrate-binding proteins

A couple of carbohydrate-binding proteins have shown promising activity as antiviral agents. Ichthyopeptins A and B, derived from Microcystis ichthyoblabe, are potential agents against influenza virus, with an IC50 value of 12.5 mg ml–1 [123]. Cyanovirin-N and scytovirin are also potent virucidal drugs that interfere with several steps of the viral fusion process. Cyanovirin-N, for example, shows both in vitro and in vivo activity against HIV and other lentiviruses in nanomolar concentrations. These 101 amino acids long, 11 kDa polypeptide derived from Nostoc ellipsosporum is being developed as a vaginal gel for preventing sexual transmission of HIV by Cellegy Pharmaceuticals, San Francisco, CA, provided its inhibitory effects upon HIV virus-CD4 cell membrane fusion [124]. Scytovirin, on the other hand, is a 95 amino acid long, 9.7 kDa polypeptide (that includes five intra-chains disulfide bonds) derived from aqueous extracts of Scytonema varium, that is able to attach to the glycoprotein envelope of HIV (gp120, gp160, and gp41), thus making the virus inactive even in low nanomolar concentrations [125].

10.3 Sulfoglycolipids

Natural cyanobacterial sulfoglycolipids show confirmed HIV-reverse transcriptase and DNA polymerase inhibitory effects [29].

11. Antibacterial activity

If bacterial resistance strengthens, the treatment may become impossible for some diseases. Nosocomial infections such as those caused by the methicillin-resistant Staphylococcus aureus or the vancomycin-resistant enterococci, caused by multi-drug-resistant bacteria, create therapeutic problems of worldwide concern [126], hence the urgency of developing new antibiotics. Accordingly, new attempts to find antibacterial activity via screening of cyanobacterial extracts have started [127], although very few cyanobacteria-related antibacterial compounds have been detected to date. Noscomin57, from Nostoc commune [128], shows antibacterial activity against Bacillus cereus, Staphylococcus Epidermidis, and Escherichia coli. Antibacterial activity of Anabaena extracts against vancomycin-resistant S. aureus with a MIC of 32–64 mg ml-1 has been reported by [129].

12. Antiprotozoal activity

The estimations of the World Health Organization indicate that >109 people over the world suffer from tropical diseases caused by Schistosoma, Trypanosoma, Leishmania, Plasmodium, and others [130]. The unsuccessful treatment of such diseases (especially malaria) is related to the growing resistance shown by these protozoa and the slow pace of drug discovery [131, 132]. In a recent project operated by the Panamanian International Co-operative Biodiversity Group, five classes of antiprotozoal compounds were isolated from cyanobacteria. Nostocarboline, an alkaloid protease inhibitor isolated from Nostoc sp. 78-12 A, displayed activity against T. cruzi, Leishmania donovani, Trypanosoma brucei, and Plasmodium falciparum [133]. Moreover, aerucyclamide C68 isolated from Microcystis aeruginosa PCC 7806 has been also detected to be active against T. brucei.

13. Protease inhibition activity

More than 120 cyanobacterial alkaloids with various biological activities (including protease inhibition) were introduced between 2001 and 2006. Some of these compounds, such as microginins (used for the treatment of high blood pressure), aeruginosins, and cyanopeptolins (a serine inhibitor used for asthma and viral infections) are described by Jaspars and Lawton [29]. Kempopeptins are other groups of protease inhibitory products, for example, kempopeptin B (with activity against trypsin, with an IC50 of 8.4 mM), kempopeptin A (a cyclodepsipeptide derived from marine Lyngbya with activity against elastase), and chymotrypsin with an IC50 of 0.32 mM and 2.6 mM, respectively [46].

14. Immunomodulatory activity

Besides the beneficial properties of cyanobacteria, their immunomodulatory activity exhibits diverse effects on immune systems, such as the increase of phagocytic activity in macrophages, the stimulation of antibody and cytokine production and the accumulation of natural killer cells into tissues, or the activation of T and B cells [134]. For instance, the effect of Spirulina in mice was investigated by Hayashi et al., who demonstrated increased phagocytic activity and antigen production. Enhanced phagocytic and natural killer cell-mediated antitumor activities, together with increased antigen production, were also shown in chicken by Qureshi and Ali [29]. Additionally, the incremental impact of cyanobacteria extracts on 13.6-fold interferon and 3-fold interleukin (IL)-1b and -4 was observed in human blood cells. Despite Spirulina has been proved to be safe, other cyanobacteria (e.g., Microcystis sp.) produce metabolites that are cytotoxic to lymphocytes and have inhibitory effects on membrane-bound leucine amino peptidase, which is related to antigen-processing and antigen presentation response [47, 135], confirmed the immune-toxicity of microcystin that presented medical competence in the lipopolysaccharide-induced lymph proliferation in mice vaccinated with sheep T-dependent antigen red blood cells.

15. Anticancer activity

The urgency of brand-new anticancer medications is an important issue provided the increasing resistance against currently available drugs (such as taxanes) and the outbreak of new types of cancer subjected to chemotherapeutic treatment failure [29]. A considerable number of highly active cyanobacterial compounds target tubulin or actin filaments in eukaryotic cells and have exhibited potent antimitotic properties, which makes them a noteworthy source of potential anticancer agents [4]. Several of them have gone through Phase I and II clinical trials such as the third generation dolastatin15 and TZT-1027 (soblidotin), a synthetic derivative of dolastatin-10 [136, 137]. They generally act by blocking cell division at the M-phase by targeting tubulin with efficacy equivalent to clinical drugs, such as vinblastine, vincristine, or paclitaxel. Some of these highly cytotoxic compounds are described below in Figure 1 [138].

Figure 1.

Potential of cyanobacterial extract as anticancer activity.

15.1 Coibamide A – An anticancer agent with a novel action mechanism

Coibamide A, extracted from a Leptolyngbya strain, shows a novedous action mechanism targeting tubulin or actin filaments. Notable cytotoxical properties against breast, central nervous system, colon, and ovary cancers have been observed [41].

15.2 Cryptophycins

Cryptophycins are examples of cyanobacteria-derived tubulin-binding compounds with potent anticancer activity. Cryptophycin A was first isolated by Schwartz and co-workers in 1990 from Nostoc sp. strains ATCC 53789 and GSV224 [22]. Microtubule dynamics suppression and blocking of G2/M phases are features making this molecule a potent anti-carcinoma metabolite [29]. Cryptophycin-52 (LY 355073), a chemical analog of cryptophycin-1, was developed to improve its hydrolytic stability but produced very slight activity in the clinical trial. The second-generation analog, cryptophycins-249 and -309, show better water solubility and stability [139]. According to a study by [140], the thioesterase derived from the cryptophycin biosynthetic pathway through the macrocyclization of a series of linear synthetic forerunners generate 16-membered cyclic depsipeptides, showed significant efficiency as anticancer agents.

15.3 Largazole- a histone deacetylase inhibitor

Largazole, an ant proliferating compound with an unusual chemical scaffold, is extracted from Symploca sp. [141], and shows a considerable histone deacetylase (HDAC) inhibitory activity [142], together with a great selectivity in human mammary epithelial and fibroblastic osteosarcoma cells. The FDA ratification of HDAC inhibitor suberoylanilide hydroxamic acid as a treatment for dermal T-cell lymphomas, besides its mood stability properties and anti-epileptic characteristics, confirms this compound for cancer treatment.

15.4 Apratoxins – signal transduction inhibitors

Apratoxins, a notable class of potent cytotoxic cyclic depsipeptides, was initially isolated from a chemically rich Lyngbya boulloni strain and, according to NCI’s Developmental Therapeutics Branch, demonstrated a unique action pattern against a panel of 60 cancer cell lines [143]. Limited findings until now indicate that the induction of G1-phase cell-cycle arrest and apoptosis is how apratoxins function as anticancer agents [39]. Apratoxin A showed moderate cytotoxicity in multiple human tumor cell lines (e.g., LoVo cell lines and KB cancer cells), although this compound is acid sensitive and decomposes when exposed to the HCl present in CDCl3. Other analogs, especially apratoxin D, have been studied in order to develop a lead structure [4].

15.5 Polypeptides- Hassallidins

Polypeptides, mostly with microbial origins, have long been used for pharmaceutical applications either as antimicrobial agents or for disinfection. A group of cyclic glycosylated lipopeptide Cyanobacteria metabolites are the hassallidins A [52] and B [144], which are purified from Hassallia; these compounds are a type of comprehensive with action against human pathogenic fungi [1].

16. Conclusions

The results in this review emphasized the significance of the probable healing function of natural bioactive products purified from cyanobacteria strains, for instance, antibacterial, antitumor, protease inhibition activity, and antiviral effects, and highlighted the necessity to restart discovering natural biological sources. However, system biology for metabolite purification, characterization, and valuation in cyanobacterial bioactive compounds that have not arrived in the clinical trials so far.

References

  1. 1. Saleem M, Nazir M, Ali MS, Hussain H, Lee YS, Riaz N, et al. Antimicrobial natural products: An update on future antibiotic drug. Natural Product Reports. 2010;27(2):238-254
  2. 2. Wase NV, Wright PC. Systems biology of cyanobacterial secondary metabolite production. Expert Opinion on Drug Discovery. 1 Aug 2008;3(8):903-929
  3. 3. Dittmann E, Gugger M, Sivonen K, Fewer DP. Natural Product Biosynthetic Diversity and Comparative Genomics of the Cyanobacteria. Trends in Microbiology. Cell Press. 1 Oct 2015;23(10):642-652
  4. 4. Tan LT. Bioactive natural products from marine cyanobacteria for drug discovery. Phytochemistry. 1 Apr 2007;68(7):954-979
  5. 5. Nowruzi B, Lorenzi AS. Production of the neurotoxin homoanatoxin-a and detection of a biosynthetic gene cluster sequence (anaC) from an Iranian isolate of Anabaena. South African Journal of Botany. 2021;139:300-305
  6. 6. Ahari H, Nowruzi B, Anvar AA, Porzani SJ. The toxicity testing of cyanobacterial toxins In vivo and In vitro by mouse bioassay: A review. Mini Reviews in Medicinal Chemistry. 1 May 2022;22(8):1131-1151
  7. 7. Nowruzi B, Bouaïcha N, Metcalf JS, Porzani SJ, Konur O. Plant-cyanobacteria interactions: Beneficial and harmful effects of cyanobacterial bioactive compounds on soil-plant systems and subsequent risk to animal and human health. Phytochemistry. 2021 Dec;1(192):112959
  8. 8. Nowruzi B, Porzani SJ. Toxic compounds produced by cyanobacteria belonging to several species of the order Nostocales: A review. Journal of Applied Toxicology. 2021;41(4):510-548
  9. 9. Baker DD et al. The value of natural products to future pharmaceutical discovery. Natural Product Reports. 2007;24(6):1225-1244
  10. 10. Singh SB, Barrett JF. Empirical antibacterial drug discovery—foundation in natural products. Biochemical pharmacology. 2006;71(7):1006-1015
  11. 11. Harvey AL. Natural products in drug discovery. Drug Discovery Today. 1 Oct 2008;13(19-20):894-901
  12. 12. Lam KS. New aspects of natural products in drug discovery. Trends in Microbiology. 1 Jun 2007;15(6):279-289
  13. 13. McChesney JD. Plant natural products: Back to the future or into extinction? Phytochemistry. 1 Jul 2007;68(14):2015-2022
  14. 14. Rishton GM. Natural products as a robust source of new drugs and drug leads: Past successes and present day issues. The American Journal of Cardiology. 2008;101(Suppl):43D-49D
  15. 15. Feher M, Schmidt JM. Property distributions: Differences betweendrugs, natural products, and molecules from combinatorial chemistry. Journal of Chemical. 2003;43:218-227
  16. 16. Grabowski K, Schneider G. Properties and architecture of drugs andnatural products revisited. Current Chemical Biology. 2007;1:115-127
  17. 17. Galm U, Shen B. Natural product drug discovery: The times have never been better. Chemistry & Biology. 2007;14:1098-1104
  18. 18. Sunazuka T et al. Efficient total synthesis of novel bioactive microbial metabolites. Accounts of Chemical Research. 2008;41:302-314
  19. 19. Boldi AM. Libraries from natural product-like scaffolds. Current Opinion in Chemical Biology. 2004;8:281-286
  20. 20. Yao N et al. Synthesis of flavonoid analogues as scaffolds for natural product-based combinatorial libraries. Journal of Combinatorial Chemistry. 2007;9:668-676
  21. 21. Jarvis LM. Liquid gold mine. Chemical and Engineering News. 2007;85:22-28
  22. 22. Molinski TF, Dalisay DS, Lievens SL, Saludes JP. Drug development from marine natural products. Jan 2009;8(1):69-85
  23. 23. Harvey AL. Natural products as a screening resource. Current Opinion in Chemical Biology. 2007;11:480-484
  24. 24. McAlpine JB et al. Microbial genomics as a guide to drug discovery andstructural elucidation: ECO-02301, a novel antifungal agent, as an example. Journal of Natural Products. 2005;68:493-496
  25. 25. Newman DJ, Hill RT. New drugs from marine microbes: The tide is turning. Journal of Industrial Microbiology & Biotechnology. 2006;33:539-544
  26. 26. Kennedy J. Mutasynthesis, chemobiosynthesis, and back to semi-synthesiscombining synthetic chemistry and biosynthetic engineering for diversifying natural products. Production Reports. 2008;25:25-34
  27. 27. Kopp F, Marahiel MA. Where chemistry meets biology: The chemoenzymatic synthesis of nonribosomal peptides and polyketides. Current Opinion in Biotechnology. 2007;18:513-520
  28. 28. Ramaswamy AV, Flatt PM, Edwards DJ, Simmons TL, Han B. The secondary metabolites and biosynthetic gene clusters of marine cyanobacteria. Applications in biotechnology. In: Proksch P, Muller WEG, editors. Frontiers in Marine Biotechnology. 2006. pp. 175-224
  29. 29. Singh RK, Tiwari SP, Rai AK, Mohapatra TM. Cyanobacteria: An emerging source for drug discovery. The Journal of Antibiotics. Jun 2011;64(6):401-412
  30. 30. Littleton J et al. Novel approaches to plant drug discovery based on high throughput pharmacological screening and genetic manipulation. Life Sciences. 2005;78:467-475
  31. 31. Freiberg C, Brotz-Oesterhelt H. The impact of transcriptome and proteome analyses on antibiotic drug discovery. Current Opinion in Microbiology. 2004;7(5):451-459
  32. 32. Elnitski L, Jin VX, Farnham PJ, Jones SJM. Locating mammalian transcription factor binding sites: A survey of computational and experimental techniques. Genome Research. 2006;16(12):1455-1464
  33. 33. Brötz-Oesterhelt H, Elisabeth J, Labischinski BH. Bacterial proteomics and its role in antibacterial drug discovery. Mass Spectrometry Reviews. 2005;24(4):549-565
  34. 34. Pullan ST, Gidley MD, Jones RA, et al. Nitric oxide in chemostat-cultured Escherichia coli is sensed by Fnr and otherglobal regulators: Unaltered methionine biosynthesis indicates lack of S nitrosation. Journal of Bacteriology. 2007;189(5):1845-1855
  35. 35. Ross PL, Huang YN, Marchese JN, et al. Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Molecular & Cellular Proteomics. 2004;3(12):1154-1169
  36. 36. Ishida K et al. Plasticity and evolution of aeruginosin biosynthesis in cyanobacteria. Applied and Environmental Microbiology. 2009;75:2017-2026
  37. 37. Williams PG. Panning for chemical gold: marine bacteria as a source of new therapeutics. Trends in biotechnology. 1 Jan 2009;27(1):45-52
  38. 38. Devillers J et al. Prediction of biological activity profiles of cyanobacterial secondary metabolites. SAR QSAR Environmental Research. 2007;18:629-643
  39. 39. Luesch H et al. A functional genomics approach to the mode of action of apratoxin. Natural Chemical Biology. 2006;2:158-167
  40. 40. Pérez Gutiérrez RM, Martínez Flores A, Vargas Solís R, Carmona Jimenez J. Two new antibacterial norabietane diterpenoids from cyanobacteria, Microcoleous lacustris, Journal of Natural Medicines. 2008;62(3):328-331
  41. 41. Medina RA et al. Coibamide A, a potent antiproliferative cyclic depsipeptide from the panamanian marine cyanobacterium Leptolyngbya sp. Journal of the American Chemical Society. 2008;130:6324-6325
  42. 42. Edwards DJ, Marquez BL, Nogle LM, McPhail K, Goeger DE, Roberts MA, Gerwick WH. Structure and biosynthesis of the jamaicamides, new mixed polyketide-peptide neurotoxins from the marine cyanobacterium Lyngbya majuscula. Chemistry & Biology, 2004;11(6):817-833
  43. 43. Burja AM, Banaigs B, Abou-Mansour E, Burgessd JG, Wrighta PC. Marine cyanobacteriaÐa prolific source of natural products. Tetrahedron. 2001;57:9347-9377
  44. 44. Lu J, Chen F, Hodson RE. Distribution, isolation, host specificity, and diversity of cyanophages infecting marine Synechococcus spp. in river estuaries. Applied and Environmental Microbiology. 2001;67(7):3285-3290
  45. 45. Mozzachiodi R, Scuri R, Roberto M, Brunelli M. Caulerpenyne, a toxin from the seaweed Caulerpa taxifolia, depresses afterhyperpolarization in invertebrate neurons. Neuroscience. 2001;107(3):519-526
  46. 46. Taori K et al. Structure and activity of largazole, a potent antiproliferative agent from the Floridian marine cyanobacterium Symploca sp. Journal of the American Chemical Society. 2008a;130:1806-1807
  47. 47. Mundt S, Kreitlow S, Novotony A, Effmert U. Biochemical and pharmacological investigations of selected cyanobacteria. International Journal of Environmental Health Research. 2001;203:327-334
  48. 48. Christiansen G, Fastner J, Erhard M, Börner T, Dittmann E. Microcystin biosynthesis in Planktothrix: genes, evolution, and manipulation. Journal of Bacteriology. 2003;185(2):564-572
  49. 49. Becker JE, Moore RE, Moore BS. Cloning, sequencing, and biochemical characterization of the nostocyclopeptide biosynthetic gene cluster: molecular basis for imine macrocyclization. Gene. 2004;325:35-42
  50. 50. Welker M, Brunke M, Preussel K, Lippert I, von Döhren H. Diversity and distribution of Microcystis (Cyanobacteria) oligopeptide chemotypes from natural communities studied by single-colony mass spectrometry. Microbiology. 2004;150(6):1785-1796
  51. 51. Nowruzi B, Khavari-Nejad RA, Sivonen K, Kazemi B, Najafi F, Nejadsattari T. Identification and toxigenic potential of a cyanobacterial strain (Stigomena sp.). Progress in Biological Sciences. 2013;3(1):79-85
  52. 52. Neuhof T, Schmieder P, Preussel K, Dieckmann R, Pham H, Bartl F, et al. Hassallidin A, a glycosylated lipopeptide with antifungal activity from the cyanobacterium Hassallia sp. Journal of Natural Products. 27 May 2005;68:695-700
  53. 53. Horgen FD, Kazmierski EB, Westenburg HE, Yoshida WY, Scheuer PJ. Malevamide D: isolation and structure determination of an isodolastatin H Analogue from the Marine Cyanobacterium Symploca h ydnoides. Journal of Natural Products. 2002;65(4):487-491
  54. 54. Luesch H, Yoshida WY, Moore RE, Paul VJ. New apratoxins of marine cyanobacterial origin from Guam and Palau. Bioorganic & Medicinal Chemistry. 2002;10(6):1973-1978
  55. 55. Sato M, Nimura-Matsune K, Watanabe S, Chibazakura T, Yoshikawa H. Expression analysis of multiple dnaK genes in the cyanobacterium Synechococcus elongatus PCC 7942. Journal of Bacteriology. 2007;189(10):3751-3758
  56. 56. Lautru S, Challis GL. Substrate recognition by nonribosomal peptide synthetase multi-enzymes. Microbiology. 2004;150(6):1629-1636
  57. 57. LePage KT, Goeger D, Yokokawa F, Asano T, Shioiri T, Gerwick WH, Murray TF. The neurotoxic lipopeptide kalkitoxin interacts with voltage-sensitive sodium channels in cerebellar granule neurons. Toxicology Letters. 2005;158(2):133-139
  58. 58. Palozza P, Torelli C, Boninsegna A, Simone R, Catalano A, Mele MC, et al. Growth-inhibitory effects of the astaxanthin-rich alga Haematococcus pluvialis in human colon cancer cells. Cancer Letters. 2009;283(1):108-117
  59. 59. Catarina Guedes A, Barbosa CR, Amaro HM, Pereira CI, Xavier Malcata F. Microalgal and cyanobacterial cell extracts for use as natural antibacterial additives against food pathogens. International Journal of Food Science & Technology. 2011;46(4):862-870
  60. 60. Lee JB, Hayashi K, Hirata M, Kuroda E, Suzuki E, Kubo Y, Hayashi T. Antiviral sulfated polysaccharide from Navicula directa, a diatom collected from deep-sea water in Toyama Bay. Biological and Pharmaceutical Bulletin. 2006;29(10):2135-2139
  61. 61. Shih SR, Tsai KN, Li YS, Chueh CC, Chan EC. Inhibition of enterovirus 71-induced apoptosis by allophycocyanin isolated from a blue-green alga Spirulina platensis. Journal of Medical Virology. 2003;70(1):119-125
  62. 62. Marquez BL, Watts KS, Yokochi A, Roberts MA, Verdier-Pinard P, Jimenez JI, et al. Structure and absolute stereochemistry of hectochlorin, a potent stimulator of actin assembly. Journal of Natural Products. 2002;65(6):866-871
  63. 63. Nie B, Chen Z. Progress in studies on naturally occurring polyacetylenes. Chinese Traditional and Herbal Drugs. 1994;11
  64. 64. Ohta S, Ono F, Shiomi Y, Nakao T, Aozasa O, Nagate T, et al. Anti-herpes simplex virus substances produced by the marine green alga, Dunaliella primolecta, Journal of Applied Phycology. 1998;10(4):349-356
  65. 65. H., Kitano. Systems biology: A brief overview. Science. 2002;2002(295):1662-1664
  66. 66. Sakharkar KR, Sakharkar MK, VTK C. A novel genomics approach for the identifi cation of drug targets in pathogens, with special reference to pseudomonas aeruginosa. In Silico Biology. 2004;4(3):355-360
  67. 67. Brahmachary M, Krishnan SPT, Koh JLY, et al. ANTIMIC: A database of antimicrobial sequences. Nucleic Acids Research. 2004;32(Suppl 1):D586-D589
  68. 68. Leikoski N et al. Highly diverse cyanobactins in strains of the genus Anabaena. Applied and Environmental Microbiology. 2010;76:701-709
  69. 69. Gademann K, Portmann C. Secondary metabolites from cyanobacteria: complex structures and powerful bioactivities. Current Organic Chemistry. 2008;12(4):326-341
  70. 70. Smith JL, Boyer GL, Zimba PV. A review of cyanobacterial odorous and bioactive metabolites: Impacts and management alternatives in aquaculture. Aquaculture. 2008;280(1-4):5-20
  71. 71. Sivonen K, Börner T. Bioactive compounds produced by cyanobacteria. In The Cyanobacteria 2008 (pp. 159-197). Caister Academic Press
  72. 72. Tripathi A, Puddick J, Prinsep MR, Lee PP, Tan LT. Hantupeptins B and C, cytotoxic cyclodepsipeptides from the marine cyanobacterium Lyngbya majuscula. Phytochemistry. 2009;71(2-3):307-311
  73. 73. Tidgewell K, Clark BR, Gerwick WH. 2.06-The natural products chemistry of cyanobacteria. In: Liu HW, Mander L, editors. Comprehensive Natural Products; 2009
  74. 74. Portmann C, Blom JF, Gademann K, Jüttner F. Aerucyclamides A and B: isolation and synthesis of toxic ribosomal heterocyclic peptides from the cyanobacterium Microcystis aeruginosa PCC 7806. Journal of Natural Products. 2008;71(7):1193-1196
  75. 75. Spolaore P, Joannis-Cassan C, Duran E, Isambert A. Commercial applications of microalgae. Journal of Bioscience and Bioengineering. 2006;101(2):87-96
  76. 76. Jaiswal P, Singh PK, Prasanna R. Cyanobacterial bioactive molecules—an overview of their toxic properties. Canadian Journal of Microbiology. 2008;54(9):701-717
  77. 77. Rastogi RP, Sinha RP. Biotechnological and industrial significance of cyanobacterial secondary metabolites. Biotechnology advances. 2009;27(4):521-539
  78. 78. Gerwick WH, Coates RC, Engene N, Gerwick L, Grindberg RV, Jones AC, Sorrels CM. Giant marine cyanobacteria produce exciting potential pharmaceuticals. Microbe-American Society for Microbiology. 2008;3(6):277
  79. 79. Kaushik P, Chauhan A. Cyanobacteria: Antibacterial Activity. New India Publishing; 2009
  80. 80. Blunt JW, Copp BR, Munro MH, Northcote PT, Prinsep MR. Marine natural products. Natural Product Reports. 2010;27(2):165-237
  81. 81. Shimizu Y. Microalgal metabolites. Current Opinion in Microbiology. 2003;6(3):236-243
  82. 82. Barchi Jr JJ, Moore RE, Patterson GM. Acutiphycin and 20, 21-didehydroacutiphycin, new antineoplastic agents from the cyanophyte Oscillatoria acutissima. Journal of the American Chemical Society. 1984;106(26):8193-8197
  83. 83. Orjala J, Gerwick WH. Barbamide, a chlorinated metabolite with molluscicidal activity from the Caribbean cyanobacterium Lyngbya majuscula. Journal of Natural Products. 1996;59(4):427-430
  84. 84. Chang Z, Sitachitta N, Rossi JV, Roberts MA, Flatt PM, Jia J, Sherman DH, Gerwick WH. Biosynthetic pathway and gene cluster analysis of Curacin A, an Antitubulin natural product from the tropical Marine Cyanobacterium Lyngbya m ajuscula. Journal of Natural Products. 2004;67(8):1356-1367
  85. 85. Yokokawa F, Fujiwara H, Shioiri T. Total synthesis and revision of absolute stereochemistry of antillatoxin, an ichthyotoxic cyclic lipopeptide from marine cyanobacterium Lyngbya majuscula. Tetrahedron. 2000;56(12):1759-1775
  86. 86. Orjala J, Nagle DG, Hsu V, Gerwick WH. Antillatoxin: an exceptionally ichthyotoxic cyclic lipopeptide from the tropical cyanobacterium Lyngbya majuscula. Journal of the American Chemical Society. 1995;117(31):8281-8282
  87. 87. Mitchell SS, Faulkner DJ, Rubins K, Bushman FD. Dolastatin 3 and two novel cyclic peptides from a palauan collection of Lyngbya m ajuscula. Journal of Natural Products. 2000;63(2):279-282
  88. 88. Saker ML, Eaglesham GK. The accumulation of cylindrospermopsin from the cyanobacterium Cylindrospermopsis raciborskii in tissues of the Redclaw crayfish Cherax quadricarinatus. Toxicon. 1999;37(7):1065-1077
  89. 89. Nakamura H, Hamer HA, Sirasani G, Balskus EP. Cylindrocyclophane biosynthesis involves functionalization of an unactivated carbon center. Journal of the American Chemical Society. 2012;134(45):18518-18521
  90. 90. Carmichael WW, Eschedor JT, Patterson GM, Moore RE. Toxicity and partial structure of a hepatotoxic peptide produced by the cyanobacterium Nodularia spumigena Mertens emend. L575 from New Zealand. Applied and Environmental Microbiology. 1988;54(9):2257-2263
  91. 91. Ramaswamy AV, Flatt PM, Edwards DJ, Simmons TL, Han B, Gerwick WH. The secondary metabolites and biosynthetic gene clusters of marine cyanobacteria. Applications in biotechnology. Frontiers in Marine Biotechnology. Horizon Bioscience. 2006:175-224
  92. 92. Schmidt EW, Nelson JT, Rasko DA, Sudek S, Eisen JA, Haygood MG, Ravel J. Patellamide A and C biosynthesis by a microcin-like pathway in Prochloron didemni, the cyanobacterial symbiont of Lissoclinum patella. Proceedings of the National Academy of Sciences. 2005;102(20):7315-7320
  93. 93. Williams AB, Jacobs RS. A marine natural product, patellamide D, reverses multidrug resistance in a human leukemic cell line. Cancer Letters. 1993;71(1-3):97-102
  94. 94. Milligan KE, Marquez BL, Williamson RT, Gerwick WH. Lyngbyabellin B, a toxic and antifungal secondary metabolite from the marine cyanobacterium Lyngbya m ajuscula. Journal of Natural Products. 2000;63(10):1440-1443
  95. 95. Esser MT, Mori T, Mondor I, Sattentau QJ, Dey B, Berger EA, Boyd MR, Lifson JD. Cyanovirin-N binds to gp120 to interfere with CD4-dependent human immunodeficiency virus type 1 virion binding, fusion, and infectivity but does not affect the CD4 binding site on gp120 or soluble CD4-induced conformational changes in gp120. Journal of Virology. 1999;73(5):4360-4371
  96. 96. Hayashi O, Ono S, Ishii K, Shi Y, Hirahashi T, Katoh T. Enhancement of proliferation and differentiation in bone marrow hematopoietic cells by Spirulina (Arthrospira) platensis in mice. Journal of Applied Phycology. 2006;18(1):47-56
  97. 97. Moore RE. Cyanobacterial secondary metabolites. In: Microbial Growth on C1 Compounds. Dordrecht: Springer; 1996. pp. 245-252
  98. 98. Harrigan GG, Luesch H, Yoshida WY, Moore RE, Nagle DG, Paul VJ. Symplostatin 2: a dolastatin 13 analogue from the marine cyanobacterium Symploca hydnoides. Journal of Natural Products. 1999;62(4):655-658
  99. 99. Wrasidlo W, Mielgo A, Torres VA, Barbero S, Stoletov K, Suyama TL, Klemke RL, Gerwick WH, Carson DA, Stupack DG. The marine lipopeptide somocystinamide A triggers apoptosis via caspase 8. Proceedings of the National Academy of Sciences. 2008;105(7):2313-2318
  100. 100. Soria-Mercado IE, Pereira A, Cao Z, Murray TF, Gerwick WH. Alotamide A, a novel neuropharmacological agent from the marine cyanobacterium Lyngbya bouillonii. Organic Letters. 2009;11(20):4704-4707
  101. 101. Han B, Gross H, Goeger DE, Mooberry SL, Gerwick WH. Aurilides B and C, Cancer Cell Toxins from a Papua New Guinea Collection of the Marine Cyanobacterium Lyngbya m ajuscula. Journal of Natural Products. 2006;69(4):572-575
  102. 102. Kwan JC, Eksioglu EA, Liu C, Paul VJ, Luesch H. Grassystatins A− C from marine cyanobacteria, potent cathepsin E inhibitors that reduce antigen presentation. Journal of Medicinal Chemistry. 2009;52(18):5732-5747
  103. 103. Graf KM, Tabor MG, Brown ML, Paige M. Synthesis of (S)-jamaicamide C carboxylic acid. Organic Letters. 2009;11(23):5382-5385
  104. 104. Li H, Liao X, Sun Y, Zhou R, Long W, Li L, et al. An economical synthesis of caulerpin and evaluation of its new anticancer activities. ChemistrySelect. 2018;3(44):12406-12409
  105. 105. Banker R, Carmeli S. Tenuecyclamides A− D, Cyclic Hexapeptides from the Cyanobacterium Nostoc s pongiaeforme var. t enue. Journal of Natural Products. 1998;61(10):1248-1251
  106. 106. Mynderse JS, Hunt AH, Moore RE. 57-Normajusculamide C, a minor cyclic depsipeptide isolated from Lyngbya majuscula. Journal of Natural Products. 1988;51(6):1299-12301
  107. 107. Nagle DG, Paul VJ, Roberts MA. Ypaoamide, a new broadly acting feeding deterrent from the marine cyanobacterium Lyngbya majuscula. Tetrahedron Letters. 1996;37(35):6263-6266
  108. 108. Ribe S, Kondru RK, Beratan DN, Wipf P. Optical rotation computation, total synthesis, and stereochemistry assignment of the marine natural product pitiamide A. Journal of the American Chemical Society. 2000;122(19):4608-4617
  109. 109. Lehtimäki J. Characterisation of cyanobacterial strains originating from the Baltic Sea with emphasis on Nodularia and its toxin, nodularin. 2000
  110. 110. Lehtimäki J, Lyra C, Suomalainen S, Sundman P, Rouhiainen L, Paulin L, Salkinoja-Salonen M, Sivonen K. Characterization of Nodularia strains, cyanobacteria from brackish waters, by genotypic and phenotypic methods. International Journal of Systematic and Evolutionary Microbiology. 2000;50(3):1043-1053
  111. 111. Bernardo PH, Chai CL, Heath GA, Mahon PJ, Smith GD, Waring P, Wilkes BA. Synthesis, electrochemistry, and bioactivity of the cyanobacterial calothrixins and related quinones. Journal of Medicinal Chemistry. 2004;47(20):4958-4963
  112. 112. 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. Journal of Natural Products. 1997;60(12):1251-1260
  113. 113. Nowruzi B, Haghighat S, Fahimi H, Mohammadi E. Nostoc cyanobacteria species: A new and rich source of novel bioactive compounds with pharmaceutical potential. Journal of Pharmaceutical Health Services Research. 2018;9(1):5-12
  114. 114. Nowruzi B, Sarvari G, Blanco S. Applications of cyanobacteria in biomedicine. In: Handbook of Algal Science, Technology and Medicine. Academic Press; 2020. pp. 441-4553
  115. 115. Villa FA, Lieske K, Gerwick L. Selective MyD88-dependent pathway inhibition by the cyanobacterial natural product malyngamide F acetate. European Journal of Pharmacology. 2010;629(1-3):140-146
  116. 116. Abed RM, Dobretsov S, Sudesh K. Applications of cyanobacteria in biotechnology. Journal of Applied Microbiology. 2009;106(1):1-2
  117. 117. Gademann K, Kobylinska J. Antimalarial natural products of marine and freshwater origin. The Chemical Record. 2009;9(3):187-198
  118. 118. Dunlap WC, Battershill CN, Liptrot CH, Cobb RE, Bourne DG, Jaspars M, Long PF, Newman DJ. Biomedicinals from the phytosymbionts of marine invertebrates: A molecular approach. Methods. 2007;42(4):358-376
  119. 119. Luescher-Mattli. M. Algae as a possible source of new antiviral agents. Current Medicinal Chemistry: Anti-Infective Agents. 2003;2:219-225
  120. 120. Jafari Porzani S, Konur O, Nowruzi B. Cyanobacterial natural products as sources for antiviral drug discovery against COVID-19. Journal of Biomolecular Structure & Dynamics. 2021;11:1-7
  121. 121. Anvar AA, Nowruzi B. Bioactive properties of spirulina: A review. Microbiology Bioactive 2021;4:134-142
  122. 122. Kenji LK et al. “Isolation of an antiviral polysacharide, nostoflan, from a terrestrial cyanobacteium.” Nostoc flagilliforme. Journal of Natural Products. 2005;68:1037
  123. 123. Zainuddin EN et al. Cyclic depsipeptides, ichthyopeptins A and B, from Microcystis ichthyoblabe. Journal of Natural Products. 2007;70:1084
  124. 124. Klasse PJ, Shattock R, Moore JP. Antiretroviral drug—Based microbicides to prevent HIV-1 sexual transmission. Annual Review of Medicine. 2008;59:455-471
  125. 125. Bokesch HR et al. A potent novel anti-HIV protein from the cultured cyanobacterium. Scytonema Varium Biochem. 2003;42:2578-2584
  126. 126. Reinert R, Donald EL, Rosi FX, Watal C, Dowzicky. M. Antimicrobial susceptibility among organisms from the Asia/Pacific Rim, Europe and Latin and North America collected as part of TEST and the in vitro activity of tigecycline. The Journal of Antimicrobial Chemotherapy. 2007;60:1018-1029
  127. 127. Biondi N et al. Cyanobacteria from benthic mats of Antarctic lakes as a new source of bioactivities. Journal of Applied Microbiology. 2008;105:105-115
  128. 128. Mundt S, Kreitlow S, Jansen R. Fatty acids with antibacterial activity from the cyanobacterium Oscillatoria redekei HUB051. Journal of Applied Phycology. 2003;15:263-267
  129. 129. Bhateja P, Mathur T, Pandya M, Fatma T, Rattan A. Activity of blue- green microalgae extracts against in vitro generated Staphylococcus aureus with reduced susceptibility to vancomycin. Fitoterpia. 2006;77:233-235
  130. 130. Simmons LT et al. Viridamides A and B, lipodepsipeptides with antiprotozoal activity from marine cyanobacterium. Oscillatoria Nigro Viridis. Natural Products. 2008;71:1544-1550
  131. 131. Lanzer MR, Rohrbach P. Subcellular pH and Ca2+ in plasmodium falciparum implications for understanding drug resistance mechanisms. Current Science. 2007;92:1561-1570
  132. 132. Sheifert K, Lunke A, Croft SL, Kayser O. Antileishmanial structure- activity relationships of synthetic phospholipids: in vitro and in vivo activities of selected derivatives. Antimicrobial Agents and Chemotherapy. 2007;51:4525-4528
  133. 133. Barbaraus D, Kaiser M, Brun R, Gademann K. Potent and selective antiplasmodial activity of the cyanobacterial alkaloid nostocarboline and its dimers. Bioorganic & Medicinal Chemistry Letters. 2008;18:4413-4415
  134. 134. Khan Z, Bhadouria P, Bisen PS. Nutritional and therapeutic potential of Spirulina. Current Pharmaceutical Biotechnology. 2005;6:373-379
  135. 135. Shen PP et al. Effects of cyanobacteria bloom extract on some parameters of immune function in mice. Toxicology Letters. 2003;143:27-36
  136. 136. Mita AC, Hammond LA, Bonate PL, Weiss G, McCreery H, Syed S, et al. Phase I and pharmacokinetic study of tasidotin hydrochloride (ILX651), a third-generation dolastatin −15 analogues, administered weekly for 3 weeks every 28 days in patients with advanced solid tumors. Clinical Cancer Research. 2006;12:5207-5215
  137. 137. Watanabe J, Minami M, Kobayashi M. Antitumor activity of TZT-1027 (soblidotin). Anticancer Research. 2006;26:1973-1981
  138. 138. Porzani SJ, Lima ST, Metcalf JS, Nowruzi B. In vivo and In vitro toxicity testing of cyanobacterial toxins: A Mini-review. Reviews of Environmental Contamination and Toxicology. 2021;258:109-150
  139. 139. Liang J et al. Cryptophycin-309249 and other cryptophycins analogs: Preclinical efficacy studies with mouse and human tumors. Investigational New Drugs. 2005;23:213-224
  140. 140. Beck ZQ , Aldrich CC, Magarvey NA, Georg GI, Sherman DH. Chemoenzymatic synthesis of cryptophycin/arenastatin natural products. Biochemistry. 2005;44:13457-13466
  141. 141. Taori K, Paul VJ, Leusch H. Kempopeptins A & B, protease inhibitors with different selectivity profiles from a marine cyanobacterium Lyngbya sp. J. Natural Products. 2008b;71:1625-1629
  142. 142. Ying Y et al. Total synthesis and molecular target of largazole,a histone deacetylase inhibitor. Journal of the American Chemical Society. 2008;130:8455-8459
  143. 143. Federici L, Woebking B, Velamakanni S, et al. New structure model for the ATP-binding cassette multidrug transporter LmrA. Biochemical Pharmacology. 2007;74(5):672-678
  144. 144. Neuhof T, Schmieder P, Seibold M. Hassallidin B—Second antifungal member of the Hassallidin family. Bioorganic and Medicinal Chemistry Letters. 2006;16:4220-4222

Written By

Bahareh Nowruzi

Submitted: 13 April 2022 Reviewed: 06 July 2022 Published: 07 October 2022