Next Article in Journal
Integrating 3D Osteocyte Culture, Microgravity Simulation, and Fluid Flow Reveals Mechanisms of Osteocyte Mechanosensation and Calcium Signaling Altered by Disuse
Previous Article in Journal
Inflammatory Biomarkers for Thrombotic Risk Assessment in Multiple Myeloma Patients on IMiD/aCD38-Based Regimens: Insights from a Prospective Observational Study
Previous Article in Special Issue
Arctic Diatoms as a Source of Antibiofilm Compounds: Identification of Methyl 3-Hydroxyoctadecanoate and Pheophorbide a
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential Role of Microalgal Antioxidant Molecules on the Microbiota–Gut Axis in Inflammatory Bowel Diseases

1
Department of Basic and Applied Sciences (DISBA), University of Basilicata, via Dell’ateneo Lucano 10, 85100 Potenza, Italy
2
AlgaeBioMed Srl, via Luigi Kossuth 7, 00149 Rome, Italy
3
Department of Agricultural, Forest and Environmental Sciences (DAFE), University of Basilicata, via Dell’ateneo Lucano 10, 85100 Potenza, Italy
4
Department of Pharmacy (DIFARMA), University of Salerno, 84084 Fisciano, Italy
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(11), 1535; https://doi.org/10.3390/biom15111535 (registering DOI)
Submission received: 10 September 2025 / Revised: 24 October 2025 / Accepted: 25 October 2025 / Published: 31 October 2025
(This article belongs to the Special Issue Recent Advances in Bioactive Compounds from Microalgae)

Abstract

Comprising multiple microorganisms, the microbiota plays a crucial role in regulating the immune system and maintaining homeostasis. The influence of genetic and environmental factors causes the composition of the microbiota to change throughout life, which is called the plasticity of the microbiota. A eubiotic microbiota promotes the immune response, reducing the risk of inflammation and diseases such as IBD and cancer. The Mediterranean diet is of fundamental importance for a healthy microbiota. On the contrary, Western diets lead to microbiota dysbiosis and inflammation. Microalgae, and, in particular, their derivatives, show promise and relevance in the search for potential anti-inflammatory and antioxidant biomolecules. This review focuses on the correlation between microbiota, nutrition, immunity and microalgal derivatives, highlighting how these may be a potential innovative therapeutic strategy for the management of chronic inflammatory diseases.

1. Introduction

The human body is composed of multiple microorganisms called the microbiota. The term microbiome, instead, defines the set of genes that the microbiota can express [1]. It is estimated that there are 10 to 100 trillion microbes in the gut, acquired at birth when the newborn is exposed to an environment rich in microorganisms. This exposure is important for the efficient maturation of the immune system [2]. Microbial colonization in the gut begins at birth, changes with age, and stabilizes during the lifetime. This highlights how the microbiota evolves throughout life and mutates. For this reason, the scientific community refers to microbiota plasticity [3]. Several factors can influence the microbial population and its stability, such as genetics, age, gender, nutrition, different diseases, the environment, and diet [4,5]. In particular, the Mediterranean diet is one of the external factors involved in maintaining microbiota homeostasis and preventing several diseases [6]. Conversely, the Western diet alters the microbiota and the immune system, contributing to the increase of various diseases [7] (Figure 1). Intestinal microorganisms are essential for maintaining health by producing fatty acids and participating in glucose and lipid metabolism. They are also involved in the differentiation of immune system cells [8]. The maintenance of homeostasis is of paramount importance, and its loss can lead to various diseases. Recent studies have shown a correlation between changes in the microbiota and diseases such as diabetes, colorectal cancer, and inflammatory bowel disease [9]. Given the enormous importance of the microbiome for human health, research groups are trying to identify the interactions between microbiota and gastrointestinal diseases, with a particular focus on their effects on the immune system [10]. Inflammatory bowel disease (IBD) is a pathological condition caused by a complex interaction of environmental, immunological, and genetic factors, which leads to chronic inflammation of the gastrointestinal tract. Among immunoregulatory molecules, the etiological agents responsible for oxidative stress, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS), are overproduced in IBD. Among the various treatments currently used for IBD therapy, a new area of research is the use of antioxidant agents. Among these, astaxanthin (AXT), a powerful antioxidant carotenoid, is noteworthy for its ability to regulate oxidative stress and inflammation, produced by many plants and microalgae, especially Haematococcus lacustris (formerly known as Haematococcus pluvialis) [11]. Microalgae, in turn, represent a promising source of bioactive compounds with therapeutic value. The innovative use of microalgae derivatives offers a possible therapeutic option for IBD, based on protecting intestinal cells from oxidative stress and modulating inflammatory reactions.

2. Materials and Methods

To identify relevant studies and publications, a literature search was conducted in PubMed (MEDLINE), Google Scholar, and Scopus databases, using keywords such as “algae,” “microalgae,” “inflammatory bowel disease,” “IBD,” “Crohn’s disease,” “ulcerative colitis,” “intestinal inflammation,” and “astaxanthin.” To increase the amount of relevant information, artificial intelligence (https://scite.ai, accessed on 14 July 2025) was used to explore and delve deeper into specific topics, expand the relevant literature, and more critically consider the various involved paths. Extracted data and relevant results were selected based on publication relevance, methodology, and search results.
A total of 285 publications were selected, including systematic reviews, research articles, and meta-analyses.

3. Microbiota and Diet

Trillions of microbes inhabit the human body. The microbiota is defined as the set of microorganisms that are quantitatively and qualitatively present in a specific environment, such as the human gastrointestinal tract, which begins in the oesophagus and ends in the anus [9]. Over the past decade, the role of the gut microbiota has attracted significant research interest due to its complexity and involvement in human health and disease. Factors influencing the richness and diversity of the human microbiota include geographical location, age, genetic composition, hormonal changes, mode of delivery at birth (caesarean or vaginal), antibiotic use, lifestyle, and diet during childhood and adulthood [4]. The plasticity of the microbiota is greatest at birth when microbial colonization of the gut begins; during childhood there is a gradual increase in microbial diversity, and with advancing age, it becomes less malleable and stabilizes. Breastfeeding plays a significant role in microbiome formation, with the predominance of certain species such as Bifidobacterium and Bacteroides. Industrialization and unhealthy lifestyles in human populations are closely associated with higher rates of horizontal gene transfer (HGT). This mechanism consists of the introgression of exogenous gene fragments dynamically, between cells that are not direct progeny, conferring functional novelty to the genome into which it is inserted. Research studies have shown that HGT frequently occurs within individual gut microbiomes, and gut bacteria thereby acquire new functionalities. Modernity has led to increased hygiene, consumption of processed foods, reduced seasonality, and excessive use of antibiotics, continuously altering the gut microbiome [12]. The maternal microbiome shapes the infant’s gut microbiome through vertical transmission at birth and horizontal gene transfer events. In particular, an interspecies transfer of mobile genetic elements from mother to infant has been discovered. The main genes identified are associated with diet and are involved in encoding functions related to carbohydrate utilization, amino acid metabolism, and iron acquisition and storage. Mother–infant HGT may have consequences not only on the gut microbiome but also on the development of the immune system [13]. The human host and microbiota have co-evolved, forming a symbiotic relationship. The host provides a place for growth, suitable conditions, and nourishment for gut bacteria, while the microbiota supports bodily functions, induces resistance to infection, and facilitates the absorption of ingested food. The microorganisms that constitute the gut microbiota belong to the genera Bacteroidota, Bacillota, Pseudomonadota, Actinomycetota, Fusobacteria, and Verrucomicrobia. Recent studies confirm that in most individuals, the microbiota can be classified into three main groups: Bacteroides, Prevotella, and Ruminococcus [14]. The function of the gut microbiota is to protect against pathogens through colonization of mucosal surfaces, production of antimicrobial substances, enhancement of the immune system, regulation of digestion and metabolism, proliferation and differentiation of epithelial cells, communication, and brain–gut functions [15]. The plasticity of the microbiota can be summarized in six stages.
  • Prenatal phase: the hypothesis of colonization in utero has been questioned, although analysis is difficult due to limited quantities of microbes and the analytical limitations of 16SrRNA sequencing.
  • Perinatal phase: the microbial pattern is established depending on the mode of delivery. With vaginal delivery, maternal vaginal microbes colonize the infant’s gut, with an enrichment of Bifidobacterium spp. and a reduction of Enterococcus and Klebsiella spp. compared to cesarean section delivery, where the infant’s gut is mainly colonized by skin microbes. These differences may disappear within a year of life.
  • Postnatal phase: the composition of the gut microbiota changes depending on the consumption of breast milk or artificial milk and markedly with weaning [16].
  • Childhood and adolescence: the pediatric gut microbiota has similar characteristics to that of adults, but a child’s gut hosts larger amounts of Firmicutes and Actinobacteria and smaller amounts of Bacteroidetes than the adult microbiota.
  • Adulthood: an equilibrium between the host and the microorganisms in the gut is reached, it can be changed through diets and the use of drugs such as antibiotics. After treatment with antibiotics, restoration of the original microbial composition can take place with the use of probiotics. Autologous fecal microbiota transplantation (FMT), on the other hand, restores the gut microbiota within a few days after antibiotic administration.
  • Older age: increased susceptibility to infections may emerge due to an altered immune system leading to chronic low-level inflammation and dysbiosis in the gut microbiota causing pathogen growth and disease outbreaks. Studies conducted on healthy older individuals suggest the maintenance of a good microbial composition over time ensuring longevity and healthy aging [3].
The gut microbiota plays a decisive role in host homeostasis and numerous gastrointestinal diseases result from imbalances in the composition of the microbiota. Qualitative and quantitative alterations of the gut microbiota are observed with irritable bowel syndrome (IBS). The main immune-mediated chronic inflammatory diseases (IBD) affecting the digestive system are ulcerative colitis (UC) and Crohn’s disease (CD). Desulfovibrio species are abundant and Escherichia coli and low levels of F. prausnitzii, Bacteroidetes, and Firmicutes. The onset and development of IBD depend on several factors, including genetic susceptibility with a total of 199 specific genes identified for IBD, immune factors, and the gut microbiota. A high risk of developing IBD has been observed in the offspring of affected parents. In IBD, the reduction in the content and diversity of the gut microbiota weakens the microbial barrier, with an increased risk for opportunistic pathogens to invade the intestinal mucosa, leading to an immune disruption between Th17 and Treg cells, aggravating the inflammatory phenomenon. In affected individuals compared to healthy individuals, the content of metabolites such as SCFA (short-chain fatty acids), and primary and secondary bile acids (BAs) in the colon is drastically reduced and the metabolism of tryptophan is altered [17]. Increased intestinal permeability plays a role in the pathogenesis of IBD. Probiotic supplementation and fecal microbiota transplantation (FMT) can be used to treat IBD [18]. Chronic intestinal inflammation has also been associated with the development of colorectal cancer (CRC) and the increased involvement of Bacteroides fragilis and Streptococcus bovis, activating immune cells to release pro-mitogenic and pro-angiogenic cytokines such as Interleukin-17 (IL-17) and the synthesis by the gut microbiota of metabolites involved in carcinogenesis such as secondary transformations of bile salts, desulphurization of bile acids, synthesis of aromatic amines by azoreductase and nitroreductase, formation of reactive oxygen species [19]. In recent years, the role of the gut microbiota as a risk factor for the development of metabolic syndrome has been discovered. Hyperglycaemia, hypertension, and dyslipidemia are related to dysbiosis of the gut microbiota [20]. There is a close relationship between circadian rhythms, gut microbiota, and metabolic dysfunction associated with the West lifestyle in promoting carcinogenesis and cancer development, particularly colorectal cancer [21]. One of the methods used to characterize the gut microbiome is stool sample analysis. DNA extraction involves purification of the sample with reagents, lysis of the bacterial cells with lysis buffer, amplification, and sequencing of the 16S amplicon (hypervariable regions of 16S ribosomal RNA). The sequencing reads are grouped into operational taxonomic units (OTU clustering algorithm) and based on sequence homology to the 16S rRNA gene databases it is possible to identify bacterial genera and species [22]. Microbial diversity plays an important role in human metabolism and their potential use in the treatment of infectious and chronic diseases and therapeutic applications [8].
Diet is the main modulator of the functions and composition of the gut microbiota. A balanced diet can ensure the formation of a good microbial flora, where all bacterial species coexist in a mutually balanced system [23]. During birth there is colonisation of the gut, which changes throughout life in microbial diversity and abundance. Many studies have shown that breastfeeding is the strongest predictor of the composition of the gut microbiota in the first months of life, promoting healthy development of the immune system and preventing pathological imprinting. A marked transformation in the gut microbial community occurs after weaning with the introduction of solid foods. The use of antibiotics has a significant impact on the evolution of the infant gut microbiota by increasing Proteobacteria and reducing Actinobacteria populations, decreasing overall diversity, and selecting drug-resistant bacteria. Epidemiological studies have shown that antibiotic consumption in early childhood leads to an increased risk of allergic diseases [24]. In early childhood, commensal intestinal microbes such as Bifidobacterium infantis, Bifidobacterium bifidum, and Lactobacillus rhamnosus can enhance the development of the intestinal barrier by strengthening tight junctions in intestinal epithelial cells. Bacteroides thetaiotaomicron and Lactobacillus reuteri attenuate host inflammatory responses by inhibiting activation of nuclear factor-κB (NF-κB) [25]. The process of urbanization has significantly shaped the gut microbiota, thus affecting the functionality of the gut microbiome. Each person’s microbiome is unique, develops rapidly during early childhood, and is relatively stable but susceptible to changes in adulthood depending on the type of diet followed. The Mediterranean diet (MD) is a dietary pattern that exerts a beneficial role on the gut microbiota in particular on its diversity and metabolic activities, promoting a state of eubiosis in terms of balance and well-being of the microbiota with a predominance of beneficial microorganisms. It is characterized by a high content of plant-based foods and habitual consumption of cereals, legumes, olive oil and nuts. These foods provide polyunsaturated fatty acids with anti-inflammatory properties, a variety of bioactive compounds with antioxidant properties such as polyphenols, vitamins, and minerals [6]. Fibre has a prebiotic action for the microbial growth of bacteria living in the human gut and is an excellent substrate for the metabolism of bacteria to produce SCFAs (short-chain fatty acids) mainly butyrate, propionate, and acetate. These metabolites play an important role in the regulation of immunity [26]. In contrast, a West diet high in saturated fats, refined carbohydrates, and animal protein, low in plant/fruit foods, and low in omega-3 PUFAs, can have a direct effect on the immune system causing intestinal barrier disruption, metabolic disorders and dysbiosis [27]. Dysbiosis is a structural and functional change in the gut microbiota, which can trigger severe inflammation through an increased number of pro-inflammatory microorganisms. Moreover, dysbiosis is directly related to the onset of colorectal cancer (CRC) [26]. A dysbiosis in the gut microbiota can lead to a reduced ability to induce a local and systemic immune response, resulting in local inflammatory diseases and diseases at distal sites [28]. The gut microbiota can produce metabolites through the fermentation of ingested food. The metabolites act as substrates and regulate the activities of epigenetic modification enzymes (DNA methylation, histone modifications and the expression of non-coding RNAs), influencing the expression of host genes and triggering immune inflammation in intestinal epithelial cells (IECs), resulting in metabolic disturbances through altered intestinal permeability [29]. A diet rich in whole grains, fruit and vegetables has beneficial effects on the bacterial composition, and reduces opportunistic bacteria, resulting in a reduction of lipopolysaccharide (LPS), trimethylamine-N-oxide (TMAO) and inflammatory cytokines. Conversely, increased SCFA production reduces inflammation and the risk of obesity and type II diabetes [30]. Diet can have a direct effect on host physiology and health and an indirect effect by altering the composition of the gut microbiota and/or its production of metabolites that in turn impact host physiology causing disease. Recent studies in animal models show a cause–effect relationship between diet and host health via the gut microbiota. Examples include strengthening barrier function through SCFA production; protection of the mucus layer through fiber acting as a nutritional substrate for the microbiota; SCFA increasing regulatory T-cells supporting immune tolerance and GALT homeostasis [31]. In addition, tryptophan metabolites such as indoles are derived from commensal fermentation of dietary tryptophan and function as ligands for the aryl hydrocarbon receptor (AhR), a receptor important in maintaining intestinal homeostasis. Loss of these metabolites is associated with the onset of inflammatory bowel disease. Nutritional intervention is a potential therapeutic approach, targeting both the gut microbiota and the immune system. Several studies discuss the relevance of immuno-nutrition, the ability to modify nutrient intake to modulate immune responses [32].
Figure 1. The Mediterranean diet involves the maintenance of the gut microbiota eubiosis and the integrity of the gut barrier and immune tolerance. West diets and ultra-processed foods result in the manifestation of permeable gut syndrome, gut dysbiosis, local inflammation and the presence of LPS in the bloodstream that will contribute to systemic endotoxemia and chronic inflammation [32].
Figure 1. The Mediterranean diet involves the maintenance of the gut microbiota eubiosis and the integrity of the gut barrier and immune tolerance. West diets and ultra-processed foods result in the manifestation of permeable gut syndrome, gut dysbiosis, local inflammation and the presence of LPS in the bloodstream that will contribute to systemic endotoxemia and chronic inflammation [32].
Biomolecules 15 01535 g001
Based on numerous research studies, it is speculated that a high-fat diet may promote gastrointestinal tumors by altering the composition of gut microbes [33]. For example, high-fat diets can lead to increased production of bile acids converted by intestinal bacteria to secondary bile acids (deoxycholic acid and lithocholic acid). Due to their genotoxic and pro-inflammatory effects, they have been implicated in the promotion of both colon and liver cancer. Conversion may also result in an immunomodulatory role in promoting liver cancer progression by inhibiting the recruitment of anti-tumor natural killer T cells in the liver [34]. In the last few years, numerous investigations revealed deep modifications of intestinal microbiota composition, firmly linked with lifestyle and altered metabolic status. More precisely, Lynch and Pedersen point out how the reduction of bacterial diversity and the depletion of beneficial species such as Faecalibacterium prausnitzii and Akkermansia muciniphila are accompanied by an expansion of bacteria typical of inflammatory and obesogenic disorders [35]. These alterations have a deep impact on systemic metabolism, like that of oxylipins, oxidative metabolites of polyunsaturated fatty acids (PUFAs) playing an important role in the regulation of inflammation and lipid homeostasis. Dysbiosis induced by high-fat diet alters the plasma oxylipin profile with an increase of pro-inflammatory arachidonic acid derivatives (e.g., 5-HETE, 12-HETE) and a reduction of anti-inflammatory omega-3 fatty acid-derived products (e.g., 17-HDHA, 14-HDHA) [36]. These changes are linked with the decrease of SCFA-producing bacteria, cementing the interaction between microbiota, lipid metabolism, and oxidative regulation. This microbiota–oxylipin axis explains the formation of a chronic, low-grade inflammatory microenvironment that is typical of obesity as well as other metabolic syndromes [37,38]. Intestinal dysbiosis alters not only microbial metabolite synthesis but also oxylipin biosynthesis and functional balance, regulating key inflammatory and metabolic processes. This supports the idea of a bidirectional dialogue between microbiota and oxidative lipid metabolism that is a potential therapeutic target in obesity and chronic inflammatory disease states [39,40]. The most important factor in preventing and fighting cancer is a well-functioning immune system. This requires a good nutritional status allowing the gut microbiome to play all its roles in metabolic control, immune control and communication with other organs [40,41].

4. IBD

The coexistence of the microbiota and the host is essential for maintaining vital host functions, and disruption of this complex coexistence has been associated with several diseases, such as IBD. IBD, which includes Crohn’s disease and ulcerative colitis, differs in symptoms, location, and histopathological features. It is characterized by chronic inflammation of the gastrointestinal tract and a clinical course alternating between phases of remission and relapse, causing significant morbidity and reduced quality of life in patients [41,42,43,44,45]. The pathogenesis of IBD is complex and still not fully understood, although it is characterized by a combination of genetic susceptibility, dysbiosis, immune system dysregulation, and environmental factors [46,47]. Recent research has highlighted the interaction between gut microbiota, immune system and genes in the pathogenesis of IBD [4]. The intestinal microbiota, in particular, has been identified as a determining factor, capable of influencing susceptibility to the disease and response to therapy [48,49]. Dysbiosis, caused by reduced microbial diversity, increased epithelial permeability, loss of anti-inflammatory bacteria and increase of pro-inflammatory ones, is a key element [50,51]. Precisely for this reason, technological advances in metagenomic sequencing have contributed to a greater understanding of microbial dysbiosis in IBD, paving the way for therapies targeting the microbiota [52,53,54]. Chronic inflammation associated with oxidative stress causes a disorganized immune response and an imbalance of the gut microbiota [55]. The role of cytokines, important in the pathogenesis and inflammatory mediation, has been extensively studied and many targeted therapies have been developed with positive outcomes in several clinical trials [56,57,58]. The imbalance between pro- and anti-inflammatory mediators is also mainly influenced by a redox imbalance, where the imbalance between ROS/RNS production and endogenous antioxidant defenses is an important pathogenic factor for these diseases [59,60,61,62]. ROS and RNS, although involved in physiological antimicrobial defense, cause oxidative damage to cellular structures, aggravating the inflammatory process and are mainly generated by activated immune cells (macrophages, neutrophils) and intestinal epithelial cells in response to pro-inflammatory stimuli such as TNF-α, IL-1β and IL-6 [63,64]. Macrophages and dendritic cells of the lamina propria, when they recognize microbial antigens via receptors such as TLR and NOD2, release proinflammatory cytokines (TNF-α, IL-1β, IL-6, IL-12/23), which recruit neutrophils and activate helper T cell responses [65,66,67,68]. In parallel, the function of Foxp3+ regulatory T cells (Treg) is compromised, losing the ability to maintain immune tolerance [69,70]. In CD, a Th1/Th17 response predominates, with increased IFN-γ and IL-17, while in UC a Th2 response is observed, with increased release of IL-4 and IL-13 [71,72,73,74,75]. The inflammatory cascade in IBD is always associated with excessive production of ROS and RNS, particularly superoxide anion (O2), hydroxyl radical (OH) and nitric oxide (NO) [76,77]. Their overproduction compromises the integrity of the intestinal barrier, thus increasing intestinal permeability, which favors bacterial translocation, which in turn further activates immune cells and strengthens inflammation [78,79,80,81,82,83,84,85]. The interaction between O2 and NO produces peroxynitrite (ONOO), a highly damaging reactive nitrogen species, which causes protein nitrification and further tissue damage [86,87]. In particular, peroxynitrite activates proinflammatory signals such as NF-κB and MAPK, increasing the secretion of cytokines [87,88]. These redox environments further regulate the immune response through NLRP3 inflammasome activation, Th1/Th17 polarization, Treg dysfunction, and mitochondrial damage [25,59,60]. Mutations in the NOD2 and ATG16L1 genes interfere with autophagy and antimicrobial defense processes, leading to increased production of proinflammatory cytokines and impaired integrity of TJs [61,62,63]. This promotes the penetration of antigens and the consequent activation of the immune response [47,89]. Dysbiosis and genetic predisposition close the vicious circle, fueling inflammation and tissue damage [90,91,92]. Foxp3+ Treg cells, particularly the RORγt+Foxp3+ subpopulation, are essential for avoiding excessive inflammation in the gut [93]. Their significant depletion has been demonstrated in the colonic lamina propria in murine models of IBD, suggesting that their loss contributes to chronic inflammation [94,95,96]. Dysbiosis, in fact, compromises immune homeostasis, as occurs, for example, in invasive-adhesive Escherichia coli (AIEC), a pathobiont often associated with IBD, which exploits the host’s genetic predisposition (e.g., polymorphisms in NOD2 and ATG16L1) to escape immune clearance and increase inflammation [97,98,99,100,101]. The intestinal epithelial barrier, protected by tight junction proteins such as claudins, is fundamental and its dysfunction, which results in increased permeability, allows luminal antigens to invade the lamina propria, activating the immune response [102,103,104,105]. Concomitantly, there is a deficiency in the production and function of regulatory T cells (Treg), particularly of the anti-inflammatory cytokines IL-10 and TGF-β, normally involved in maintaining immune homeostasis [106,107]. This situation facilitates the migration of neutrophils, macrophages and dendritic cells into the lamina propria, which release inflammatory mediators, ROS and RNS [107,108]. Dysbiosis and genetic predisposition close the vicious circle, fueling inflammation and tissue damage [90,91,92]. Foxp3+ Treg cells, particularly the RORγt+Foxp3+ subpopulation, are essential for avoiding excessive inflammation in the gut [93]. Their depletion has been demonstrated in the colonic lamina propria in murine models of IBD, suggesting that their loss contributes to chronic inflammation [94,95,96]. The intestinal epithelial barrier, protected by tight junction proteins such as claudins, is also fundamental [102]. Its dysfunction, in the form of increased permeability, allows luminal antigens to invade the lamina propria, activating the immune response [103,104,105]. Dysbiosis exacerbates the damage, as demonstrated by the reduction of microbial diversity and the proliferation of proinflammatory taxa [64,80,84]. The primary target of IBD therapy is TNF-α, and cyclical treatments with anti-TNF-α agents have been shown to reduce inflammation and induce remission [109,110]. Recently, research has also focused on blocking other cytokine pathways, such as IL-23 and IL-12, in the process of T-helper cell activation and differentiation, for example, with ustekinumab, as an alternative treatment for patients refractory to anti-TNF-α [111,112,113,114,115]. Traditional therapies, such as aminosalicylates, corticosteroids and immunosuppressants, have been the main therapeutic approaches, but they have significant side effects and unpredictable efficacy [116,117,118]. Despite their high cost, biological therapies have revolutionized the management of IBD, improving clinical outcomes in most patients while highlighting treatment refractoriness in some patients, underscoring the need for alternative and innovative therapeutic approaches. [119,120]. Recent studies suggest that redox balance intervention through antioxidant treatments can reduce inflammation in IBD [121]. To this end, carotenoids have attracted the attention of the scientific community as adjuvants in the treatment of IBD [122,123]. Carotenoids are a highly diverse class of pigments that play an important role in the biological processes of plants, algae and some bacteria, contributing to the bright colours of fruits and vegetables and participating in photosynthesis [124,125]. Carotenoids have long been recognized for their single oxygen neutralization and free radical scavenging activity, thus protecting cells from oxidative stress [126,127,128]. Their activity is particularly relevant in chronic diseases such as cancer, diabetes, obesity, cardiovascular and neurodegenerative diseases, where oxidative stress and inflammation are key factors [76,129,130]. In recent years, there has been a growing interest in xanthophylls and microalgae, a subclass of carotenoids that includes astaxanthin (AXT), lutein and zeaxanthin [131]. AXT, in particular, has attracted great interest due to its antioxidant and anti-inflammatory properties, making it a suitable candidate for the treatment of inflammatory diseases such as rheumatoid arthritis [132,133]. In addition to their antioxidant and anti-inflammatory actions, carotenoids also contribute to intestinal health [134,135]. The interaction between carotenoids and gut microbiota is a new area of research, with important therapeutic implications for IBD [134,135,136]. Dietary interventions, particularly those rich in carotenoids, have shown potential benefits in modulating intestinal inflammation and improving clinical response in patients with IBD [137,138].

5. Microbiota and Immune System

The microbiota consists of a series of microorganisms (viruses, bacteria, fungi) that cooperate with the host by regulating various functions including the immune response to pathogens. To ensure homeostasis, gut bacteria use molecules produced during their replication to control microbial population density and gene expression. Alterations in the microbiota cause inflammation, infection and disease onset [28]. The organ that hosts the most immune cells is the gut. This has dendritic cells, macrophages, T-cells and B-cells capable of inducing an immune response against pathogens without altering the microorganisms present [139]. Bacteria in the gut interact with the immune system by conditioning the maturation of immune cells, regulating the immune response and supporting its development in childhood [140]. Microbial occupation begins simultaneously with the maturation of the immune system during childhood. The microbiota–immune system correlation is crucial for immune tolerance and the prevention of various diseases [49]. The bacteria in the gut produce vitamins, fatty acids and various compounds essential for maintaining health. In addition to defending against the attack of pathogens, intestinal bacteria can keep the intestinal wall intact [141]. Furthermore, essential compounds such as vitamins are key in the innate and adaptive immune response. The loss of just one essential nutrient could alter immunity [142]. In detail, the production of fatty acids induces an enhancement of the anti-inflammatory response with the production of antibodies by B cells and an enhancement of the action of T cells [143]. Alterations in the microbiota are causative of alterations in the immune response resulting in disease. Underlying this is the restoration of homeostasis [144]. Also at the genetic level, through transcriptomics studies on adaptive and innate cells and through epigenetic regulation of cytokines, it has been possible to study the microbiota–immune system interaction. Studies in mice show that microbiota–immune system imbalance underlies multifactorial diseases, such as cancer, infectious diseases, metabolic diseases, diabetes, and inflammatory and neurodegenerative diseases [145]. Among the latter, relevant are Parkinson’s disease and Tourette’s syndrome [146]. The variety of diseases results from a variable microbial composition. For example, there are different microbial populations along the gastrointestinal tract. Each of these can be considered on its own and limited by nutrients and organs. Thus, alteration of one population may be causative of a given disease [147]. In a healthy subject, Toll Like Receptor (TLR) levels are low. These increase in the case of an imbalance between the microbiota and the immune system resulting in the activation of monocytes, macrophages, and natural killer cells involved in innate immunity [148]. Alteration of homeostasis resulting from disruption of the microbiota–immune system balance is causative of intestinal diseases. Underlying the imbalance is an inflammatory context. The continued presence of an inflammatory state can be causative of neoplasms [149]. The stability of the intestinal barrier is due to fermentation products obtained from the microbiota. In general, TLRs recognize bacterial-specific components that are presented to CD4 T cells and B cells [150]. The microbiota can induce the activation and differentiation of B cells resulting in the production of antibodies [151]. These can bind the antigen, blocking pathogenic action and thus infection. Alterations in the process can lead to an increase in pathogenic Gram-negative bacteria resulting in an alteration of the intestinal barrier and increased infection [150]. Among these, inflammatory bowel diseases are very common. In the altered microbiota–immune system correlation, both T-helper cells (pro-inflammatory) and regulatory T-cells (anti-inflammatory) are activated. Active T-helper cells produce cytokines and molecules causing inflammatory states with a proliferation of pro-inflammatory bacteria called pathobacteria [152]. Pathobacteria, through specific virulence mechanisms, can evade the immune system by generating toxins. The transformation of bacteria in the microbiota into pathobacteria induces a loss of beneficial responses in the body resulting in the risk of developing critical illnesses that can cause mortality [153]. Innate response cells regulate the homeostasis–inflammation transition and are involved in the development of cancer. In detail, in a tumor context, innate cells undergo functional changes and lose their ability to induce and aid the immune response [154]. The disruption of the balance of the intestinal microbiota, increasing pathogenic bacteria, is called intestinal dysbiosis. The resulting inflammatory state can be causative for tumor development. Inflammation arises from chemokine ligand 5 (CCL5), which attracts lymphocytes to the gut. Local inflammation induces the activation of interleukin-6 (IL-6) and the proliferation of intestinal epithelial cells resulting in tumor formation (Figure 2) [155].
Figure 2. Tumor development from the inflammatory state [153].
Figure 2. Tumor development from the inflammatory state [153].
Biomolecules 15 01535 g002
Several studies have shown that certain gut bacteria can be used as biomarkers for digestive cancers such as pancreatic cancer and hepatocellular carcinoma. Treatments for gastrointestinal diseases include microbiota transplantation, the use of prebiotics and probiotics, and the possibility of using engineered bacteria [156]. Moreover, since the microbiota is able to induce the activation of anti-inflammatory and pro-inflammatory cells, its modulation can be effective in cancer immunotherapy [157].

6. Microbiota and Cancer

In 2020, there were 19.3 million cancer cases and 10 million deaths. Infection with Helicobacter pylori, a Gram-negative bacterium, is considered the leading cause of gastric cancer [158]. Tumor initiation and progression caused by infection could be induced by mutated gene expression and activation of signaling pathways [159]. It was observed that some tumor cases were attributable to other infectious contexts of papilloma virus and hepatitis B and C virus [160]. The cancer–microbiota interaction is complex and requires further study, but in about 20% of cancers, microorganisms are thought to be involved and take part in the tumor environment. Carcinogenesis is triggered by a disruption of the homeostasis of the colon epithelium due to both tumor cell proliferation and host cell death [161]. Tumor cells originate from acquired genetic modifications, undergoing uncontrolled proliferation and escaping immune control. Certain microorganisms can thus promote tumourigenesis by altering the immune response and creating a favorable environment for tumor development [162]. The gut microbiota and its bioactive metabolites do not directly cause cancer but may promote and modulate tumor progression and therapeutic efficacy [163]. Colorectal cancer (CRC) is considered one of the most devastating and widespread malignancies worldwide. CRC is the third most commonly diagnosed cancer, and a genetic predisposition underlies its development. In the cancer patient, the gut microbiota evolves with the pathogenesis of the disease, the etiology of the tumor being due to an infectious insult or high consumption of alcohol, smoking, obesity and diabetes [164]. The main tumor-promoting mechanisms are pathogenic bacterial virulence factors and/or toxins, bacterial metabolic products and dysregulation of immune responses [165]. Microbiota-related carcinogenesis is due to dysbiosis-related inflammation and carcinogen formation. Cancer-associated intestinal dysbiosis in individuals genetically predisposed to IBD can induce an aberrant immune response and the proliferation of pro-inflammatory bacteria, which increase inflammation leading to altered intestinal permeability [152]. A diet rich in saturated fats and simple sugars can induce microbial dysbiosis in the gut and increase the risk of CRC with the production of carcinogenic metabolites such as NOC (N-nitroso compounds), TMAO and secondary bile acids [166]. The most common bacterial species in CRC patients detected in biopsies is F. nucleatum, which besides being involved in inflammation interacts with E-cadherin expressed on both epithelial cells and tumor cells through its virulence factor FadA. This bacterium upregulates oncogenes and the subsequent phosphorylation of β-catenin results in the activation of NF-KB by upregulating the synthesis of inflammatory cytokines and cell proliferation [167]. Metabolites may inhibit or promote tumor development by inducing pro-inflammatory and immunosuppressive effects. Short-chain fatty acids (SCFAs) and bile acids such as ursodeoxycholic acid (UDCA) are the main metabolites of the gut microbiota with a protective role. They inhibit inflammation, promote apoptosis of cancer cells and defend colon epithelial barriers from cancerous invasion [168]. For example, butyrate has antitumor activity. Recent studies have found that the effects of butyrate vary depending on its concentration, high concentrations of butyrate inhibit tumourigenesis, while low concentrations promote tumor evolution; however, further studies are needed [169]. Some bile acids and TMAO, on the other hand, promote cancer cell proliferation, metastatic spread, DNA damage and genomic instability. Moreover, the presence of bacteria in the gastrointestinal tract induces the breakdown of heme leading to the production of hydrogen sulfide and nitric oxide, these compounds also cause DNA damage and promote carcinogenesis [170]. It has been reported how epigenetic alterations in the colorectal epithelium such as DNA methylation, histone modifications and non-coding RNAs are due to the gut microbiota, and its metabolites act as a potential driving force for the initiation and promotion of tumourigenesis in CRC [171]. Host-gut microbiota communication is required to maintain gut homeostasis through a regulatory network in which microRNAs (miRNAs) have emerged as major mediators. Diet-induced microbial metabolites can influence miRNA expression in host cells and promote or inhibit tumors in CRC. Host-derived miRNAs such as miR-515-5p and miR-1226-5p may also affect the microbiota and promote bacterial growth of F.nucleatum and E. coli, suggesting a possible influence on tumor growth by modifying bacterial activities and the gut microenvironment [172]. Patients with CRC and healthy individuals have different gut microbiota in their feces, which is why the gut microbiota can serve as a microbial marker to assess CRC risk and can be used to screen for precancerous lesions and CRC [173]. Also, the gut microbiota influences antitumor immunity through the production of inosine and hypoxanthine involving the proliferation of T-helper 1 cells by inducing the death of transformed cells, inhibition of inflammatory cytokines and blockade of checkpoint sites, which could be of interest in therapeutic applications [174]. Modulation of the gut microbiota represents an innovative and effective strategy in cancer treatments [175]. Precision and customized cancer treatments are possible by combining bacteria with nanotechnology, optimizing drug targeting and reducing toxic and side effects [176]. Numerous studies have shown improved health in cancer patients through the use of probiotics, which can inhibit tumor growth and modulate the individual’s inflammatory state [177].

7. Microalgae

In order to regulate oxidative stress and the immune response, interest in microalgae is growing significantly as potential sustainable alternatives. Microalgae are photoautotrophic plant organisms due to the presence of photosynthetic pigments, such as chlorophyll and carotenoids. Microalgae exhibit a high ability to colonize various environments and to respond very rapidly to environmental changes, making them organisms with remarkable adaptability thanks to their high metabolic plasticity [178]. Moreover, microalgae are extremely efficient organisms due to their excellent biochemical composition in:
  • Proteins, which serve both biological and structural functions. Microalgae tend to accumulate proteins in amounts ranging from 10% to 70% of the dry biomass weight.
  • Lipids and microalgal biomass contain both structural (polar) and storage (neutral) lipids. Microalgae are rich in long-chain fatty acids with up to 20 or 22 carbon atoms, and in polyunsaturated fatty acids (PUFAs) such as docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and gamma-linolenic acid (GLA). The most relevant are omega-3 and omega-6 fatty acids. Ossilipins can be obtained from the oxidation of PUFAs. These are lipophilic signalling molecules mainly involved in inflammatory processes and stress responses [179]. The activity of new families of bioactive mediators derived from omega-3 polyunsaturated fatty acids, such as eicosapentaenoic acid and docosahexaenoic acid, is significant. These mediators, called resolvins, docosatriens and protectins, have protective and anti-inflammatory properties [180]. Their role is promising in the resolution of inflammation and, in general, in the development of new therapeutic approaches [181].
  • Carbohydrates, the composition is species-specific in microalgae and includes both monomeric sugars (monosaccharides) and polymeric forms (di-, oligo-, and polysaccharides). The most abundant sugars are glucose, rhamnose, xylose, and mannose. They serve various functions such as storing metabolic energy and are also major structural components of the cell wall, along with glycoproteins. It is estimated that sugar content can account for up to 60% of the dry biomass weight.
  • Photosynthetic pigments with potential applications in cosmetics, nutraceuticals, and pharmaceuticals. They exhibit remarkable properties, including antioxidant, anti-inflammatory, neuroprotective, hepatoprotective, anti-mutagenic, and anti-allergic effects. The three main classes of pigments are: phycobiliproteins (PBPs), carotenoids (carotenes and xanthophylls) and chlorophylls, the green pigments typical of plants [182].
  • Vitamins, the accumulation and synthesis of vitamins in photosynthetic organisms are highly variable and closely linked to physiological responses to environmental changes. Microalgae are producers of many essential vitamins such as vitamin A, B-group vitamins, vitamin C, vitamin D, vitamin K, and vitamin E [183].
Haematococcus lacustris is a unicellular green microalga belonging to the Chloropyta. This is a great source of fatty acids and protein, and it is the largest producer of astaxanthin (AXT). AXT is a xanthophyll belonging to the red carotenoids [184]. Structurally, it has β-ionic rings containing oxygenated ketone and hydroxyl groups linked together by a polyene chain. For this reason, AXT has antioxidant properties and reduces the production of free radicals. AXT is widely used as a protective agent in many diseases caused by oxidative stress. In addition to its antioxidant properties, it is also an anti-inflammatory and anti-tumour agent [184]. Thanks to its properties, AXT is very interesting in the treatment of chronic inflammatory diseases such as IBD.

8. Potential Therapies and Biomolecules from Microalgae

Several classes of drugs, from glucocorticoids to biologics, are used for the treatment of IBD. Biological treatments have revolutionized disease management, targeting selective inflammatory molecules and improving the quality of life of many patients [185,186]. For a long time, IBD therapy has focused on controlling pain and associated symptoms, using various drugs such as aminosalicylates and corticosteroids, although the latter, while beneficial for many patients, have significant side effects [187,188,189]. However, treatment resistance and relapses remain common problems, highlighting the need for new or adjuvant therapeutic options [190]. Probiotics, as modulators of the intestinal microbiota, have been considered for their potential to restore intestinal homeostasis and reduce inflammation [191,192]. The production of probiotics with specific strains, such as Limosilactobacillus fermentum, represents an area of research to improve their therapeutic potential [193]. The synergistic use of probiotics with other compounds, such as hydroxyectoine, can amplify the beneficial effects on the intestinal environment. Hydroxyectoine, in fact, forms a protective and hydrating envelope around probiotic cells, stabilizing their proteins and membranes as they pass through the intestine, preventing damage caused by dehydration, acids, and bile salts, increasing the survival of probiotics and amplifying their beneficial effects on the intestinal environment [194]. Recent studies have identified new therapeutic molecules useful in the treatment of IBD such as oroxyloside, a chemical compound that improves DSS-induced colitis through the inhibition of endoplasmic reticulum stress by activating PPARγ and polyphenolic agents such as resveratrol and curcumin which have shown the ability to act on cellular signaling pathways and suppress intestinal inflammation [119,120,195,196,197]. These results indicate that the search for new molecules, natural or otherwise, can expand therapeutic options to restore intestinal homeostasis and improve the quality of life in IBD patients. For this reason, natural bioactive compounds are used as supplements or as therapeutic alternatives to alleviate IBD-related problems, such as carotenoids. These are, in fact, natural pigments with notable antioxidant and anti-inflammatory properties. For example, β-carotene has demonstrated anti-inflammatory activity in animal models of DSS-induced ulcerative colitis [198]. Among the carotenoid xanthophylls, the most interesting for antioxidant activity is AXT, produced by various algae and microorganisms [199]. L’AXT (3,3′-dihydroxy-β,β-carotene-4,4′-dione) is a highly effective keto-carotenoid belonging to the class of xanthophylls. Its chemical structure comprises two hydroxyl groups and two carbonyl groups, which give it a powerful antioxidant activity: it is in fact 10 to 550 times more effective than other antioxidants such as vitamin E, β-carotene and lycopene in neutralizing free radicals [199,200,201]. Its chemical nature makes it very stable and lipophilic, thus increasing its bioavailability and accumulation in biological systems [202,203]. This carotenoid is biologically synthesized by microalgae, in particular Haematococcus lacustris, which can accumulate AXT up to 4–5% of the dry weight under stress conditions (high light intensity or nutrient scarcity) [204,205,206]. In addition to these green algae, AXT is also found in some yeasts (e.g., Phaffia rhodozyma), some mushrooms (Xanthophyllomyces dendrorhous), and several seafoods such as salmon and shrimp [207,208,209,210,211]. Other microalgae, such as Chlorella zofingiensis and Scenedesmus sp., have also been studied, but produce lower amounts of AXT [131]. Due to its bioactivity, AXT is of great interest in the pharmaceutical and nutraceutical sectors, with applications ranging from the treatment of inflammation to use as a food supplement [100,212,213]. Recent technological advances in cultivation processes have allowed the optimization of biomass production and AXT yield [214]. Furthermore, genetic modification processes have been employed to optimize AXT biosynthesis pathways in microalgae, with the aim of increasing yield and reducing production costs [215]. AXT is a highly effective antioxidant, able to neutralize free radicals and reduce oxidative stress, thus effectively preventing cellular damage in long-term chronic diseases. Furthermore, it possesses anti-inflammatory effects, as demonstrated by numerous in vitro and in vivo studies, acting on a broad spectrum of chronic diseases [212,214,216,217,218]. Its action is mainly expressed through the interaction with key cellular pathways such as NF-κB, MAPK, JAK-STAT and Nrf2, which regulate both the inflammatory response and antioxidant defense mechanisms [219,220]. In some studies, the role of AXT as a protective agent in the treatment of diseases, including colitis, has been investigated: in murine models, astaxanthin modulated inflammatory responses and preserved the intestinal barrier function [217,218]. Positive effects of AXT have also been observed at the cardiovascular level, with improvements in lipid metabolism, endothelial function and a reduction in the risk of atherosclerosis, as confirmed by a meta-analysis [205,221]. Its ability to inhibit the expression of the metalloproteinase MMP-9 and to reduce lipid deposits in macrophages make it an ideal candidate for the prevention of atherosclerosis [222,223,224]. The neuroprotective role of AXT is of particular interest, since it is able to preserve cognitive functions and improve synaptic plasticity, as demonstrated by studies conducted on young and old mice [225]. The modulation of neuroinflammatory pathways and the reduction of oxidative stress suggest a possible therapeutic use of AXT in neurodegenerative diseases [226,227]. In models of Alzheimer’s disease and Parkinson’s disease, astaxanthin has shown protective effects, reducing neuroinflammation and neuronal apoptosis, thanks to the activation of pathways such as PI3K/Akt and Nrf2 [222,226,228,229,230,231,232,233,234]. By selectively inhibiting NF-κB activation by reducing IκB-α degradation, thus preventing subsequent nuclear translocation of NF-κB, the transcription of key pro-inflammatory genes such as TNF-α, IL-1β, IL-6, COX-2 and iNOS is blocked [231,232,234,235,236,237]. Studies conducted on cellular and animal models indicate that AXT is particularly effective in inhibiting the chemokine MCP-1, responsible for macrophage recruitment, as well as modulating the immune response by affecting the balance between Th1 and Th2 [219,238,239]. In parallel, AXT acts as a potent antioxidant, both directly, by neutralizing free radicals, and indirectly, through the activation of the Nrf2 signaling pathway and the expression of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase, thus protecting cells from oxidative stress [151,165,166,167]. Experimental studies have confirmed these properties and, in particular, treatments with AXT have shown a clinically significant reduction in the levels of C-reactive protein (CRP) and 8-OHdG (8-hydroxy-2′-deoxyguanosine), a marker of oxidative DNA damage [240,241]. Furthermore, positive regulations of the immune response, such as the activity of T and B lymphocytes, have also been observed [199,242]. AXT represents a promising therapeutic candidate for the treatment of IBD [238]. Its molecular structure has been shown to be important in neutralizing reactive oxygen species (ROS) and reducing oxidative stress, decreasing inflammation and redox imbalance as well as related intestinal damage [208,209,243,244]. In vitro studies have shown that AXT reduces oxidative damage (measured by lipid peroxidation levels) and, at the same time, increases the activity of antioxidant enzymes such as SOD and catalase, thus protecting intestinal epithelial cells from oxidative damage [243,245,246]. The mechanisms of action of AXT, based on the inhibition of NF-κB, a key factor in the release of pro-inflammatory cytokines such as TNF-α and other interleukins, show a decrease in key inflammatory biomarkers, which reflects and confirms an additional anti-inflammatory action [208,209,243,247]. Furthermore, AXT promotes the activation of the transcription factor Nrf2, which protects against oxidative damage and stimulates the expression of detoxifying and antioxidant enzymes, thus providing an additional level of protection during inflammatory processes [243]. As mentioned above, the action of AXT has also been associated with the modulation of important interleukin pathways, such as TNF-α, IL-1, IL-6, IL-17 and IL-23 [209,248]. Kim et al. have shown that AXT treatment reduced the production of TNF-α and IL-6 in macrophages, thanks to the inhibition of the NF-κB pathway [249]. AXT has also been observed to reduce the overproduction of IL-1β, a pro-inflammatory cytokine crucial in the pathogenesis of IBD [250]. AXT action has shown a significant effect on the differentiation of helper T cells (Th) in IBD, modulating the ratio between Th1 and Th2 responses [251,252]. In IBD, in fact, a dominant Th1 response is generally associated with colonic inflammation, while a deviation towards Th2 favors tissue repair and damage interruption [72,253,254]. Other observations have highlighted an immunomodulatory activity, suppressing pro-inflammatory cytokines such as TNF-α and inducing the production of anti-inflammatory cytokines such as IL-4 and IL-10 [123,219]. These data suggest that AXT is an effective therapeutic agent for the treatment of IBD, thanks to its ability to selectively modulate T cell differentiation and cytokine profile [123,219]. Its biological activity is expressed not only through a protective antioxidant mechanism but also through a balancing system between Th1 and Th2 responses, relevant for the reduction of IBD-associated inflammation [123,218,219]. AXT also acts on intestinal epithelial cells, helping to preserve the integrity of the intestinal barrier [255,256]. Indeed, the benefits of AXT include improving intestinal barrier function and strengthening epithelial cells to maintain the integrity of tight junctions, thus preventing increased intestinal permeability [123,257,258]. Its influence on the expression of proteins such as occludin and claudin strengthens barrier function, preventing the onset of IBD symptoms and reducing the severity of the disease, through the inhibition of mucosal erosion and ulceration [245,246]. The action of AXT allows the maintenance of the mucosal barrier, essential to prevent the translocation of antigens and pro-inflammatory agents from the intestinal lumen to the underlying tissues [255,259,260]. Numerous in vitro and in vivo models have confirmed its anti-inflammatory and protective effects against colitis [123,261,262]. In animal models, supplementation with AXT inhibited the development of colitis symptoms, significantly reducing the expression of pro-inflammatory cytokines such as TNF-α and IL-6, improving colonic histology and increasing mucosal integrity [123,205,261,262,263]. AXT treatment also reduced disease activity and mucosal damage in mice with DSS-induced colitis, correlated with lower levels of oxidative stress markers and pro-inflammatory cytokines [123,205,224,261,262,263,264]. Finally, AXT reduces NO release from macrophages, as excessive NO production can prolong inflammation and increase tissue damage, reducing oxidative stress and the resulting inflammatory burden [235,249]. Further confirmation shows that AXT treatment led to a marked decrease in inflammatory biomarkers and the activation of proteins involved in the formation of tight junctions in colonic epithelial cells, further strengthening its protective effect against inflammation and oxidative stress [265,266,267]. The positive effects of AXT use in IBD have also focused on its effects on the microbiota. Fang Liu et al. showed that krill oil, rich in ω-3 PUFA and AXT, modulates the intestinal microbiome and metabolome by attenuating inflammation and promoting the resolution of colitis in in vitro and in vivo models and normal intestinal eubiosis [268]. In several in vivo studies on murine models, AXT obtained from algal extracts promoted the propagation of beneficial microbial populations such as Bacteroidia, Bacilli, Clostridia, and Verrucomicrobia in the intestine, modulating microbial signatures and regulating metabolic homeostasis [269,270]. Wang et al. investigated not only the therapeutic but also preventive effects of AXT which, with the additional use of probiotics, prevented intestinal dysbiosis, oxidative stress, inflammation, and metabolic disorders, all key factors in IBD [270,271]. In murine models of diet-induced obesity, AXT improved lipid profiles and liver function, increased the activity of antioxidant enzymes, and reduced levels of reactive oxygen species. In parallel, a modulation of the intestinal microbiota has been observed, with an increase in microorganisms with beneficial effects on intestinal homeostasis [271]. Other evidence has highlighted the role of AXT as a modulator of numerous intracellular pathways related to oxidative stress and inflammation, such as NF-κB, Nrf2, PI3K/Akt, JAK-STAT, and MAPK signaling, reducing the production of pro-inflammatory cytokines, increasing the antioxidant response, and limiting apoptotic processes [231,272]. Finally, AXT has been shown to be useful in preserving the integrity of the intestinal mucosa in immunosuppressed mouse models, reducing oxidative stress and promoting a more balanced microbial flora, with higher levels of beneficial bacteria and an increased production of short-chain fatty acids [272]. AXT, therefore, has demonstrated promising therapeutic potential for regulating IBD symptoms and related pathologies, with a pronounced protective activity of the intestinal environment [123,273,274]. In conclusion, AXT is an interesting candidate for the treatment of IBD, thanks to the regulation of interleukin pathways, the strengthening of the intestinal barrier, and the suppression of NO production in macrophages [244,264,275]. Evidence suggests that AXT could be used as a dietary adjuvant alongside current IBD therapies [276]. The numerous mechanisms of action of AXT—in particular its antioxidant and anti-inflammatory activity—confirm its potential as a therapeutic agent for the management of IBD [238,244]. In recent years, the therapeutic role of microalgae in modulating the intestinal microbiota as a source of prebiotics has been emerging, thanks to their rich nutritional profile. Bioactive compounds derived from microalgae include polysaccharides, bioactive proteins and peptides, sterols, terpenoids, polyphenols and cyclic polysulphide compounds. Many studies highlight those microalgae activate the immune system, protect the defensive capacity and increase anti-infective activities. Microalgae derivatives have antiviral, antibacterial, anticancer, immunomodulatory and antioxidant properties [277]. Among the most important microalgal derivatives with high antioxidant properties are terpenes [278]. Studies show that β-carotene is capable of suppressing lipid peroxidation, inflammation caused by oxidative stress, and tumour formation [279]. For example, Red algae produce halogen, a monoterpene with high anti-tumour activity [280]. In addition, microalgae produce high amounts of vitamins, including α-tocopherols, which, in addition to being the most potent antioxidants, play a key role in the prevention of neurodegenerative diseases [279]. Short-chain fatty acids (SCFAs) play a very important role in the interaction between the microbiome and the immune system. These metabolites are produced during the colon filtration process by the microbiota. In an in vitro simulated gut model, the fermentation activity of gut microbes of the microalgae Chlorella vulgaris and its compounds increased SCFA production. Studies show that the presence of SCFAs, particularly acetic, propionic, butyric and isobutyric acids, produced by beneficial gut bacteria, is indicative of good health. A decrease in bacteria causing dysbiosis, such as Staphylococcus, Enterococcus and Enterobacteriaceae, has been observed with fatty acid production [281]. On the other hand, polyunsaturated fatty acids (PUFAs) produced by microalgae, especially omega-3 and DHA, have attracted considerable interest in the potential treatment of IBD. PUFAs have been observed to block inflammation, normalise the immune response, and protect intestinal epithelial cells, thereby helping to maintain microbiome balance [273]. Other microalgal compounds with therapeutic potential for IBD include tocopherols and phycobiliproteins. Furthermore, the therapeutic effect of phycocyanin and phycocyanobilin has been studied in mouse models of colitis, demonstrating anti-inflammatory and antioxidant activity. The inhibitory effect of phycocyanin and phycocyanobilin on IgE activity, reduction of vascular permeability and inflammation through low expression levels of inflammatory genes and pro-inflammatory cytokines (TNF-α) in the gut and systemic circulation was observed [281]. IBD represents an interesting model for the complex interactions between the microbiota and the immune system in the context of chronic inflammation. In this regard, microalgae, thanks to their high levels of bioactive compounds, have emerged as agents for managing inflammatory conditions. Some microalgae species, such as Spirulina platensis, Chlorella vulgaris, and Phaeodactylum tricornutum, are rich in bioactive compounds such as phycocyanin, polysaccharides, polyphenols, and unsaturated fatty acids, with anti-inflammatory, antioxidant, and immunomodulatory properties and useful for managing IBD [281,282]. Among the most commonly present compounds, AXT is a promising candidate for modulating inflammation and oxidative stress. Haematococcus lacustris is currently the main natural source of this carotenoid, representing a significant percentage of its dry weight.
The possibility of engineering microalgae to produce larger quantities of metabolites makes these organisms of particular scientific and biotechnological importance. Clinical studies have shown that oral administration of astaxanthin is the most common, with a recommended dose of 4–12 mg to evaluate its anxiolytic effects, which can be adjusted depending on the desired effect [133]. Clinical studies have also shown that doses of 12–24 mg have been used without signs of marked toxicity.
Preliminary studies suggest that dietary supplementation with microalgae may promote interactions with the microbiota and stimulate the development of anti-inflammatory molecules derived from fermentation processes. Recent studies have suggested that, despite the high carbohydrate content of seaweed and microalgae, most of them are not digested in the gastrointestinal tract and therefore act as dietary fiber. Alginates, fucoids, carrageenans, xylans, etc., are soluble fibers that are not completely fermented by the microbiota into short-chain fatty acids, despite their ability to positively regulate intestinal metabolism. Other algal polysaccharides (EPs) can instead act as insoluble fibers (cellulose, mannans, β-glucans, lignin, etc.). Microalgal PSs may provide health benefits and can be used as emerging prebiotics [283].
In recent years, oxylipins have attracted the attention of the scientific community. These molecules are derived from the enzymatic or non-enzymatic oxidation of PUFAs and play a dual role in IBD. While some species are pro-inflammatory, others, such as the resolvins discovered by Serhan et al., actively promote its resolution [181]. Pro-resolving oxylipins, in fact, modulate the immune system and the intestinal microbiota, as evidenced by an in vivo study by Arita et al. [284]. In this context, microalgae represent a sustainable source of precursor fatty acids and a natural reservoir for novel lipid metabolites such as oxylipins, offering a nutritional strategy to modulate inflammation and the microbiota [285,286].

9. Conclusions

The gut microbiota plays a key role in the proper functioning of the body. Several studies have confirmed how the microbiota influences the onset, rate of progression and treatment of tumors. The interaction of the microbiota and cells of the immune system generates a microenvironment that when altered causes loss of homeostasis and tumor initiation. Tumors develop through cells that ignore tolerance rules and escape tumor surveillance mechanisms. Studies have identified defensive mechanisms by the microbiota such as antigen mimicry and transformation of chemotherapeutic agents. Despite numerous studies, the microbiota–immune system–tumor interaction presents complex mechanisms that bring different research teams to attention. Insight into these processes would lead to a clearer understanding and better management of treatments.

Author Contributions

Conceptualization, R.P.R., A.S. and G.M.; methodology, V.D.F., V.I. and F.P.; software, V.D.F.; validation, R.P.R. and M.D.; formal analysis, V.I. and F.P.; investigation V.D.F.; resources, V.I., F.P. and V.D.F.; data curation, R.P.R.; writing—original draft preparation, V.I., F.P., V.D.F. and M.D.; writing—review and editing, R.P.R. and M.D.; visualization, R.P.R.; supervision, A.S.; project administration, G.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

Author Rosa Paola Radice was employed by the company AlgaeBioMed Srl. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Frost, F. Einführung in das Mikrobiom. Introduction to the microbiome. Die Inn. Med. 2022, 63, 1015–1021. [Google Scholar] [CrossRef]
  2. Ross, S.M. Microbiota and the Immune System, Part 2: Mediterranean Dietary Influences on Microbiota. Holist. Nurs. Pract. 2024, 38, 58–61. [Google Scholar] [CrossRef]
  3. Thriene, K.; Michels, K.B. Human Gut Microbiota Plasticity throughout the Life Course. Int. J. Environ. Res. Public Health 2023, 20, 1463. [Google Scholar] [CrossRef]
  4. Ahrodia, T.; Das, S.; Bakshi, S.; Das, B. Structure, functions, and diversity of the healthy human microbiome. Prog. Mol. Biol. Transl. Sci. 2022, 191, 53–82. [Google Scholar] [CrossRef]
  5. Kort, R.; Caspers, M.; van de Graaf, A.; van Egmond, W.; Keijser, B.; Roeselers, G. Shaping the oral microbiota through intimate kissing. Microbiome 2014, 2, 41. [Google Scholar] [CrossRef]
  6. Pérez-Cano, F.J. Mediterranean Diet, Microbiota and Immunity. Nutrients 2022, 14, 273. [Google Scholar] [CrossRef]
  7. García-Montero, C.; Fraile-Martínez, O.; Gómez-Lahoz, A.M.; Pekarek, L.; Castellanos, A.J.; Noguerales-Fraguas, F.; Coca, S.; Guijarro, L.G.; García-Honduvilla, N.; Asúnsolo, A.; et al. Nutritional components in western diet versus mediterranean diet at the gut microbiota-immune system interplay. implications for health and disease. Nutrients 2021, 13, 699. [Google Scholar] [CrossRef]
  8. Ghosh, S.; Pramanik, S. Structural diversity, functional aspects and future therapeutic applications of human gut microbiome. Arch. Microbiol. 2021, 203, 5281–5308. [Google Scholar] [CrossRef]
  9. Ruan, W.; Engevik, M.A.; Spinler, J.K.; Versalovic, J. Healthy Human Gastrointestinal Microbiome: Composition and Function After a Decade of Exploration. Dig. Dis. Sci. 2020, 65, 695–705. [Google Scholar] [CrossRef]
  10. Mertz, L. Finding New Ways to Analyze the Microbiome. IEEE Pulse 2022, 13, 26–30. [Google Scholar] [CrossRef]
  11. Marcolungo, L.; Bellamoli, F.; Cecchin, M.; Lopatriello, G.; Rossato, M.; Cosentino, E.; Rombauts, S.; Delledonne, M.; Ballottari, M. Haematococcus lacustris genome assembly and annotation reveal diploid genetic traits and stress-induced gene expression patterns. Algal Res. 2024, 80, 103567. [Google Scholar] [CrossRef]
  12. Groussin, M.; Poyet, M.; Sistiaga, A.; Kearney, S.M.; Moniz, K.; Noel, M.; Hooker, J.; Gibbons, S.M.; Segurel, L.; Froment, A.; et al. Elevated rates of horizontal gene transfer in the industrialized human microbiome. Cell 2021, 184, 2053–2067.e18. [Google Scholar] [CrossRef]
  13. Vatanen, T.; Jabbar, K.S.; Ruohtula, T.; Honkanen, J.; Avila-Pacheco, J.; Siljander, H.; Stražar, M.; Oikarinen, S.; Hyöty, H.; Ilonen, J.; et al. Mobile genetic elements from the maternal microbiome shape infant gut microbial assembly and metabolism. Cell 2022, 185, 4921–4936.e15. [Google Scholar] [CrossRef]
  14. Colella, M.; Charitos, I.A.; Ballini, A.; Cafiero, C.; Topi, S.; Palmirotta, R.; Santacroce, L. Microbiota revolution: How gut microbes regulate our lives. World J. Gastroenterol. 2023, 29, 4368–4383. [Google Scholar] [CrossRef]
  15. Gomaa, E.Z. Human gut microbiota/microbiome in health and diseases: A review. Antonie Van Leeuwenhoek 2020, 113, 2019–2040. [Google Scholar] [CrossRef]
  16. Álvarez, J.; Real, J.M.F.; Guarner, F.; Gueimonde, M.; Rodríguez, J.M.; de Pipaon, M.S.; Sanz, Y. Microbiota intestinal y salud. Gut microbes and health. Gastroenterol. Y Hepatol. 2021, 44, 519–535. [Google Scholar] [CrossRef]
  17. Hu, Y.; Chen, Z.; Xu, C.; Kan, S.; Chen, D. Disturbances of the Gut Microbiota and Microbiota-Derived Metabolites in Inflammatory Bowel Disease. Nutrients 2022, 14, 5140. [Google Scholar] [CrossRef]
  18. Qiu, P.; Ishimoto, T.; Fu, L.; Zhang, J.; Zhang, Z.; Liu, Y. The Gut Microbiota in Inflammatory Bowel Disease. Front. Cell. Infect. Microbiol. 2022, 12, 733992. [Google Scholar] [CrossRef]
  19. Tojo, R.; Suárez, A.; Clemente, M.G.; de los Reyes-Gavilán, C.G.; Margolles, A.; Gueimonde, M.; Ruas-Madiedo, P. Intestinal microbiota in health and disease: Role of bifidobacteria in gut homeostasis. World J. Gastroenterol. 2014, 20, 15163–15176. [Google Scholar] [CrossRef]
  20. Wang, P.X.; Deng, X.R.; Zhang, C.H.; Yuan, H.J. Gut microbiota and metabolic syndrome. Chin. Med J. 2020, 133, 808–816. [Google Scholar] [CrossRef]
  21. Bishehsari, F.; Voigt, R.M.; Keshavarzian, A. Circadian rhythms and the gut microbiota: From the metabolic syndrome to cancer. Nat. Rev. Endocrinol. 2020, 16, 731–739. [Google Scholar] [CrossRef]
  22. Al Bander, Z.; Nitert, M.D.; Mousa, A.; Naderpoor, N. The gut microbiota and inflammation: An overview. Int. J. Environ. Res. Public Health 2020, 17, 7618. [Google Scholar] [CrossRef]
  23. Zhang, P. Influence of Foods and Nutrition on the Gut Microbiome and Implications for Intestinal Health. Int. J. Mol. Sci. 2022, 23, 9588. [Google Scholar] [CrossRef]
  24. Illiano, P.; Brambilla, R.; Parolini, C. The mutual interplay of gut microbiota, diet and human disease. FEBS J. 2020, 287, 833–855. [Google Scholar] [CrossRef]
  25. Ames, S.R.; Lotoski, L.C.; Azad, M.B. Comparing early life nutritional sources and human milk feeding practices: Personalized and dynamic nutrition supports infant gut microbiome development and immune system maturation. Gut Microbes 2023, 15, 2190305. [Google Scholar] [CrossRef]
  26. Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; De Lorenzo, A. Influence of mediterranean diet on human gut microbiota. Nutrients 2021, 13, 7. [Google Scholar] [CrossRef]
  27. Rinninella, E.; Tohumcu, E.; Raoul, P.; Fiorani, M.; Cintoni, M.; Mele, M.C.; Cammarota, G.; Gasbarrini, A.; Ianiro, G. The role of diet in shaping human gut microbiota. Best Pr. Res. Clin. Gastroenterol. 2023, 62–63, 101828. [Google Scholar] [CrossRef]
  28. Wiertsema, S.P.; van Bergenhenegouwen, J.; Garssen, J.; Knippels, L.M.J. The interplay between the gut microbiome and the immune system in the context of infectious diseases throughout life and the role of nutrition in optimizing treatment strategies. Nutrients 2021, 13, 886. [Google Scholar] [CrossRef]
  29. Li, D.; Li, Y.; Yang, S.; Lu, J.; Jin, X.; Wu, M. Diet-gut microbiota-epigenetics in metabolic diseases: From mechanisms to therapeutics. Biomed. Pharmacother. 2022, 153, 113290. [Google Scholar] [CrossRef]
  30. Beam, A.; Clinger, E.; Hao, L. Effect of diet and dietary components on the composition of the gut microbiota. Nutrients 2021, 13, 2795. [Google Scholar] [CrossRef]
  31. Perler, B.K.; Friedman, E.S.; Wu, G.D. The Role of the Gut Microbiota in the Relationship Between Diet and Human Health. Annu. Rev. Physiol. 2023, 85, 449–468. [Google Scholar] [CrossRef]
  32. Tong, Y.; Gao, H.; Qi, Q.; Liu, X.; Li, J.; Gao, J.; Li, P.; Wang, Y.; Du, L.; Wang, C. High fat diet, gut microbiome and gastrointestinal cancer. Theranostics 2021, 11, 5889–5910. [Google Scholar] [CrossRef]
  33. Daschner, P.J.; Ross, S.; Seifried, H.; Kumar, A.; Flores, R. Nutrition and Microbiome Interactions in Human Cancer. J. Acad. Nutr. Diet. 2023, 123, 504–514. [Google Scholar] [CrossRef]
  34. Tanaka, Y.; Shimizu, S.; Shirotani, M.; Yorozu, K.; Kitamura, K.; Oehorumu, M.; Kawai, Y.; Fukuzawa, Y. Nutrition and cancer risk from the viewpoint of the intestinal microbiome. Nutrients 2021, 13, 3326. [Google Scholar] [CrossRef]
  35. Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N. Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef]
  36. Ávila-Román, J.; Arreaza-Gil, V.; Cortés-Espinar, A.J.; Soliz-Rueda, J.R.; Mulero, M.; Muguerza, B.; Arola-Arnal, A.; Arola, L.; Torres-Fuentes, C. Impact of gut microbiota on plasma oxylipins profile under healthy and obesogenic conditions. Clin. Nutr. 2021, 40, 1475–1486. [Google Scholar] [CrossRef]
  37. Yu, Y.; Raka, F.; Adeli, K. The Role of the Gut Microbiota in Lipid and Lipoprotein Metabolism. J. Clin. Med. 2019, 8, 2227. [Google Scholar] [CrossRef]
  38. Wang, Z.; Koonen, D.; Hofker, M.; Fu, J. Gut microbiome and lipid metabolism: From associations to mechanisms. Curr. Opin. Infect. Dis. 2016, 27, 216–224. [Google Scholar] [CrossRef]
  39. Lv, J.; Zhao, H.-P.; Yu, Y.; Wang, J.-H.; Zhang, X.-J.; Guo, Z.-Q.; Jiang, W.-Y.; Wang, K.; Guo, L. From gut microbial ecology to lipid homeostasis: Decoding the role of gut microbiota in dyslipidemia pathogenesis and intervention. World J. Gastroenterol. 2025, 31, 108680. [Google Scholar] [CrossRef]
  40. Serhan, C.N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014, 510, 92–101. [Google Scholar] [CrossRef]
  41. Dai, C.; Huang, Y.-H.; Jiang, M. Combination therapy in inflammatory bowel disease: Current evidence and perspectives. Int. Immunopharmacol. 2022, 114, 109545. [Google Scholar] [CrossRef]
  42. Brizzi, A.; Rispoli, R.M.; Autore, G.; Marzocco, S. Anti-Inflammatory Effects of Algae-Derived Biomolecules in Gut Health: A Review. Int. J. Mol. Sci. 2025, 26, 885. [Google Scholar] [CrossRef]
  43. Wehkamp, J.; Götz, M.; Herrlinger, K.; Steurer, W.; Stange, E.F. Inflammatory Bowel Disease: Crohn’s disease and ulcerative colitis. Dtsch. Arztebl. Int. 2016, 113, 72–82. [Google Scholar] [CrossRef]
  44. Ngo, B.; Farrell, C.P.; Barr, M.; Wolov, K.; Bailey, R.; Mullin, J.M.; Thornton, J.J. Tumor Necrosis Factor Blockade for Treatment of Inflammatory Bowel Disease: Efficacy and Safety. Curr. Mol. Pharmacol. 2010, 3, 145–152. [Google Scholar] [CrossRef]
  45. Van Assche, G.; Herrmann, K.A.; Louis, E.; Everett, S.M.; Colombel, J.-F.; Rahier, J.-F.; Vanbeckevoort, D.; Meunier, P.; Tolan, D.; Ernst, O.; et al. Effects of infliximab therapy on transmural lesions as assessed by magnetic resonance enteroclysis in patients with ileal Crohn’s disease. J. Crohn’s Colitis 2013, 7, 950–957. [Google Scholar] [CrossRef]
  46. Singh, N.; Bernstein, C.N. Environmental risk factors for inflammatory bowel disease. United Eur. Gastroenterol. J. 2022, 10, 1047–1053. [Google Scholar] [CrossRef]
  47. Santana, P.T.; Rosas, S.L.B.; Ribeiro, B.E.; Marinho, Y.; de Souza, H.S.P. Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 3464. [Google Scholar] [CrossRef]
  48. Becker, H.E.F.; Demers, K.; Derijks, L.J.J.; Jonkers, D.M.A.E.; Penders, J. Current evidence and clinical relevance of drug-microbiota interactions in inflammatory bowel disease. Front. Microbiol. 2023, 14, 1107976. [Google Scholar] [CrossRef]
  49. Aldars-García, L.; Marin, A.C.; Chaparro, M.; Gisbert, J.P. The interplay between immune system and microbiota in inflammatory bowel disease: A narrative review. Int. J. Mol. Sci. 2021, 22, 3076. [Google Scholar] [CrossRef]
  50. Hrncir, T. Gut Microbiota Dysbiosis: Triggers, Consequences, Diagnostic and Therapeutic Options. Microorganisms 2022, 10, 578. [Google Scholar] [CrossRef]
  51. Zhao, M.; Chu, J.; Feng, S.; Guo, C.; Xue, B.; He, K.; Li, L. Immunological mechanisms of inflammatory diseases caused by gut microbiota dysbiosis: A review. Biomed. Pharmacother. 2023, 164, 114985. [Google Scholar] [CrossRef]
  52. Plichta, D.R.; Graham, D.B.; Subramanian, S.; Xavier, R.J. Therapeutic Opportunities in Inflammatory Bowel Disease: Mechanistic Dissection of Host-Microbiome Relationships. Cell 2019, 178, 1041–1056. [Google Scholar] [CrossRef]
  53. Hemmati, M.A.; Monemi, M.; Asli, S.; Mohammadi, S.; Foroozanmehr, B.; Haghmorad, D.; Oksenych, V.; Eslami, M. Using New Technologies to Analyze Gut Microbiota and Predict Cancer Risk. Cells 2024, 13, 1987. [Google Scholar] [CrossRef]
  54. Maheshwari, K.; Musyuni, P.; Moulick, A.; Mishra, H.; Ekielski, A.; Mishra, P.K.; Aggarwal, G. Unveiling the microbial symphony: Next-Gen sequencing and bioinformatics insights into the human gut microbiome. Health Sci. Rev. 2024, 11, 100173. [Google Scholar] [CrossRef]
  55. Li, L.; Peng, P.; Ding, N.; Jia, W.; Huang, C.; Tang, Y. Oxidative Stress, Inflammation, Gut Dysbiosis: What Can Polyphenols Do in Inflammatory Bowel Disease? Antioxidants 2023, 12, 967. [Google Scholar] [CrossRef]
  56. Neurath, M.F. Targeting cytokines in inflammatory bowel disease. Sci. Transl. Med. 2022, 14, eabq4473. [Google Scholar] [CrossRef]
  57. Sanchez-Muñoz, F.; Dominguez-Lopez, A.; Yamamoto-Furusho, J.K. Role of cytokines in inflammatory bowel disease. World J. Gastroenterol. 2008, 14, 4280–4288. [Google Scholar] [CrossRef]
  58. Waldner, M.J.; Neurath, M.F. Novel cytokine-targeted therapies and intestinal inflammation. Curr. Opin. Pharmacol. 2009, 9, 702–707. [Google Scholar] [CrossRef]
  59. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxidative Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  60. Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef]
  61. Vona, R.; Pallotta, L.; Cappelletti, M.; Severi, C.; Matarrese, P. The impact of oxidative stress in human pathology: Focus on gastrointestinal disorders. Antioxidants 2021, 10, 201. [Google Scholar] [CrossRef]
  62. Afzal, S.; Manap, A.S.A.; Attiq, A.; Albokhadaim, I.; Kandeel, M.; Alhojaily, S.M. From imbalance to impairment: The central role of reactive oxygen species in oxidative stress-induced disorders and therapeutic exploration. Front. Pharmacol. 2023, 14, 1269581. [Google Scholar] [CrossRef]
  63. Morris, G.; Gevezova, M.; Sarafian, V.; Maes, M. Redox regulation of the immune response. Cell. Mol. Immunol. 2022, 19, 1079–1101. [Google Scholar] [CrossRef]
  64. Tian, T.; Wang, Z.; Zhang, J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. Oxidative Med. Cell. Longev. 2017, 2017, 4535194. [Google Scholar] [CrossRef]
  65. Steinbach, E.C.; Plevy, S.E. The role of macrophages and dendritic cells in the initiation of inflammation in IBD. Inflamm. Bowel Dis. 2014, 20, 166–175. [Google Scholar] [CrossRef]
  66. Ramos, G.P.; Papadakis, K.A. Mechanisms of Disease: Inflammatory Bowel Diseases. Mayo Clin. Proc. 2019, 94, 155–165. [Google Scholar] [CrossRef]
  67. Bates, J.; Diehl, L. Dendritic cells in IBD pathogenesis: An area of therapeutic opportunity? J. Pathol. 2014, 232, 112–120. [Google Scholar] [CrossRef]
  68. Liu, Z.; Zhang, Y.; Jin, T.; Yi, C.; Ocansey, D.K.W.; Mao, F. The role of NOD2 in intestinal immune response and microbiota modulation: A therapeutic target in inflammatory bowel disease. Int. Immunopharmacol. 2022, 113, 109466. [Google Scholar] [CrossRef]
  69. Pedros, C.; Duguet, F.; Saoudi, A.; Chabod, M. Disrupted regulatory T cell homeostasis in inflammatory bowel diseases. World J. Gastroenterol. 2016, 22, 974–995. [Google Scholar] [CrossRef]
  70. Laukova, M.; Zaretsky, A.G. Regulatory T cells as a therapeutic approach for inflammatory bowel disease. Eur. J. Immunol. 2023, 53, 2250007. [Google Scholar] [CrossRef]
  71. Heller, F.; Florian, P.; Bojarski, C.; Richter, J.; Christ, M.; Hillenbrand, B.; Mankertz, J.; Gitter, A.H.; Bürgel, N.; Fromm, M.; et al. Interleukin-13 Is the Key Effector Th2 Cytokine in Ulcerative Colitis That Affects Epithelial Tight Junctions, Apoptosis, and Cell Restitution. Gastroenterology 2005, 129, 550–564. [Google Scholar] [CrossRef]
  72. Bamias, G.; Cominelli, F. Role of Th2 immunity in intestinal inflammation. Curr. Opin. Gastroenterol. 2015, 31, 471–476. [Google Scholar] [CrossRef]
  73. Siakavellas, S.I.; Bamias, G. Role of the IL-23/IL-17 axis in Crohn’s disease. Discov. Med. 2012, 14, 253–262. Available online: https://www.discoverymedicine.com/Spyros-I-Siakavellas/2012/10/26/role-of-the-il-23-il-17-axis-in-crohns-disease/ (accessed on 14 July 2025).
  74. Padoan, A.; Musso, G.; Contran, N.; Basso, D. Inflammation, Autoinflammation and Autoimmunity in Inflammatory Bowel Diseases. Curr. Issues Mol. Biol. 2023, 45, 5534–5557. [Google Scholar] [CrossRef]
  75. Paroni, M.; Leccese, G.; Ranzani, V.; Moschetti, G.; Chiara, M.; Perillo, F.; Ferri, S.; Clemente, F.; Noviello, D.; Conforti, F.S.; et al. An Intestinal Th17 Subset is Associated with Inflammation in Crohn’s Disease and Activated by Adherent-invasive Escherichia coli. J. Crohn’s Colitis 2023, 17, 1988–2001. [Google Scholar] [CrossRef]
  76. Jomova, K.; Raptova, R.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch. Toxicol. 2023, 97, 2499–2574. [Google Scholar] [CrossRef]
  77. Muro, P.; Zhang, L.; Li, S.; Zhao, Z.; Jin, T.; Mao, F.; Mao, Z. The emerging role of oxidative stress in inflammatory bowel disease. Front. Endocrinol. 2024, 15, 1390351. [Google Scholar] [CrossRef]
  78. Wang, Y.; Chen, Y.; Zhang, X.; Lu, Y.; Chen, H. New insights in intestinal oxidative stress damage and the health intervention effects of nutrients: A review. J. Funct. Foods 2020, 75, 104248. [Google Scholar] [CrossRef]
  79. Lin, P.Y.; Stern, A.; Peng, H.H.; Chen, J.H.; Yang, H.C. Redox and Metabolic Regulation of Intestinal Barrier Function and Associated Disorders. Int. J. Mol. Sci. 2022, 23, 14463. [Google Scholar] [CrossRef]
  80. Conlin, V.S.; Wu, X.; Nguyen, C.; Dai, C.; Vallance, B.A.; Buchan, A.M.J.; Boyer, L.; Jacobson, K. Vasoactive intestinal peptide ameliorates intestinal barrier disruption associated with Citrobacter rodentium-induced colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 297, G735–G750. [Google Scholar] [CrossRef]
  81. Zhu, L.; Han, J.; Li, L.; Wang, Y.; Li, Y.; Zhang, S. Claudin family participates in the pathogenesis of inflammatory bowel diseases and colitis-associated colorectal cancer. Front. Immunol. 2019, 10, 1441. [Google Scholar] [CrossRef]
  82. Landy, J.; Ronde, E.; English, N.; Clark, S.K.; Hart, A.L.; Knight, S.C.; Ciclitira, P.J.; Al-Hassi, H.O. Tight junctions in inflammatory bowel diseases and inflammatory bowel disease associated colorectal cancer. World J. Gastroenterol. 2016, 22, 3117–3126. [Google Scholar] [CrossRef]
  83. Keshavarzian, A.; Banan, A.; Farhadi, A.; Komanduri, S.; Mutlu, E.; Zhang, Y.; Fields, J.Z. Increases in free radicals and cytoskeletal protein oxidation and nitration in the colon of patients with inflammatory bowel disease. Gut 2003, 52, 720–728. [Google Scholar] [CrossRef]
  84. Linares, R.; Francés, R.; Gutiérrez, A.; Juanola, O. Bacterial Translocation as Inflammatory Driver in Crohn’s Disease. Front. Cell Dev. Biol. 2021, 9, 703310. [Google Scholar] [CrossRef]
  85. Luchetti, M.M.; Ciccia, F.; Avellini, C.; Benfaremo, D.; Rizzo, A.; Spadoni, T.; Svegliati, S.; Marzioni, D.; Santinelli, A.; Costantini, A.; et al. Gut epithelial impairment, microbial translocation and immune system activation in inflammatory bowel disease–Associated spondyloarthritis. Rheumatology 2021, 60, 92–102. [Google Scholar] [CrossRef]
  86. Dijkstra, G.; Moshage, H.; Van Dullemen, H.M.; de Jager-Krikken, A.; Tiebosch, A.T.M.G.; Kleibeuker, J.H.; Jansen, P.L.M.; Van Goor, H. Expression of nitric oxide synthases and formation of nitrotyrosine and reactive oxygen species in inflammatory bowel disease. J. Pathol. 1998, 186, 416–421. [Google Scholar] [CrossRef]
  87. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef]
  88. Ziegler, K.; Kunert, A.T.; Reinmuth-Selzle, K.; Leifke, A.L.; Widera, D.; Weller, M.G.; Schuppan, D.; Fröhlich-Nowoisky, J.; Lucas, K.; Pöschl, U. Chemical modification of pro-inflammatory proteins by peroxynitrite increases activation of TLR4 and NF-κB: Implications for the health effects of air pollution and oxidative stress. Redox Biol. 2020, 37, 101581. [Google Scholar] [CrossRef]
  89. Homer, C.R.; Richmond, A.L.; Rebert, N.A.; Achkar, J.-P.; McDonald, C. W1239 ATG16L1 and NOD2 Interact in an Autophagy-Dependent Anti-Bacterial Pathway Implicated in Crohn’s Disease Pathogenesis. Gastroenterology 2010, 138, S-681. [Google Scholar] [CrossRef]
  90. Sultan, S.; El-Mowafy, M.; Elgaml, A.; Ahmed, T.A.E.; Hassan, H.; Mottawea, W. Metabolic Influences of Gut Microbiota Dysbiosis on Inflammatory Bowel Disease. Front. Physiol. 2021, 12, 715506. [Google Scholar] [CrossRef]
  91. Graham, D.B.; Xavier, R.J. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 2020, 578, 527–539. [Google Scholar] [CrossRef]
  92. Nishida, A.; Inoue, R.; Inatomi, O.; Bamba, S.; Naito, Y.; Andoh, A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin. J. Gastroenterol. 2018, 11, 1–10. [Google Scholar] [CrossRef]
  93. Bhaumik, S.; Mickael, M.E.; Moran, M.; Spell, M.; Basu, R. RORγt Promotes Foxp3 Expression by Antagonizing the Effector Program in Colonic Regulatory T Cells. J. Immunol. 2021, 207, 2027–2038. [Google Scholar] [CrossRef]
  94. Chen, Y.T.; Chang, Y.M.; Chen, Y.L.; Su, Y.H.; Liao, C.C.; Chiang, T.H.; Chen, W.Y.; Su, Y.C. N-ethyl-N-nitrosourea (ENU)-induced C-terminal truncation of Runx3 results in autoimmune colitis associated with Th17/Treg imbalance. Immunol. Lett. 2024, 268, 106869. [Google Scholar] [CrossRef]
  95. Singh, A.K.; Kumar, R.; Yin, J.; Ii, J.F.B.; Kathania, M.; Mukherjee, S.; Kumar, J.; Conlon, K.P.; Basrur, V.; Chen, Z.; et al. RORγt-Raftlin1 complex regulates the pathogenicity of Th17 cells and colonic inflammation. Nat. Commun. 2023, 14, 4972. [Google Scholar] [CrossRef]
  96. Kanai, T.; Mikami, Y.; Sujino, T.; Hisamatsu, T.; Hibi, T. RORγt-dependent IL-17A-producing cells in the pathogenesis of intestinal inflammation. Mucosal Immunol. 2012, 5, 240–247. [Google Scholar] [CrossRef]
  97. Mousa, W.K.; Chehadeh, F.; Husband, S. Microbial dysbiosis in the gut drives systemic autoimmune diseases. Front. Immunol. 2022, 13, 906258. [Google Scholar] [CrossRef]
  98. Liu, Y.; Freeborn, J.; Armbrister, S.A.; Tran, D.Q.; Rhoads, J.M. Treg-associated monogenic autoimmune disorders and gut microbial dysbiosis. Pediatr. Res. 2022, 91, 35–43. [Google Scholar] [CrossRef]
  99. Toor, D.; Wsson, M.K.; Kumar, P.; Karthikeyan, G.; Kaushik, N.K.; Goel, C.; Singh, S.; Kumar, A.; Prakash, H. Dysbiosis disrupts gut immune homeostasis and promotes gastric diseases. Int. J. Mol. Sci. 2019, 20, 2432. [Google Scholar] [CrossRef]
  100. Khan, I.A.; Nayak, B.; Markandey, M.; Bajaj, A.; Verma, M.; Kumar, S.; Singh, M.K.; Kedia, S.; Ahuja, V. Differential prevalence of pathobionts and host gene polymorphisms in chronic inflammatory intestinal diseases: Crohn’s disease and intestinal tuberculosis. PLoS ONE 2021, 16, e0256098. [Google Scholar] [CrossRef]
  101. Zheng, L.; Duan, S.-L.; Dai, Y.-C.; Wu, S.-C. Role of adherent invasive Escherichia coli in pathogenesis of inflammatory bowel disease. World J. Clin. Cases 2022, 10, 11671–11689. [Google Scholar] [CrossRef]
  102. Dunleavy, K.A.; Raffals, L.E.; Camilleri, M. Intestinal Barrier Dysfunction in Inflammatory Bowel Disease: Underpinning Pathogenesis and Therapeutics. Dig. Dis. Sci. 2023, 68, 4306–4320. [Google Scholar] [CrossRef]
  103. Di Tommaso, N.; Gasbarrini, A.; Ponziani, F.R. Intestinal barrier in human health and disease. Int. J. Environ. Res. Public Health 2021, 18, 12836. [Google Scholar] [CrossRef]
  104. Bischoff, S.C.; Barbara, G.; Buurman, W.; Ockhuizen, T.; Schulzke, J.-D.; Serino, M.; Tilg, H.; Watson, A.; Wells, J.M.; Pihlsgård, M.; et al. Intestinal permeability—A new target for disease prevention and therapy. BMC Gastroenterol. 2014, 14, 189. [Google Scholar] [CrossRef]
  105. McGuckin, M.A.; Eri, R.; Simms, L.A.; Florin, T.H.; Radford-Smith, G. Intestinal barrier dysfunction in inflammatory bowel diseases. Inflamm. Bowel Dis. 2009, 15, 100–113. [Google Scholar] [CrossRef]
  106. Okeke, E.B.; Uzonna, J.E. The pivotal role of regulatory T cells in the regulation of innate immune cells. Front. Immunol. 2019, 10, 680. [Google Scholar] [CrossRef]
  107. Goswami, T.K.; Singh, M.; Dhawan, M.; Mitra, S.; Bin Emran, T.; Rabaan, A.A.; Al Mutair, A.; Al Alawi, Z.; Alhumaid, S.; Dhama, K. Regulatory T cells (Tregs) and their therapeutic potential against autoimmune disorders—Advances and challenges. Hum. Vaccines Immunother. 2022, 18, 2035117. [Google Scholar] [CrossRef]
  108. Shu, P.; Liang, H.; Zhang, J.; Lin, Y.; Chen, W.; Zhang, D. Reactive oxygen species formation and its effect on CD4+ T cell-mediated inflammation. Front. Immunol. 2023, 14, 1199233. [Google Scholar] [CrossRef]
  109. Zhao, S.S.; Robertson, S.; Reich, T.; Harrison, N.L.; Moots, R.J.; Goodson, N.J. Prevalence and impact of comorbidities in axial spondyloarthritis: Systematic review and meta-analysis. Rheumatology 2020, 59, iv47–iv57. [Google Scholar] [CrossRef]
  110. Peyrin-Biroulet, L.; Sandborn, W.J.; Panaccione, R.; Domènech, E.; Pouillon, L.; Siegmund, B.; Danese, S.; Ghosh, S. Tumour necrosis factor inhibitors in inflammatory bowel disease: The story continues. Ther. Adv. Gastroenterol. 2021, 14, 17562848211059954. [Google Scholar] [CrossRef]
  111. González-Lama, Y.; Vera, M.I.; Calvo, M.; Abreu, L. Marcadores de evolución de la enfermedad inflamatoria intestinal tratada con inmunomoduladores o agentes biológicos. Markers of the course of inflammatory bowel disease treated with immunomodulators or biological agents. Gastroenterol. Y Hepatol. 2010, 33, 449–460. [Google Scholar] [CrossRef]
  112. Pugliese, D.; Privitera, G.; Fiorani, M.; Parisio, L.; Calvez, V.; Papa, A.; Gasbarrini, A.; Armuzzi, A. Targeting IL12/23 in ulcerative colitis: Update on the role of ustekinumab. Ther. Adv. Gastroenterol. 2022, 15, 17562848221102283. [Google Scholar] [CrossRef]
  113. Verstockt, B.; Salas, A.; Sands, B.E.; Abraham, C.; Leibovitzh, H.; Neurath, M.F.; Vande Casteele, N.; Alimentiv Translational Research Consortium (ATRC). IL-12 and IL-23 pathway inhibition in inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 433–446. [Google Scholar] [CrossRef]
  114. Toussirot, E. The IL23/Th17 pathway as a therapeutic target in chronic inflammatory diseases. Inflamm. Allergy-Drug Targets 2012, 11, 159–168. [Google Scholar] [CrossRef]
  115. Tang, C.; Chen, S.; Qian, H.; Huang, W. Interleukin-23: As a drug target for autoimmune inflammatory diseases. Immunology 2012, 135, 112–124. [Google Scholar] [CrossRef]
  116. Hazel, K.; O’Connor, A. Emerging treatments for inflammatory bowel disease. Ther. Adv. Chronic Dis. 2020, 11, 2040622319899297. [Google Scholar] [CrossRef]
  117. Higashiyama, M.; Hokaria, R. New and Emerging Treatments for Inflammatory Bowel Disease. Digestion 2022, 104, 74–81. [Google Scholar] [CrossRef]
  118. Dai, A.; Kim, S.J. Systemic calcineurin inhibitors tacrolimus and voclosporin: A review of off-label dermatologic uses. J. Am. Acad. Dermatol. 2024, 90, 358–367. [Google Scholar] [CrossRef]
  119. Paramsothy, S.; Rosenstein, A.K.; Mehandru, S.; Colombel, J.-F. The current state of the art for biological therapies and new small molecules in inflammatory bowel disease. Mucosal Immunol. 2018, 11, 1558–1570. [Google Scholar] [CrossRef]
  120. Parigi, T.L.; D’AMico, F.; Abreu, M.T.; Dignass, A.; Dotan, I.; Magro, F.; Griffiths, A.M.; Jairath, V.; Iacucci, M.; Mantzaris, G.J.; et al. Difficult-to-treat inflammatory bowel disease: Results from an international consensus meeting. Lancet Gastroenterol. Hepatol. 2023, 8, 853–859. [Google Scholar] [CrossRef]
  121. Moura, F.A.; de Andrade, K.Q.; dos Santos, J.C.F.; Araújo, O.R.P.; Goulart, M.O.F. Antioxidant therapy for treatment of inflammatory bowel disease: Does it work? Redox Biol. 2015, 6, 617–639. [Google Scholar] [CrossRef]
  122. Pantalos, G.; Vaou, N.; Papachristidou, S.; Stavropoulou, E.; Tsigalou, C.; Voidarou, C.; Bezirtzoglou, E. Antioxidant and Anti-Inflammatory Phytochemicals for the Treatment of Inflammatory Bowel Disease: A Systematic Review. Appl. Sci. 2024, 14, 2177. [Google Scholar] [CrossRef]
  123. Sakai, S.; Nishida, A.; Ohno, M.; Inatomi, O.; Bamba, S.; Sugimoto, M.; Kawahara, M.; Andoh, A. Astaxanthin, a xanthophyll carotenoid, prevents development of dextran sulphate sodium-induced murine colitis. J. Clin. Biochem. Nutr. 2019, 64, 66–72. [Google Scholar] [CrossRef]
  124. Maoka, T. Carotenoids as natural functional pigments. J. Nat. Med. 2020, 74, 1–16. [Google Scholar] [CrossRef]
  125. Swapnil, P.; Meena, M.; Singh, S.K.; Dhuldhaj, U.P.; Harish; Marwal, A. Vital roles of carotenoids in plants and humans to deteriorate stress with its structure, biosynthesis, metabolic engineering and functional aspects. Curr. Plant Biol. 2021, 26, 100203. [Google Scholar] [CrossRef]
  126. Mordi, R.C.; Ademosun, O.T.; Ajanaku, C.O.; Olanrewaju, I.O.; Walton, J.C. Free radical mediated oxidative degradation of carotenes and xanthophylls. Molecules 2020, 25, 1038. [Google Scholar] [CrossRef]
  127. Ramel, F.; Birtic, S.; Cuiné, S.; Triantaphylidès, C.; Ravanat, J.-L.; Havaux, M. Chemical quenching of singlet oxygen by carotenoids in plants. Plant Physiol. 2012, 158, 1267–1278. [Google Scholar] [CrossRef]
  128. Pérez-Gálvez, A.; Viera, I.; Roca, M. Carotenoids and chlorophylls as antioxidants. Antioxidants 2020, 9, 505. [Google Scholar] [CrossRef]
  129. Singh, L.; Prusty, D.; Behera, M.; Perveen, K.; Bukhari, N.A. Optimizing pH and Light for Enhanced Carotenoid Synthesis and Antioxidant Properties in Sub-Aerial Cyanobacteria. J. Basic Microbiol. 2024, 65, e2400570. [Google Scholar] [CrossRef]
  130. Bakac, E.R.; Percin, E.; Gunes-Bayir, A.; Dadak, A. A Narrative Review: The Effect and Importance of Carotenoids on Aging and Aging-Related Diseases. Int. J. Mol. Sci. 2023, 24, 15199. [Google Scholar] [CrossRef]
  131. Pereira, A.G.; Otero, P.; Echave, J.; Carreira-Casais, A.; Chamorro, F.; Collazo, N.; Jaboui, A.; Lourenço-Lopes, C.; Simal-Gandara, J.; Prieto, M.A. Xanthophylls from the Sea: Algae as Source of Bioactive Carotenoids. Mar. Drugs 2021, 19, 188. [Google Scholar] [CrossRef]
  132. Pružinská, K.; Chrastina, M.; Khademnematolahi, S.; Vyletelová, V.; Gajdošová, L.; Pastvová, L.; Dráfi, F.; Poništ, S.; Pašková, Ľ.; Kucharská, J.; et al. Astaxanthin, Compared to Other Carotenoids, Increases the Efficacy of Methotrexate in Rat Adjuvant Arthritis. Int. J. Mol. Sci. 2024, 25, 8710. [Google Scholar] [CrossRef]
  133. Medoro, A.; Davinelli, S.; Milella, L.; Willcox, B.J.; Allsopp, R.C.; Scapagnini, G.; Willcox, D.C. Dietary Astaxanthin: A Promising Antioxidant and Anti-Inflammatory Agent for Brain Aging and Adult Neurogenesis. Mar. Drugs 2023, 21, 643. [Google Scholar] [CrossRef]
  134. Eroglu, A.; Al’aBri, I.S.; Kopec, R.E.; Crook, N.; Bohn, T. Carotenoids and Their Health Benefits as Derived via Their Interactions with Gut Microbiota. Adv. Nutr. Int. Rev. J. 2023, 14, 238–255. [Google Scholar] [CrossRef]
  135. Rocha, H.R.; Coelho, M.C.; Gomes, A.M.; Pintado, M.E. Carotenoids Diet: Digestion, Gut Microbiota Modulation, and Inflammatory Diseases. Nutrients 2023, 15, 2265. [Google Scholar] [CrossRef]
  136. Zhang, Y.; Song, J.; Wang, H.; Li, Y.; Wu, C. Advances in Carotenoid Based Delivery Systems for Alleviating Inflammatory Bowel Disease. Food Rev. Int. 2023, 40, 2427–2447. [Google Scholar] [CrossRef]
  137. Furfaro, F.; Alfarone, L.; Gilardi, D.; Correale, C.; Allocca, M.; Fiorino, G.; Argollo, M.; Zilli, A.; Zacharopoulou, E.; Loy, L.; et al. TL1A: A New Potential Target in the Treatment of Inflammatory Bowel Disease. Curr. Drug Targets 2021, 22, 760–769. [Google Scholar] [CrossRef]
  138. Naik, R.G.; Purcell, S.A.; Gold, S.L.; Christiansen, V.; D’aloisio, L.D.; Raman, M.; Haskey, N. From Evidence to Practice: A Narrative Framework for Integrating the Mediterranean Diet into Inflammatory Bowel Disease Management. Nutrients 2025, 17, 470. [Google Scholar] [CrossRef]
  139. Kogut, M.H.; Lee, A.; Santin, E. Microbiome and pathogen interaction with the immune system. Poult. Sci. 2020, 99, 1906–1913. [Google Scholar] [CrossRef]
  140. Hitch, T.C.A.; Hall, L.J.; Walsh, S.K.; Leventhal, G.E.; Slack, E.; de Wouters, T.; Walter, J.; Clavel, T. Microbiome-based interventions to modulate gut ecology and the immune system. Mucosal Immunol. 2022, 15, 1095–1113. [Google Scholar] [CrossRef]
  141. Lukacova, I.; Ambro, L.; Dubayova, K.; Marekova, M. The gut microbiota, its relationship to the immune system, and possibilities of its modulation. Epidemiol. Mikrobiol. Imunol. 2023, 72, 40–53. Available online: https://www.prolekare.cz/en/journals/epidemiology-microbiology-immunology/2023-1-23/the-gut-microbiota-its-relationship-to-the-immune-system-and-possibilities-of-its-modulation-134140 (accessed on 14 July 2025).
  142. Barrea, L.; Muscogiuri, G.; Frias-Toral, E.; Laudisio, D.; Pugliese, G.; Castellucci, B.; Garcia-Velasquez, E.; Savastano, S.; Colao, A. Nutrition and immune system: From the Mediterranean diet to dietary supplementary through the microbiota. Crit. Rev. Food Sci. Nutr. 2020, 61, 3066–3090. [Google Scholar] [CrossRef]
  143. Said, E.A.; Al-Jabri, Z.; Al-Reesi, I.; Al-Shizawi, N.; Al-Balushi, M.S.; Al-Jabri, A.A. Effects of Bacterial Metabolites on the Immune System: Enemies and Friends. Endocrine, Metab. Immune Disord.—Drug Targets 2022, 22, 1167–1177. [Google Scholar] [CrossRef]
  144. Amoroso, C.; Perillo, F.; Strati, F.; Fantini, M.; Caprioli, F.; Facciotti, F. The Role of Gut Microbiota Biomodulators on Mucosal Immunity and Intestinal Inflammation. Cells 2020, 9, 1234. [Google Scholar] [CrossRef]
  145. Boccuto, L.; Tack, J.; Ianiro, G.; Abenavoli, L.; Scarpellini, E. Human Genes Involved in the Interaction between Host and Gut Microbiome: Regulation and Pathogenic Mechanisms. Genes 2023, 14, 857. [Google Scholar] [CrossRef]
  146. Mahjoub, Y.; Martino, D. Immunology and microbiome: Implications for motor systems. Handb. Clin. Neurol. 2023, 195, 135–157. [Google Scholar] [CrossRef]
  147. Graham, D.B.; Xavier, R.J. Conditioning of the immune system by the microbiome. Trends Immunol. 2023, 44, 499–511. [Google Scholar] [CrossRef]
  148. Negi, S.; Das, D.K.; Pahari, S.; Nadeem, S.; Agrewala, J.N. Potential role of gut microbiota in induction and regulation of innate immune memory. Front. Immunol. 2019, 10, 2441. [Google Scholar] [CrossRef]
  149. Rio, P.; Gasbarrini, A.; Gambassi, G.; Cianci, R. Pollutants, microbiota and immune system: Frenemies within the gut. Front. Public Health 2024, 12, 1285186. [Google Scholar] [CrossRef]
  150. Yoo, J.Y.; Groer, M.; Dutra, S.V.O.; Sarkar, A.; McSkimming, D.I. Gut microbiota and immune system interactions. Microorganisms 2020, 8, 1587. [Google Scholar] [CrossRef]
  151. Pabst, O.; Nowosad, C.R. B cells and the intestinal microbiome in time, space and place. Semin. Immunol. 2023, 69, 101806. [Google Scholar] [CrossRef]
  152. Giambra, V.; Pagliari, D.; Rio, P.; Totti, B.; Di Nunzio, C.; Bosi, A.; Giaroni, C.; Gasbarrini, A.; Gambassi, G.; Cianci, R. Gut Microbiota, Inflammatory Bowel Disease, and Cancer: The Role of Guardians of Innate Immunity. Cells 2023, 12, 2654. [Google Scholar] [CrossRef]
  153. Miniet, A.A.; Grunwell, J.R.; Coopersmith, C.M. The microbiome and the immune system in critical illness. Curr. Opin. Crit. Care 2021, 27, 157–163. [Google Scholar] [CrossRef]
  154. Mattiola, I.; Diefenbach, A. Regulation of innate immune system function by the microbiome: Consequences for tumor immunity and cancer immunotherapy. Semin. Immunol. 2023, 66, 101724. [Google Scholar] [CrossRef]
  155. Ge, Y.; Wang, X.; Guo, Y.; Yan, J.; Abuduwaili, A.; Aximujiang, K.; Yan, J.; Wu, M. Gut microbiota influence tumor development and Alter interactions with the human immune system. J. Exp. Clin. Cancer Res. 2021, 40, 42. [Google Scholar] [CrossRef]
  156. Xie, Y.; Liu, F. The role of the gut microbiota in tumor, immunity, and immunotherapy. Front. Immunol. 2024, 15, 1410928. [Google Scholar] [CrossRef]
  157. Park, J.S.; Gazzaniga, F.S.; Kasper, D.L.; Sharpe, A.H. Microbiota-dependent regulation of costimulatory and coinhibitory pathways via innate immune sensors and implications for immunotherapy. Exp. Mol. Med. 2023, 55, 1913–1921. [Google Scholar] [CrossRef]
  158. Ait-Zenati, F.; Djoudi, F.; Mehelleb, D.; Madaoui, M. Involvement of the human microbiome in frequent cancers, current knowledge and carcinogenesis mechanisms. Bull. Du Cancer 2023, 110, 776–789. [Google Scholar] [CrossRef]
  159. Wang, M.; Yang, G.; Tian, Y.; Zhang, Q.; Liu, Z.; Xin, Y. The role of the gut microbiota in gastric cancer: The immunoregulation and immunotherapy. Front. Immunol. 2023, 14, 1183331. [Google Scholar] [CrossRef]
  160. Herrera-Quintana, L.; Vázquez-Lorente, H.; Lopez-Garzon, M.; Cortés-Martín, A.; Plaza-Diaz, J. Cancer and the Microbiome of the Human Body. Nutrients 2024, 16, 2790. [Google Scholar] [CrossRef]
  161. Garrett, W.S. Cancer and the microbiota. Science 2015, 348, 80–86. [Google Scholar] [CrossRef]
  162. El Tekle, G.; Garrett, W.S. Bacteria in cancer initiation, promotion and progression. Nat. Rev. Cancer 2023, 23, 600–618. [Google Scholar] [CrossRef]
  163. Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, eabc4552. [Google Scholar] [CrossRef]
  164. Jia, W.; Rajani, C.; Xu, H.; Zheng, X. Gut microbiota alterations are distinct for primary colorectal cancer and hepatocellular carcinoma. Protein Cell 2021, 12, 374–393. [Google Scholar] [CrossRef]
  165. Wang, Y.; Li, H. Gut microbiota modulation: A tool for the management of colorectal cancer. J. Transl. Med. 2022, 20, 178. [Google Scholar] [CrossRef]
  166. Loke, Y.L.; Chew, M.T.; Ngeow, Y.F.; Lim, W.W.D.; Peh, S.C. Colon Carcinogenesis: The Interplay Between Diet and Gut Microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 603086. [Google Scholar] [CrossRef]
  167. Schwartz, D.J.; Rebeck, O.N.; Dantas, G. Complex interactions between the microbiome and cancer immune therapy. Crit. Rev. Clin. Lab. Sci. 2019, 56, 567–585. [Google Scholar] [CrossRef]
  168. Qu, R.; Zhang, Y.; Ma, Y.; Zhou, X.; Sun, L.; Jiang, C.; Zhang, Z.; Fu, W. Role of the Gut Microbiota and Its Metabolites in Tumorigenesis or Development of Colorectal Cancer. Adv. Sci. 2023, 10, e2205563. [Google Scholar] [CrossRef]
  169. Guo, Q.; Qin, H.; Liu, X.; Zhang, X.; Chen, Z.; Qin, T.; Chang, L.; Zhang, W. The Emerging Roles of Human Gut Microbiota in Gastrointestinal Cancer. Front. Immunol. 2022, 13, 915047. [Google Scholar] [CrossRef]
  170. Godley, F.A.; Shogan, B.D.; Hyman, N.H. Role of the Microbiome in Malignancy. Surg. Infect. 2023, 24, 271–275. [Google Scholar] [CrossRef]
  171. Zhao, Y.; Wang, C.; Goel, A. Role of gut microbiota in epigenetic regulation of colorectal Cancer. Biochim. Biophys. Acta (BBA) Rev. Cancer 2021, 1875, 188490. [Google Scholar] [CrossRef]
  172. Dong, J.; Tai, J.W.; Lu, L.F. miRNA–Microbiota Interaction in Gut Homeostasis and Colorectal Cancer. Trends Cancer 2019, 5, 666–669. [Google Scholar] [CrossRef]
  173. Fan, X.; Jin, Y.; Chen, G.; Ma, X.; Zhang, L. Gut Microbiota Dysbiosis Drives the Development of Colorectal Cancer. Digestion 2021, 102, 508–515. [Google Scholar] [CrossRef]
  174. Samami, E.; Aleebrahim-Dehkordi, E.; Mohebalizadeh, M.; Yaribash, S.; Saghazadeh, A.; Rezaei, N. Inosine, gut microbiota, and cancer immunometabolism. Am. J. Physiol. Metab. 2023, 324, E1–E8. [Google Scholar] [CrossRef]
  175. Barbosa, A.M.; Gomes-Gonçalves, A.; Castro, A.G.; Torrado, E. Immune System Efficiency in Cancer and the Microbiota Influence. Pathobiology 2021, 88, 170–186. [Google Scholar] [CrossRef]
  176. Wang, W.; Fan, J.; Zhang, C.; Huang, Y.; Chen, Y.; Fu, S.; Wu, J. Targeted modulation of gut and intra-tumor microbiota to improve the quality of immune checkpoint inhibitor responses. Microbiol. Res. 2024, 282, 127668. [Google Scholar] [CrossRef]
  177. Cheng, W.Y.; Wu, C.-Y.; Yu, J. The role of gut microbiota in cancer treatment: Friend or foe? Gut 2020, 69, 1867–1876. [Google Scholar] [CrossRef]
  178. Dolganyuk, V.; Belova, D.; Babich, O.; Prosekov, A.; Ivanova, S.; Katserov, D.; Patyukov, N.; Sukhikh, S. Microalgae: A promising source of valuable bioproducts. Biomolecules 2020, 10, 1153. [Google Scholar] [CrossRef]
  179. Mosblech, A.; Feussner, I.; Heilmann, I. Oxylipins: Structurally diverse metabolites from fatty acid oxidation. Plant Physiol. Biochem. 2009, 47, 511–517. [Google Scholar] [CrossRef]
  180. Serhan, C.N. Novel eicosanoid and docosanoid mediators: Resolvins, docosatrienes, and neuroprotectins. Curr. Opin. Clin. Nutr. Metab. Care 2005, 8, 115–121. [Google Scholar] [CrossRef]
  181. Serhan, C.N.; Hong, S.; Gronert, K.; Colgan, S.P.; Devchand, P.R.; Mirick, G.; Moussignac, R.-L. Resolvins: A family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J. Exp. Med. 2002, 196, 1025–1037. [Google Scholar] [CrossRef]
  182. Zittelli, G.C.; Lauceri, R.; Faraloni, C.; Benavides, A.M.S.; Torzillo, G. Valuable pigments from microalgae: Phycobiliproteins, primary carotenoids, and fucoxanthin. Photochem. Photobiol. Sci. 2023, 22, 1733–1789. [Google Scholar] [CrossRef]
  183. Del Mondo, A.; Smerilli, A.; Sané, E.; Sansone, C.; Brunet, C. Challenging microalgal vitamins for human health. Microb. Cell Factories 2020, 19, 201. [Google Scholar] [CrossRef]
  184. Mularczyk, M.; Michalak, I.; Marycz, K. Astaxanthin and other nutrients from haematococcus pluvialis—Multifunctional applications. Mar. Drugs 2020, 18, 459. [Google Scholar] [CrossRef]
  185. De Mattos, B.R.R.; Garcia, M.P.G.; Nogueira, J.B.; Paiatto, L.N.; Albuquerque, C.G.; Souza, C.L.; Fernandes, L.G.R.; Tamashiro, W.M.D.S.C.; Simioni, P.U. Inflammatory bowel disease: An overview of immune mechanisms and biological treatments. Mediat. Inflamm. 2015, 2015, 493012. [Google Scholar] [CrossRef]
  186. Juillerat, P.; Grueber, M.M.; Ruetsch, R.; Santi, G.; Vuillèmoz, M.; Michetti, P. Positioning biologics in the treatment of IBD: A practical guide—Which mechanism of action for whom? Curr. Res. Pharmacol. Drug Discov. 2022, 3, 100104. [Google Scholar] [CrossRef]
  187. Lautenschläger, C.; Schmidt, C.; Fischer, D.; Stallmach, A. Drug delivery strategies in the therapy of inflammatory bowel disease. Adv. Drug Deliv. Rev. 2014, 71, 58–76. [Google Scholar] [CrossRef]
  188. Jeong, D.Y.; Kim, S.; Son, M.J.; Son, C.Y.; Kim, J.Y.; Kronbichler, A.; Lee, K.H.; Shin, J.I. Induction and maintenance treatment of inflammatory bowel disease: A comprehensive review. Autoimmun. Rev. 2019, 18, 439–454. [Google Scholar] [CrossRef]
  189. Cai, Z.; Wang, S.; Li, J. Treatment of Inflammatory Bowel Disease: A Comprehensive Review. Front. Med. 2021, 8, 2681. [Google Scholar] [CrossRef]
  190. Jha, M.; Waheed, A.; Al Hooti, J.; Nair, S.; Najam, A.; Mal, M.; Tummala, N.; Shariq, A.S.; Hurairah, A.; Daniel, M. Advancements in Immunomodulatory Therapies for IBD and Their Interplay with the Gut–Brain Axis: An Updated Review of Current Literature and Beyond. Health Sci. Rep. 2025, 8, e71157. [Google Scholar] [CrossRef]
  191. Kushkevych, I.; Dvořáková, M.; Dordevic, D.; Futoma-Kołoch, B.; Gajdács, M.; Al-Madboly, L.A.; El-Salam, M.A. Advances in gut microbiota functions in inflammatory bowel disease: Dysbiosis, management, cytotoxicity assessment, and therapeutic perspectives. Comput. Struct. Biotechnol. J. 2025, 27, 851–868. [Google Scholar] [CrossRef]
  192. Zhou, J.; Li, M.; Chen, Q.; Li, X.; Chen, L.; Dong, Z.; Zhu, W.; Yang, Y.; Liu, Z.; Chen, Q. Programmable probiotics modulate inflammation and gut microbiota for inflammatory bowel disease treatment after effective oral delivery. Nat. Commun. 2022, 13, 3432. [Google Scholar] [CrossRef]
  193. Ann, S.; Choi, Y.; Yoon, Y. Comparative Genomic Analysis and Physiological Properties of Limosilactobacillus fermentum SMFM2017-NK2 with Ability to Inflammatory Bowel Disease. Microorganisms 2023, 11, 547. [Google Scholar] [CrossRef]
  194. Dabous, A.; D’ambrosio, S.; Cimini, D.; Schiraldi, C. Novel hydroxyectoines based formulations are suitable for preserving viability of Limosilactobacillus fermentum, Levilactobacillus brevis SP-48 and Bifidobacterium lactis HN019 during freeze-drying and storage, and in simulated gastrointestinal fluids. Dry. Technol. 2023, 41, 2062–2073. [Google Scholar] [CrossRef]
  195. Sahoo, D.K.; Heilmann, R.M.; Paital, B.; Patel, A.; Yadav, V.K.; Wong, D.; Jergens, A.E. Oxidative stress, hormones, and effects of natural antioxidants on intestinal inflammation in inflammatory bowel disease. Front. Endocrinol. 2023, 14, 1217165. [Google Scholar] [CrossRef]
  196. Laurindo, L.F.; Santos, A.R.d.O.d.; de Carvalho, A.C.A.; Bechara, M.D.; Guiguer, E.L.; Goulart, R.d.A.; Sinatora, R.V.; Araújo, A.C.; Barbalho, S.M. Phytochemicals and Regulation of NF-kB in Inflammatory Bowel Diseases: An Overview of In Vitro and In Vivo Effects. Metabolites 2023, 13, 96. [Google Scholar] [CrossRef]
  197. Blagov, A.V.; Orekhova, V.A.; Sukhorukov, V.N.; Melnichenko, A.A.; Orekhov, A.N. Potential Use of Antioxidant Compounds for the Treatment of Inflammatory Bowel Disease. Pharmaceuticals 2023, 16, 1150. [Google Scholar] [CrossRef]
  198. Zhu, L.; Song, Y.; Liu, H.; Wu, M.; Gong, H.; Lan, H.; Zheng, X. Gut microbiota regulation and anti-inflammatory effect of β-carotene in dextran sulfate sodium-stimulated ulcerative colitis in rats. J. Food Sci. 2021, 86, 2118–2130. [Google Scholar] [CrossRef]
  199. Ambati, R.R.; Phang, S.-M.; Ravi, S.; Aswathanarayana, R.G. Astaxanthin: Sources, extraction, stability, biological activities and its commercial applications—A review. Mar. Drugs 2014, 12, 128–152. [Google Scholar] [CrossRef]
  200. Davinelli, S.; Nielsen, M.E.; Scapagnini, G. Astaxanthin in skin health, repair, and disease: A comprehensive review. Nutrients 2018, 10, 522. [Google Scholar] [CrossRef]
  201. Konisky, H.; Bowe, W.P.; Yang, P.; Kobets, K. A clinical evaluation of the efficacy and tolerability of a novel topical antioxidant formulation featuring vitamin C, astaxanthin, and fermented turmeric. J. Cosmet. Dermatol. 2023, 22, 3088–3094. [Google Scholar] [CrossRef]
  202. Galarza, J.I.; Vega, B.O.A.; Villón, J.; Henríquez, V. Deesterification of astaxanthin and intermediate esters from Haematococcus pluvialis subjected to stress. Biotechnol. Rep. 2019, 23, e00351. [Google Scholar] [CrossRef]
  203. Odeberg, J.M.; Lignell, Å.; Pettersson, A.; Höglund, P. Oral bioavailability of the antioxidant astaxanthin in humans is enhanced by incorporation of lipid based formulations. Eur. J. Pharm. Sci. 2003, 19, 299–304. [Google Scholar] [CrossRef]
  204. 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]
  205. Yuan, J.P.; Peng, J.; Yin, K.; Wang, J.H. Potential health-promoting effects of astaxanthin: A high-value carotenoid mostly from microalgae. Mol. Nutr. Food Res. 2011, 55, 150–165. [Google Scholar] [CrossRef]
  206. Fei, Z.; Fan, F.; Liao, J.; Wan, M.; Bai, W.; Wang, W.; He, M.; Li, Y. Improving astaxanthin production of Haematococcus pluvialis on the outdoor large scale cultivation by optimizing the disinfection strategy of photobioreactor. Algal Res. 2022, 64, 102708. [Google Scholar] [CrossRef]
  207. 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]
  208. Hu, J.; Lu, W.; Lv, M.; Wang, Y.; Ding, R.; Wang, L. Extraction and purification of astaxanthin from shrimp shells and the effects of different treatments on its content. Rev. Bras. Farm. 2019, 29, 24–29. [Google Scholar] [CrossRef]
  209. Pan, X.; Wang, B.; Duan, R.; Jia, J.; Li, J.; Xiong, W.; Ling, X.; Chen, C.; Huang, X.; Zhang, G.; et al. Enhancing astaxanthin accumulation in Xanthophyllomyces dendrorhous by a phytohormone: Metabolomic and gene expression profiles. Microb. Biotechnol. 2020, 13, 1446–1460. [Google Scholar] [CrossRef]
  210. Mussagy, C.U.; Pereira, J.F.B.; Dufossé, L.; Raghavan, V.; Santos-Ebinuma, V.C.; Pessoa, A. Advances and trends in biotechnological production of natural astaxanthin by Phaffia rhodozyma yeast. Crit. Rev. Food Sci. Nutr. 2021, 63, 1862–1876. [Google Scholar] [CrossRef]
  211. Luna-Flores, C.H.; Wang, A.; von Hellens, J.; Speight, R.E. Towards commercial levels of astaxanthin production in Phaffia rhodozyma. J. Biotechnol. 2022, 350, 42–54. [Google Scholar] [CrossRef]
  212. Riccioni, G.; D’orazio, N.; Franceschelli, S.; Speranza, L. Marine carotenoids and cardiovascular risk markers. Mar. Drugs 2011, 9, 1166–1175. [Google Scholar] [CrossRef]
  213. Lim, S.R.; Kim, D.W.; Sung, J.; Kim, T.H.; Choi, C.H.; Lee, S.J. Astaxanthin inhibits autophagic cell death induced by bisphenol a in human dermal fibroblasts. Antioxidants 2021, 10, 1273. [Google Scholar] [CrossRef]
  214. Patel, A.K.; Tambat, V.S.; Chen, C.-W.; Chauhan, A.S.; Kumar, P.; Vadrale, A.P.; Huang, C.-Y.; Dong, C.-D.; Singhania, R.R. Recent advancements in astaxanthin production from microalgae: A review. Bioresour. Technol. 2022, 364, 128030. [Google Scholar] [CrossRef]
  215. Wan, X.; Zhou, X.-R.; Moncalian, G.; Su, L.; Chen, W.-C.; Zhu, H.-Z.; Chen, D.; Gong, Y.-M.; Huang, F.-H.; Deng, Q.-C. Reprogramming microorganisms for the biosynthesis of astaxanthin via metabolic engineering. Prog. Lipid Res. 2021, 81, 101083. [Google Scholar] [CrossRef]
  216. Gao, D.; Yao, S.; Sun, Y.; Zheng, D.; Zhang, Q.; Li, W. Astaxanthin attenuate iohexol-induced human proximal renal tubular epithelial cells injury via the ROS/NLRP3 inflammasome signal pathway. SDRP J. Food Sci. Technol. 2019, 4, 687–695. [Google Scholar] [CrossRef]
  217. Luo, M.; Yuan, Q.; Liu, M.; Song, X.; Xu, Y.; Zhang, T.; Zeng, X.; Wu, Z.; Pan, D.; Guo, Y. Astaxanthin nanoparticles ameliorate dextran sulfate sodium-induced colitis by alleviating oxidative stress, regulating intestinal flora, and protecting the intestinal barrier. Food Funct. 2023, 14, 9567–9579. [Google Scholar] [CrossRef]
  218. Talukdar, J.; Bhadra, B.; Dattaroy, T.; Nagle, V.; Dasgupta, S. Potential of natural astaxanthin in alleviating the risk of cytokine storm in COVID-19. Biomed. Pharmacother. 2020, 132, 110886. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  219. Chang, M.X.; Xiong, F. Astaxanthin and its Effects in Inflammatory Responses and Inflammation-Associated Diseases: Recent Advances and Future Directions. Molecules 2020, 25, 5342. [Google Scholar] [CrossRef]
  220. He, J.; Zhong, Y.; Li, Y.; Liu, S.; Pan, X. Astaxanthin Alleviates Oxidative Stress in Mouse Preantral Follicles and Enhances Follicular Development Through the AMPK Signaling Pathway. Int. J. Mol. Sci. 2025, 26, 2241. [Google Scholar] [CrossRef]
  221. Cheng, C.; Xu, F.; Pan, X.-F.; Wang, C.; Fan, J.; Yang, Y.; Liu, Y.; Sun, L.; Liu, X.; Xu, Y.; et al. Genetic mapping of serum metabolome to chronic diseases among Han Chinese. Cell Genom. 2025, 5, 100743. [Google Scholar] [CrossRef]
  222. Pereira, C.P.M.; Souza, A.C.R.; Vasconcelos, A.R.; Prado, P.S.; Name, J.J. Antioxidant and anti-inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review). Int. J. Mol. Med. 2020, 47, 37–48. [Google Scholar] [CrossRef]
  223. Zhang, X.-S.; Zhang, X.; Zhang, Q.-R.; Wu, Q.; Li, W.; Jiang, T.-W.; Hang, C.-H. Astaxanthin reduces matrix metalloproteinase-9 expression and activity in the brain after experimental subarachnoid hemorrhage in rats. Brain Res. 2015, 1624, 113–124. [Google Scholar] [CrossRef]
  224. Kishimoto, Y.; Yoshida, H.; Kondo, K. Potential Anti-Atherosclerotic Properties of Astaxanthin. Mar. Drugs 2016, 14, 35. [Google Scholar] [CrossRef]
  225. Grimmig, B.; Hudson, C.; Moss, L.; Peters, M.; Subbarayan, M.; Weeber, E.J.; Bickford, P.C. Astaxanthin supplementation modulates cognitive function and synaptic plasticity in young and aged mice. GeroScience 2019, 41, 77–87. [Google Scholar] [CrossRef]
  226. Wang, S.; Qi, X. The Putative Role of Astaxanthin in Neuroinflammation Modulation: Mechanisms and Therapeutic Potential. Front. Pharmacol. 2022, 13, 916653. [Google Scholar] [CrossRef]
  227. Gökçeoğlu, A.; Yarim, G.F. The Effects Of Astaxanthin On The Nervous System. Samsun Sağlık Bilim. Derg. 2022, 7, 657–674. [Google Scholar] [CrossRef]
  228. Brasil, F.B.; Gobbo, R.C.B.; de Almeida, F.J.S.; Luckachaki, M.D.; Dall’oglio, E.L.; de Oliveira, M.R. The signaling pathway PI3K/Akt/Nrf2/HO-1 plays a role in the mitochondrial protection promoted by astaxanthin in the SH-SY5Y cells exposed to hydrogen peroxide. Neurochem. Int. 2021, 146, 105024. [Google Scholar] [CrossRef]
  229. Abbaszadeh, F.; Jorjani, M.; Joghataei, M.T.; Mehrabi, S. Astaxanthin Modulates Autophagy, Apoptosis, and Neuronal Oxidative Stress in a Rat Model of Compression Spinal Cord Injury. Neurochem. Res. 2022, 47, 2043–2051. [Google Scholar] [CrossRef]
  230. Si, P.; Zhu, C. Biological and neurological activities of astaxanthin (Review). Mol. Med. Rep. 2022, 26, 300. [Google Scholar] [CrossRef]
  231. Davinelli, S.; Saso, L.; D’angeli, F.; Calabrese, V.; Intrieri, M.; Scapagnini, G. Astaxanthin as a Modulator of Nrf2, NF-κB, and Their Crosstalk: Molecular Mechanisms and Possible Clinical Applications. Molecules 2022, 27, 502. [Google Scholar] [CrossRef]
  232. Lai, T.-T.; Yang, C.-M.; Yang, C.-H. Astaxanthin protects retinal photoreceptor cells against high glucose-induced oxidative stress by induction of antioxidant enzymes via the pi3k/akt/nrf2 pathway. Antioxidants 2020, 9, 729. [Google Scholar] [CrossRef]
  233. Li, K.; Wang, W.; Xiao, W. Astaxanthin: A promising therapeutic agent for organ fibrosis. Pharmacol. Res. 2023, 188, 106657. [Google Scholar] [CrossRef]
  234. Guo, Q.; Jin, Y.; Chen, X.; Ye, X.; Shen, X.; Lin, M.; Zeng, C.; Zhou, T.; Zhang, J. NF-κB in biology and targeted therapy: New insights and translational implications. Signal Transduct. Target. Ther. 2024, 9, 53. [Google Scholar] [CrossRef]
  235. Lee, S.-J.; Bai, S.-K.; Lee, K.-S.; Namkoong, S.; Na, H.-J.; Ha, K.-S.; Han, J.-A.; Yim, S.-V.; Chang, K.; Kwon, Y.-G.; et al. Astaxanthin inhibits nitric oxide production and inflammatory gene expression by suppressing IκB kinase-dependent NF-κB activation. Mol. Cells 2003, 16, 97–105. [Google Scholar] [CrossRef]
  236. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
  237. Laflamme, N.; Lacroix, S.; Rivest, S. An essential role of interleukin-1β in mediating NF-κB activity and COX-2 transcription in cells of the blood-brain barrier in response to a systemic and localized inflammation but not during endotoxemia. J. Neurosci. 1999, 19, 10923–10930. [Google Scholar] [CrossRef]
  238. Kohandel, Z.; Farkhondeh, T.; Aschner, M.; Pourbagher-Shahri, A.M.; Samarghandian, S. Anti-inflammatory action of astaxanthin and its use in the treatment of various diseases. Biomed. Pharmacother. 2022, 145, 112179. [Google Scholar] [CrossRef]
  239. Kitahara, A.; Takahashi, K.; Morita, N.; Murashima, T.; Onuma, H.; Sumitani, Y.; Tanaka, T.; Kondo, T.; Hosaka, T.; Ishida, H. The novel mechanisms concerning the inhibitions of palmitate-induced proinflammatory factor releases and endogenous cellular stress with astaxanthin on MIN6 β-cells. Mar. Drugs 2017, 15, 185. [Google Scholar] [CrossRef]
  240. Xia, W.; Tang, N.; Kord-Varkaneh, H.; Low, T.Y.; Tan, S.C.; Wu, X.; Zhu, Y. The effects of astaxanthin supplementation on obesity, blood pressure, CRP, glycemic biomarkers, and lipid profile: A meta-analysis of randomized controlled trials. Pharmacol. Res. 2020, 161, 105113. [Google Scholar] [CrossRef]
  241. Yeh, P.T.; Huang, H.W.; Yang, C.M.; Yang, W.S.; Yang, C.H. Astaxanthin inhibits expression of retinal oxidative stress and inflammatory mediators in streptozotocin-induced diabetic rats. PLoS ONE 2016, 11, e0146438. [Google Scholar] [CrossRef]
  242. Jyonouchi, H.; Zhang, L.; Tomita, Y. Studies of immunomodulating actions of carotenoids. ii. astaxanthin enhances in vitro antibody production to t-dependent antigens without facilitating polyclonal b-cell activation. Nutr. Cancer 1993, 19, 269–280. [Google Scholar] [CrossRef]
  243. Ferro, M.; Graubard, A.; Beauchesne, T. Astaxanthin: The possible effects of this carotenoid on disease, inflammation and aging control. A meta-analysis. Int. J. Food Nutr. Res. 2021, 5, 44. [Google Scholar] [CrossRef]
  244. Lee, J.; Kim, M.-H.; Kim, H. Anti-Oxidant and Anti-Inflammatory Effects of Astaxanthin on Gastrointestinal Diseases. Int. J. Mol. Sci. 2022, 23, 15471. [Google Scholar] [CrossRef]
  245. Nakagawa, K.; Kiko, T.; Miyazawa, T.; Burdeos, G.C.; Kimura, F.; Satoh, A.; Miyazawa, T. Antioxidant effect of astaxanthin on phospholipid peroxidation in human erythrocytes. Br. J. Nutr. 2011, 105, 1563–1571. [Google Scholar] [CrossRef]
  246. Iizuka, M.; Ayaori, M.; Uto-Kondo, H.; Yakushiji, E.; Takiguchi, S.; Nakaya, K.; Hisada, T.; Sasaki, M.; Komatsu, T.; Yogo, M.; et al. Astaxanthin enhances ATP-binding cassette transporter A1/G1 expressions and cholesterol efflux from macrophages. J. Nutr. Sci. Vitaminol. 2012, 58, 96–104. [Google Scholar] [CrossRef]
  247. Yang, C.; Hassan, Y.I.; Liu, R.; Zhang, H.; Chen, Y.; Zhang, L.; Tsao, R. Anti-Inflammatory Effects of Different Astaxanthin Isomers and the Roles of Lipid Transporters in the Cellular Transport of Astaxanthin Isomers in Caco-2 Cell Monolayers. J. Agric. Food Chem. 2019, 67, 6222–6231. [Google Scholar] [CrossRef]
  248. Yin, Y.; Xu, N.; Qin, T.; Zhou, B.; Shi, Y.; Zhao, X.; Ma, B.; Xu, Z.; Li, C. Astaxanthin provides antioxidant protection in lps-induced dendritic cells for inflammatory control. Mar. Drugs 2021, 19, 534. [Google Scholar] [CrossRef]
  249. Kim, Y.H.; Koh, H.-K.; Kim, D.-S. Down-regulation of IL-6 production by astaxanthin via ERK-, MSK-, and NF-κB-mediated signals in activated microglia. Int. Immunopharmacol. 2010, 10, 1560–1572. [Google Scholar] [CrossRef]
  250. Speranza, L.; Pesce, M.; Patruno, A.; Franceschelli, S.; de Lutiis, M.A.; Grilli, A.; Felaco, M. Astaxanthin treatment reduced oxidative induced pro-inflammatory cytokines secretion in U937: SHP-1 as a novel biological target. Mar. Drugs 2012, 10, 890–899. [Google Scholar] [CrossRef]
  251. Davinelli, S.; Melvang, H.M.; Andersen, L.P.; Scapagnini, G.; Nielsen, M.E. Astaxanthin from shrimp cephalothorax stimulates the immune response by enhancing IFN-γ, IL-10, and IL-2 secretion in splenocytes of Helicobacter pylori-infected mice. Mar. Drugs 2019, 17, 382. [Google Scholar] [CrossRef]
  252. Hwang, Y.H.; Hong, S.G.; Mun, S.K.; Kim, S.J.; Lee, S.J.; Kim, J.J.; Kang, K.Y.; Yee, S.T. The protective effects of astaxanthin on the OVA-induced asthma mice model. Molecules 2017, 22, 2019. [Google Scholar] [CrossRef]
  253. Olsen, T.; Rismo, R.; Cui, G.; Goll, R.; Christiansen, I.; Florholmen, J. TH1 and TH17 interactions in untreated inflamed mucosa of inflammatory bowel disease, and their potential to mediate the inflammation. Cytokine 2011, 56, 633–640. [Google Scholar] [CrossRef]
  254. Cao, H.; Diao, J.; Liu, H.; Liu, S.; Liu, J.; Yuan, J.; Lin, J. The Pathogenicity and Synergistic Action of Th1 and Th17 Cells in Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2023, 29, 818–829. [Google Scholar] [CrossRef]
  255. Lv, X.; Obianwuna, U.E.; Wang, L.; Xie, S.; An, K.; Han, L.; Wu, D.; Qi, X.; Wu, S.; Xia, Z. Astaxanthin supplementation enhances carcass performance, meat quality, and intestinal barrier function by modulating cecal microbiota and metabolomics in overfed Pekin ducks. Poult. Sci. 2025, 104, 105310. [Google Scholar] [CrossRef]
  256. Lv, X.; Obianwuna, U.E.; Yang, W.; Zhang, Z.; An, K.; Shi, B.; Dong, Y.; Wu, S.; Xia, Z. Astaxanthin supplementation mitigated intestinal damage and immunity in overfed Pekin ducks by regulating gut morphology, intestinal inflammation, and antioxidant balance. Anim. Nutr. 2025, 21, 426–438. [Google Scholar] [CrossRef]
  257. Chen, Y.; Zhao, S.; Jiao, D.; Yao, B.; Yang, S.; Li, P.; Long, M. Astaxanthin Alleviates Ochratoxin A-Induced Cecum Injury and Inflammation in Mice by Regulating the Diversity of Cecal Microbiota and TLR4/MyD88/NF-κB Signaling Pathway. Oxidative Med. Cell. Longev. 2021, 2021, 8894491. [Google Scholar] [CrossRef]
  258. Lin, X.; Bo, H.; Gu, J.; Yi, X.; Zhang, P.; Liu, R.; Li, H.; Sun, G.; Lin, C.-H. Astaxanthin, a carotenoid antioxidant, pretreatment alleviates cognitive deficits in aircraft noised mice by attenuating inflammatory and oxidative damage to the gut, heart and hippocampus. Biomed. Pharmacother. 2022, 148, 112777. [Google Scholar] [CrossRef]
  259. Zhang, L.; Cao, W.; Gao, Y.; Yang, R.; Zhang, X.; Xu, J.; Tang, Q. Astaxanthin (ATX) enhances the intestinal mucosal functions in immunodeficient mice. Food Funct. 2020, 11, 3371–3381. [Google Scholar] [CrossRef]
  260. Kayama, H.; Takeda, K. Emerging roles of host and microbial bioactive lipids in inflammatory bowel diseases. Eur. J. Immunol. 2023, 53, e2249866. [Google Scholar] [CrossRef]
  261. Yasui, Y.; Hosokawa, M.; Mikami, N.; Miyashita, K.; Tanaka, T. Dietary astaxanthin inhibits colitis and colitis-associated colon carcinogenesis in mice via modulation of the inflammatory cytokines. Chem. Interactions 2011, 193, 79–87. [Google Scholar] [CrossRef]
  262. Li, C.; Zhou, Y.; Yuan, M.; Yang, Y.; Song, R.; Xu, G.; Chen, G. Astaxanthin-loaded polylactic acid-glycolic acid nanoparticles ameliorate ulcerative colitis through antioxidant effects. Front. Nutr. 2023, 10, 1267274. [Google Scholar] [CrossRef]
  263. Ni, Y.; Nagashimada, M.; Zhuge, F.; Zhan, L.; Nagata, N.; Tsutsui, A.; Nakanuma, Y.; Kaneko, S.; Ota, T. Astaxanthin prevents and reverses diet-induced insulin resistance and steatohepatitis in mice: A comparison with vitamin E. Sci. Rep. 2015, 5, 17192. [Google Scholar] [CrossRef]
  264. Zhang, H.; Wang, M.; Zhou, Y.; Bao, S.; Wang, F.; Li, C. Protective Effects of Astaxanthin against Oxidative Stress: Attenuation of TNF-α-Induced Oxidative Damage in SW480 Cells and Azoxymethane/Dextran Sulfate Sodium-Induced Colitis-Associated Cancer in C57BL/6 Mice. Mar. Drugs 2024, 22, 469. [Google Scholar] [CrossRef]
  265. Tominaga, K.; Hongo, N.; Fujishita, M.; Takahashi, Y.; Adachi, Y. Protective effect of astaxanthin on skin deterioration. J. Clin. Biochem. Nutr. 2017, 61, 33–39. [Google Scholar] [CrossRef]
  266. Mohammadi, S.G.; Feizi, A.; Bagherniya, M.; Shafie, D.; Ahmadi, A.-R.; Kafeshani, M. The effect of astaxanthin supplementation on inflammatory markers, oxidative stress indices, lipid profile, uric acid level, blood pressure, endothelial function, quality of life, and disease symptoms in heart failure subjects. Trials 2024, 25, 518. [Google Scholar] [CrossRef]
  267. Ma, B.; Lu, J.; Kang, T.; Zhu, M.; Xiong, K.; Wang, J. Astaxanthin supplementation mildly reduced oxidative stress and inflammation biomarkers: A systematic review and meta-analysis of randomized controlled trials. Nutr. Res. 2022, 99, 40–50. [Google Scholar] [CrossRef]
  268. Liu, F.; Smith, A.D.; Solano-Aguilar, G.; Wang, T.T.Y.; Pham, Q.; Beshah, E.; Tang, Q.; Urban, J.F.; Xue, C.; Li, R.W. Mechanistic insights into the attenuation of intestinal inflammation and modulation of the gut microbiome by krill oil using in vitro and in vivo models. Microbiome 2020, 8, 83. [Google Scholar] [CrossRef]
  269. Pratap, K.; Majzoub, M.E.; Taki, A.C.; Hernandez, S.M.; Magnusson, M.; Glasson, C.R.K.; de Nys, R.; Thomas, T.; Lopata, A.L.; Kamath, S.D. The Algal Polysaccharide Ulvan and Carotenoid Astaxanthin Both Positively Modulate Gut Microbiota in Mice. Foods 2022, 11, 565. [Google Scholar] [CrossRef]
  270. Wu, L.; Lyu, Y.; Srinivasagan, R.; Wu, J.; Ojo, B.; Tang, M.; El-Rassi, G.D.; Metzinger, K.; Smith, B.J.; A Lucas, E.; et al. Astaxanthin-Shifted gut microbiota is associated with inflammation and metabolic homeostasis in mice. J. Nutr. 2020, 150, 2687–2698. [Google Scholar] [CrossRef]
  271. Wang, M.; Xu, W.; Yu, J.; Liu, Y.; Ma, H.; Ji, C.; Zhang, C.; Xue, J.; Li, R.; Cui, H. Astaxanthin from Haematococcus pluvialis Prevents High-Fat Diet-Induced Hepatic Steatosis and Oxidative Stress in Mice by Gut-Liver Axis Modulating Properties. Front. Nutr. 2022, 9, 840648. [Google Scholar] [CrossRef]
  272. Ren, P.; Yue, H.; Tang, Q.; Wang, Y.; Xue, C. Astaxanthin exerts an adjunctive anti-cancer effect through the modulation of gut microbiota and mucosal immunity. Int. Immunopharmacol. 2024, 128, 111553. [Google Scholar] [CrossRef]
  273. Yuan, Z.; Zhang, Y.; Zhang, X.; Wang, L.; Ling, G.; Zhang, P. Microalgae and Bioactive Substances Therein: Unveiling Therapeutic Promise against Inflammatory Bowel Disease. J. Agric. Food Chem. 2025, 73, 18497–18510. [Google Scholar] [CrossRef]
  274. Wang, J.; Chen, Y.; Han, L.; Xia, S.; Zhang, X.; Xue, P.; Kang, Y.; Ming, J.; Xu, Z. Microenvironment responsive pod-structured astaxanthin nanocarrier for ameliorating inflammatory bowel disease. Chin. Chem. Lett. 2024, 35, 109029. [Google Scholar] [CrossRef]
  275. Wu, Y.; Bashir, M.A.; Shao, C.; Wang, H.; Zhu, J.; Huang, Q. Astaxanthin targets IL-6 and alleviates the LPS-induced adverse inflammatory response of macrophages. Food Funct. 2024, 15, 4207–4222. [Google Scholar] [CrossRef] [PubMed]
  276. Zhu, X.; Chen, X.; Wang, M.; Hu, H. Astaxanthin: A Compound in the Prevention of Chronic Diseases and as a Potential Adjuvant Treatment Agent. Antioxidants 2025, 14, 715. [Google Scholar] [CrossRef] [PubMed]
  277. Ma, K.; Chen, S.; Wu, Y.; Ma, Y.; Qiao, H.; Fan, J.; Wu, H. Dietary supplementation with microalgae enhances the zebrafish growth performance by modulating immune status and gut microbiota. Appl. Microbiol. Biotechnol. 2022, 106, 773–788. [Google Scholar] [CrossRef]
  278. Costa, J.A.V.; Cassuriaga, A.P.A.; Moraes, L.; Morais, M.G. Biosynthesis and potential applications of terpenes produced from microalgae. Bioresour. Technol. Rep. 2022, 19, 101166. [Google Scholar] [CrossRef]
  279. Kim, S.-W.; Chojnacka, K. Marine Algae Extracts: Processes, Products, and Applications; John Wiley & Sons: Hoboken, NJ, USA, 2015; Available online: https://books.google.it/books?hl=it&lr=&id=LhB1BgAAQBAJ&oi=fnd&pg=PR5&dq=K.+C.+Se-Kwon+Kim,+Marine+Algae+Extracts:+Processes,+Products,+and+Applications.+2015.&ots=qAOUPKQrd0&sig=ds6pYJgV4RXw6UEzDxAcR15sQ1U#v=onepage&q=K.%20C.%20Se-Kwon%20Kim%2C%20Marine%20Algae%20Extracts%3A%20Processes%2C%20Products%2C%20and%20Applications.%202015.&f=false (accessed on 28 August 2025).
  280. Repici, A.; Hasan, A.; Capra, A.P.; Scuderi, S.A.; Paterniti, I.; Campolo, M.; Ardizzone, A.; Esposito, E. Marine Algae and Deriving Biomolecules for the Management of Inflammatory Bowel Diseases: Potential Clinical Therapeutics to Decrease Gut Inflammatory and Oxidative Stress Markers? Mar. Drugs 2024, 22, 336. [Google Scholar] [CrossRef]
  281. Eilam, Y.; Khattib, H.; Pintel, N.; Avni, D. Microalgae—Sustainable Source for Alternative Proteins and Functional Ingredients Promoting Gut and Liver Health. Glob. Challenges 2023, 7, 2200177. [Google Scholar] [CrossRef] [PubMed]
  282. Shoham, S.; Pintel, N.; Avni, D. Oxidative Stress, Gut Bacteria, and Microalgae: A Holistic Approach to Manage Inflammatory Bowel Diseases. Antioxidants 2025, 14, 697. [Google Scholar] [CrossRef] [PubMed]
  283. De Jesus Raposo, M.F.; De Morais, A.M.M.B.; De Morais, R.M.S.C. Emergent sources of prebiotics: Seaweeds and microalgae. Mar. Drugs 2016, 14, 27. [Google Scholar] [CrossRef]
  284. Arita, M.; Yoshida, M.; Hong, S.; Tjonahen, E.; Glickman, J.N.; Petasis, N.A.; Blumberg, R.S.; Serhan, C.N. Resolvin E1, an endogenous lipid mediator derived from omega-3 eicosapentaenoic acid, protects against 2,4,6-trinitrobenzene sulfonic acid-induced colitis. Proc. Natl. Acad. Sci. USA 2005, 102, 7671–7676. [Google Scholar] [CrossRef]
  285. Robertson, R.C.; Guihéneuf, F.; Bahar, B.; Schmid, M.; Stengel, D.B.; Fitzgerald, G.F.; Ross, R.; Stanton, C. The anti-inflammatory effect of algae-derived lipid extracts on lipopolysaccharide (LPS)-stimulated human THP-1 macrophages. Mar. Drugs 2015, 13, 5402–5424. [Google Scholar] [CrossRef] [PubMed]
  286. Colas, R.A.; Shinohara, M.; Dalli, J.; Chiang, N.; Serhan, C.N. Identification and signature profiles for pro-resolving and inflammatory lipid mediators in human tissue. Am. J. Physiol. Physiol. 2014, 307, C39–C54. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Radice, R.P.; Iannelli, V.; Padula, F.; De Fabrizio, V.; Drosos, M.; Scopa, A.; Martelli, G. The Potential Role of Microalgal Antioxidant Molecules on the Microbiota–Gut Axis in Inflammatory Bowel Diseases. Biomolecules 2025, 15, 1535. https://doi.org/10.3390/biom15111535

AMA Style

Radice RP, Iannelli V, Padula F, De Fabrizio V, Drosos M, Scopa A, Martelli G. The Potential Role of Microalgal Antioxidant Molecules on the Microbiota–Gut Axis in Inflammatory Bowel Diseases. Biomolecules. 2025; 15(11):1535. https://doi.org/10.3390/biom15111535

Chicago/Turabian Style

Radice, Rosa Paola, Valeria Iannelli, Francesca Padula, Vincenzo De Fabrizio, Marios Drosos, Antonio Scopa, and Giuseppe Martelli. 2025. "The Potential Role of Microalgal Antioxidant Molecules on the Microbiota–Gut Axis in Inflammatory Bowel Diseases" Biomolecules 15, no. 11: 1535. https://doi.org/10.3390/biom15111535

APA Style

Radice, R. P., Iannelli, V., Padula, F., De Fabrizio, V., Drosos, M., Scopa, A., & Martelli, G. (2025). The Potential Role of Microalgal Antioxidant Molecules on the Microbiota–Gut Axis in Inflammatory Bowel Diseases. Biomolecules, 15(11), 1535. https://doi.org/10.3390/biom15111535

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