Next Article in Journal
Valorization of Spent Coffee Grounds as a Substrate for Fungal Laccase Production and Biosorbents for Textile Dye Decolorization
Previous Article in Journal
Diversity and Biological Activity of Secondary Metabolites Produced by the Endophytic Fungus Penicillium ochrochlorae
Previous Article in Special Issue
Improving Organic Acid Secretion of Aspergillus niger by Overexpression C4-Dicarboxylic Acid Transporters
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pigments from Microorganisms: A Sustainable Alternative for Synthetic Food Coloring

1
Department of Botany, The Institute of Science, 15, Madame Cama Road, Mumbai 400032, India
2
Chimie et Biotechnologie des Produits Naturels (CHEMBIOPRO Lab) & ESIROI Agroalimentaire, Université de la Réunion, 15 Avenue René Cassin, 97744 Saint-Denis, France
3
TERI-Deakin Nano Biotechnology Centre, The Energy and Resources Institute, Darbari Seth Block, IHC Complex, Lodhi Road, New Delhi 110003, India
*
Author to whom correspondence should be addressed.
Fermentation 2025, 11(7), 395; https://doi.org/10.3390/fermentation11070395
Submission received: 21 May 2025 / Revised: 30 June 2025 / Accepted: 5 July 2025 / Published: 10 July 2025
(This article belongs to the Special Issue Metabolic Engineering in Microbial Synthesis)

Abstract

Microbial pigments are gaining acceptance as a green, sustainable substitute for synthetic food pigments due to growing health issues and their adverse health impacts. This review provides an overview of the potential of microbial pigments as natural food colorants and the advantages of microbial pigments over synthetic pigments. Microbial pigments are a natural source of color with medicinal properties like anticancer, antimicrobial, and antioxidant activity. Important pigments covered are astaxanthin, phycocyanin, prodigiosin, riboflavin, β-carotene, violacein, melanin, and lycopene, and their microbial origins and characteristics. The review also covers commercial production of microbial pigments, i.e., strain development and fermentation processes. Microbial pigments also find extensive applications in food industries, including preservatives for food. Also covered are their pharmacological activity and other applications, such as in the formation of nanoparticles. Finally, the challenges and future directions of microbial pigment production are covered, including the need for cost-effective production, regulatory acceptability, and the potential of genetic engineering and fermentation-based technologies to enhance pigment yield and quality.

1. Introduction

Colors are utilized to enhance the appearance of different commercial products such as food, clothing, and drugs. Food color is both a visual and practical cue, as well as an indicator of freshness and safety [1]. Although color is an important sensory characteristic of food, it is not a texture of food or a nutrient, or a flavor indicator [2]. Most importantly, color can affect how consumers perceive and anticipate these characteristics.
Application of synthetic colors in foods is prevalent due to the fact that they give permanent effects and result in strong color without altering the taste [3]. Some of the most common synthetic food colors include Erythrosine, Sunset Yellow FCF (E110), Allura Red, Tartrazine, Blue No. 1, and Blue No. 2, as shown in Figure 1. These food colors enhance the visual appeal and acceptability of various food products, including candy, sodas, and meat [4,5].
Nonetheless, the use of synthetic food dyes has raised considerable health concerns. As noted in recent research, studies on the effects on individuals consuming excessive amounts of synthetic food additives have shown that excessive intake can result in cardiovascular disease, gastrointestinal complications, respiratory diseases, dermatologic problems, and neurologic adverse reactions [6]. There is significant evidence indicating an association between food additive intolerance and hypersensitivity, hyperactivity in children, physical and mental ailments, allergies, and cancer [7].
Synthetic food colorings, especially azo dyes such as Allura Red and Tartrazine, have been linked to oral problems, diseases of the cardiovascular system, and carcinogenic activity [3]. These dyes can stain dentin and increase bacterial growth in dental crowns, contributing to plaque formation. Additionally, certain bacterial pathogens can elicit inflammatory reactions in the body and enter the bloodstream, thus potentially contributing to plaque build-up along major arteries of the heart and a host of cardiovascular diseases, including ischemic heart disease, strokes, coronary artery disease, and heart attacks [6].
Gupta et al. reported that synthetic food colorants, such as Tartrazine, Amaranth, and Erythrosine, can indeed have toxic effects on human lymphocyte cells in vitro and can interact directly with DNA [8]. Significant cytotoxicity and genotoxicity were observed with an (8 mM) treatment of high-concentration Amaranth; the frequency of sister chromatid exchanges increased by 1.7-fold compared to the control. At 2–8 mM, Erythrosine showed potent cytotoxicity and cytostaticity, while Tartrazine was hazardous at 4–8 mM dosages. Remarkably, using spectroscopy and electrophoresis, these dyes were demonstrated to bind to calf thymus DNA, resulting in the degradation of linear double-stranded DNA. PCR was used for DNA amplification (treated with a colorant) and showed reduced and delayed electrophoretic mobility, demonstrating that dye molecules can directly bind to DNA [9].
Because they are unsafe for health, pigments from microorganisms are a good option and a fine substitute for food colorants [10,11]. Microbial sources are more advantageous than plant sources for natural food colorants because they can be mass-produced, are readily available, provide high quantities, and are easy to handle [12,13]. For example, genetically modified Yarrowia lipolytica strains yield β-carotene at concentrations of up to 400 mg/L within 72–96 h, while the extraction of β-carotene from marigold flowers provides only 15–20 mg/kg of petals and the flowers take weeks to grow [13,14]. Therefore, the production of β-carotene with microbes not only increases the yield by a significant amount but also decreases the cost of production by 25–35% compared to traditional plant extraction techniques using optimized bioreactor systems [15].
There are different sources of pigments from microorganisms such as unicellular fungi, bacteria, photosynthetic bacteria, microalgae, and cyanobacteria [16]. Photosynthetic microorganisms, particularly microalgae and cyanobacteria, are excellent sources for pigment production while simultaneously capturing CO2, offering a dual benefit of valuable product generation and greenhouse gas mitigation [17,18,19]. Many microbial pigments can serve as colorants, as well as providing medicinal health benefits. Compared to synthetic colors, natural pigments can offer multiple physiological and biochemical properties, including antioxidant, antimicrobial, anticancer, and immunomodulatory properties, and can serve as substitutes in various industries [20]. The pigments produced by various fungi, such as azaphilones, carotenoids, quinones, and melanin, have various pharmacological activities and may help to treat infections, cancers, Alzheimer’s disease, and cardiovascular disorders [21]. Also, their behavior towards the host’s immune system can result in favorable characteristics in the control of fungal infections [22]. Because of their many advantages over synthetic colorants, microbial pigments have attracted considerable interest in the food industry. They offer a safe, eco-friendly, and sustainable alternative to synthetic dyes [13,23].
Microbial pigments have been reviewed in many reviews, but most of them have dealt primarily with their general advantages, biosynthetic processes, and drug properties [13,24,25,26,27,28]. To completely understand the industrial feasibility of microbial pigments, including their benefits over synthetic pigments and regulatory considerations, more research is necessary. This review integrates a comprehensive classification of microbial pigments based on their structure, industrial relevance, and various uses, such as in food industries and nanoparticle synthesis. It also explores commercialization challenges and recent advances in bioprocess optimization. This review offers a new perspective that enhances and extends previous studies.

2. Importance of Microbial Pigments

Microbial pigments are gaining consumer popularity because of their low environmental footprint and potential health benefits. One of the main benefits is that microbial pigments are fully biodegradable; therefore, the disposal of these products does not harm the environment [29]. In addition, few or no chemical processes are involved in the manufacturing of microbial pigments, thus minimizing adverse effects and environmental damage [14]. Moreover, in addition to their environmental benefits, microbial pigments provide a colorful range of muted, relaxing colors, making them popular among consumers looking for a more natural and harmonious aesthetic [30].
Synthetic colors are resistant to typical reduction and oxidation processes, making them highly challenging to remove from industrial effluents [31]. Their destruction produces byproducts that have been linked to health risks, while natural color degradation byproducts have not been found to include such toxic substances [32]. Synthetic color-based effluents can pose severe harm to water streams and ecosystems because of their synthetic origin and complicated molecular structures. For example, degradation of azo dyes, accounting for over half of all dyes manufactured annually, can result in the discharge of carcinogenic aromatic amines [33]. The aromatic amines, primarily azo dye degradation products, are recognized as primary carcinogenic compounds and pose serious risks to human health and the environment [34]. Microbial pigments are safer and more environmentally friendly because they originate from renewable sources, have a simple structure, and are biodegradable [29]. Unlike synthetic dyes, which may release harmful degradation byproducts, microbial pigments are increasingly in demand in a broad array of manufacturing industries because of their eco-friendliness and bioactivity [16,35]. They offer several advantages over synthetic colorants, including their low or non-toxicity in ecotoxicological and cytotoxicity tests [23]. This gives them the capacity to be a safer alternative when applied in food, pharmaceuticals, textiles, and other industries [20,36].
Even though microbial pigments are well known for being biodegradable and environmentally friendly, their full environmental evaluation requires strong life cycle assessment (LCA). LCA research indicates that microbial pigments, as much as they are currently constrained by high water and energy requirements during fermentation and extraction, have substantially smaller environmental impacts in terms of greenhouse gas emissions, toxicity, and fossil resource depletion compared to their synthetic equivalents [11,20]. The utilization of renewable substrates and the optimization of bioprocesses can further enhance the circular economy-based sustainability of microbial pigment production [37]. These findings have been confirmed by recent studies through comparative LCA of microbial and synthetic pigments [38,39].
Many hazardous and allergenic synthetic colors are currently prohibited. Upon reviewing the advantages and disadvantages of microbial food colorants in comparison with synthetic food colorants, it is clear that microbial alternatives provide environmentally sustainable and potentially beneficial health options (Table 1) [40].
However, there have been numerous hindrances to their large-scale application in industries. The production cost is high and the yield is low compared to synthetic pigments. Large-scale industrial production of pigments based on microorganisms to meet industrial requirements remains a challenge, necessitating developments in biotechnology, genetic engineering, and new fermentation techniques to enhance viability and cost-effectiveness [41]. Food laws are also a hindrance to the use of new sources of natural pigments, as they require costly toxicology tests and production processes [42].

3. Classification and Some Important Food-Grade Pigments

Pigments are classified into synthetic and natural colorants. Natural pigments are classified based on their source, chemical structure, solubility, and color systems. Natural pigments are widely classified into three groups based on their sources: plant pigments, animal pigments, and microbial pigments [43]. In the current research, we specifically emphasize the pigments of microorganisms. Among the major microbial pigments used as food colorants, astaxanthin, riboflavin, phycocyanin, lycopene, prodigiosin, β-carotene, violacein, and melanin are some of the major ones (Figure 2). Table 2 provides a comprehensive list of pigments, including their sources and characteristics.

3.1. Lycopene

Lycopene is an all-trans molecule with 2 non-conjugated and 11 conjugated double bonds for its red color and bio-protective activity induced by plants and microorganisms [44]. Its biosynthesis in microorganisms occurs primarily through the MEP or MVA pathway, leading to the formation of GGPP, a primary precursor to carotenoids (Figure 3) [45,46]. Blakeslea trispora, a filamentous fungus, is a commercial producer of lycopene [47]. Lycopene production in microbes can be greatly enhanced by optimizing culture conditions as well as through metabolic engineering strategies [48]. Lycopene production in B. trispora can be enhanced by the addition of inhibitors like pyridine or imidazole that inhibit lycopene cyclase activity [49]. Escherichia coli was genetically engineered to produce lycopene effectively by overexpression of the carotenoid biosynthetic gene cluster. Strain LYC010, for instance, produced 3.52 g/L (50.6 mg/g DCW) of lycopene in fed-batch fermentation [50]. Microbial lycopene has gained attention for food, pharmaceutical, and cosmetic applications because of its natural origin, stability, and health-protective activity. It exhibits antimicrobial, antioxidant, anti-inflammatory, and anticancer activity. It was shown to have preventive activity against a wide array of diseases, including diabetes and cardiovascular diseases [51].

3.2. β-Carotene

This reddish-orange pigment is derived primarily from Flavibacterium multivorum, Dunaliella salina, Phycomyces, Phaffia rhodozyma, Rhodosporidium, Rhodotorula mucilagenosa, Sporobolomyces, Blakeslea trispora, and Sporidiobolus [52]. Microbial biosynthesis of β-carotene occurs through the isoprenoid pathway via the methylerythritol phosphate (MEP) or the mevalonate (MVA) pathway to give the essential precursor geranylgeranyl pyrophosphate (GGPP) [53]. They both merge to give isopentenyl diphosphate (IPP) and the latter’s isomer, dimethylallyl diphosphate (DMAPP), the universal precursors to all the isoprenoids, including β-carotene (Figure 3) [46]. β-carotene, a major provitamin A carotenoid, has received tremendous attention due to its antioxidant activity, coloring property, and widespread applications in food, pharmaceutical, and nutraceutical industries [54]. It is blessed with many health benefits such as immunity boosting, preventing cardiovascular disease and cancer, and causing anti-aging effects [55].

3.3. Canthaxanthin

Microbial biosynthesis of canthaxanthin includes the transformation of β-carotene to canthaxanthin through ketolation reactions catalyzed by β-carotene ketolase enzymes. The enzymes at the 4 and 4′ positions of the β-ionone rings of β-carotene introduce keto groups to give the red-orange color of canthaxanthin (Figure 3) [56,57]. Various microorganisms, including Saccharomyces cerevisiae, Mucor circinelloides, and Escherichia coli, can naturally produce canthaxanthin [56,57,58]. The chemistry of canthaxanthin is such that its antioxidant activity is due to its singlet oxygen quenching ability and free radical scavenging activity [59]. Its antioxidant activity is due to its conjugated double-bond chemistry, which allows it to effectively inactivate reactive oxygen species [60]. Amazingly, canthaxanthin has been shown to protect LDL cholesterol from oxidation and induce antioxidant enzymes like catalase and superoxide dismutase [59].

3.4. Astaxanthin

Astaxanthin is a lipophilic diketo carotenoid pigment [61]. It is a red-orange xanthophyll carotenoid pigment, primarily synthesized by microalgae and red unicellular fungi. Microorganisms commercially used to manufacture astaxanthin include Haematococcus pluvialis, a green microalga, and the heterobasidiomycetous unicellular fungus Xanthophyllomyces dendrorhous (Phaffia rhodozyma teleomorph) [62,63]. It is a highly effective antioxidant carotenoid and has attracted significant attention due to its health potential. Its lipophilic character and poor water solubility limit its bioavailability and application in industries [64,65]. Its bioavailability has been improved by employing various strategies in recent research, i.e., glycosylation. Glycosylation was found to enhance the water solubility, bioavailability, photostability, and biological activities of astaxanthin to a significant level [66]. Heterologous glycosylated astaxanthin production was first reported in Yarrowia lipolytica with a yield of 1.47 mg/L. The process can be improved to increase water solubility, photostability, and biological activity [66].

3.5. Phycocyanin

Phycocyanin is a natural blue pigment mainly isolated from microalgae, particularly cyanobacteria such as Spirulina platensis and Aphanizomenon flosaquae [67]. Phycocyanin is an important accessory light-harvesting pigment extending the light absorption spectrum of photosynthesis. It consists of α and β protein subunits that are covalently linked to phycocyanobilin chromophores, which are responsible for its blue color and fluorescence [68]. It has attracted a lot of attention in the food sector because of its positive impact on health, including anti-inflammatory, antitumor, and antioxidant properties [69]. It is used in dietary supplements, foods, and drinks that are high in protein.

3.6. Prodigiosin

Prodigiosin is a red tripyrrole pigment that is biosynthesized mainly by Serratia marcescens and other bacterial species like Vibrio psychoeerytrus, Streptomyces sp., Pseudomonas sp., Rugamonas rubra, Pseudoalteromonas rubra, Actinomycetes sp., and Streptoverticillium rubrireticuli [70,71,72]. Microbial biosynthesis of prodigiosin is a bipartite process involving independent biosynthesis of two major precursors, 2-methyl-3-n-amyl-pyrrole (MAP) and 4-methoxy-2,2′-bipyrrole-5-carbaldehyde (MBC), which are enzymatically coupled to form the final pigment [73]. It is characterized by its unique pyrrolylpyrromethane skeleton and has a molecular weight of 323–324 Da [74]. It exhibits antineoplastic, antibacterial, antibiotic, antimalarial, anti-inflammatory, anticancer, and antitumor properties [75,76,77].

3.7. Riboflavin

The yellow water-soluble bacterial pigment or vitamin B2 is produced by the ascomycete Ashbya gossypii; Candida famata, a filamentous fungus; and the bacterium Bacillus subtilis [78]. It plays a crucial role as a precursor for the essential cofactors flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN), both of which are essential in numerous redox reactions in all of the kingdoms of life [79]. The isoalloxazine ring structure of flavins is responsible for their characteristic yellow fluorescence and redox properties [80]. They are used in many products, including fruit drinks, energy drinks, baby foods, dairy products, cereals, sauces, and vitamin-fortified milk [81].

3.8. Violacein

Violacein is a purple pigment that is biosynthesized by a number of bacteria through the condensation of two tryptophan units [82,83]. The pigment, which was initially isolated from Chromobacterium violaceum, has been a focus of extensive research interest owing to its variability in biological activities and potential applications in the future [84,85]. This indole-derived pigment possesses a typical UV-visible spectrum with maximum light absorbance at 575 nm, which remains unchanged in a range of bacterial sources [86]. Different bacteria of different genera, including Pseudoalteromonas, Iodobacter, Duganella, Janthinobacterium, Massilia, and Collimonas, are able to produce violacein [87].

3.9. Melanin

Melanins are heterogeneous pigments found in all types of life which are synthesized via different biochemical pathways. Two of the most common pathways for melanin synthesis are the DOPA pathway, which is initiated through conversion of L-tyrosine, and the DHN pathway, which is initiated through malonyl-coenzyme A (Figure 4) [88,89]. Melanin is a common and heterogeneous pigment found in all biological kingdoms, i.e., animals, plants, and microbes [90,91]. Melanin is a high-molecular weight pigment and usually dark brown to black in color, but reddish-yellow in color under some conditions [92]. Melanin is classified into five classes on the basis of the chemical structure of the precursor: neuromelanin (not typically produced by microorganisms), eumelanin, allomelanin, pheomelanin, and pyomelanin [93]. Melanin is a multifunctional molecule in a diverse range of microbial species, like Burkholderia cenocepacia, Rubrivivax benzoatilyticus JA2, Streptomyces cyaneus, Aspergillus fumigatus, Cryptococcus neoformans, Histoplasma capsulatum, and Paracoccidioides brasiliensis [94,95,96,97,98]. Melanin possesses certain physicochemical characteristics, is biocompatible and biostable, and is a candidate compound for various applications [99]. Melanin and melanin nanoparticles (MNPs) have been reported to have desirable applications in cancer theranostics due to their unique properties. MNPs have high photothermal conversion efficiency as photothermal agents under near-infrared (NIR) light, which can destroy tumors under photothermal therapy (PTT) efficiently [100]. The photothermal ability of MNPs can also trigger immunogenic cell death and tumor-associated antigen release, which may activate antitumor immune responses [101].
Table 2. Microbial pigments, sources, characteristics, and applications.
Table 2. Microbial pigments, sources, characteristics, and applications.
PigmentSource Organism(s)Major Industrial Producer (Biotechnological Source)Mol. Formula
and Solubility
ApplicationStability Profile (pH/Temp. Light)Refs.
Astaxanthin
(orange-pink-red)
Serratia marcescens,
Halobacterium salinarium,
Agrobacterium aurantiacum,
Paracoccus carotinifaciens,
Pseudoalteromonas rubra
Haematococcus pluvialis [GRAS GRN 294/580], Xanthophyllomyces dendrorhous [No GRAS], and engineered E. coli (introducing genes from H. pluvialis, like β-carotenoid hydroxylase and ketolase) [no GRAS]C40H52O4
596.8 g/mol
lipid soluble
Animal and fish food,
food colorants, anti-aging, and memory improvement
pH 5–8,
stable up to 70 °C,
moderately light-sensitive
[102,103,104,105,106,107,108,109,110]
Phycocyanin
(blue)
Pseudomonas aeruginosa,
Aphanizomenon flos-aquae,
Spirulina sp.
Spirulina platensis [GRAS GRN 424]C33H38N4O6
water-soluble
Ice creams and sweetspH 6.5–7.5,
unstable > 45 °C,
very light-sensitive
[111,112,113,114,115,116,117]
Prodigiosin
(red)
Pseudoalteromonas rubra,
Serratia marcescens
Recombinant E. coli (Sau3A fragments of S. marcescens DNA were introduced into E. coli K-12), Pseudomonas putida KT2440 C20H25N3O
323.4 g/mol
water-insoluble
Colorants in yogurt, milk, and carbonated beveragespH 4–6, degrades > 60 °C, light-sensitive[70,118,119,120,121,122]
Riboflavin
(yellow)
Debaryomyces subglobosus,
Ashbya gossypii,
Clostridium acetobutylicum
Ashbya gossypii, Bacillus subtilis [GRAS under 21 CFR 184.1695] C17H20N4O6
376.4 g/mol
water-soluble
Food industries and dietary supplementspH 5–7,
stable up to 100 °C,
extremely light-sensitive (photo-labile)
[78,123,124,125,126,127]
β-carotene
(yellow)
Rhodotorula gracilis,
Rhodotorula rubra,
Blakeslea trispora
Yarrowia lipolytica [GRAS GRN632 and BSL1 status]C40H56
536.9 g/mol
water-insoluble
Vitamin A sources, food industries, and boosts immunitypH 4–8,
degrades > 60 °C,
highly light-sensitive
[128,129,130,131,132]
Violacein
(purple)
Chromobacterium violaceum,
Janthinobacterium lividum,
Pseudoalteromonas spp.,
Pseudoalteromonas tunicata
E. coli strain (B8/pTRPH1-pVio-VioE), Citrobacter freundii, Corynebacterium glutamicum, and Yarrowia lipolyticaC20H13N3O3
343.3 g/mol
Cosmetics, textiles, medicine, and food industriespH 5–7,
stable < 60 °C,
light-sensitive
[84,124,133,134]
Melanin
(black)
Burkholderia cenocepacia, Rubrivivax benzoatilyticus JA2Gliocephalotrichum simplex, MEL1 mutant of Aspergillus nidulans, Streptomyces kathirae strain SC-1 C18H10N2O4
318.3 g/mol
Cosmetic creams, food industries, and anti-HIV activitypH 2–10,
highly heat-stable, light-stable
[135,136,137,138,139]
Lycopene
(red)
Fusarium sp.,
Blakeslea trispora
Blakeslea trispora [no GRAS but approved in EU (2006/721/EC)] C40H56
536.9 g/mol
Meat colorantspH 4–7,
degrades > 60 °C,
highly light-sensitive
[140,141,142]
Canthaxanthin
(orange-pink)
Halobacterium sp.,
Bradyrhizobium sp.,
Fusarium Sporotrichioides
Haematococcus pluvialis [GRAS GRN 294], genetically engineered Mucor circinelloides, and Saccharomyces cerevisiae [no GRAS] C40H52O2
564.8 g/mol
Food colorant, salmon food and poultry feedpH 4–9,
stable up to 80 °C,
relatively light-stable
[143,144,145]
GRAS: U.S. FDA Generally Recognized as Safe. GRN: GRAS Notification Number. “No GRAS” = no official FDA recognition, but the organism or product may be accepted elsewhere (e.g., EU Novel Food Catalogue).] [Note: Some listed pigment-producing microorganisms, e.g., Serratia marcescens, Burkholderia cenocepacia, Pseudomonas aeruginosa, Citrobacter freundii, and Chromobacterium violaceum, are Risk Group 2 (RG2) organisms and opportunistic pathogens [146,147]. Their use in food, pharmaceutical, or cosmetic industries should be preceded by strict biosafety screening and regulatory approval. Biosafety Level 1 and GRAS-listed strains should be used in large-scale applications.

4. Economic Evaluation of Microbial Pigments

Microbial pigment production measurement through bioprocesses is intricate due to low-technology goods, insufficient statistical information at a regional level, and the scattered nature of production among many small enterprises globally. There is greater interest in the use of microbial pigments, however. Natural pigments are sometimes more costly than synthetic pigments [148]. Typically, synthetic colors have a lower price range of USD 700–800 for each 100 g. In the food sectors, the benefits of microbial pigments are longer-term considerations, other than just direct cost considerations [149]. The production cost of bacterial β-carotene is around USD 1000 per kilogram and that of chemically synthesized β-carotene is around USD 500 per kilogram [150]. The current market prices place microbial (natural) β-carotene between USD 350 and USD 7500 per kilogram and synthetic β-carotene between USD 200 and USD 2000 per kilogram depending upon purity, formulation, and production scale [151]. The β-carotene market worldwide was worth approximately USD 1.03 billion in 2023, and the natural β-carotene market alone was estimated to be worth around USD 1.28 billion in 2024 [152]. The synthetic β-carotene market was worth around USD 256 million in 2024 [153]. Advanced fermentation techniques and genetically modified strains can be used to reduce the production cost of substrates. Some microbes can utilize agro-industrial waste as a substrate, considerably reducing costs for raw materials as well as promoting sustainability by reducing waste.
Cortes et al. (2017) forecasted that the market for natural food pigments would expand at a compound annual growth rate (CAGR) of 6.22% during 2015–2019 [154]. Gürses et al. (2016) projected that the overall size and amount of consumption of pigments and dyes in the global market would be 9.9 million tons and amount to USD 26.53 billion in 2017, boosted by expansion in key end-use markets [43]. The β-carotene market was expected to grow from USD 247 million by the end of 2007 to USD 285 million by the end of 2015, with a CAGR of 1.8%. The moderate growth rate indicates a steady demand for β-carotene across industries [154]. The astaxanthin market’s projected growth rate of 16.2% CAGR and the anticipated value of USD 3.4 billion by 2027 align with the rapid expansion observed in several related industries [155]. The industrial production of astaxanthin is carried out utilizing Paracoccus sp. It was anticipated that, by 2021, the volume of astaxanthin sales would amount to USD 1.1 billion. With a CAGR of 16.2%, the astaxanthin market is predicted to attain a value of USD 3.4 billion by 2027 [156]. The color market is not only growing but evolving. The increasing demand for natural colorants, particularly those derived from microorganisms, is reshaping the industry.

5. Industrial Considerations in the Use of Bacterial and Fungal Pigment Producers

Bacteria and fungi are widely exploited industrially for pigment production primarily due to their faster growth rates, resilience to different substrates, and high yields of micronutrients and pigments [16]. Interestingly, while the two have industrial advantages in terms of providing numerous sources of pigments, they also present certain challenges that require special considerations.
Fungi offer bioavailability, cost, and scalability advantages in terms of cell culture and downstream processing. They can produce a variety of pigments, such as carotenoids, melanin, polyketides, and azaphilones [157]. Pigment biosynthesis in microorganisms can be increased under various stress conditions. Monascus ruber CGMCC 10910 led to a significant increase in yellow pigment production after being subjected to high-glucose stress conditions. The intracellular and extracellular pigment yield increased by 26.31% and 94.86%, respectively [158]. Also, high salt stress resulted in a 40% increment in pigment yield [159]. Rhodotorula mucilaginosa AJB01 showed an increase in carotenoid production under osmotic stress and UV light exposure [160].
Additional pigment and secondary metabolite yields could be obtained using genera such as Talaromyces, Monascus, and Fusarium. They are eco-sustainable and can be grown on low-budget agro-industrial wastes (solid-state fermentation), and thus represent a low-cost form of production. Certain fungi strains present biosafety concerns, e.g., the co-production of mycotoxins by Monascus purpureus citrinin [161]. This can be addressed by screening of the strains, mutagenesis, or genome editing. Filamentous growth also imposes mixing and oxygen transfer issues in bioreactors, which necessitate special reactor designs or agitation regimes [162].
Bacterial pigments are generally used because of their easy growth, rapid production, and simplicity of pigment extraction. In particular, Serratia marcescens and Streptomyces spp. are more or less flexible than fungi that grow rapidly and have low nutrient requirements, which makes them ideal for continuous fermentation systems [36]. These processes facilitate the production of pigments due to their simplified metabolic pathways, relatively short life cycles, and tractability to genetic manipulation. However, they are relatively vulnerable to contamination and may have stringent aseptic requirements, particularly in open systems. High pigment yields, in addition to considerations such as high biosafety (GRAS status) requirements, strain genetic stability, growth patterns, and the aerobic fermentation systems exploited, are knowledge perspectives that are relatively helpful for industrial management based on the criteria of strain selection [16,41].
Contamination management must be of concern at scale and across industries. Solid-state fermentation (SSF) systems, which use fungi, are less likely to be contaminated since they work under low-water activity conditions, in contrast to submerged fermentation (SmF) systems, primarily for bacteria, which need rigorous sterilization and closed systems. Automated monitoring and online analytics minimize variability and batch inconsistency [163].

6. Advances in Large-Scale Production and Genetic Engineering of Microbial Pigments

6.1. Industrial Production of Microbial Pigments

Before producing large quantities of pigments, it is critical to characterize the pigment producer, pigment, and pigment properties. Screening methods have also been developed for this purpose. One of the fastest and simplest methods for detecting pigments is the use of a portable handheld Raman spectrometer [27]. Moreover, it is important to have prior knowledge of the metabolic pathways of pigment producers to identify and eliminate toxic and pathogenic pigment producers. For example, this information has been useful in the manufacture of food products such as QuornTM, as it can also identify cytotoxic compounds (e.g., 4,15-diacetoxyscirpenol) produced by Fusarium venenatum [164].
Monascus purpureus produces red pigments for food coloring, but also produces citrinin, a nephrotoxic mycotoxin that can harm humans and animals. Furthermore, researchers have used several genetic engineering techniques in an attempt to reduce or eliminate citrinin production in order not to impact pigment generation (essentially increasing the amount of pigment) [165]. The CRISPR/Cas9 system was developed to delete large genomic regions such as biosynthetic gene clusters of polykaryotic fungi such as M. purpureus. The researchers successfully deleted the citrinin biosynthetic gene cluster (15 kb) in a commercial strain and characterized stable homokaryotic mutants that no longer produce citrinin. Moreover, their genetic engineering increased the production of pigment by 2–5% [166].
Robust analytical techniques are critical in the industrial production of microbial pigments to achieve product quality, safety, and regulatory acceptance. High-Performance Liquid Chromatography (HPLC) in combination with Mass Spectrometry (MS), and techniques like UV-Visible Spectroscopy, LC-MS, FTIR, and NMR enable precise structural identification and quality control of microbial pigments. Utilizing such analytical methods, industries can maintain routine pigment quality, provide batch-to-batch analysis, and maintain high levels of purity for mass application and regulatory approvals. Such comprehensive validation is essential for full utilization of microbial pigments in food, pharmaceutical, and cosmetic industries [167].
The pigment manufacturing industry must use a large-scale production method that aligns with emerging fermentation techniques. For instance, SSF and SmF use fermentation principles to produce pigments, but they differ depending on the situation or the preset variables. Each of these two techniques uses different variables to determine which substrates are available, the microorganism type, and desired pigment attributes. SSF offers advantages, such as lower energy requirements and reduced wastewater production, making it an environmentally friendly option [168].
SmF can use more energy with energy systems; however, it is often preferred for many large-scale industrial applications owing to the control over the processes and technologies available for later scale-up and automation of larger production [169]. Additionally, SmF allows for the growth of microorganisms in nutrient-rich liquid media at an optimal pH, and aeration facilitates higher yields. SmF can easily be scaled; however, it incurs higher capital and operational costs. Alternatively, SSF can use solid substrates based on agro-industrial byproducts, making them eco-friendly and more cost-effective by leveraging low-cost materials [170].
The Response Surface Methodology (RSM) has been found to be an effective tool for optimizing the conditions of media to maximize pigment yield in a variety of fermentation processes [171]. Several studies have demonstrated that RSM can be applied to determine optimal operating conditions to enhance production. For instance, in the production of red and orange pigments, RSM was applied to optimize the conditions for submerged fermentation of Talaromyces albobiverticillius 30548. Optimum pigment and biomass yields were established at a pH of 6.4, with an agitation rate of 164 rpm, a temperature of 24 °C, and a fermentation period of 149 h [172].
Wild-type strains generally require more time to produce pigments and often do not yield sufficient quantities to make the process commercially viable, highlighting the importance of strain development [173]. To improve a strain, it is treated with common mutagens such as ethyl methyl sulfonate (EMS), 1-methyl-3-nitro-1-nitrosoguanidine (NTG), and ultraviolet (UV) to cause an increase in pigment production. After being treated with EMS, UV, NTG, and microwave radiation, there was an enhancement in pigment production by Serratia marcescens and Haematococcus pluvialis [174].
To achieve industrial feasibility, a bioreactor needs to be designed with respect to the microbial strain, type of pigment produced, and yield. Bubble column reactors, stirred-tank bioreactors (STRs), and airlift bioreactors are usually used based on the type of pigment and microbial strain. STRs are the most popular, as oxygen transfer is efficient, while airlift bioreactors are appropriate for shear-sensitive microorganisms that require enhanced oxygen transfer. Bubble column reactors have high mass transfer rates and are frequently used in aerobic microbial fermentation [175]. Bioreactors used at an industrial level can range in volume from 10 L to 100,000 L, and working parameters, such as pH, agitation, temperature, feed of nutrients, and dissolved oxygen, must be monitored continuously [176].
Pigment synthesis is significantly impacted by nutritional enrichment or shortage. Pigment synthesis may be impeded by organic acids generated during Monascus ruber culture under oxygenated conditions [177]. Introducing rice and wheat feed (either mixed grains or broken leftovers) has demonstrated the enrichment of carotenoid production in fungi and unicellular fungi [178]. It was demonstrated that Acinetobacter wofii produced more pink pigment when given methanol as its only carbon source [179]. Carotenoids are produced by Mycobacterium tuberculosis in response to acidic stress (pH 5.0–6.0) and prolonged anaerobic growth [180]. When succinate was supplied as the only carbon source, Pseudomonas fluorescens produced a significant amount of water-soluble, yellow-green, fluorescent pigment; however, when citric or malic acids were employed as substrates, no pigment was produced [112]. Several substrates have been found to be strong stimulators for different pigment molecules, including phenylalanine, tryptophan, and, most importantly, tyrosine. However, further research is needed to determine whether other substrates are also effective in improving pigmentation [181].
Pigment extraction is essential for producing high-quality products after fermentation. Traditionally, extraction is carried out with a solvent and has historically been laborious and complicated. Except for steam distillation, other methodologies use ethanol to extract pigments, and when traditional solvents are used, this process is not economical. However, in modern extraction techniques, non-toxic solvents, including supercritical CO2 and ethyl acetate, have been increasingly utilized in alignment with green chemistry principles [14,182]. Some techniques include membrane filtration, a non-solvent process; ultrasound-assisted extraction, which enhances the extraction of the pigment and reduces the amount of solvent; and supercritical fluid extraction techniques, which utilize CO2 as a solvent in the extraction process [183].
Safety and stability are also paramount in the scaling of the production of commercial applications in food, cosmetics, and pharmaceuticals [184]. Stabilizing processes for pigments include encapsulating treatments, for example, biopolymer coatings and nano-emulsions, which are utilized to improve the stability of pigments throughout transportation, handling, and storage [185]. On the other hand, regulatory approval is also required for industrial-scale production. Organizations such as the European Food Safety Authority (EFSA) and the Food and Drug Administration (FDA) require rigorous quality control procedures, including toxicity tests, stability tests, and compliance with Generally Recognized as Safe (GRAS) regulations [186]. These tests ensure that microbial pigments are safe for consumption.

6.2. Genetically Engineered Microorganisms for Pigment Production

Recent advances in genetic engineering have enabled the use of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology to enhance pigment production in microbes. The approach facilitates easy microorganism engineering to produce natural colors by expressing color-producing genes in their genomes using the CRISPR-Cas9 system [187]. For instance, in Escherichia coli recombinant cultures, the MEP pathway has been enhanced through co-expression of enzymes such as isopentenyl pyrophosphate isomerase (FNI) and 1-deoxy-D-xylulose-5-phosphate synthase (DXS) and has resulted in significantly elevated β-carotene production [46]. At the same time, overexpression of dxs encoding the very first committed step in the MEP pathway enabled production of lycopene in Corynebacterium glutamicum [188].
Microorganism-based synthesis of anthocyanins has proven to be an effective alternative to conventional plant extraction techniques. This method has a number of advantages, including reduced production time, purer yield, reduced amount of waste, and greater batch-to-batch consistency in the yields. Different microbial systems have been engineered to produce anthocyanins, both prokaryotic and eukaryotic organisms [189]. For instance, Escherichia coli has been genetically engineered to produce cyanidin-3-O-glucoside and pelargonidin-3-O-glucoside efficiently [190]. Anthocyanins have also been produced with other microorganisms such as Saccharomyces cerevisiae and Corynebacterium glutamicum [191]. Although issues are still present, including enhancing enzyme efficiency and stabilizing the products, the process of using microorganisms to make anthocyanins is evolving, showing potential for large-scale production of these pigments of interest [192]. Some genetically engineered microorganisms and processes for pigment manufacture are listed in Table 3.
One notable success was the engineering of Yarrowia lipolytica using CRISPR-Cas9 to enhance β-carotene production by optimizing the MVA pathway. In one study, researchers achieved a remarkable 24-fold (408 mg/L) increase in β-carotene production. This was accomplished by overexpressing several genes related to the MVA pathway, including GGS1, ERG13, and HMG, as well as multiple copies of carB and carp [193]. Future experiments utilizing CRISPR-Cas9 and genetic engineering should focus on achieving cost-effective and high-yield pigment production at a commercial scale.
Though CRISPR-Cas9 technology is applied in genetically modified microorganisms (GEMs) within the food sector, it is encountering severe challenges regarding regulation, biosafety, and acceptability for consumers. Most countries have regulatory systems that categorize CRISPR-edited strains applied for food purposes in the form of genetically modified organism (GMO) regulations. This categorization calls for elaborate risk assessments, traceability, and labeling procedures, which may prove to be cumbersome and contentious [194]. Moreover, consumer attitudes have been conservative due to poor awareness and ethical objections to gene editing technologies.
Successful commercialization of genetically modified microbes (GEMs) in the pigment industry relies on the convergence of socio-technical factors that take into account biosafety, regulatory matters, and public engagement. These are critical to the usability and acceptability of GEMs in the pigment industry.
There has to be an appropriate regulatory system. It should include safety, environmental, and ethical evaluations to secure public confidence and government approval [195]. This will guarantee that GEMs comply with existing environmental and health and safety policies. Open and science-based policies help reduce GEM risks and give the public and stakeholders confidence in the safety of these technologies.
Public acceptance is also an important socio-technical factor. Activists and consumers are stakeholders who must be addressed and persuaded in a way that will promote their acceptance of GEMs. Public campaigns, education, and debate on the benefits and safety of GEMs will minimize fear and resistance [196]. Effective communication campaigns that address public ethical and safety concerns can create a good image and build a more positive climate for commercialization.
Finally, considering socio-economic impacts, such as job creation and enabling a circular bioeconomy, can be beneficial for the commercialization of GEMs. Focusing on the overall economic impact, these innovations can be supported even further by policymakers and investors [197].
Table 3. Some genetically modified microorganisms.
Table 3. Some genetically modified microorganisms.
Pigment Microorganism Genetic Engineering Strategy Yield Before ModificationReported Yield
After Modification
Refs.
Astaxanthin E. coliOptimization of the astaxanthin biosynthetic pathway with multivariate modular methodsNot reported Not reported [198]
β-caroteneE. coliOverexpression of dxs (1-deoxy-D-xylulose-5-phosphate synthase) and idi (isopentenyl diphosphate isomerase), CrtE (GGPP synthase), CrtB (phytoene synthase), and CrtI (phytoene desaturase)~20 mg/L~450 mg/L[199]
ViolaceinE. coliViolacein biosynthesis genes’ heterologous expression Not reported Not reported[200]
AstaxanthinY. lipolyticaCombination of HpCrtZ and HpCrtW from H. pluvialis Not reported 3.3 g/L or 41.3 mg/g DCW under fed-batch fer mentation conditions[201]
β-caroteneY. lipolyticaOptimized promoter gene pairs based on Golden Gate DNA assembly17 mg/L408 mg/L[131,193]
ViolaceinY. lipolyticaGolden Gate Assembly method Not reported70.04 mg/L[202]
Astaxanthin S. cerevisiaeRecombination of CrtW and CrtZ genes in vitro and in vivoNot reported 4.7 mg/g DCW[203]
β-caroteneS. cerevisiaeIntegration of CrtI, CrtE, and CrtYB (bifunctional phytoene synthase and lycopene cyclase) and additional copies of tHMG1 from X. dendrorhous using CRISPR-Cas9Not reported 5.9 mg/g dry weight[204]
LycopeneS. cerevisiaeAssembly of IDI, CrtE, and CrtBNot reported 41.8 mg/g DCW[205,206]
β-caroteneS. cerevisiaeLipase expression and introduction of the β-carotene synthetic pathway from X. dendrorhous Not reported772.8 mg/L [207,208]
Dry cell weight (DCW). Note: Yields are reported either as volumetric titer (mg/L or g/L) or specific productivity (mg/g DCW), depending on the source. These units reflect different aspects of production performance and are not directly comparable.

6.3. Issues and Implications of Different Pigment-Producing Microorganisms

Selection of microbial hosts for pigment manufacture must be performed carefully in light of their divergent biosafety profiles, metabolic characteristics, and downstream processing considerations. Microbial systems offer scalable, environmentally benign substitutes to synthetic pigments, but their use in industry must be compatible with safety, reproducibility, and acceptability to regulators. These considerations must be given special emphasis when industries request natural pigment replacements, where microbial producers must be selected and regulated with careful regard to their intrinsic risks, limitations, and practicability.
Bacterial pigment producers such as Serratia marcescens, Pseudomonas aeruginosa, and Chromobacterium violaceum are severe biosafety concerns. Serratia marcescens, a widely reported prodigiosin producer, is an opportunistic pathogen with nosocomial infection and antibiotic resistance linkages [209]. Its quorum-sensing system regulates virulence factors and biofilm formation, which is a severe biosafety risk factor [210]. Although photodynamic inactivation (PDI) and gene manipulations such as ptrA gene disruption have been fruitful in curtailing virulence at the expense of pigment production, these approaches emphasize the imperative need for stringent safety evaluations [211,212]. S. marcescens is economical to culture on inexpensive substrates such as oilseed cakes and waste from industry, providing a window of opportunity for sustainable pigment production if biosafety concerns are addressed. Pseudomonas aeruginosa is a Biosafety Level 2 (BSL-2) pathogen, primarily due to its production of pyocyanin (a blue-green pigment), a virulence factor that plays a widespread role in its contribution to pathogenicity [213]. Pyocyanin is a redox-active phenazine compound that not only contributes to the bacterium’s virulence but also the danger it poses to host tissues, particularly in immunocompromised hosts or cystic fibrosis patients [214,215]. Chromobacterium violaceum, the natural violacein producer, is also a safety concern due to its established virulence in humans, primarily through its type III secretion systems (T3SSs), implicated in cytotoxicity and tissue destruction. These risks restrict its industrial applications to non-ingestible or topical products under stringent regulatory control [216]. Janthinobacterium lividum, which produces violacein and deoxyviolacein, also possesses pathogenicity, and hence safety screening is necessary [217]. Bacterial pigment synthesis is generally strain-specific, the differences in yield, stability, and regulation of biosynthesis complicating standardization. Overcoming these challenges depends on developments in strain enhancement and metabolic engineering. Through manipulation of gene pathways and fermentation conditions, improved yields and more stable pigment biosynthesis are possible [184]. Utilization of biotechnological innovations, e.g., heterologous hosts or purified enzymes, can improve the reliability and efficiency of pigment biosynthesis. These methods not only enhance yields but also offer improved control of pigment character, i.e., color and chemical composition [184]. The fast growth of bacteria also poses the risk of contamination, which requires strict aseptic control. Use of closed systems and advanced filtration processes can ensure sterility at the time of production, especially in continuous or large-scale fermentative systems [218]. These issues are major hurdles to regulatory approval, scalability, and commercial feasibility.
In contrast, fungal pigment producers are known for producing a wide range of natural pigments, but their industrial use presents several safety, scalability, and regulatory concerns. Most critical among these are the high co-production rates of mycotoxins with pigments. For example, Monascus purpureus yields citrinin, a nephrotoxic metabolite, restricting its application for food and pharmaceutical applications [219]. Even after attempts at removal of citrinin by means of strain improvement, mutagenesis, or gene editing, residual mycotoxins need ongoing safety assessment, despite genetic or screening remediation [166,220]. Similarly, Penicillium oxalicum and Talaromyces atroroseus are bioactive pigment-producing species but phylogenetically closely related to mycotoxin-producing strains like Penicillium citrinum and Aspergillus flavus and need extensive screening [221]. Fungi like Fusarium fujikuroi that can also produce bikaverin and fusarubin belong to genera which have phytopathogenic or toxic strains and hence are industrially problematic [222,223].
Filamentous fungi present engineering challenges because of their mycelial morphology, resulting in high broth viscosity, inadequate mixing, and oxygen transfer problems [224,225]. These factors hinder scalability and necessitate specially designed bioreactors. Pigment yield and composition are extremely sensitive to cultural conditions of pH, temperature, and nutrient ratio, resulting in batch-to-batch variability and variable product quality [226]. Moreover, the intracellular site of the pigment necessitates mechanical or enzymatic cell breakage and solvent extraction, which results in increased downstream costs and also raises the problem of solvent residues and environmental toxicity.
Microalgae are also promising sources of high-value pigments like astaxanthin, β-carotene, lutein, and phycocyanin [227]. Their large-scale use is, however, restricted by some technical and economic limitations. Cultivation demands costly photobioreactors or well-regulated open ponds with strict control of light, CO2, temperature, and supply of nutrients to achieve maximum pigment yield. Environmental fluctuation, contamination with competing microorganisms, and culture crashes often undermine yield and operational efficiency [228]. Open ponds, although less expensive, are plagued by contamination, low yields, and high water losses [229].
The majority of microalgal pigments are intracellular, requiring energy-prone harvesting, drying, cell disruption, and solvent extraction, all contributing to processing expense and environmental footprint [230]. Though some species such as Haematococcus pluvialis and Dunaliella salina possess GRAS status, numerous other strains lack defined safety profiles and need extensive toxicological evaluation. Spirulina platensis, a phycocyanin source, has the propensity to accumulate heavy metals when grown in uncontrolled conditions, raising further safety issues [231]. Microalgae’s biomass and pigment composition are highly stress-sensitive to extrinsic conditions, which is a cause of batch-to-batch variation and quality control problems [12]. Furthermore, genetically engineered microalgal strains also face additional regulatory scrutiny and limited public acceptability in some markets [232]. All of these factors compromise the cost-effectiveness, scale-up, and regulatory acceptability of microalgal pigments in the context of commercial interest. Therefore, organism-specific issues must be addressed by targeted strain selection, process optimization, biosafety validation, and regulatory compliance to realize the full industrial potential of microbial pigment production.

7. Applications in the Food Industry

Most bacteria and fungi have been thoroughly studied for their ability to produce food colors. Table 4 provides a comprehensive collection of commercialized microbial pigments, together with information about their source organisms. Many microbial pigments are already used at the industrial level in the food industry.

7.1. Pigment-Producing Microorganisms and Applications in the Food Industry

Monascus purpureus has been shown to produce a diverse range of pigments, such as red, orange, and yellow compounds. The primary pigments are dimeric yellow (ankaflavin and monascin), orange (rubropunctatin and monascorubrin), and red (rubropunctamine and monascorubramine) molecules [234]. The pigments are commonly applied in food, especially in Asian countries, and are also referred to as “red yeast rice”. These pigments are also found in processed meats and sausage [235]. Talaromyces atroroseus is another non-toxigenic fungal species that can produce azaphilone biosynthetic families of colors, such as mitorubrins and Monascus pigments, while not producing mycotoxins, which are suitable for beverages and confectionery products and for coloring in other applications.
Microbial pigments also have a great impact on the dairy industry regarding the color and quality of countless products. One example is strawberry milk, which is colored by β-carotene, a natural pigment synthesized by Blakeslea trispora [236]. Brevibacterium linens produces carotenoid pigments that provide washed-rind cheeses with their characteristic orange-red color. Penicillium purpurogenum and Talaromyces spp. are sources of reddish and orange azaphilones, which are incorporated into a variety of baked goods [21].
Actinorhodin, a blue-pigmented secondary metabolite with antibiotic qualities, is produced by Streptomyces coelicolor and may represent a novel food coloring [237]. Carotenoids are biopigments that are synthesized by various microorganisms, such as bacteria like Micrococcus luteus. These pigments are extensively used in food as food colorants of natural origin, especially in meat [235].
Microbial pigments employed in the food and beverage industry are of particular interest as natural colorants because of their biodegradability and possible health benefits. Talaromyces sp. has been found to be capable of synthesizing water-soluble red pigments, which find applications in fruit juices [238]. Astaxanthin, used in health-oriented foods, nutraceuticals, sports nutrition drinks, and beverage products, is synthesized by Paracoccus carotinifaciens and Xanthophyllomyces dendrorhous.

7.2. Food Preservatives

Microbial pigments are also favored in the food industry as preservatives because of their antibacterial and antioxidant properties, as well as consumer demand, safety, and potential health benefits [174]. Prodigiosin, produced by Serratia marcescens, exhibits strong antioxidant and antimicrobial activities. It is a potential scavenger of DPPH and ABTS radicals at low levels (10 mg/L) and also a growth inhibitor of foodborne microorganisms, increasing food product shelf life [239].
Several pigments, especially anthocyanins, flavonoid water-soluble pigments, have also been reported to exhibit considerable antibacterial activity against a variety of pathogens. These natural constituents have been known to demonstrate potential in increasing shelf life and preservation of food against spoilage. Anthocyanins are pH-sensitive, and this allows them to sense chemical and microbial alterations in foods. They have high antioxidant activities and antimicrobial activities, thereby increasing food product shelf life [240].
Carotenoids have numerous activities, such as harvesting light energy, neutralizing oxidants, and functioning as virulence agents and efficient antioxidants capable of scavenging singlet oxygen and peroxyl radicals generated during photooxidation [241]. Because of these activities, they help to preserve food quality by preventing light-induced oxidative damage.

7.3. Pharmacological Activities

Microbial pigments show a wide range of pharmacological activities, including antioxidant, antibacterial, anti-inflammatory, antiviral, immunomodulatory, and anticancer activity; hence, they are regarded as future drug candidates [36].
Prodigiosin, isolated from Serratia marcescens JSSCPM1, has been reported to exhibit antibacterial activity against Gram-negative bacteria [242]. Key research has validated that prodigiosin causes bactericidal activity through induction of programmed cell death (PCD) in bacterial cells. It involves DNA fragmentation, production of reactive oxygen species (ROS), and induction of proteases with caspase-like substrate specificity. Prodigiosin is internalized by bacterial cells, and the majority of it is present in the membrane and nuclear fractions, thus ensuring that it comes into contact with bacterial DNA and allowing intracellular mobility. Furthermore, it greatly inhibits bacterial motility [243].
Monascus ruber pigments were also found to be active against Staphylococcus aureus and Escherichia coli (MIC: 10–20 mg/mL) [244]. Their mode of action operates mainly through membrane disruption. The orange pigment of Monascus ruber inhibits the cytoplasmic membranes of bacteria, enhancing the electric conductivity of bacterial suspensions and leading to cell death. The pigment can be incorporated into phospholipid bilayers, which also suggests that it can disrupt bacterial membranes if the pigment is bound to cellular membranes [245].
The antioxidant activity of pigments such as prodigiosin from Streptomyces sp. WMA-LM31 reached 62.5% against protein and lipid oxidative stress [246]. Carotenoids such as β-carotene, astaxanthin from Xanthophyllomyces dendrorhous, and torularhodin from Rhodotorula spp. neutralize ROS, prevent lipid peroxidation, and increase cellular antioxidant defense [247,248,249]. Melanin from Aspergillus niger and Cryptococcus neoformans possesses redox buffering activity and UV protection [250]. Melanin is also involved in photoprotection and immunomodulation by absorption of UV and degradation of ROS in UV-induced oxidative stress, and therefore a potential sun screen ingredient found in nature [251,252,253].
Violacein, derived from Chromobacterium violaceum, has received significant attention because of its broad range of biological activity, including antiviral activity. It activates the human Toll-like Receptor 8 (TLR8) signaling pathway and thus enhances innate immunity. The receptor-dependent process indicates that violacein can enhance the body’s own antiviral defense; however, the exact interactions of this process with particular viruses like HSV or DENV are not yet fully known [254].
Torularhodin and naphthoquinone derivatives have proven to be very promising as anti-inflammatory drugs through different mechanisms. One of the significant mechanisms through which torularhodin exerts its anti-inflammatory effect is by activating the Nrf2/NF-κB signaling pathways. Oxidative stress and neuroinflammation are key factors in a very large number of degenerative diseases, and torularhodin reverses the issues by facilitating the translocation of Nrf2, thereby upregulating antioxidant enzymes such as HO-1. Concurrently, it inhibits the NF-κB pathway, which is proven to mediate inflammatory cytokine expression [255].
Talaromyces sp. SK-S009 produces naphthoquinone analogs, which have been reported to inhibit cyclooxygenase and lipoxygenase pathways. These pathways are crucial for the biosynthesis of pro-inflammatory mediators, and therefore such analogs are promising for the treatment of inflammatory disorders [256].
Certain microbial pigments demonstrated anticancer activity through apoptosis induction, metastasis inhibition, and the targeting of crucial cellular pathways. Indole-type pigments were active against inhibiting CDC25 phosphatases, SIRT1, ferriprotoporphyrin IX, and DNA binding [253]. Prodigiosin and analogs were cytotoxic to 60 human cancer cell lines and blocked Wnt/β-catenin signaling through the inhibition of LRP6, DVL, and GSK3β [75,257]. Prodigiosin was also described as an in vivo tumor growth and metastasis inhibitor, an MMP-2 inhibitor, and a RhoA pathway modulator [258]. Rubropunctatin, a Monascus pigment, demonstrated anticancer activity against human cell lines HepG2, BGC-823, MKN45, SH-SY5Y, and AGS [259]. A summary or table of some microbial pigments and their bioactivity is illustrated in Table 5.

7.4. Alternative Applications

Various applications, apart from bioactivity, such as numerous microbes, display a range of pigment synthesis with varied applications. For example, Rhodotorula paludigena produces a carotenoid pigment that is used to enhance the skin pigmentation of ornamental fish [292].
Talaromyces ruber generates an extracellular fluorescent pigment that is used for wood coloring [293]. Vibrio sp., a marine bacterium, produces a red pigment that can facilitate wound healing and exhibits antibacterial properties [294]. Nigrospora aurantiaca produces a red pigment called bostrycin, which is well known for its remarkable staining resistance in cotton fabric [295]. Isaria spp. and Fusarium spp. produce purple and ruby-red pigments, respectively, which are specifically used in the leather dyeing process [296]. Yarrowia lipolytica W29 produces pyomelanin, a brown pigment that functions as an antioxidant and boosts the sun protection factor (SPF) in sunscreens [297]. Serratia rubidaea can produce prodigiosin, which is used to dye fabrics and is also antibacterial [298].
Various microbes, such as Candida lipolytica, Trichosporon cutaneum, Lipomyces starkeyi, Cryptococcus curvatus, and Yarrowia lipolytica, can be used for pigment production when they are grown on agro-industrial residues, especially lignocellulosic residues. Colors derived from these microorganisms possess outstanding photoprotective, antimicrobial, and antioxidant properties. These properties make them extremely suitable for skincare products, limiting the occurrence of wrinkles and acting as a defense against sunburn [299].
Furthermore, using agro-industrial waste, Actinomucor elegans and Umbelopsis isabellina synthesized carotenoids that are recognized for their antioxidant properties [300]. Talaromyces albobiverticillius utilizes pineapple peels as a substrate to generate a red pigment. This pigment has various functions, like dyeing cotton fabric, and it also has antibacterial and antioxidant properties [301].

8. Emerging Application: Microbial Pigment-Mediated Nanoparticle Synthesis

Different microbial pigments have been used to prepare nanoparticles with diverse chemical and physical characteristics. Bacterial pigment nanoparticles of different sizes ranged between 10 and 100 nm, but in a single case, Au particles were 2000 nm in size. Algal and fungal pigment-mediated metal nanoparticles have size ranges of 5–25 nm and 1–60 nm, respectively [302]. Fungal pigments have significant potential for producing smaller nanoparticles.
However, the process is not yet understood. El-Baz et al. have postulated a likely process for the increase in redox potential with an increase in concentration of red pigment of Monascus extract. Redox potential has been found to play a critical role in the stabilization and reduction of AgNPs [303]. Microbial pigments have several negatively charged functional groups like hydroxyl, ketone, and carboxyl groups, which allow Ag+ ions to adsorb [304]. These pigment-metal complexes initiate the formation of superoxide free radicals (O2) from dissolved oxygen during photolytic activity, leading to the reduction of Ag+ to Ag0 (Figure 5) [305].
Rubropunctatin, a water-insoluble pigment, was purified and utilized as a blocking and reducing agent [306]. The rubropunctatin AgNPs (R-AgNPs) synthesized were well dispersed in solution, spherical in morphology, and had an average size of 13.54 nm. The R-AgNPs are highly active against bacteria, and they have even reduced in vitro toxicity. This indicates that bioactive rubropunctatin functionalized on AgNPs enhances bioavailability and synergistically boosts their activity [307]. Table 6 shows the numerous microbes and pigments used in the production of nanoparticles, as well as their uses.

9. Challenges and Future Perspectives

Microorganisms can produce colors both externally and internally. Although the industrial synthesis of food pigments from microorganisms has several benefits, the species employed must be culturable and productive within a restricted region and time [13]. They should be non-pathogenic, non-toxic, capable of growth on a variety of low-cost raw materials, and stable in extreme physical and chemical conditions [112]. Despite the availability of many varieties, the industrial use of natural pigments from microbiological sources is challenging. Some microorganisms are easy to screen but difficult to scale-up commercially. Microbial pigment purification and isolation from fermentation broth is a time-consuming, low-yielding, and costly process [174]. Additionally, these pigments may interact with other food components, leading to undesirable odors and flavors [27]. Temperature, pH, light, UV, oxygen, and heat all have an impact on microbial color, which can cause fading and shorten shelf life. Also, environmental factors such as metal ions, proteins, and organic compounds might influence their natural color [27]. Ending the present lag in microbial pigment production is a priority in an effort to unleash their full potential as bioactive, environmentally friendly alternatives to synthetic dyes. Solving the issues of low yields, high expense, and low scalability through the advancements in metabolic engineering, strain improvement, and process design will assist in making production commercially viable.
In summary, microbial pigments are an extremely promising group of natural pigments with applications ranging from the food, pharmaceutical, and cosmetic industries to the environment. Microbial pigments are bioactive, biodegradable, and safe for manufacture on renewable substrates, and hence are suitable alternatives to synthetic dyes. But full industrial realization will involve a coordinated effort in strain performance optimization, yield improvement through metabolic and process engineering, and solving stability and regulatory problems. Future research will be required to promote life cycle analyses, multi-omics approaches to pathway elucidation, and scalable downstream processing technology. With ongoing inter-disciplinary innovation and policy support, microbial pigments have the potential to play a central part in the transition to cleaner and greener industrial applications.

Author Contributions

Conceptualization, U.K., J.P. and S.K.D.; Methodology, A.C.; Software, U.K.; Validation, M.V., M.F. and S.K.D.; Formal Analysis, A.C.; Investigation, A.C.; Writing—Original Draft Preparation, U.K.; Writing—Review and Editing, S.K.D. and L.D.; Supervision, L.D.; Project Administration, U.K.; Funding Acquisition, U.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We appreciate all the individuals and organizations that have contributed to this review paper. We would also like to express our sincere gratitude to the authors of the papers we have reviewed, whose insights and research have informed and shaped our understanding of the topic. We would also like to thank our colleagues and partners for their helpful feedback and assistance throughout the evaluation process.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. De Mejia, E.G.; Zhang, Q.; Penta, K.; Eroglu, A.; Lila, M.A. The Colors of Health: Chemistry, Bioactivity, and Market Demand for Colorful Foods and Natural Food Sources of Colorants. Annu. Rev. Food Sci. Technol. 2020, 11, 145–182. [Google Scholar] [CrossRef]
  2. Poorniammal, R.; Prabhu, S.; Dufossé, L.; Kannan, J. Safety Evaluation of Fungal Pigments for Food Applications. J. Fungi 2021, 7, 692. [Google Scholar] [CrossRef]
  3. Barciela, P.; Perez-Vazquez, A.; Prieto, M.A. Azo Dyes in the Food Industry: Features, Classification, Toxicity, Alternatives, and Regulation. Food Chem. Toxicol. 2023, 178, 113935. [Google Scholar] [CrossRef] [PubMed]
  4. Okafor, S.N.; Obonga, W.; Ezeokonkwo, M.A.; Nurudeen, J.; Orovwigho, U.; Ahiabuike, J. Assessment of the Health Implications of Synthetic and Natural Food Colourants—A Critical Review. Pharm. Biosci. J. 2016, 4, 1–11. [Google Scholar] [CrossRef]
  5. Stachová, I.; Lhotská, I.; Solich, P.; Šatínský, D. Determination of Green, Blue and Yellow Artificial Food Colorants and Their Abuse in Herb-Coloured Green Easter Beers on Tap. Food Addit. Contam. Part A 2016, 33, 1139–1146. [Google Scholar] [CrossRef] [PubMed]
  6. John, A.; Yang, H.-H.; Muhammad, S.; Khan, Z.I.; Yu, H.; Luqman, M.; Tofail, M.; Hussain, M.I.; Awan, M.U.F. Cross Talk between Synthetic Food Colors (Azo Dyes), Oral Flora, and Cardiovascular Disorders. Appl. Sci. 2022, 12, 7084. [Google Scholar] [CrossRef]
  7. Trasande, L.; Shaffer, R.M.; Sathyanarayana, S. Food Additives and Child Health. Pediatrics 2018, 142, e20181410. [Google Scholar] [CrossRef]
  8. Gupta, R.; Ranjan, S.; Yadav, A.; Verma, B.; Malhotra, K.; Madan, M.; Chopra, O.; Jain, S.; Gupta, S.; Joshi, A.; et al. Toxic Effects of Food Colorants Erythrosine and Tartrazine on Zebrafish Embryo Development. Curr. Res. Nutr. Food Sci. 2019, 7, 876–885. [Google Scholar] [CrossRef]
  9. Mpountoukas, P.; Pantazaki, A.; Kostareli, E.; Christodoulou, P.; Kareli, D.; Poliliou, S.; Mourelatos, C.; Lambropoulou, V.; Lialiaris, T. Cytogenetic Evaluation and DNA Interaction Studies of the Food Colorants Amaranth, Erythrosine and Tartrazine. Food Chem. Toxicol. 2010, 48, 2934–2944. [Google Scholar] [CrossRef]
  10. Kiki, M.J. Biopigments of Microbial Origin and Their Application in the Cosmetic Industry. Cosmetics 2023, 10, 47. [Google Scholar] [CrossRef]
  11. Sharma, N.; Shekhar, P.; Kumar, V.; Kaur, H.; Jayasena, V. Microbial Pigments: Sources, Current Status, Future Challenges in Cosmetics and Therapeutic Applications. J. Basic. Microbiol. 2024, 64, 4–21. [Google Scholar] [CrossRef] [PubMed]
  12. Nwoba, E.G.; Ogbonna, C.N.; Ishika, T.; Vadiveloo, A. Microalgal Pigments: A Source of Natural Food Colors. In Microalgae Biotechnology for Food, Health and High Value Products; Alam, M.A., Xu, J.-L., Wang, Z., Eds.; Springer: Singapore, 2020; pp. 81–123. ISBN 9789811501685. [Google Scholar]
  13. Rana, B.; Bhattacharyya, M.; Patni, B.; Arya, M.; Joshi, G.K. The Realm of Microbial Pigments in the Food Color Market. Front. Sustain. Food Syst. 2021, 5, 603892. [Google Scholar] [CrossRef]
  14. Pailliè-Jiménez, M.E.; Stincone, P.; Brandelli, A. Natural Pigments of Microbial Origin. Front. Sustain. Food Syst. 2020, 4, 590439. [Google Scholar] [CrossRef]
  15. Jacobsen, I.H.; Ledesma-Amaro, R.; Martinez, J.L. Recombinant β-Carotene Production by Yarrowia lipolytica—Assessing the Potential of Micro-Scale Fermentation Analysis in Cell Factory Design and Bioreaction Optimization. Front. Bioeng. Biotechnol. 2020, 8, 29. [Google Scholar] [CrossRef] [PubMed]
  16. Ramesh, C.; Prasastha, V.R.; Venkatachalam, M.; Dufossé, L. Natural Substrates and Culture Conditions to Produce Pigments from Potential Microbes in Submerged Fermentation. Fermentation 2022, 8, 460. [Google Scholar] [CrossRef]
  17. Hu, J.; Meng, W.; Su, Y.; Qian, C.; Fu, W. Emerging Technologies for Advancing Microalgal Photosynthesis and Metabolism toward Sustainable Production. Front. Mar. Sci. 2023, 10, 1260709. [Google Scholar] [CrossRef]
  18. Morocho-Jácome, A.L.; Ruscinc, N.; Martinez, R.M.; De Carvalho, J.C.M.; Santos De Almeida, T.; Rosado, C.; Costa, J.G.; Velasco, M.V.R.; Baby, A.R. (Bio)Technological Aspects of Microalgae Pigments for Cosmetics. Appl. Microbiol. Biotechnol. 2020, 104, 9513–9522. [Google Scholar] [CrossRef]
  19. Barone, G.D.; Ferizović, D.; Biundo, A.; Lindblad, P. Hints at the Applicability of Microalgae and Cyanobacteria for the Biodegradation of Plastics. Sustainability 2020, 12, 10449. [Google Scholar] [CrossRef]
  20. Venil, C.K.; Velmurugan, P.; Dufossé, L.; Renuka Devi, P.; Veera Ravi, A. Fungal Pigments: Potential Coloring Compounds for Wide Ranging Applications in Textile Dyeing. J. Fungi 2020, 6, 68. [Google Scholar] [CrossRef]
  21. Afroz Toma, M.; Rahman, M.H.; Rahman, M.S.; Arif, M.; Nazir, K.H.M.N.H.; Dufossé, L. Fungal Pigments: Carotenoids, Riboflavin, and Polyketides with Diverse Applications. J. Fungi 2023, 9, 454. [Google Scholar] [CrossRef]
  22. Lin, L.; Xu, J. Fungal Pigments and Their Roles Associated with Human Health. J. Fungi 2020, 6, 280. [Google Scholar] [CrossRef] [PubMed]
  23. Agarwal, H.; Bajpai, S.; Mishra, A.; Kohli, I.; Varma, A.; Fouillaud, M.; Dufossé, L.; Joshi, N.C. Bacterial Pigments and Their Multifaceted Roles in Contemporary Biotechnology and Pharmacological Applications. Microorganisms 2023, 11, 614. [Google Scholar] [CrossRef]
  24. Huang, X.; Gan, L.; He, Z.; Jiang, G.; He, T. Bacterial Pigments as a Promising Alternative to Synthetic Colorants: From Fundamentals to Applications. J. Microbiol. Biotechnol. 2024, 34, 2153–2165. [Google Scholar] [CrossRef]
  25. Aman Mohammadi, M.; Ahangari, H.; Mousazadeh, S.; Hosseini, S.M.; Dufossé, L. Microbial Pigments as an Alternative to Synthetic Dyes and Food Additives: A Brief Review of Recent Studies. Bioprocess Biosyst. Eng. 2022, 45, 1–12. [Google Scholar] [CrossRef] [PubMed]
  26. Anshi; Kapil, S.; Goswami, L.; Sharma, V. Unveiling the Intricacies of Microbial Pigments as Sustainable Alternatives to Synthetic Colorants: Recent Trends and Advancements. Micro 2024, 4, 621–640. [Google Scholar] [CrossRef]
  27. Sen, T.; Barrow, C.J.; Deshmukh, S.K. Microbial Pigments in the Food Industry—Challenges and the Way Forward. Front. Nutr. 2019, 6, 7. [Google Scholar] [CrossRef] [PubMed]
  28. Patel, N.; Maharshi, A.; Gohil, N. Microbial Pigments: A Potential Substitute of Synthetic Colorants in the Food and Healthcare Sectors. In Microbial Products for Health and Nutrition; Kothari, V., Ray, S., Kumar, P., Eds.; Springer Nature Singapore: Singapore, 2024; pp. 191–220. ISBN 978-981-9742-34-9. [Google Scholar]
  29. Barreto, J.V.D.O.; Casanova, L.M.; Junior, A.N.; Reis-Mansur, M.C.P.P.; Vermelho, A.B. Microbial Pigments: Major Groups and Industrial Applications. Microorganisms 2023, 11, 2920. [Google Scholar] [CrossRef]
  30. Kumar Roy Choudhury, A. Eco—Friendly Dyes and Dyeing. Adv. Mat. Technol. Environ. 2018, 2, 145–176. [Google Scholar]
  31. Piaskowski, K.; Świderska-Dąbrowska, R.; Zarzycki, P.K. Dye Removal from Water and Wastewater Using Various Physical, Chemical, and Biological Processes. J. AOAC Int. 2018, 101, 1371–1384. [Google Scholar] [CrossRef]
  32. Rane, A.; Joshi, S.J. Biodecolorization and Biodegradation of Dyes: A Review. Open Biotechnol. J. 2021, 15, 97–108. [Google Scholar] [CrossRef]
  33. Popli, S.; Patel, U.D. Destruction of Azo Dyes by Anaerobic-Aerobic Sequential Biological Treatment: A Review. Int. J. Environ. Sci. Technol. 2015, 12, 405–420. [Google Scholar] [CrossRef]
  34. Alzain, H.; Kalimugogo, V.; Hussein, K. A Review of Environmental Impact of Azo Dyes. Int. J. Res. Rev. 2023, 10, 64–689. [Google Scholar] [CrossRef]
  35. Chatragadda, R.; Dufossé, L. Ecological and Biotechnological Aspects of Pigmented Microbes: A Way Forward in Development of Food and Pharmaceutical Grade Pigments. Microorganisms 2021, 9, 637. [Google Scholar] [CrossRef] [PubMed]
  36. Numan, M.; Bashir, S.; Mumtaz, R.; Tayyab, S.; Rehman, N.U.; Khan, A.L.; Shinwari, Z.K.; Al-Harrasi, A. Therapeutic Applications of Bacterial Pigments: A Review of Current Status and Future Opportunities. 3 Biotech 2018, 8, 207. [Google Scholar] [CrossRef]
  37. Qin, Z.; Wang, X.; Gao, S.; Li, D.; Zhou, J. Production of Natural Pigments Using Microorganisms. J. Agric. Food Chem. 2023, 71, 9243–9254. [Google Scholar] [CrossRef] [PubMed]
  38. Aldaghi, S.A.; Ubais, R.; Schmitt, I.; Wendisch, V.F.; Costamagna, M.; Perucca, M. Life Cycle Assessment of Bacterial, Algal, and Synthetic Approaches for Astaxanthin Production at a Laboratory Scale: Comparative Environmental Analysis and Sensitivity of Energy Sources. Processes 2023, 11, 2911. [Google Scholar] [CrossRef]
  39. Delgove, M.A.F.; Laurent, A.; Woodley, J.M.; De Wildeman, S.M.A.; Bernaerts, K.V.; van der Meer, Y. A Prospective Life Cycle Assessment (LCA) of Monomer Synthesis: Comparison of Biocatalytic and Oxidative Chemistry. ChemSusChem 2019, 12, 1349–1360. [Google Scholar] [CrossRef]
  40. Renita, A.A.; Gajaria, T.K.; Sathish, S.; Kumar, J.A.; Lakshmi, D.S.; Kujawa, J.; Kujawski, W. Progress and Prospective of the Industrial Development and Applications of Eco-Friendly Colorants: An Insight into Environmental Impact and Sustainability Issues. Foods 2023, 12, 1521. [Google Scholar] [CrossRef]
  41. Díez, B.H.; Torres, C.A.V.; Gaudêncio, S.P. Actinomycete-Derived Pigments: A Path Toward Sustainable Industrial Colorants. Mar. Drugs 2025, 23, 39. [Google Scholar] [CrossRef]
  42. Durán, N.; Teixeira, M.F.S.; De Conti, R.; Esposito, E. Ecological-Friendly Pigments From Fungi. Crit. Rev. Food Sci. Nutr. 2002, 42, 53–66. [Google Scholar] [CrossRef]
  43. Gürses, A.; Açıkyıldız, M.; Güneş, K.; Gürses, M.S. Classification of Dye and Pigments. In Dyes and Pigments; Gürses, A., Açıkyıldız, M., Güneş, K., Gürses, M.S., Eds.; Springer International Publishing: Cham, Switzerland, 2016; pp. 31–45. ISBN 978-3-319-33890-3. [Google Scholar]
  44. Li, M.; Xia, Q.; Zhang, H.; Zhang, R.; Yang, J. Metabolic Engineering of Different Microbial Hosts for Lycopene Production. J. Agric. Food Chem. 2020, 68, 14104–14122. [Google Scholar] [CrossRef] [PubMed]
  45. Yoshida, R.; Yoshimura, T.; Hemmi, H. Reconstruction of the “Archaeal” Mevalonate Pathway from the Methanogenic Archaeon Methanosarcina Mazei in Escherichia Coli Cells. Appl. Environ. Microbiol. 2020, 86, e02889–19. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, J.; Guo, L. Biosynthesis of β-Carotene in Engineered E. Coli Using the MEP and MVA Pathways. Microb. Cell Fact. 2014, 13, 160. [Google Scholar] [CrossRef]
  47. Mehta, B.J.; Cerdá-Olmedo, E. Mutants of Carotene Production in Blakeslea Trispora. Appl. Microbiol. Biotechnol. 1995, 42, 836–838. [Google Scholar] [CrossRef]
  48. Wang, Z.; Sun, J.; Yang, Q.; Yang, J. Metabolic Engineering Escherichia coli for the Production of Lycopene. Molecules 2020, 25, 3136. [Google Scholar] [CrossRef]
  49. López-Nieto, M.J.; Costa, J.; Peiro, E.; Méndez, E.; Rodríguez-Sáiz, M.; De La Fuente, J.L.; Cabri, W.; Barredo, J.L. Biotechnological Lycopene Production by Mated Fermentation of Blakeslea Trispora. Appl. Microbiol. Biotechnol. 2004, 66, 153–159. [Google Scholar] [CrossRef] [PubMed]
  50. Sun, T.; Miao, L.; Li, Q.; Dai, G.; Lu, F.; Liu, T.; Zhang, X.; Ma, Y. Production of Lycopene by Metabolically-Engineered Escherichia coli. Biotechnol. Lett. 2014, 36, 1515–1522. [Google Scholar] [CrossRef]
  51. Kong, K.-W.; Khoo, H.-E.; Prasad, K.N.; Ismail, A.; Tan, C.-P.; Rajab, N.F. Revealing the Power of the Natural Red Pigment Lycopene. Molecules 2010, 15, 959–987. [Google Scholar] [CrossRef]
  52. Mannazzu, I.; Landolfo, S.; Da Silva, T.L.; Buzzini, P. Red Yeasts and Carotenoid Production: Outlining a Future for Non-Conventional Yeasts of Biotechnological Interest. World J. Microbiol. Biotechnol. 2015, 31, 1665–1673. [Google Scholar] [CrossRef]
  53. Paniagua-Michel, J.; Olmos-Soto, J.; Ruiz, M.A. Pathways of Carotenoid Biosynthesis in Bacteria and Microalgae. In Microbial Carotenoids from Bacteria and Microalgae; Barredo, J.-L., Ed.; Humana Press: Totowa, NJ, USA, 2012; Volume 892, pp. 1–12. ISBN 978-1-61779-878-8. [Google Scholar]
  54. Gupta, I.; Adin, S.N.; Panda, B.P.; Mujeeb, M. β-Carotene—Production Methods, Biosynthesis from Phaffia Rhodozyma, Factors Affecting Its Production during Fermentation, Pharmacological Properties: A Review. Biotechnol. Appl. Biochem. 2022, 69, 2517–2529. [Google Scholar] [CrossRef]
  55. Berman, J.; Zorrilla-López, U.; Farré, G.; Zhu, C.; Sandmann, G.; Twyman, R.M.; Capell, T.; Christou, P. Nutritionally Important Carotenoids as Consumer Products. Phytochem. Rev. 2015, 14, 727–743. [Google Scholar] [CrossRef]
  56. Chen, M.; Li, M.; Ye, L.; Yu, H. Construction of Canthaxanthin-Producing Yeast by Combining Spatiotemporal Regulation and Pleiotropic Drug Resistance Engineering. ACS Synth. Biol. 2022, 11, 325–333. [Google Scholar] [CrossRef]
  57. Steiger, S.; Sandmann, G. Cloning of Two Carotenoid Ketolase Genes from Nostoc Punctiforme for the Heterologous Production of Canthaxanthin and Astaxanthin. Biotechnol. Lett. 2004, 26, 813–817. [Google Scholar] [CrossRef] [PubMed]
  58. Papp, T.; Csernetics, Á.; Nagy, G.; Bencsik, O.; Iturriaga, E.A.; Eslava, A.P.; Vágvölgyi, C. Canthaxanthin Production with Modified Mucor circinelloides Strains. Appl. Microbiol. Biotechnol. 2013, 97, 4937–4950. [Google Scholar] [CrossRef]
  59. Esatbeyoglu, T.; Rimbach, G. Canthaxanthin: From Molecule to Function. Mol. Nutr. Food Res. 2017, 61, 1600469. [Google Scholar] [CrossRef] [PubMed]
  60. Nakano, T.; Wiegertjes, G. Properties of Carotenoids in Fish Fitness: A Review. Mar. Drugs 2020, 18, 568. [Google Scholar] [CrossRef]
  61. Goswami, G.; Chaudhuri, S.; Dutta, D. The Present Perspective of Astaxanthin with Reference to Biosynthesis and Pharmacological Importance. World J. Microbiol. Biotechnol. 2010, 26, 1925–1939. [Google Scholar] [CrossRef]
  62. Davinelli, S.; Nielsen, M.E.; Scapagnini, G. Astaxanthin in Skin Health, Repair, and Disease: A Comprehensive Review. Nutrients 2018, 10, 522. [Google Scholar] [CrossRef]
  63. Domínguez-Bocanegra, A.R.; Ponce-Noyola, T.; Torres-Muñoz, J.A. Astaxanthin Production by Phaffia rhodozyma and Haematococcus pluvialis: A Comparative Study. Appl. Microbiol. Biotechnol. 2007, 75, 783–791. [Google Scholar] [CrossRef]
  64. Liao, Y.; Wang, H.; Li, S.; Xue, Y.; Chen, Y.; Adu-Frimpong, M.; Xu, Y.; Yu, J.; Xu, X.; Smyth, H.D.C.; et al. Preparation of Astaxanthin-loaded Composite Micelles with Coaxial Electrospray Technology for Enhanced Oral Bioavailability and Improved Antioxidation Capability. J. Sci. Food Agric. 2024, 104, 1408–1419. [Google Scholar] [CrossRef]
  65. Martínez-Álvarez, Ó.; Calvo, M.M.; Gómez-Estaca, J. Recent Advances in Astaxanthin Micro/Nanoencapsulation to Improve Its Stability and Functionality as a Food Ingredient. Mar. Drugs 2020, 18, 406. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, J.; Zhang, R.; Zhang, G.; Liu, Z.; Jiang, H.; Mao, X. Heterologous Expression of the Plant-Derived Astaxanthin Biosynthesis Pathway in Yarrowia Lipolytica for Glycosylated Astaxanthin Production. J. Agric. Food Chem. 2023, 71, 2943–2951. [Google Scholar] [CrossRef] [PubMed]
  67. Santos, M.C.D.; Bicas, J.L. Natural Blue Pigments and Bikaverin. Microbiol. Res. 2021, 244, 126653. [Google Scholar] [CrossRef] [PubMed]
  68. Eisenberg, I.; Caycedo-Soler, F.; Harris, D.; Yochelis, S.; Huelga, S.F.; Plenio, M.B.; Adir, N.; Keren, N.; Paltiel, Y. Regulating the Energy Flow in a Cyanobacterial Light-Harvesting Antenna Complex. J. Phys. Chem. B 2017, 121, 1240–1247. [Google Scholar] [CrossRef]
  69. Zhong, Y.; Sun, S.; Dai, T.; Zhang, H.; Wu, J.; Gong, E.S. Phycocyanin-Chitosan Complex Stabilized Emulsion: Preparation, Characteristics, Digestibility, and Stability. Int. J. Biol. Macromol. 2024, 260, 129253. [Google Scholar] [CrossRef]
  70. Setiyono, E.; Adhiwibawa, M.A.S.; Indrawati, R.; Prihastyanti, M.N.U.; Shioi, Y.; Brotosudarmo, T.H.P. An Indonesian Marine Bacterium, Pseudoalteromonas rubra, Produces Antimicrobial Prodiginine Pigments. ACS Omega 2020, 5, 4626–4635. [Google Scholar] [CrossRef]
  71. Han, R.; Xiang, R.; Li, J.; Wang, F.; Wang, C. High-level Production of Microbial Prodigiosin: A Review. J. Basic. Microbiol. 2021, 61, 506–523. [Google Scholar] [CrossRef]
  72. Kuo, Y.-Y.; Li, S.-Y. Solid-State Fermentation of Food Waste by Serratia marcescens NCHU05 for Prodigiosin Production. J. Taiwan. Inst. Chem. Eng. 2024, 160, 105260. [Google Scholar] [CrossRef]
  73. Williamson, N.R.; Simonsen, H.T.; Ahmed, R.A.A.; Goldet, G.; Slater, H.; Woodley, L.; Leeper, F.J.; Salmond, G.P.C. Biosynthesis of the Red Antibiotic, Prodigiosin, in Serratia: Identification of a Novel 2-methyl-3-n-amyl-pyrrole (MAP) Assembly Pathway, Definition of the Terminal Condensing Enzyme, and Implications for Undecylprodigiosin Biosynthesis in Streptomyces. Mol. Microbiol. 2005, 56, 971–989. [Google Scholar] [CrossRef]
  74. Lapenda, J.C.L.; Alves, V.P.; Adam, M.L.; Rodrigues, M.D.; Nascimento, S.C. Cytotoxic Effect of Prodigiosin, Natural Red Pigment, Isolated from Serratia marcescens UFPEDA 398. Indian J. Microbiol. 2020, 60, 182–195. [Google Scholar] [CrossRef]
  75. Wang, Z.; Li, B.; Zhou, L.; Yu, S.; Su, Z.; Song, J.; Sun, Q.; Sha, O.; Wang, X.; Jiang, W.; et al. Prodigiosin Inhibits Wnt/β-Catenin Signaling and Exerts Anticancer Activity in Breast Cancer Cells. Proc. Natl. Acad. Sci. USA 2016, 113, 13150–13155. [Google Scholar] [CrossRef]
  76. Li, D.; Liu, J.; Wang, X.; Kong, D.; Du, W.; Li, H.; Hse, C.-Y.; Shupe, T.; Zhou, D.; Zhao, K. Biological Potential and Mechanism of Prodigiosin from Serratia marcescens Subsp. Lawsoniana in Human Choriocarcinoma and Prostate Cancer Cell Lines. Int. J. Mol. Sci. 2018, 19, 3465. [Google Scholar] [CrossRef]
  77. Lapenda, J.C.; Silva, P.A.; Vicalvi, M.C.; Sena, K.X.F.R.; Nascimento, S.C. Antimicrobial Activity of Prodigiosin Isolated from Serratia marcescens UFPEDA 398. World J. Microbiol. Biotechnol. 2015, 31, 399–406. [Google Scholar] [CrossRef] [PubMed]
  78. Stahmann, K.-P.; Revuelta, J.L.; Seulberger, H. Three Biotechnical Processes Using Ashbya gossypii, Candida famata, or Bacillus subtilis Compete with Chemical Riboflavin Production. Appl. Microbiol. Biotechnol. 2000, 53, 509–516. [Google Scholar] [CrossRef]
  79. Macheroux, P.; Kappes, B.; Ealick, S.E. Flavogenomics—A Genomic and Structural View of Flavin-dependent Proteins. FEBS J. 2011, 278, 2625–2634. [Google Scholar] [CrossRef] [PubMed]
  80. Rhee, H.-W.; Choi, S.J.; Yoo, S.H.; Jang, Y.O.; Park, H.H.; Pinto, R.M.; Cameselle, J.C.; Sandoval, F.J.; Roje, S.; Han, K.; et al. A Bifunctional Molecule as an Artificial Flavin Mononucleotide Cyclase and a Chemosensor for Selective Fluorescent Detection of Flavins. J. Am. Chem. Soc. 2009, 131, 10107–10112. [Google Scholar] [CrossRef] [PubMed]
  81. Powers, H.J. Riboflavin (Vitamin B-2) and Health. Am. J. Clin. Nutr. 2003, 77, 1352–1360. [Google Scholar] [CrossRef]
  82. Choi, S.Y.; Yoon, K.; Lee, J.I.; Mitchell, R.J. Violacein: Properties and Production of a Versatile Bacterial Pigment. BioMed Res. Int. 2015, 2015, 1–8. [Google Scholar] [CrossRef]
  83. Balibar, C.J.; Walsh, C.T. In Vitro Biosynthesis of Violacein from l-Tryptophan by the Enzymes VioA−E from Chromobacterium Violaceum. Biochemistry 2006, 45, 15444–15457. [Google Scholar] [CrossRef]
  84. Ahmed, A.; Ahmad, A.; Li, R.; AL-Ansi, W.; Fatima, M.; Mushtaq, B.S.; Basharat, S.; Li, Y.; Bai, Z. Recent Advances in Synthetic, Industrial and Biological Applications of Violacein and Its Heterologous Production. J. Microbiol. Biotechnol. 2021, 31, 1465–1480. [Google Scholar] [CrossRef]
  85. Park, H.; Park, S.; Yang, Y.-H.; Choi, K.-Y. Microbial Synthesis of Violacein Pigment and Its Potential Applications. Crit. Rev. Biotechnol. 2021, 41, 879–901. [Google Scholar] [CrossRef] [PubMed]
  86. Yada, S.; Wang, Y.; Zou, Y.; Nagasaki, K.; Hosokawa, K.; Osaka, I.; Arakawa, R.; Enomoto, K. Isolation and Characterization of Two Groups of Novel Marine Bacteria Producing Violacein. Mar. Biotechnol. 2008, 10, 128–132. [Google Scholar] [CrossRef] [PubMed]
  87. Aranda, S.; Montes-Borrego, M.; Landa, B.B. Purple-Pigmented Violacein-Producing Duganella Spp. Inhabit the Rhizosphere of Wild and Cultivated Olives in Southern Spain. Microb. Ecol. 2011, 62, 446–459. [Google Scholar] [CrossRef]
  88. Chang, P.-K.; Cary, J.W.; Lebar, M.D. Biosynthesis of Conidial and Sclerotial Pigments in Aspergillus Species. Appl. Microbiol. Biotechnol. 2020, 104, 2277–2286. [Google Scholar] [CrossRef]
  89. Rzepka, Z.; Buszman, E.; Beberok, A.; Wrześniok, D. From Tyrosine to Melanin: Signaling Pathways and Factors Regulating Melanogenesis. Postep. Hig. Med. Dosw. 2016, 70, 695–708. [Google Scholar] [CrossRef]
  90. Eisenman, H.C.; Casadevall, A. Synthesis and Assembly of Fungal Melanin. Appl. Microbiol. Biotechnol. 2012, 93, 931–940. [Google Scholar] [CrossRef]
  91. Glagoleva, A.Y.; Shoeva, O.Y.; Khlestkina, E.K. Melanin Pigment in Plants: Current Knowledge and Future Perspectives. Front. Plant Sci. 2020, 11, 770. [Google Scholar] [CrossRef] [PubMed]
  92. Cavallini, C.; Vitiello, G.; Adinolfi, B.; Silvestri, B.; Armanetti, P.; Manini, P.; Pezzella, A.; d’Ischia, M.; Luciani, G.; Menichetti, L. Melanin and Melanin-Like Hybrid Materials in Regenerative Medicine. Nanomaterials 2020, 10, 1518. [Google Scholar] [CrossRef]
  93. Song, W.; Yang, H.; Liu, S.; Yu, H.; Li, D.; Li, P.; Xing, R. Melanin: Insights into Structure, Analysis, and Biological Activities for Future Development. J. Mater. Chem. B 2023, 11, 7528–7543. [Google Scholar] [CrossRef]
  94. Keith, K.E.; Killip, L.; He, P.; Moran, G.R.; Valvano, M.A. Burkholderia cenocepacia C5424 Produces a Pigment with Antioxidant Properties Using a Homogentisate Intermediate. J. Bacteriol. 2007, 189, 9057–9065. [Google Scholar] [CrossRef]
  95. Ahmad, S.; Mohammed, M.; Mekala, L.P.; Chintalapati, S.; Chintalapati, V.R. Tryptophan, a Non-Canonical Melanin Precursor: New L-Tryptophan Based Melanin Production by Rubrivivax benzoatilyticus JA2. Sci. Rep. 2020, 10, 8925. [Google Scholar] [CrossRef] [PubMed]
  96. El-Sayyad, G.S.; Mosallam, F.M.; El-Sayed, S.S.; El-Batal, A.I. Facile Biosynthesis of Tellurium Dioxide Nanoparticles by Streptomyces cyaneus Melanin Pigment and Gamma Radiation for Repressing Some Aspergillus Pathogens and Bacterial Wound Cultures. J. Clust. Sci. 2020, 31, 147–159. [Google Scholar] [CrossRef]
  97. Valiante, V.; Baldin, C.; Hortschansky, P.; Jain, R.; Thywißen, A.; Straßburger, M.; Shelest, E.; Heinekamp, T.; Brakhage, A.A. The Aspergillus Fumigatus Conidial Melanin Production Is Regulated by the Bifunctional bHLH DevR and MADS-box RlmA Transcription Factors. Mol. Microbiol. 2016, 102, 321–335. [Google Scholar] [CrossRef]
  98. Gómez, B.L.; Nosanchuk, J.D. Melanin and Fungi. Curr. Opin. Infect. Dis. 2003, 16, 91–96. [Google Scholar] [CrossRef]
  99. Wang, Z.; Tschirhart, T.; Schultzhaus, Z.; Kelly, E.E.; Chen, A.; Oh, E.; Nag, O.; Glaser, E.R.; Kim, E.; Lloyd, P.F.; et al. Melanin Produced by the Fast-Growing Marine Bacterium Vibrio natriegens through Heterologous Biosynthesis: Characterization and Application. Appl. Environ. Microbiol. 2020, 86, e02749-19. [Google Scholar] [CrossRef] [PubMed]
  100. Sun, J.; Xu, W.; Li, L.; Fan, B.; Peng, X.; Qu, B.; Wang, L.; Li, T.; Li, S.; Zhang, R. Ultrasmall Endogenous Biopolymer Nanoparticles for Magnetic Resonance/Photoacoustic Dual-Modal Imaging-Guided Photothermal Therapy. Nanoscale 2018, 10, 10584–10595. [Google Scholar] [CrossRef]
  101. Guo, K.; Jiao, Z.; Zhao, X.; Hu, Y.; Zhao, N.; Xu, F.-J. Melanin-Based Immunoregulatory Nanohybrids Enhance Antitumor Immune Responses in Breast Cancer Mouse Model. ACS Nano 2023, 17, 10792–10805. [Google Scholar] [CrossRef]
  102. Pogorzelska, E.; Godziszewska, J.; Brodowska, M.; Wierzbicka, A. Antioxidant Potential of Haematococcus pluvialis Extract Rich in Astaxanthin on Colour and Oxidative Stability of Raw Ground Pork Meat during Refrigerated Storage. Meat Sci. 2018, 135, 54–61. [Google Scholar] [CrossRef]
  103. Asker, D. Isolation and Characterization of a Novel, Highly Selective Astaxanthin-Producing Marine Bacterium. J. Agric. Food Chem. 2017, 65, 9101–9109. [Google Scholar] [CrossRef]
  104. Dufossé, L. Current and Potential Natural Pigments from Microorganisms (Bacteria, Yeasts, Fungi, Microalgae). In Handbook on Natural Pigments in Food and Beverages; Schweiggert, R., Ed.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 337–354. ISBN 978-0-08-100371-8. [Google Scholar]
  105. Capelli, B.; Cysewki, G.R. Natural Astaxanthin the World’s Best Kept Health Secret; Cyanotech Corporation: Kailua-Kona, HI, USA, 2012; p. 202. [Google Scholar]
  106. Schmidt, I.; Schewe, H.; Gassel, S.; Jin, C.; Buckingham, J.; Hümbelin, M.; Sandmann, G.; Schrader, J. Biotechnological Production of Astaxanthin with Phaffia rhodozyma/Xanthophyllomyces dendrorhous. Appl. Microbiol. Biotechnol. 2011, 89, 555–571. [Google Scholar] [CrossRef]
  107. Zhou, P.; Ye, L.; Xie, W.; Lv, X.; Yu, H. Highly Efficient Biosynthesis of Astaxanthin in Saccharomyces cerevisiae by Integration and Tuning of Algal crtZ and Bkt. Appl. Microbiol. Biotechnol. 2015, 99, 8419–8428. [Google Scholar] [CrossRef] [PubMed]
  108. Le-Feuvre, R.; Moraga-Suazo, P.; Gonzalez, J.; Martin, S.S.; Henríquez, V.; Donoso, A.; Agurto-Muñoz, C. Biotechnology Applied to Haematococcus pluvialis Fotow: Challenges and Prospects for the Enhancement of Astaxanthin Accumulation. J. Appl. Phycol. 2020, 32, 3831–3852. [Google Scholar] [CrossRef]
  109. Lemuth, K.; Steuer, K.; Albermann, C. Engineering of a Plasmid-Free Escherichia coli Strain for Improved in Vivo Biosynthesis of Astaxanthin. Microb. Cell Factories 2011, 10, 29. [Google Scholar] [CrossRef] [PubMed]
  110. Liu, X.; McClements, D.J.; Cao, Y.; Xiao, H. Chemical and Physical Stability of Astaxanthin-Enriched Emulsion-Based Delivery Systems. Food Biophys. 2016, 11, 302–310. [Google Scholar] [CrossRef]
  111. Eriksen, N.T. Production of Phycocyanin—A Pigment with Applications in Biology, Biotechnology, Foods and Medicine. Appl. Microbiol. Biotechnol. 2008, 80, 1–14. [Google Scholar] [CrossRef]
  112. Dufossé; Chatragadda, R.; Nambali Valsalan, V.; Ramalingam, K.; Chidambaram Kulandaisamy, V.; Laurent, D. Multifaceted Applications of Microbial Pigments: Current Knowledge, Challenges and Future Directions for Public Health Implications. Microorganisms 2019, 7, 186. [Google Scholar] [CrossRef]
  113. Cuellar-Bermudez, S.P.; Aguilar-Hernandez, I.; Cardenas-Chavez, D.L.; Ornelas-Soto, N.; Romero-Ogawa, M.A.; Parra-Saldivar, R. Extraction and Purification of High-value Metabolites from Microalgae: Essential Lipids, Astaxanthin and Phycobiliproteins. Microb. Biotechnol. 2015, 8, 190–209. [Google Scholar] [CrossRef] [PubMed]
  114. Oddities and Curiosities in the Algal World. In Algae; CRC Press: Boca Raton, FL, USA, 2014; pp. 325–342. ISBN 978-0-429-10718-4.
  115. Abdelaziz, A.A.; Kamer, A.M.A.; Al-Monofy, K.B.; Al-Madboly, L.A. Pseudomonas aeruginosa’s Greenish-Blue Pigment Pyocyanin: Its Production and Biological Activities. Microb. Cell Factories 2023, 22, 110. [Google Scholar] [CrossRef]
  116. Julianti, E.; Susanti, S.; Singgih, M.; Mulyani, L.N. Optimization of Extraction Method and Characterization of Phycocyanin Pigment from Spirulina platensis. J. Math. Fundam Sci. 2019, 51, 168–176. [Google Scholar] [CrossRef]
  117. Braga, A.R.C.; Figueira, F.D.S.; Silveira, J.T.D.; Morais, M.G.D.; Costa, J.A.V.; Kalil, S.J. Improvement of Thermal Stability of C-Phycocyanin by Nanofiber and Preservative Agents: C-Phycocyanin Stability: Preservative and Nanofiber. J. Food Process. Preserv. 2016, 40, 1264–1269. [Google Scholar] [CrossRef]
  118. Namazkar, S.; Ahmad, W.A. Spray-Dried Prodigiosin from Serratia marcescens As A Colorant. Biosci. Biotechnol. Res. Asia 2013, 10, 69–76. [Google Scholar] [CrossRef]
  119. Dauenhauer, S.A.; Hull, R.A.; Williams, R.P. Cloning and Expression in Escherichia coli of Serratia Marcescens Genes Encoding Prodigiosin Biosynthesis. J. Bacteriol. 1984, 158, 1128–1132. [Google Scholar] [CrossRef] [PubMed]
  120. Domröse, A.; Klein, A.S.; Hage-Hülsmann, J.; Thies, S.; Svensson, V.; Classen, T.; Pietruszka, J.; Jaeger, K.-E.; Drepper, T.; Loeschcke, A. Efficient Recombinant Production of Prodigiosin in Pseudomonas putida. Front. Microbiol. 2015, 6, 972. [Google Scholar] [CrossRef]
  121. Pan, X.; Tang, M.; You, J.; Liu, F.; Sun, C.; Osire, T.; Fu, W.; Yi, G.; Yang, T.; Yang, S.-T.; et al. Regulator RcsB Controls Prodigiosin Synthesis and Various Cellular Processes in Serratia marcescens JNB5-1. Appl. Environ. Microbiol. 2021, 87, e02052–20. [Google Scholar] [CrossRef]
  122. Dos Santos, R.A.; Rodríguez, D.M.; Da Silva, L.A.R.; De Almeida, S.M.; De Campos-Takaki, G.M.; De Lima, M.A.B. Enhanced Production of Prodigiosin by Serratia marcescens UCP 1549 Using Agrosubstrates in Solid-State Fermentation. Arch. Microbiol. 2021, 203, 4091–4100. [Google Scholar] [CrossRef]
  123. Hong, M.Y.; Seeram, N.P.; Zhang, Y.; Heber, D. Anticancer Effects of Chinese Red Yeast Rice versus Monacolin K Alone on Colon Cancer Cells. J. Nutr. Biochem. 2008, 19, 448–458. [Google Scholar] [CrossRef] [PubMed]
  124. Dufossé, L. Microbial Pigments from Bacteria, Yeasts, Fungi, and Microalgae for the Food and Feed Industries. In Natural and Artificial Flavoring Agents and Food Dyes; Grumezescu, A.M., Holban, A.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2018; Volume 7, pp. 113–132. ISBN 978-0-12-811518-3. [Google Scholar]
  125. Averianova, L.A.; Balabanova, L.A.; Son, O.M.; Podvolotskaya, A.B.; Tekutyeva, L.A. Production of Vitamin B2 (Riboflavin) by Microorganisms: An Overview. Front. Bioeng. Biotechnol. 2020, 8, 570828. [Google Scholar] [CrossRef]
  126. Schwechheimer, S.K.; Park, E.Y.; Revuelta, J.L.; Becker, J.; Wittmann, C. Biotechnology of Riboflavin. Appl. Microbiol. Biotechnol. 2016, 100, 2107–2119. [Google Scholar] [CrossRef]
  127. Hustad, S.; Eussen, S.; Midttun, Ø.; Ulvik, A.; Van De Kant, P.M.; Mørkrid, L.; Gislefoss, R.; Ueland, P.M. Kinetic Modeling of Storage Effects on Biomarkers Related to B Vitamin Status and One-Carbon Metabolism. Clin. Chem. 2012, 58, 402–410. [Google Scholar] [CrossRef]
  128. Sigurdson, G.T.; Tang, P.; Giusti, M.M. Natural Colorants: Food Colorants from Natural Sources. Annu. Rev. Food Sci. Technol. 2017, 8, 261–280. [Google Scholar] [CrossRef]
  129. Kot, A.M.; Błażejak, S.; Kurcz, A.; Gientka, I.; Kieliszek, M. Rhodotorula Glutinis—Potential Source of Lipids, Carotenoids, and Enzymes for Use in Industries. Appl. Microbiol. Biotechnol. 2016, 100, 6103–6117. [Google Scholar] [CrossRef] [PubMed]
  130. Eroglu, A.; Hruszkewycz, D.P.; Dela Sena, C.; Narayanasamy, S.; Riedl, K.M.; Kopec, R.E.; Schwartz, S.J.; Curley, R.W.; Harrison, E.H. Naturally Occurring Eccentric Cleavage Products of Provitamin A β-Carotene Function as Antagonists of Retinoic Acid Receptors. J. Biol. Chem. 2012, 287, 15886–15895. [Google Scholar] [CrossRef] [PubMed]
  131. Larroude, M.; Celinska, E.; Back, A.; Thomas, S.; Nicaud, J.; Ledesma-Amaro, R. A Synthetic Biology Approach to Transform Yarrowia lipolytica into a Competitive Biotechnological Producer of Β-carotene. Biotechnol. Bioeng. 2018, 115, 464–472. [Google Scholar] [CrossRef]
  132. Aman, R.; Schieber, A.; Carle, R. Effects of Heating and Illumination on Trans.Cis Isomerization and Degradation of β-Carotene and Lutein in Isolated Spinach Chloroplasts. J. Agric. Food Chem. 2005, 53, 9512–9518. [Google Scholar] [CrossRef] [PubMed]
  133. Zhou, Y.; Fang, M.-Y.; Li, G.; Zhang, C.; Xing, X.-H. Enhanced Production of Crude Violacein from Glucose in Escherichia coli by Overexpression of Rate-Limiting Key Enzyme(S) Involved in Violacein Biosynthesis. Appl. Biochem. Biotechnol. 2018, 186, 909–916. [Google Scholar] [CrossRef]
  134. Rabasco Alvarez, A.M.; González Rodríguez, M.L. Lipids in Pharmaceutical and Cosmetic Preparations. Grasas Aceites 2000, 51, 74–96. [Google Scholar] [CrossRef]
  135. Surwase, S.N.; Jadhav, S.B.; Phugare, S.S.; Jadhav, J.P. Optimization of Melanin Production by Brevundimonas Sp. SGJ Using Response Surface Methodology. 3 Biotech 2013, 3, 187–194. [Google Scholar] [CrossRef]
  136. Jalmi, P.; Bodke, P.; Wahidullah, S.; Raghukumar, S. The Fungus Gliocephalotrichum simplex as a Source of Abundant, Extracellular Melanin for Biotechnological Applications. World J. Microbiol. Biotechnol. 2012, 28, 505–512. [Google Scholar] [CrossRef]
  137. Campanhol, B.S.; Ribeiro, B.D.; Casellato, F.; Medina, K.J.D.; Sponchiado, S.R.P. Improvement of DOPA-Melanin Production by Aspergillus nidulans Using Eco-Friendly and Inexpensive Substrates. J. Fungi 2023, 9, 714. [Google Scholar] [CrossRef]
  138. Guo, J.; Rao, Z.; Yang, T.; Man, Z.; Xu, M.; Zhang, X. High-Level Production of Melanin by a Novel Isolate of Streptomyces kathirae. FEMS Microbiol. Lett. 2014, 357, 85–91. [Google Scholar] [CrossRef]
  139. Mavridi-Printezi, A.; Guernelli, M.; Menichetti, A.; Montalti, M. Bio-Applications of Multifunctional Melanin Nanoparticles: From Nanomedicine to Nanocosmetics. Nanomaterials 2020, 10, 2276. [Google Scholar] [CrossRef]
  140. Giovannucci, E.; Rimm, E.B.; Liu, Y.; Stampfer, M.J.; Willett, W. A Prospective Study of Tomato Products, Lycopene, and Prostate Cancer Risk. J. Natl. Cancer Inst. 2002, 94, 391–398. [Google Scholar] [CrossRef]
  141. Papaioannou, E.H.; Liakopoulou-Kyriakides, M.; Karabelas, A.J. Natural Origin Lycopene and Its “Green” Downstream Processing. Crit. Rev. Food Sci. Nutr. 2016, 56, 686–709. [Google Scholar] [CrossRef] [PubMed]
  142. Silva, D.F.; Favaro-Trindade, C.S.; Rocha, G.A.; Thomazini, M. Microencapsulation of Lycopene by Gelatin-Pectin Complex Coacervation. J. Food Process. Preserv. 2012, 36, 185–190. [Google Scholar] [CrossRef]
  143. Hamidi, M.; Hejazi, M.S.; Nazemyieh, H.; Hejazi, M.A.; Naziri, D. Halorubrum Sp. TBZ112, an Extremely Halophilic Carotenoid- Producing Archaeon Isolated from Urmia Lake. Pharm. Sci. 2017, 23, 150–158. [Google Scholar] [CrossRef]
  144. Naz, T.; Yang, J.; Nosheen, S.; Sun, C.; Nazir, Y.; Mohamed, H.; Fazili, A.B.A.; Ullah, S.; Li, S.; Yang, W.; et al. Genetic Modification of Mucor circinelloides for Canthaxanthin Production by Heterologous Expression of β-Carotene Ketolase Gene. Front. Nutr. 2021, 8, 756218. [Google Scholar] [CrossRef]
  145. Promdonkoy, P.; Watcharawipas, A.; Bubphasawan, S.; Sansatchanon, K.; Suwanakitti, N.; Kocharin, K.; Runguphan, W. Metabolic Engineering of Saccharomyces cerevisiae for Production of Canthaxanthin, Zeaxanthin, and Astaxanthin. J. Fungi 2024, 10, 433. [Google Scholar] [CrossRef]
  146. Neza, E.; Centini, M. Microbiologically Contaminated and Over-Preserved Cosmetic Products According Rapex 2008–2014. Cosmetics 2016, 3, 3. [Google Scholar] [CrossRef]
  147. Durán, N.; Menck, C.F.M. Chromobacterium violaceum: A Review of Pharmacological and Industiral Perspectives. Crit. Rev. Microbiol. 2001, 27, 201–222. [Google Scholar] [CrossRef]
  148. Tuli, H.S.; Chaudhary, P.; Beniwal, V.; Sharma, A.K. Microbial Pigments as Natural Color Sources: Current Trends and Future Perspectives. J. Food Sci. Technol. 2015, 52, 4669–4678. [Google Scholar] [CrossRef]
  149. Mummaleti, G.; Udo, T.; Mohan, A.; Kong, F. Synthesis, Characterization and Application of Microbial Pigments in Foods as Natural Colors. Crit. Rev. Food Sci. Nutr. 2024, 1–30. [Google Scholar] [CrossRef] [PubMed]
  150. Venil, C.K.; Zakaria, Z.A.; Ahmad, W.A. Bacterial Pigments and Their Applications. Process Biochem. 2013, 48, 1065–1079. [Google Scholar] [CrossRef]
  151. Raita, S.; Feldmane, L.; Kusnere, Z.; Spalvins, K.; Kuzmika, I.; Berzina, I.; Mika, T. Microbial Carotenoids Production: Strains, Conditions, and Yield Affecting Factors. Environ. Clim. Technol. 2023, 27, 1027–1048. [Google Scholar] [CrossRef]
  152. Grand View Research. Beta-Carotene Market Size, Share & Trends Analysis Report by Type (Natural, Synthetic), by End-Use (Food & Beverages, Pharmaceutical, Dietary Supplements), by Region (North America, Asia Pacific), And Segment Forecasts, 2024–2030; Grand View Research: San Francisco, CA, USA, 2024. [Google Scholar]
  153. Fact.MR. Synthetic Beta-Carotene Market Outlook (2024–2034); Fact.MR: Rockville, MD, USA, 2024. [Google Scholar]
  154. Cortez, R.; Luna-Vital, D.A.; Margulis, D.; Gonzalez De Mejia, E. Natural Pigments: Stabilization Methods of Anthocyanins for Food Applications. Comp. Rev. Food Sci. Food Safe 2017, 16, 180–198. [Google Scholar] [CrossRef]
  155. Jardine, A.; Sayed, S. Valorisation of Chitinous Biomass for Antimicrobial Applications. Pure Appl. Chem. 2018, 90, 293–304. [Google Scholar] [CrossRef]
  156. Nawaz, A.; Chaudhary, R.; Shah, Z.; Dufossé, L.; Fouillaud, M.; Mukhtar, H.; Ul Haq, I. An Overview on Industrial and Medical Applications of Bio-Pigments Synthesized by Marine Bacteria. Microorganisms 2020, 9, 11. [Google Scholar] [CrossRef]
  157. Lin, L.; Xu, J. Production of Fungal Pigments: Molecular Processes and Their Applications. J. Fungi 2022, 9, 44. [Google Scholar] [CrossRef]
  158. Huang, T.; Wang, M.; Shi, K.; Chen, G.; Tian, X.; Wu, Z. Metabolism and Secretion of Yellow Pigment under High Glucose Stress with Monascus ruber. AMB Expr. 2017, 7, 79. [Google Scholar] [CrossRef] [PubMed]
  159. Chen, G.; Yang, S.; Wang, C.; Shi, K.; Zhao, X.; Wu, Z. Investigation of the Mycelial Morphology of Monascus and the Expression of Pigment Biosynthetic Genes in High-Salt-Stress Fermentation. Appl. Microbiol. Biotechnol. 2020, 104, 2469–2479. [Google Scholar] [CrossRef]
  160. Garcia-Cortes, A.; Garcia-Vásquez, J.A.; Aranguren, Y.; Ramirez-Castrillon, M. Pigment Production Improvement in Rhodotorula mucilaginosa AJB01 Using Design of Experiments. Microorganisms 2021, 9, 387. [Google Scholar] [CrossRef]
  161. Carvalho, J.C.D.; Oishi, B.O.; Pandey, A.; Soccol, C.R. Biopigments from Monascus: Strains Selection, Citrinin Production and Color Stability. Braz. Arch. Biol. Technol. 2005, 48, 885–894. [Google Scholar] [CrossRef]
  162. Li, Y.-P.; Pan, Y.-F.; Zou, L.-H.; Xu, Y.; Huang, Z.-B.; He, Q.-H. Lower Citrinin Production by Gene Disruption of ctnB Involved in Citrinin Biosynthesis in Monascus aurantiacus Li AS3.4384. J. Agric. Food Chem. 2013, 61, 7397–7402. [Google Scholar] [CrossRef] [PubMed]
  163. Hölker, U.; Höfer, M.; Lenz, J. Biotechnological Advantages of Laboratory-Scale Solid-State Fermentation with Fungi. Appl. Microbiol. Biotechnol. 2004, 64, 175–186. [Google Scholar] [CrossRef]
  164. Hashempour-Baltork, F.; Hosseini, S.M.; Assarehzadegan, M.; Khosravi-Darani, K.; Hosseini, H. Safety Assays and Nutritional Values of Mycoprotein Produced by Fusarium venenatum IR372C from Date Waste as Substrate. J. Sci. Food Agric. 2020, 100, 4433–4441. [Google Scholar] [CrossRef] [PubMed]
  165. Liang, B.; Du, X.-J.; Li, P.; Sun, C.-C.; Wang, S. Investigation of Citrinin and Pigment Biosynthesis Mechanisms in Monascus purpureus by Transcriptomic Analysis. Front. Microbiol. 2018, 9, 1374. [Google Scholar] [CrossRef]
  166. Liu, W.; An, C.; Shu, X.; Meng, X.; Yao, Y.; Zhang, J.; Chen, F.; Xiang, H.; Yang, S.; Gao, X.; et al. A Dual-Plasmid CRISPR/Cas System for Mycotoxin Elimination in Polykaryotic Industrial Fungi. ACS Synth. Biol. 2020, 9, 2087–2095. [Google Scholar] [CrossRef]
  167. Silva, T.R.E.; Silva, L.C.F.; De Queiroz, A.C.; Alexandre Moreira, M.S.; De Carvalho Fraga, C.A.; De Menezes, G.C.A.; Rosa, L.H.; Bicas, J.; De Oliveira, V.M.; Duarte, A.W.F. Pigments from Antarctic Bacteria and Their Biotechnological Applications. Crit. Rev. Biotechnol. 2021, 41, 809–826. [Google Scholar] [CrossRef]
  168. Cerda, A.; Artola, A.; Barrena, R.; Font, X.; Gea, T.; Sánchez, A. Innovative Production of Bioproducts From Organic Waste Through Solid-State Fermentation. Front. Sustain. Food Syst. 2019, 3, 63. [Google Scholar] [CrossRef]
  169. Chmelová, D.; Legerská, B.; Kunstová, J.; Ondrejovič, M.; Miertuš, S. The Production of Laccases by White-Rot Fungi under Solid-State Fermentation Conditions. World J. Microbiol. Biotechnol. 2022, 38, 21. [Google Scholar] [CrossRef]
  170. Egbune, E.O.; Ezedom, T.; Orororo, O.C.; Egbune, O.U.; Avwioroko, O.J.; Aganbi, E.; Anigboro, A.A.; Tonukari, N.J. Solid-State Fermentation of Cassava (Manihot Esculenta Crantz): A Review. World J. Microbiol. Biotechnol. 2023, 39, 259. [Google Scholar] [CrossRef]
  171. Wang, Y.; Yang, G.; Sage, V.; Xu, J.; Sun, G.; He, J.; Sun, Y. Optimization of Dark Fermentation for Biohydrogen Production Using a Hybrid Artificial Neural Network (ANN) and Response Surface Methodology (RSM) Approach. Environ. Prog. Sustain. Energy 2021, 40, e13485. [Google Scholar] [CrossRef]
  172. Venkatachalam, M.; Shum-Chéong-Sing, A.; Dufossé, L.; Fouillaud, M. Statistical Optimization of the Physico-Chemical Parameters for Pigment Production in Submerged Fermentation of Talaromyces Albobiverticillius 30548. Microorganisms 2020, 8, 711. [Google Scholar] [CrossRef] [PubMed]
  173. Kumar, G.; Shekh, A.; Jakhu, S.; Sharma, Y.; Kapoor, R.; Sharma, T.R. Bioengineering of Microalgae: Recent Advances, Perspectives, and Regulatory Challenges for Industrial Application. Front. Bioeng. Biotechnol. 2020, 8, 914. [Google Scholar] [CrossRef]
  174. Nigam, P.S.; Luke, J.S. Food Additives: Production of Microbial Pigments and Their Antioxidant Properties. Curr. Opin. Food Sci. 2016, 7, 93–100. [Google Scholar] [CrossRef]
  175. Uyar, B.; Ali, M.D.; Uyar, G.E.O. Design Parameters Comparison of Bubble Column, Airlift and Stirred Tank Photobioreactors for Microalgae Production. Bioprocess. Biosyst. Eng. 2024, 47, 195–209. [Google Scholar] [CrossRef]
  176. Yang, J.; Lu, C.; Stasny, B.; Henley, J.; Guinto, W.; Gonzalez, C.; Gleason, J.; Fung, M.; Collopy, B.; Benjamino, M.; et al. Fed-batch Bioreactor Process Scale-up from 3-L to 2,500-L Scale for Monoclonal Antibody Production from Cell Culture. Biotechnol. Bioeng. 2007, 98, 141–154. [Google Scholar] [CrossRef]
  177. Hajjaj, H.; Blanc, P.; Groussac, E.; Uribelarrea, J.-L.; Goma, G.; Loubiere, P. Kinetic Analysis of Red Pigment and Citrinin Production by Monascus ruber as a Function of Organic Acid Accumulation. Enzym. Microb. Technol. 2000, 27, 619–625. [Google Scholar] [CrossRef]
  178. De Carvalho, J.C.; Cardoso, L.C.; Ghiggi, V.; Woiciechowski, A.L.; De Souza Vandenberghe, L.P.; Soccol, C.R. Microbial Pigments. In Biotransformation of Waste Biomass into High Value Biochemicals; Brar, S.K., Dhillon, G.S., Soccol, C.R., Eds.; Springer: New York, NY, USA, 2014; pp. 73–97. ISBN 978-1-4614-8004-4. [Google Scholar]
  179. Ghosh, A.; Goyal, A.; Jain, R.K. Study of Methanol-Induced Phenotypic Changes in a Novel Strain of Acinetobacter lwoffii. Arch. Microbiol. 2007, 188, 533–539. [Google Scholar] [CrossRef]
  180. Robledo, J.A.; Murillo, A.M.; Rouzaud, F. Physiological Role and Potential Clinical Interest of Mycobacterial Pigments. IUBMB Life 2011, 63, 71–78. [Google Scholar] [CrossRef]
  181. Maeda, H.; Dudareva, N. The Shikimate Pathway and Aromatic Amino Acid Biosynthesis in Plants. Annu. Rev. Plant Biol. 2012, 63, 73–105. [Google Scholar] [CrossRef]
  182. Zhang, X.; Heinonen, S.; Levänen, E. Applications of Supercritical Carbon Dioxide in Materials Processing and Synthesis. RSC Adv. 2014, 4, 61137–61152. [Google Scholar] [CrossRef]
  183. Fabrowska, J.; Messyasz, B.; Szyling, J.; Walkowiak, J.; Łęska, B. Isolation of Chlorophylls and Carotenoids from Freshwater Algae Using Different Extraction Methods. Phycol. Res. 2018, 66, 52–57. [Google Scholar] [CrossRef]
  184. Venil, C.K.; Dufossé, L.; Renuka Devi, P. Bacterial Pigments: Sustainable Compounds with Market Potential for Pharma and Food Industry. Front. Sustain. Food Syst. 2020, 4, 100. [Google Scholar] [CrossRef]
  185. Da Silva, A.F.; Moreira, A.F.; Miguel, S.P.; Coutinho, P. Recent Advances in Microalgae Encapsulation Techniques for Biomedical Applications. Adv. Colloid Interface Sci. 2024, 333, 103297. [Google Scholar] [CrossRef] [PubMed]
  186. Dinu, V.; Kilic, A.; Wang, Q.; Ayed, C.; Fadel, A.; Harding, S.E.; Yakubov, G.E.; Fisk, I.D. Policy, Toxicology and Physicochemical Considerations on the Inhalation of High Concentrations of Food Flavour. Npj Sci. Food 2020, 4, 15. [Google Scholar] [CrossRef]
  187. Zhang, B.; Lin, J.; Perčulija, V.; Li, Y.; Lu, Q.; Chen, J.; Ouyang, S. Structural Insights into Target DNA Recognition and Cleavage by the CRISPR-Cas12c1 System. Nucleic Acids Res. 2022, 50, 11820–11833. [Google Scholar] [CrossRef]
  188. Heider, S.A.E.; Wolf, N.; Hofemeier, A.; Peters-Wendisch, P.; Wendisch, V.F. Optimization of the IPP Precursor Supply for the Production of Lycopene, Decaprenoxanthin and Astaxanthin by Corynebacterium glutamicum. Front. Bioeng. Biotechnol. 2014, 2, 28. [Google Scholar] [CrossRef]
  189. Shu, C.; Zhang, S.; Wu, S.; Liu, S.; Xu, J.; Zhao, J.; Li, B. Microorganism-Mediated Production of Anthocyanins: Current Progress and Future Prospects. Food Res. Int. 2025, 201, 115550. [Google Scholar] [CrossRef]
  190. Yan, Y.; Chemler, J.; Huang, L.; Martens, S.; Koffas, M.A.G. Metabolic Engineering of Anthocyanin Biosynthesis in Escherichia coli. Appl. Environ. Microbiol. 2005, 71, 3617–3623. [Google Scholar] [CrossRef]
  191. Zha, J.; Zang, Y.; Mattozzi, M.; Plassmeier, J.; Gupta, M.; Wu, X.; Clarkson, S.; Koffas, M.A.G. Metabolic Engineering of Corynebacterium Glutamicum for Anthocyanin Production. Microb. Cell Fact. 2018, 17, 143. [Google Scholar] [CrossRef]
  192. Jones, J.A.; Vernacchio, V.R.; Collins, S.M.; Shirke, A.N.; Xiu, Y.; Englaender, J.A.; Cress, B.F.; McCutcheon, C.C.; Linhardt, R.J.; Gross, R.A.; et al. Complete Biosynthesis of Anthocyanins Using E. coli Polycultures. mBio 2017, 8, e00621-17. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, X.-K.; Wang, D.-N.; Chen, J.; Liu, Z.-J.; Wei, L.-J.; Hua, Q. Metabolic Engineering of β-Carotene Biosynthesis in Yarrowia lipolytica. Biotechnol. Lett. 2020, 42, 945–956. [Google Scholar] [CrossRef]
  194. Ahmad, A.; Jamil, A.; Munawar, N. GMOs or Non-GMOs? The CRISPR Conundrum. Front. Plant Sci. 2023, 14, 1232938. [Google Scholar] [CrossRef] [PubMed]
  195. Ravi, R.; Janodia, M.D. Factors Affecting Technology Transfer and Commercialization of University Research in India: A Cross-Sectional Study. J. Knowl. Econ. 2022, 13, 787–803. [Google Scholar] [CrossRef]
  196. Flammini, S.; Arcese, G.; Lucchetti, M.C.; Mortara, L. Business Model Configuration and Dynamics for Technology Commercialization in Mature Markets. Br. Food J. 2017, 119, 2340–2358. [Google Scholar] [CrossRef]
  197. Mendoza-Moheno, J.; Cruz-Coria, E.; González-Cruz, T.F. Socio-Technical Innovation in Community-Based Tourism Organizations: A Proposal for Local Development. Technol. Forecast. Social. Change 2021, 171, 120949. [Google Scholar] [CrossRef]
  198. Zhou, P.; Xie, W.; Yao, Z.; Zhu, Y.; Ye, L.; Yu, H. Development of a Temperature-responsive Yeast Cell Factory Using Engineered Gal4 as a Protein Switch. Biotechnol. Bioeng. 2018, 115, 1321–1330. [Google Scholar] [CrossRef]
  199. Kim, S.-W.; Kim, J.-B.; Jung, W.-H.; Kim, J.-H.; Jung, J.-K. Over-Production of β-Carotene from Metabolically Engineered Escherichia Coli. Biotechnol. Lett. 2006, 28, 897–904. [Google Scholar] [CrossRef]
  200. Xu, X.; Chu, X.; Du, B.; Huang, C.; Xie, C.; Zhang, Z.; Jiang, L. Functional Characterization of a Novel Violacein Biosynthesis Operon from Janthinobacterium Sp. B9–8. Appl. Microbiol. Biotechnol. 2022, 106, 2903–2916. [Google Scholar] [CrossRef]
  201. Zhu, H.-Z.; Jiang, S.; Wu, J.-J.; Zhou, X.-R.; Liu, P.-Y.; Huang, F.-H.; Wan, X. Production of High Levels of 3 S,3′ S -Astaxanthin in Yarrowia lipolytica via Iterative Metabolic Engineering. J. Agric. Food Chem. 2022, 70, 2673–2683. [Google Scholar] [CrossRef]
  202. Tong, Y.; Zhou, J.; Zhang, L.; Xu, P. A Golden-Gate Based Cloning Toolkit to Build Violacein Pathway Libraries in Yarrowia lipolytica. ACS Synth. Biol. 2021, 10, 115–124. [Google Scholar] [CrossRef]
  203. Qi, D.-D.; Jin, J.; Liu, D.; Jia, B.; Yuan, Y.-J. In Vitro and in Vivo Recombination of Heterologous Modules for Improving Biosynthesis of Astaxanthin in Yeast. Microb. Cell Fact. 2020, 19, 103. [Google Scholar] [CrossRef]
  204. Verwaal, R.; Wang, J.; Meijnen, J.-P.; Visser, H.; Sandmann, G.; Van Den Berg, J.A.; Van Ooyen, A.J.J. High-Level Production of Beta-Carotene in Saccharomyces cerevisiae by Successive Transformation with Carotenogenic Genes from Xanthophyllomyces dendrorhous. Appl. Environ. Microbiol. 2007, 73, 4342–4350. [Google Scholar] [CrossRef] [PubMed]
  205. Kang, W.; Ma, T.; Liu, M.; Qu, J.; Liu, Z.; Zhang, H.; Shi, B.; Fu, S.; Ma, J.; Lai, L.T.F.; et al. Modular Enzyme Assembly for Enhanced Cascade Biocatalysis and Metabolic Flux. Nat. Commun. 2019, 10, 4248. [Google Scholar] [CrossRef] [PubMed]
  206. Hong, J.; Park, S.-H.; Kim, S.; Kim, S.-W.; Hahn, J.-S. Efficient Production of Lycopene in Saccharomyces cerevisiae by Enzyme Engineering and Increasing Membrane Flexibility and NAPDH Production. Appl. Microbiol. Biotechnol. 2019, 103, 211–223. [Google Scholar] [CrossRef] [PubMed]
  207. Fathi, Z.; Tramontin, L.R.R.; Ebrahimipour, G.; Borodina, I.; Darvishi, F. Metabolic Engineering of Saccharomyces cerevisiae for Production of β-Carotene from Hydrophobic Substrates. FEMS Yeast Res. 2021, 21, foaa068. [Google Scholar] [CrossRef]
  208. Sun, L.; Atkinson, C.A.; Lee, Y.; Jin, Y. High-level Β-carotene Production from Xylose by Engineered Saccharomyces cerevisiae without Overexpression of a Truncated HMG1 (t HMG1). Biotechnol. Bioeng. 2020, 117, 3522–3532. [Google Scholar] [CrossRef]
  209. Ferreira, R.L.; Rezende, G.S.; Damas, M.S.F.; Oliveira-Silva, M.; Pitondo-Silva, A.; Brito, M.C.A.; Leonardecz, E.; Góes, F.R.D.; Campanini, E.B.; Malavazi, I.; et al. Characterization of KPC-Producing Serratia marcescens in an Intensive Care Unit of a Brazilian Tertiary Hospital. Front. Microbiol. 2020, 11, 956. [Google Scholar] [CrossRef]
  210. Khayyat, A.N.; Hegazy, W.A.H.; Shaldam, M.A.; Mosbah, R.; Almalki, A.J.; Ibrahim, T.S.; Khayat, M.T.; Khafagy, E.-S.; Soliman, W.E.; Abbas, H.A. Xylitol Inhibits Growth and Blocks Virulence in Serratia marcescens. Microorganisms 2021, 9, 1083. [Google Scholar] [CrossRef]
  211. Tavares-Carreon, F.; De Anda-Mora, K.; Rojas-Barrera, I.C.; Andrade, A. Serratia marcescens Antibiotic Resistance Mechanisms of an Opportunistic Pathogen: A Literature Review. PeerJ 2023, 11, e14399. [Google Scholar] [CrossRef]
  212. Hamilton, J.J.; Marlow, V.L.; Owen, R.A.; Costa, M.D.A.A.; Guo, M.; Buchanan, G.; Chandra, G.; Trost, M.; Coulthurst, S.J.; Palmer, T.; et al. A Holin and an Endopeptidase Are Essential for Chitinolytic Protein Secretion in Serratia marcescens. J. Cell Biol. 2014, 207, 615–626. [Google Scholar] [CrossRef] [PubMed]
  213. Mudaliar, S.B.; Bharath Prasad, A.S. A Biomedical Perspective of Pyocyanin from Pseudomonas aeruginosa: Its Applications and Challenges. World J. Microbiol. Biotechnol. 2024, 40, 90. [Google Scholar] [CrossRef] [PubMed]
  214. Lau, G.W.; Ran, H.; Kong, F.; Hassett, D.J.; Mavrodi, D. Pseudomonas aeruginosa Pyocyanin Is Critical for Lung Infection in Mice. Infect. Immun. 2004, 72, 4275–4278. [Google Scholar] [CrossRef]
  215. Ran, H.; Hassett, D.J.; Lau, G.W. Human Targets of Pseudomonas aeruginosa Pyocyanin. Proc. Natl. Acad. Sci. USA 2003, 100, 14315–14320. [Google Scholar] [CrossRef]
  216. Venkatramanan, M.; Nalini, E. Regulation of Virulence in Chromobacterium violaceum and Strategies to Combat It. Front. Microbiol. 2024, 15, 1303595. [Google Scholar] [CrossRef]
  217. Oh, W.T.; Giri, S.S.; Yun, S.; Kim, H.J.; Kim, S.G.; Kim, S.W.; Kang, J.W.; Han, S.J.; Kwon, J.; Jun, J.W.; et al. Janthinobacterium lividum as An Emerging Pathogenic Bacterium Affecting Rainbow Trout (Oncorhynchus Mykiss) Fisheries in Korea. Pathogens 2019, 8, 146. [Google Scholar] [CrossRef] [PubMed]
  218. Narsing Rao, M.P.; Xiao, M.; Li, W.-J. Fungal and Bacterial Pigments: Secondary Metabolites with Wide Applications. Front. Microbiol. 2017, 8, 1113. [Google Scholar] [CrossRef]
  219. Gong, P.; Shi, R.; Tang, J.; Wang, J.; Luo, Q.; Zhang, J.; Ruan, X.; Wang, C.; Chen, W. Effect of Exogenous and Endogenous Ectoine on Monascus Development, Metabolism, and Pigment Stability. Foods 2023, 12, 3217. [Google Scholar] [CrossRef]
  220. Wang, J.-J.; Lee, C.-L.; Pan, T.-M. Modified Mutation Method for Screening Low Citrinin-Producing Strains of Monascus purpureus on Rice Culture. J. Agric. Food Chem. 2004, 52, 6977–6982. [Google Scholar] [CrossRef]
  221. Reverberi, M.; Ricelli, A.; Zjalic, S.; Fabbri, A.A.; Fanelli, C. Natural Functions of Mycotoxins and Control of Their Biosynthesis in Fungi. Appl. Microbiol. Biotechnol. 2010, 87, 899–911. [Google Scholar] [CrossRef]
  222. Nikitin, D.A.; Ivanova, E.A.; Semenov, M.V.; Zhelezova, A.D.; Ksenofontova, N.A.; Tkhakakhova, A.K.; Kholodov, V.A. Diversity, Ecological Characteristics and Identification of Some Problematic Phytopathogenic Fusarium in Soil: A Review. Diversity 2023, 15, 49. [Google Scholar] [CrossRef]
  223. Zain, M.; Yasmin, S.; Hafeez, F. Isolation and Characterization of Plant Growth Promoting Antagonistic Bacteria from Cotton and Sugarcane Plants for Suppression of Phytopathogenic Fusarium Species. Iran. J. Biotechnol. 2019, 17, 61–70. [Google Scholar] [CrossRef]
  224. Gong, Z.; Zhang, S.; Liu, J. Recent Advances in Chitin Biosynthesis Associated with the Morphology and Secondary Metabolite Synthesis of Filamentous Fungi in Submerged Fermentation. J. Fungi 2023, 9, 205. [Google Scholar] [CrossRef]
  225. Lu, Z.; Chen, Z.; Liu, Y.; Hua, X.; Gao, C.; Liu, J. Morphological Engineering of Filamentous Fungi: Research Progress and Perspectives. J. Microbiol. Biotechnol. 2024, 34, 1197–1205. [Google Scholar] [CrossRef]
  226. Méndez, A.; Pérez, C.; Montañéz, J.C.; Martínez, G.; Aguilar, C.N. Red Pigment Production by Penicillium purpurogenum GH2 Is Influenced by pH and Temperature. J. Zhejiang Univ. Sci. B 2011, 12, 961–968. [Google Scholar] [CrossRef] [PubMed]
  227. Borowitzka, M.A. High-Value Products from Microalgae—Their Development and Commercialisation. J. Appl. Phycol. 2013, 25, 743–756. [Google Scholar] [CrossRef]
  228. Dębowski, M.; Świca, I.; Kazimierowicz, J.; Zieliński, M. Large Scale Microalgae Biofuel Technology—Development Perspectives in Light of the Barriers and Limitations. Energies 2022, 16, 81. [Google Scholar] [CrossRef]
  229. Uduman, N.; Qi, Y.; Danquah, M.K.; Forde, G.M.; Hoadley, A. Dewatering of Microalgal Cultures: A Major Bottleneck to Algae-Based Fuels. J. Renew. Sustain. Energy 2010, 2, 012701. [Google Scholar] [CrossRef]
  230. De Souza, M.P.; Hoeltz, M.; Gressler, P.D.; Benitez, L.B.; Schneider, R.C.S. Potential of Microalgal Bioproducts: General Perspectives and Main Challenges. Waste Biomass Valorization 2019, 10, 2139–2156. [Google Scholar] [CrossRef]
  231. Diaconu, M.; Soreanu, G.; Balan, C.D.; Buciscanu, I.I.; Maier, V.; Cretescu, I. Study of Spirulina platensis (Arthrospira) Development under the Heavy Metals Influence, as a Potential Promoter of Wastewater Remediation. Water 2023, 15, 3962. [Google Scholar] [CrossRef]
  232. Spicer, A.; Molnar, A. Gene Editing of Microalgae: Scientific Progress and Regulatory Challenges in Europe. Biology 2018, 7, 21. [Google Scholar] [CrossRef] [PubMed]
  233. Dufossé, L. Back to Nature, Microbial Production of Pigments and Colorants for Food Use. In Advances in Food and Nutrition Research; Elsevier: Amsterdam, The Netherlands, 2022; Volume 102, pp. 93–122. ISBN 978-0-323-99084-4. [Google Scholar]
  234. Patakova, P. Monascus Secondary Metabolites: Production and Biological Activity. J. Ind. Microbiol. Biotechnol. 2013, 40, 169–181. [Google Scholar] [CrossRef]
  235. Vendruscolo, F.; Bühler, R.M.M.; De Carvalho, J.C.; De Oliveira, D.; Moritz, D.E.; Schmidell, W.; Ninow, J.L. Monascus: A Reality on the Production and Application of Microbial Pigments. Appl. Biochem. Biotechnol. 2016, 178, 211–223. [Google Scholar] [CrossRef]
  236. Zhang, Y.; Navarro, E.; Cánovas-Márquez, J.T.; Almagro, L.; Chen, H.; Chen, Y.Q.; Zhang, H.; Torres-Martínez, S.; Chen, W.; Garre, V. A New Regulatory Mechanism Controlling Carotenogenesis in the Fungus Mucor circinelloides as a Target to Generate β-Carotene over-Producing Strains by Genetic Engineering. Microb. Cell Fact. 2016, 15, 99. [Google Scholar] [CrossRef]
  237. Mak, S.; Nodwell, J.R. Actinorhodin Is a Redox-active Antibiotic with a Complex Mode of Action against Gram-positive Cells. Mol. Microbiol. 2017, 106, 597–613. [Google Scholar] [CrossRef] [PubMed]
  238. De Oliveira, F.; Pedrolli, D.B.; Teixeira, M.F.S.; De Carvalho Santos-Ebinuma, V. Water-Soluble Fluorescent Red Colorant Production by Talaromyces amestolkiae. Appl. Microbiol. Biotechnol. 2019, 103, 6529–6541. [Google Scholar] [CrossRef]
  239. Arivizhivendhan, K.V.; Mahesh, M.; Boopathy, R.; Swarnalatha, S.; Regina Mary, R.; Sekaran, G. Antioxidant and Antimicrobial Activity of Bioactive Prodigiosin Produces from Serratia marcescens Using Agricultural Waste as a Substrate. J. Food Sci. Technol. 2018, 55, 2661–2670. [Google Scholar] [CrossRef] [PubMed]
  240. Eghbaljoo, H.; Alizadeh Sani, M.; Sani, I.K.; Maragheh, S.M.; Sain, D.K.; Jawhar, Z.H.; Pirsa, S.; Kadi, A.; Dadkhodayi, R.; Zhang, F.; et al. Development of Smart Packaging Halochromic Films Embedded with Anthocyanin Pigments; Recent Advances. Crit. Rev. Food Sci. Nutr. 2025, 65, 770–786. [Google Scholar] [CrossRef]
  241. Nupur, L.N.U.; Vats, A.; Dhanda, S.K.; Raghava, G.P.S.; Pinnaka, A.K.; Kumar, A. ProCarDB: A Database of Bacterial Carotenoids. BMC Microbiol. 2016, 16, 96. [Google Scholar] [CrossRef]
  242. Arivuselvam, R.; Dera, A.A.; Parween Ali, S.; Alraey, Y.; Saif, A.; Hani, U.; Arumugam Ramakrishnan, S.; Azeeze, M.S.T.A.; Rajeshkumar, R.; Susil, A.; et al. Isolation, Identification, and Antibacterial Properties of Prodigiosin, a Bioactive Product Produced by a New Serratia marcescens JSSCPM1 Strain: Exploring the Biosynthetic Gene Clusters of Serratia Species for Biological Applications. Antibiotics 2023, 12, 1466. [Google Scholar] [CrossRef]
  243. Darshan, N.; Manonmani, H.K. Prodigiosin Inhibits Motility and Activates Bacterial Cell Death Revealing Molecular Biomarkers of Programmed Cell Death. AMB Expr. 2016, 6, 50. [Google Scholar] [CrossRef] [PubMed]
  244. Vendruscolo, F.; Tosin, I.; Giachini, A.J.; Schmidell, W.; Ninow, J.L. Antimicrobial Activity of M. onascus Pigments Produced in Submerged Fermentation: Antimicrobial Activity of Monascus Pigments. J. Food Process. Preserv. 2014, 38, 1860–1865. [Google Scholar] [CrossRef]
  245. Zhao, G.-P.; Li, Y.-Q.; Yang, J.; Cui, K.-Y. Antibacterial Characteristics of Orange Pigment Extracted from Monascus pigments against Escherichia coli. Czech J. Food Sci. 2016, 34, 197–203. [Google Scholar] [CrossRef]
  246. Lu, W.; Shi, Y.; Wang, R.; Su, D.; Tang, M.; Liu, Y.; Li, Z. Antioxidant Activity and Healthy Benefits of Natural Pigments in Fruits: A Review. Int. J. Mol. Sci. 2021, 22, 4945. [Google Scholar] [CrossRef] [PubMed]
  247. Igielska-Kalwat, J.; Gościańska, J.; Nowak, I. Carotenoids as Natural Antioxidants. Postep. Hig. Med. Dosw. 2015, 69, 418–428. [Google Scholar] [CrossRef] [PubMed]
  248. Brotosudarmo, T.H.P.; Limantara, L.; Setiyono, E.; Heriyanto. Structures of Astaxanthin and Their Consequences for Therapeutic Application. Int. J. Food Sci. 2020, 2020, 2156582. [Google Scholar] [CrossRef]
  249. Li, C.; Cheng, P.; Li, Z.; Xu, Y.; Sun, Y.; Qin, D.; Yu, G. Transcriptomic and Metabolomic Analyses Provide Insights into the Enhancement of Torulene and Torularhodin Production in Rhodotorula glutinis ZHK under Moderate Salt Conditions. J. Agric. Food Chem. 2021, 69, 11523–11533. [Google Scholar] [CrossRef]
  250. Hamilton, A.J.; Holdom, M.D. Antioxidant Systems in the Pathogenic Fungi of Man and Their Role in Virulence. Med. Mycol. 1999, 37, 375–389. [Google Scholar] [CrossRef]
  251. Solano, F. Photoprotection and Skin Pigmentation: Melanin-Related Molecules and Some Other New Agents Obtained from Natural Sources. Molecules 2020, 25, 1537. [Google Scholar] [CrossRef]
  252. Oh, J.-J.; Kim, J.Y.; Son, S.H.; Jung, W.-J.; Kim, D.H.; Seo, J.-W.; Kim, G.-H. Fungal Melanin as a Biocompatible Broad-Spectrum Sunscreen with High Antioxidant Activity. RSC Adv. 2021, 11, 19682–19689. [Google Scholar] [CrossRef]
  253. Panathur, N.; Gokhale, N.; Dalimba, U.; Koushik, P.V.; Yogeeswari, P.; Sriram, D. New Indole–Isoxazolone Derivatives: Synthesis, Characterisation and in Vitro SIRT1 Inhibition Studies. Bioorganic Med. Chem. Lett. 2015, 25, 2768–2772. [Google Scholar] [CrossRef]
  254. Venegas, F.A.; Köllisch, G.; Mark, K.; Diederich, W.E.; Kaufmann, A.; Bauer, S.; Chavarría, M.; Araya, J.J.; García-Piñeres, A.J. The Bacterial Product Violacein Exerts an Immunostimulatory Effect Via TLR8. Sci. Rep. 2019, 9, 13661. [Google Scholar] [CrossRef]
  255. Zhang, W.; Hua, H.; Guo, Y.; Cheng, Y.; Pi, F.; Yao, W.; Xie, Y.; Qian, H. Torularhodin from Sporidiobolus pararoseus Attenuates d-Galactose/AlCl3 Induced Cognitive Impairment, Oxidative Stress, and Neuroinflammation via the Nrf2/NF-κB Pathway. J. Agric. Food Chem. 2020, 68, 6604–6614. [Google Scholar] [CrossRef] [PubMed]
  256. Corsi, F.; Deidda Tarquini, G.; Urbani, M.; Bejarano, I.; Traversa, E.; Ghibelli, L. The Impressive Anti-Inflammatory Activity of Cerium Oxide Nanoparticles: More than Redox? Nanomaterials 2023, 13, 2803. [Google Scholar] [CrossRef] [PubMed]
  257. Regourd, J.; Al-Sheikh Ali, A.; Thompson, A. Synthesis and Anti-Cancer Activity of C-Ring-Functionalized Prodigiosin Analogues. J. Med. Chem. 2007, 50, 1528–1536. [Google Scholar] [CrossRef]
  258. Zhang, J.; Shen, Y.; Liu, J.; Wei, D. Antimetastatic Effect of Prodigiosin through Inhibition of Tumor Invasion. Biochem. Pharmacol. 2005, 69, 407–414. [Google Scholar] [CrossRef]
  259. Zheng, Y.; Xin, Y.; Shi, X.; Guo, Y. Cytotoxicity of Monascus Pigments and Their Derivatives to Human Cancer Cells. J. Agric. Food Chem. 2010, 58, 9523–9528. [Google Scholar] [CrossRef] [PubMed]
  260. Reyes, F.G.R.; Valim, M.F.C.F.A.; Vercesi, A.E. Effect of Organic Synthetic Food Colours on Mitochondrial Respiration. Food Addit. Contam. 1996, 13, 5–11. [Google Scholar] [CrossRef]
  261. Di Mascio, P.; Kaiser, S.; Sies, H. Lycopene as the Most Efficient Biological Carotenoid Singlet Oxygen Quencher. Arch. Biochem. Biophys. 1989, 274, 532–538. [Google Scholar] [CrossRef]
  262. Chen, C.-Y.; Jesisca; Hsieh, C.; Lee, D.-J.; Chang, C.-H.; Chang, J.-S. Production, Extraction and Stabilization of Lutein from Microalga Chlorella sorokiniana MB-1. Bioresour. Technol. 2016, 200, 500–505. [Google Scholar] [CrossRef]
  263. Ventura Pinto, A.; Lisboa De Castro, S. The Trypanocidal Activity of Naphthoquinones: A Review. Molecules 2009, 14, 4570–4590. [Google Scholar] [CrossRef]
  264. Fuhrman, B.; Elis, A.; Aviram, M. Hypocholesterolemic Effect of Lycopene and β-Carotene Is Related to Suppression of Cholesterol Synthesis and Augmentation of LDL Receptor Activity in Macrophages. Biochem. Biophys. Res. Commun. 1997, 233, 658–662. [Google Scholar] [CrossRef] [PubMed]
  265. Jeong, S.W.; Yang, J.E.; Choi, Y.J. Isolation and Characterization of a Yellow Xanthophyll Pigment-Producing Marine Bacterium, Erythrobacter Sp. SDW2 Strain, in Coastal Seawater. Mar. Drugs 2022, 20, 73. [Google Scholar] [CrossRef] [PubMed]
  266. Aruldass, C.A.; Dufossé, L.; Ahmad, W.A. Current Perspective of Yellowish-Orange Pigments from Microorganisms- a Review. J. Clean. Prod. 2018, 180, 168–182. [Google Scholar] [CrossRef]
  267. Sikkandar, S.; Murugan, K.; Al-Sohaibani, S.; Rayappan, F.; Nair, A.; Tilton, F. Halophilic Bacteria-A Potent Source of Carotenoids with Antioxidant and Anticancer Potentials. J. Pure Appl. Microbiol. 2013, 7, 2825–2830. [Google Scholar]
  268. Suresh, R.; Uma, G.; Gokulakrishnan, R. Antibacterial Property of Halobacterial Carotenoids against Human Bacterial Pathogens. J. Sci. Ind. Res. 2016, 75, 253–257. [Google Scholar]
  269. Rezaeeyan, Z.; Safarpour, A.; Amoozegar, M.A.; Babavalian, H.; Tebyanian; Shakeri, F. High Carotenoid Production by a Halotolerant Bacterium, Kocuria Sp. Strain QWT-12 and Anticancer Activity of Its Carotenoid. EXCLI J. 2017, 16, 840–851. [Google Scholar] [CrossRef]
  270. Megha, S.; Varalakshmi Kilingar, N. Yellow Pigment from a Novel Bacteria, Micrococcus terreus, Activates Caspases and Leads to Apoptosis of Cervical and Liver Cancer Cell Lines. J. Appl. Pharm. Sci. 2021, 11, 77–84. [Google Scholar] [CrossRef]
  271. Hsu, L.-C.; Hsu, Y.-W.; Liang, Y.-H.; Kuo, Y.-H.; Pan, T.-M. Anti-Tumor and Anti-Inflammatory Properties of Ankaflavin and Monaphilone A from Monascus purpureus NTU 568. J. Agric. Food Chem. 2011, 59, 1124–1130. [Google Scholar] [CrossRef]
  272. Malik, K.; Tokkas, J.; Goyal, S. Microbial Pigments: A Review. Int. J. Microb. Resour. Technol. 2012, 1, 361–365. [Google Scholar]
  273. Tsubokura, A.; Yoneda, H.; Mizuta, H. Paracoccus Carotinifaciens Sp. Nov., a New Aerobic Gram-Negative Astaxanthin-Producing Bacterium. Int. J. Syst. Evol. Microbiol. 1999, 49, 277–282. [Google Scholar] [CrossRef] [PubMed]
  274. Correa-Llantén, D.N.; Amenábar, M.J.; Blamey, J.M. Antioxidant Capacity of Novel Pigments from an Antarctic Bacterium. J. Microbiol. 2012, 50, 374–379. [Google Scholar] [CrossRef]
  275. Ramírez, J.; Nuñez, M.L.; Valdivia, R. Increased Astaxanthin Production by a Phaffia rhodozyma Mutant Grown on Date Juice from Yucca fillifera. J. Ind. Microbiol. Biotechnol. 2000, 24, 187–190. [Google Scholar] [CrossRef]
  276. Sonani, R.R.; Singh, N.K.; Kumar, J.; Thakar, D.; Madamwar, D. Concurrent Purification and Antioxidant Activity of Phycobiliproteins from Lyngbya Sp. A09DM: An Antioxidant and Anti-Aging Potential of Phycoerythrin in Caenorhabditis elegans. Process Biochem. 2014, 49, 1757–1766. [Google Scholar] [CrossRef]
  277. Melvin, M.S.; Tomlinson, J.T.; Saluta, G.R.; Kucera, G.L.; Lindquist, N.; Manderville, R.A. Double-Strand DNA Cleavage by Copper·Prodigiosin. J. Am. Chem. Soc. 2000, 122, 6333–6334. [Google Scholar] [CrossRef]
  278. Wang, Y.; Nakajima, A.; Hosokawa, K.; Soliev, A.B.; Osaka, I.; Arakawa, R.; Enomoto, K. Cytotoxic Prodigiosin Family Pigments from Pseudoalteromonas Sp. 1020R Isolated from the Pacific Coast of Japan. Biosci. Biotechnol. Biochem. 2012, 76, 1229–1232. [Google Scholar] [CrossRef]
  279. Konzen, M.; De Marco, D.; Cordova, C.A.S.; Vieira, T.O.; Antônio, R.V.; Creczynski-Pasa, T.B. Antioxidant Properties of Violacein: Possible Relation on Its Biological Function. Bioorganic Med. Chem. 2006, 14, 8307–8313. [Google Scholar] [CrossRef]
  280. Antonisamy, P.; Ignacimuthu, S. Immunomodulatory, Analgesic and Antipyretic Effects of Violacein Isolated from Chromobacterium violaceum. Phytomedicine 2010, 17, 300–304. [Google Scholar] [CrossRef]
  281. Baron, S.S.; Rowe, J.J. Antibiotic Action of Pyocyanin. Antimicrob. Agents Chemother. 1981, 20, 814–820. [Google Scholar] [CrossRef]
  282. Wahyudi, A.T.; Nursari, R.; Purwaningtyas, W.E.; Cahlia, U.; Priyanto, J.A. Crude Extract of Blue-Green Pigment Derived from the Marine Bacterium Pseudomonas aeruginosa P1.S9 Has Antibacterial, Antioxidant and Cytotoxic Activities. OnLine J. Biol. Sci. 2022, 22, 118–125. [Google Scholar] [CrossRef]
  283. Asmae, Z.; Jaouad, A.; Jamal, G.; Khalid, S.; El Hassouni, M. Antioxidant and Antimicrobial Activities of Melanin Produced by a Pseudomonas balearica Strain. J. Biotechnol. Lett. 2014, 5, 87–94. [Google Scholar]
  284. Elsayed, Y.; Refaat, J.; R Abdelmohsen, U.; A Fouad, M. The Genus Rhodococcus as a Source of Novel Bioactive Substances: A Review. J. Pharmacogn. Phytochem. 2017, 6, 83–92. [Google Scholar]
  285. Xie, Z.-T.; Mi, B.-Q.; Lu, Y.-J.; Chen, M.-T.; Ye, Z.-W. Research Progress on Carotenoid Production by Rhodosporidium toruloides. Appl. Microbiol. Biotechnol. 2024, 108, 7. [Google Scholar] [CrossRef]
  286. Vinarov, A.; Robucheva, Z.; Sidorenko, T.; Dirina, E. Microbial Biosynthesis and Making of Pigment Melanin. Commun. Agric. Appl. Biol. Sci. 2003, 68, 325–326. [Google Scholar]
  287. Kavitha, R.; Aiswariya, S.; Ratnavali, C.M.G. Anticancer Activity of Red Pigment from Serratia marcescens in Human Cervix Carcinoma. Int. J. PharmTech Res. 2010, 2, 784–787. [Google Scholar]
  288. Clauditz, A.; Resch, A.; Wieland, K.-P.; Peschel, A.; Götz, F. Staphyloxanthin Plays a Role in the Fitness of Staphylococcus aureus and Its Ability to Cope with Oxidative Stress. Infect. Immun. 2006, 74, 4950–4953. [Google Scholar] [CrossRef]
  289. Balagurunathan, R.; Selvameenal, L.; Radhakrishnan, M. Antibiotic Pigment from Desert Soil Actinomycetes; Biological Activity, Purification and Chemical Screening. Indian. J. Pharm. Sci. 2009, 71, 499–504. [Google Scholar] [CrossRef]
  290. Darshan, N.; Manonmani, H.K. Prodigiosin and Its Potential Applications. J. Food Sci. Technol. 2015, 52, 5393–5407. [Google Scholar] [CrossRef]
  291. Sibero, M.T.; Bachtiarini, T.U.; Trianto, A.; Lupita, A.H.; Sari, D.P.; Igarashi, Y.; Harunari, E.; Sharma, A.R.; Radjasa, O.K.; Sabdono, A. Characterization of a Yellow Pigmented Coral-Associated Bacterium Exhibiting Anti-Bacterial Activity Against Multidrug Resistant (MDR) Organism. Egypt. J. Aquat. Res. 2019, 45, 81–87. [Google Scholar] [CrossRef]
  292. Rekha, R.; Nimsi, K.A.; Manjusha, K.; Sirajudheen, T.K. Marine Yeast Rhodotorula paludigena VA 242 a Pigment Enhancing Feed Additive for the Ornamental Fish Koi Carp. Aquac. Fish. 2024, 9, 66–70. [Google Scholar] [CrossRef]
  293. Vaidya, A.A.; O’Callahan, D.; Donaldson, L.; West, M.; Campion, S.; Singh, T. A Closed-Loop Circularity in Wood Sugar as a Renewable Carbon Source for Fungal Pigment Production and Application of Pigments in Wood Colouration. Bioresour. Technol. Rep. 2023, 24, 101648. [Google Scholar] [CrossRef]
  294. Shiva Krishna, P.; Sudha, S.; Reddy, K.A.; Al-Dhabaan, F.A.; Meher; Prakasham, R.S.; Singara Charya, M.A. Studies on Wound Healing Potential of Red Pigment Isolated from Marine Bacterium Vibrio Sp. Saudi J. Biol. Sci. 2019, 26, 723–729. [Google Scholar] [CrossRef]
  295. Suwannarach, N.; Kumla, J.; Nishizaki, Y.; Sugimoto, N.; Meerak, J.; Matsui, K.; Lumyong, S. Optimization and Characterization of Red Pigment Production from an Endophytic Fungus, Nigrospora aurantiaca CMU-ZY2045, and Its Potential Source of Natural Dye for Use in Textile Dyeing. Appl. Microbiol. Biotechnol. 2019, 103, 6973–6987. [Google Scholar] [CrossRef]
  296. Velmurugan, P.; Kamala-Kannan, S.; Balachandar, V.; Lakshmanaperumalsamy, P.; Chae, J.-C.; Oh, B.-T. Natural Pigment Extraction from Five Filamentous Fungi for Industrial Applications and Dyeing of Leather. Carbohydr. Polym. 2010, 79, 262–268. [Google Scholar] [CrossRef]
  297. Ben Tahar, I.; Kus-Liśkiewicz, M.; Lara, Y.; Javaux, E.; Fickers, P. Characterization of a Nontoxic Pyomelanin Pigment Produced by the Yeast Yarrowia lipolytica. Biotechnol. Prog. 2020, 36, 2912. [Google Scholar] [CrossRef]
  298. Metwally, R.A.; El Sikaily, A.; El-Sersy, N.A.; Ghozlan, H.A.; Sabry, S.A. Antimicrobial Activity of Textile Fabrics Dyed with Prodigiosin Pigment Extracted from Marine Serratia rubidaea RAM_Alex Bacteria. Egypt. J. Aquat. Res. 2021, 47, 301–305. [Google Scholar] [CrossRef]
  299. Rocha Balbino, T.; Sánchez-Muñoz, S.; Díaz-Ruíz, E.; Moura Rocha, T.; Mier-Alba, E.; Custódio Inácio, S.; Jose Castro-Alonso, M.; De Carvalho Santos-Ebinuma, V.; Fernando Brandão Pereira, J.; César Santos, J.; et al. Lignocellulosic Biorefineries as a Platform for the Production of High-Value Yeast Derived Pigments—A Review. Bioresour. Technol. 2023, 386, 129549. [Google Scholar] [CrossRef]
  300. Dulf, F.V.; Vodnar, D.C.; Dulf, E.-H. Solid-State Fermentation with Zygomycetes Fungi as a Tool for Biofortification of Apple Pomace with γ-Linolenic Acid, Carotenoid Pigments and Phenolic Antioxidants. Food Res. Int. 2023, 173, 113448. [Google Scholar] [CrossRef]
  301. Umesh, M.; Suresh, S.; Santosh, A.S.; Prasad, S.; Chinnathambi, A.; Al Obaid, S.; Jhanani, G.K.; Shanmugam, S. Valorization of Pineapple Peel Waste for Fungal Pigment Production Using Talaromyces albobiverticillius: Insights into Antibacterial, Antioxidant and Textile Dyeing Properties. Environ. Res. 2023, 229, 115973. [Google Scholar] [CrossRef]
  302. Koli, S.H.; Patil, S.V.; Mohite, B.V.; Otari, S.V. Microbial Pigment-Mediated Synthesis of Metal Nanoparticles. J. Clust. Sci. 2024, 35, 1625–1642. [Google Scholar] [CrossRef]
  303. El-Baz, A.F.; El-Batal, A.I.; Abomosalam, F.M.; Tayel, A.A.; Shetaia, Y.M.; Yang, S. Extracellular biosynthesis of anti-Candida silver nanoparticles using Monascus purpureus. J. Basic Microbiol. 2016, 56, 531–540. [Google Scholar] [CrossRef]
  304. Lian, Q.; Yao, L.; Uddin Ahmad, Z.; Gang, D.D.; Konggidinata, M.I.; Gallo, A.A.; Zappi, M.E. Enhanced Pb(II) Adsorption onto Functionalized Ordered Mesoporous Carbon (OMC) from Aqueous Solutions: The Important Role of Surface Property and Adsorption Mechanism. Environ. Sci. Pollut. Res. 2020, 27, 23616–23630. [Google Scholar] [CrossRef]
  305. Fu, H.; Liu, H.; Mao, J.; Chu, W.; Li, Q.; Alvarez, P.J.J.; Qu, X.; Zhu, D. Photochemistry of Dissolved Black Carbon Released from Biochar: Reactive Oxygen Species Generation and Phototransformation. Environ. Sci. Technol. 2016, 50, 1218–1226. [Google Scholar] [CrossRef]
  306. Chen, G.; Wu, Z. Production and Biological Activities of Yellow Pigments from Monascus Fungi. World J. Microbiol. Biotechnol. 2016, 32, 136. [Google Scholar] [CrossRef]
  307. Lin, G.; Zhao, C.; Liao, W.; Yang, J.; Zheng, Y. Eco-Friendly Green Synthesis of Rubropunctatin Functionalized Silver Nanoparticles and Evaluation of Antibacterial Activity. Nanomaterials 2022, 12, 4052. [Google Scholar] [CrossRef]
  308. Bhatnagar, S.; Kobori, T.; Ganesh, D.; Ogawa, K.; Aoyagi, H. Biosynthesis of Silver Nanoparticles Mediated by Extracellular Pigment from Talaromyces purpurogenus and Their Biomedical Applications. Nanomaterials 2019, 9, 1042. [Google Scholar] [CrossRef]
  309. Chavan, A.; Pawar, J.; Kakde, U. Fungal Pigment and Its Potential in Nanoparticle Synthesis, Characterization and Its Application. Adv. Biores. 2023, 14, 349–355. [Google Scholar] [CrossRef]
  310. Rahul, S.; Chandrashekhar, P.; Hemant, B.; Bipinchandra, S.; Mouray, E.; Grellier, P.; Satish, P. In Vitro Antiparasitic Activity of Microbial Pigments and Their Combination with Phytosynthesized Metal Nanoparticles. Parasitol. Int. 2015, 64, 353–356. [Google Scholar] [CrossRef]
  311. Manikprabhu, D.; Lingappa, K. Antibacterial Activity of Silver Nanoparticles against Methicillin-Resistant Staphylococcus aureus Synthesized Using Model Streptomyces Sp. Pigment by Photo-Irradiation Method. J. Pharm. Res. 2013, 6, 255–260. [Google Scholar] [CrossRef]
  312. Salunkhe, N.S.; Koli, S.H.; Mohite, B.V.; Patil, V.S.; Patil, S.V. Xanthomonadin Mediated Synthesis of Biocidal and Photo-Protective Silver Nanoparticles (XP-AgNPs). Results Chem. 2022, 4, 100663. [Google Scholar] [CrossRef]
  313. Baraka, A.; Dickson, S.; Gobara, M.; El-Sayyad, G.S.; Zorainy, M.; Awaad, M.I.; Hatem, H.; Kotb, M.M.; Tawfic, A.F. Synthesis of Silver Nanoparticles Using Natural Pigments Extracted from Alfalfa Leaves and Its Use for Antimicrobial Activity. Chem. Pap. 2017, 71, 2271–2281. [Google Scholar] [CrossRef]
  314. Zhang, R.; Yu, J.; Guo, X.; Li, W.; Xing, Y.; Wang, Y. Monascus Pigment-mediated Green Synthesis of Silver Nanoparticles: Catalytic, Antioxidant, and Antibacterial Activity. Appl. Organomet. Chem. 2021, 35, e6120. [Google Scholar] [CrossRef]
  315. Poorniammal, R.; Prabhu, S. Exploitation of Thermomyces Fungal Pigment for the Synthesis of Silver Nanoparticles for Textile Application. Emergent Life Sci. Res. 2023, 09, 40–48. [Google Scholar] [CrossRef]
  316. Nair, V.; Sambre, D.; Joshi, S.; Bankar, A.; Ravi Kumar, A.; Zinjarde, S. Yeast-Derived Melanin Mediated Synthesis of Gold Nanoparticles. J. Bionanosci. 2013, 7, 159–168. [Google Scholar] [CrossRef]
  317. El-Sayyad, G.S.; Mosallam, F.M.; El-Batal, A.I. One-Pot Green Synthesis of Magnesium Oxide Nanoparticles Using Penicillium Chrysogenum Melanin Pigment and Gamma Rays with Antimicrobial Activity against Multidrug-Resistant Microbes. Adv. Powder Technol. 2018, 29, 2616–2625. [Google Scholar] [CrossRef]
  318. Sowani, H.; Mohite, P.; Damale, S.; Kulkarni, M.; Zinjarde, S. Carotenoid Stabilized Gold and Silver Nanoparticles Derived from the Actinomycete Gordonia Amicalis HS-11 as Effective Free Radical Scavengers. Enzym. Microb. Technol. 2016, 95, 164–173. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of synthetic food dyes: (A) Erythrosine, (B) Allura Red, (C) Tartrazine, (D) Sunset Yellow, (E) Brilliant Blue, and (F) Indigo Carmine.
Figure 1. Chemical structures of synthetic food dyes: (A) Erythrosine, (B) Allura Red, (C) Tartrazine, (D) Sunset Yellow, (E) Brilliant Blue, and (F) Indigo Carmine.
Fermentation 11 00395 g001
Figure 2. Chemical structures of some important microbial pigments (The color of each molecular structure in the image corresponds to the actual color of the respective pigment in nature.)
Figure 2. Chemical structures of some important microbial pigments (The color of each molecular structure in the image corresponds to the actual color of the respective pigment in nature.)
Fermentation 11 00395 g002
Figure 3. Schematic representation of biosynthetic pathway of lycopene, β-carotene, canthaxanthin, and astaxanthin.
Figure 3. Schematic representation of biosynthetic pathway of lycopene, β-carotene, canthaxanthin, and astaxanthin.
Fermentation 11 00395 g003
Figure 4. Melanin biosynthesis pathway in microorganisms.
Figure 4. Melanin biosynthesis pathway in microorganisms.
Fermentation 11 00395 g004
Figure 5. The schematic diagram represents the synthesis of AgNPs using a microbial pigment.
Figure 5. The schematic diagram represents the synthesis of AgNPs using a microbial pigment.
Fermentation 11 00395 g005
Table 1. Advantages and disadvantages of microbial and synthetic food colorants.
Table 1. Advantages and disadvantages of microbial and synthetic food colorants.
Synthetic Food Colorants Microbial Food Colorants
Advantages High stability against oxygen, light, and heat (>100 °C)
High pH stability (stable across pH 2–9)
High tinctorial strength
Availability of all shades of color
Synthetic food colorants are less expensive than natural food colorants
Widely approved by FDA/EFSA, e.g., Tartrazine E102, Allura Red E129
Biodegradable and environmentally friendly
Generally regarded as safe and non-toxic
Exhibit bioactivities such as anticancer, antimicrobial, and antioxidant activities, among others
Can be produced using agro-industrial waste as a substrate (support the circular economy)
Disadvantages Often linked to adverse health effects (carcinogenicity, hyperactivity in children, allergies)
Environmental persistence and non-biodegradability
Relatively low stability towards oxygen, light, and heat (degrade above 60–80 °C)
Low pH stability (stable between pH 4 and 8)
Relatively low color saturation
Not all shades of color are available
Higher production costs and regulatory constraints
Regulatory approval and large-scale standardization remain challenges
Limited approvals by FDA/EFSA (e.g., β-carotene, Monascus pigment)
Table 4. Commercially utilized microbial pigments in industries [233].
Table 4. Commercially utilized microbial pigments in industries [233].
Pigment Name Color Industry Name Producing Microorganism
Phycocyanin BlueFermentalgArthrospira sp. (formerly Spirulina sp.) and Galdieria sulphuraria
Lycopene Red DSM Nutritional ProductsBlakeslea trispora
β-CaroteneYellow-orangeDSM Nutritional ProductsBlakeslea trispora
β-Carotene Yellow-orange Henkel-Cognis AustraliaDunaliella salina
Astaxanthin Pink-red JX Nippon Oil & EnergyParacoccus carotinifaciens
Astaxanthin Pink-red AstaReal Haematococcus pluvialis
AnthraquinonesRedNatural Red Penicillium oxalicum and many others
Table 5. Pigment-producing microorganisms and their bioactivity.
Table 5. Pigment-producing microorganisms and their bioactivity.
MicroorganismsPigmentColorNotable Functional BenefitRef.
Agrobacterium aurantiacumAstaxanthinPink-redAntioxidant, anticancer, anti-inflammatory[260]
Ashbya gossypiRiboflavinYellowAnticancer, antioxidant[81]
Blakeslea trisporaB-carotene, LycopeneCream/redAnticancer, antioxidant[261]
Chlorococcum, ChlorellaLuteinYellowAntioxidant[262]
Cordyceps unilateralisNaphtoquinoneDeep blood-redAnticancer, antibacterial, trypanocidal[263]
Dunaliella salinaB-CaroteneRed, orangeSuppression of cholesterol synthesis, antioxidant, anticancer[264]
Erythrobacter sp. SDW2XanthophyllYellow Antioxidant activity and potential for foods, cosmetics, and pharmaceuticals[265]
Flavobacterium sp., Paracoccus zeaxanthinifaciensZeaxanthinYellowAntioxidant, photoprotectant[266]
Fusarium sporotrichioidesLycopeneRedAnticancer, antioxidant[261]
Haloferax volcaniiCarotenoidsYellow Anticancer activity (human liver carcinoma cell HepG2)[267]
Halomonas sp.CarotenoidYellow Antimicrobial activity [268]
Kocuria marina, MeiothermusCarotenoidsYellow Antioxidant activity[269]
Kocuria sp. QWT-12Carotenoids Yellow Anticancer (breast cancer cell lines MCF-7)[269]
Micrococcus terreus--------Yellow Anticancer activity against cervical and liver cancer[270]
Monascus purpureusMonascin AnkaflavinRed-yellowAntitumor, anti-inflammatory[271]
Pacilomyces farinosusAnthraquinoneRedAntifungal, virucidal[272]
Paracoccus carotinifaciensAstaxanthinPink-redAntioxidant, anticancer, anti-inflammatory[273]
PedobacterCarotenoids Yellow Antioxidant activity[274]
Phaffia rhodozymaAstaxanthinPink-redAnti-inflammatory, antioxidant, anticancer [275]
Porphyridium cruentumPhycoerythrinRedAntioxidant, antitumor, immunoregulatory[276]
Pseudoalteromonas rubraProdigiosinRedImmunosuppressant, anticancer[277]
Pseudoalteromonas sp.Prodigiosin Red Cytotoxicity against U937 leukemia cells[278]
PseudoalteromonasVioloaceinPurple Antioxidant activity[279,280]
Pseudomonas aeruginosaPhyocyaninBlue-greenCiliary dysmotility, cytotoxicity[281]
Pseudomonas Aeruginosa P1.S9--------Blue-greenCytotoxic activities, antibacterial, antioxidant[282]
Pseudomonas balearicaMelaninBlack Antimicrobial activity against phytopathogenic strains[283]
Rhodococcus maris--------Yellow The risk of breast cancer was shown to be reduced[284]
Rhodosporidium toruloidesCarotenoidYellow–orange-redAntioxidant activity[285]
Saccharomyces neoformans var. nigricansMelaninBlack melaninAntimicrobial, antibiofilm, antioxidant[286]
Pseudomoalteromonas rubraProdigiosin Red Anticancer activity against human cervix carcinoma[287]
Staphylococcus aureusStaphyloxanthin ZeaxanthinGolden yellowAntioxidant[288]
Streptomyces hygroscopicus--------Yellow Antibacterial activity against MRSA, VRSA, and ESBL cultures[289]
Streptoverticillium rubrireticuliProdigiosinRedAntibacterial, antimalarial, antineoplastic[290]
Vibrio owensii TNKJ.CR.24-7 (MH488980.1)B Carotene Yellow Antibacterial activity [291]
Xanthophyllomyces dendrorhousAstaxanthinPink-redAntioxidant, anticancer, anti-inflammatory[275]
Table 6. Microbial pigments used in the synthesis of nanoparticles.
Table 6. Microbial pigments used in the synthesis of nanoparticles.
MicroorganismPigment NPsBiomedical ApplicationsRef.
Talaromyces purpurogenus-----AgNPsAntimicrobial and anticancer[308]
Talaromyces australis-----AgNPsAntibacterial[309]
Serratia nematodophila (NMCC 76), C. violaceum (KM226331)-----AgNPs, AuNPsAntiparasitic, antiplasmodial[310]
Streptomyces coelicolorActinorhodin AgNPs Antibacterial [311]
Xanthomonas sp. Xanthomonadin AgNPs Photoprotecting, antioxidant [312]
Monascus purpureus NRRL 1992-----AgNPs Antibacterial, antifungal [313]
Monascus ruber-----AgNPs Antibacterial, antioxidant, catalytic degradation of toxic dyes [314]
Thermomyces sp. Yellow AgNPs Textile application [315]
Yarrowia lipolytica NCYC789 Melanin AuNPs Antibacterial [316]
Penicillium chrysogenumMelanin MgONPs Antibacterial, antifungal [317]
Gordonia amicalis HS-11Carotenoids AuNPsFree radical scavenging activity [318]
Streptomycetes coelicolorActinorhodinAgNPs Enhancement of antibacterial activity [311]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chavan, A.; Pawar, J.; Kakde, U.; Venkatachalam, M.; Fouillaud, M.; Dufossé, L.; Deshmukh, S.K. Pigments from Microorganisms: A Sustainable Alternative for Synthetic Food Coloring. Fermentation 2025, 11, 395. https://doi.org/10.3390/fermentation11070395

AMA Style

Chavan A, Pawar J, Kakde U, Venkatachalam M, Fouillaud M, Dufossé L, Deshmukh SK. Pigments from Microorganisms: A Sustainable Alternative for Synthetic Food Coloring. Fermentation. 2025; 11(7):395. https://doi.org/10.3390/fermentation11070395

Chicago/Turabian Style

Chavan, Akshay, Jaya Pawar, Umesh Kakde, Mekala Venkatachalam, Mireille Fouillaud, Laurent Dufossé, and Sunil Kumar Deshmukh. 2025. "Pigments from Microorganisms: A Sustainable Alternative for Synthetic Food Coloring" Fermentation 11, no. 7: 395. https://doi.org/10.3390/fermentation11070395

APA Style

Chavan, A., Pawar, J., Kakde, U., Venkatachalam, M., Fouillaud, M., Dufossé, L., & Deshmukh, S. K. (2025). Pigments from Microorganisms: A Sustainable Alternative for Synthetic Food Coloring. Fermentation, 11(7), 395. https://doi.org/10.3390/fermentation11070395

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