Skip to main content
Comparative Immunology Reports logoLink to Comparative Immunology Reports
. 2025 May 29;9:200230. doi: 10.1016/j.cirep.2025.200230

Abiotic and biotic factors affecting the immune system of aquatic species: A review

Zulhisyam Abdul Kari a,b
PMCID: PMC12173003

Highlights

  • Abiotic environmental factors, including temperature, salinity, pH, hypoxia, and pollutants, significantly influence immune functions in aquatic animals.

  • Pathogenic and parasitic infections, as biotic stressors, markedly inhibit both innate and adaptive immune responses, thereby heightening susceptibility to disease.

  • Aquatic species' immune systems demonstrate structural and functional diversity, influenced by evolutionary adaptations to specific ecological niches.

  • Integrating environmental and immunological knowledge is essential for enhancing aquaculture resilience, disease management, and the sustainability of aquatic ecosystem health.

Keywords: Aquatic animals, Immune response, Biotic factors, Abiotic factors, Aquaculture

Abstract

The immune response of aquatic organisms is affected by biotic and abiotic elements, determining their health and survival. Thus, this review aimed to integrate existing research on how environmental factors (temperature, salinity, pH, pollution) and biotic factors (infections, parasites, and the microbiome) shape immune function in aquatic animals. Aquatic animals, which include fish, crustaceans, and molluscs, have innate and adaptive immune systems that are predominant in non-mammalian species. Abiotic stresses such as temperature variations and pollution can compromise immune responses, putting them at higher risk of infection. Biotic interactions also influence immunological responses, resulting in immunosuppression under elevated pathogen burden and interspecies competition. Therefore, understanding these aspects in aquaculture, ecosystem management, and conservation initiatives can offer insights into the impact of environmental changes and stresses on the immunological resilience of aquatic animals.

Introduction

The immune response of aquatic organisms is a complex and dynamic mechanism essential for their survival and overall health, affected by various biotic and abiotic variables. Biotic factors comprise interactions with diseases, parasites, and competitors, whereas abiotic factors involve environmental variables such as temperature, salinity, pH, and pollutant presence [1]. Understanding the influence of these elements on immune responses is crucial for managing aquatic ecosystems, enhancing aquaculture methods, and evaluating environmental health. Fish, crustaceans, molluscs, and amphibians exhibit various immunological responses tailored to their distinct environments and behaviours, which can be classified into innate and adaptive immunity [[2], [3], [4]]. Innate immunity is the primary defence mechanism that offers rapid, non-specific protection against infections. Meanwhile, adaptive immunity is a more targeted and enduring response to protect against previously encountered infections, often observed in teleost fish and certain amphibians.

Pathogen load and interspecies interactions are biotic factors that affect the immune response of aquatic organisms [[5], [6], [7], [8], [9], [10], [11]]. High pathogen densities suppress their immune responses, making these animals vulnerable to infections [12]. Additionally, competing for resources may induce stress in aquatic species, potentially compromising their immunological systems [13]. Abiotic variables such as excessive temperatures, salinity variations, and pollution can also impair immune processes, increasing the species’ susceptibility to illnesses [[14], [15], [16]]. Exposure to heavy metals and endocrine-disrupting chemicals has been demonstrated to hamper immune system efficacy in numerous aquatic species [17].

In this review, the literature on the interaction between biotic and abiotic stimuli and the immune systems of aquatic organisms was synthesised to highlight their significant influence on immune responses. In addition, this review elucidated the interactions of these variables that affect health outcomes in aquatic ecosystems and aquaculture, offering insights for the stakeholders to improve the health of aquatic species and the sustainability of aquatic resources.

Overview of immune systems in aquatic animals

The immune system, namely innate (non-specific) and adaptive (specific) immunity, is critical in fighting infections in aquatic animals [[18], [19], [20]]. The two systems function together to preserve the organism despite significant variances in their approaches and prevalence among species (Fig. 1). Innate immunity is more pivotal for non-mammals, such as aquatic animals, while adaptive immunity may be rudimentary or non-existent, depending on the species. Innate immunity serves as the primary defence mechanism in all animals, including aquatic species [21]. It is non-specific and targets a wide array of pathogens without the need for prior exposure. Furthermore, innate immune responses are instantaneous and involve a variety of cellular and humoral elements.

Fig. 1.

Fig 1:

Immune systems in aquatic animals.

Aquatic animals display a notable diversity in the structures and functions of their immune systems. All aquatic animals exhibit innate immunity as a non-specific initial defence mechanism; however, the presence and complexity of adaptive immunity differ among species. Jawed fish are an example of vertebrates that exhibit advanced adaptive immune systems, characterised by specific immune responses and the capacity for immunological memory [[22], [23], [24]]. In contrast, invertebrates depend solely on innate immunity, employing diverse cellular and humoral components to identify and eradicate pathogens. Several aquatic invertebrates do not possess an adaptive immune system, depending solely on their innate immune defences for protection against infections [25]. The innate immune systems in molluscs, echinoderms, and crustaceans are well-developed to compensate for the lack of adaptive immunity. This immunological diversity in aquatic animals illustrates their evolutionary adaptations to distinct environments, with the immune system being essential for health and survival.

According to Guryanova and Ovchinnikova [26], innate immunity consists of several immune cells responsible for recognising, phagocytosing, and combating pathogens. Phagocytosis is a process in which cells consume and digest pathogens and is part of an animal’s primary defence mechanism. Phagocytic cells, such as macrophages and neutrophils, are the primary effectors of the innate immune systems in fish and other aquatic vertebrates [27,28]. Macrophages serve as the initial defence against pathogens and regulate the inflammatory responses. Meanwhile, natural killer (NK) cells, found in most vertebrates, including fish, identify and eliminate diseased or abnormal cells [29]. These cells do not require antigen recognition to operate, enabling them to function swiftly against infections. Hemocytes are essential in the immune response of aquatic invertebrates, including molluscs and crustaceans. These immune cells are crucial for organisms that lack an adaptive immune response and must depend only on innate defences. Hematocytes exert their protective functions through phagocytosis, encapsulation, and the synthesis of antimicrobial compounds [30,31].

Apart from cellular responses, aquatic animals have humoral components for immune protection in the form of soluble molecules in the blood or body fluids. The humoral factors include lysozyme, an enzyme present in the blood, tissues, and mucus of fish and invertebrates, crucial for fighting bacterial infections [[32], [33], [34]]. This enzyme degrades the peptidoglycan layer in bacterial cell walls, resulting in bacterial cell death [33]. The complement system is another component of the innate immune response in fish and other aquatic vertebrates, which functions similarly to that in mammals to recognise and clear pathogens. This system consists of a cascade of proteins that mark pathogens to facilitate their elimination through cell lysis and enhance inflammation [35]. Antimicrobial peptides (AMPs) are small molecules that demonstrate broad-spectrum activity against bacteria, viruses, and fungi [[36], [37], [38]]. These peptides are present in the mucus, blood, and tissues of various aquatic species, such as fish and amphibians. Defensins and hepcidins are examples of AMPs in aquatic vertebrates for a swift and efficient defence against microbial invasions [39]. Furthermore, carbohydrate-binding proteins, such as lectins, are essential in invertebrates and lower vertebrates for pathogen recognition and clearance. This peptide aids in pathogen elimination by recognising its surface carbohydrates [40,41].

Adaptive immunity is more advanced in vertebrates such as fish due to its specificity to antigens and the ability to form immunological memory [42], allowing the animal to respond more efficiently to future encounters with identical pathogens [43]. It depends on the function of lymphocytes, specifically B cells and T cells, and the synthesis of antibodies. Adaptive immunity is well developed in jawed vertebrates, including bony fish (teleosts) and cartilaginous fish (sharks and rays). Teleost fish possess a functional adaptive immune system similar to mammals but with distinct characteristics. In contrast, Jawless vertebrates, including lampreys and hagfish, have adaptive immune system that operates independently of immunoglobulins. These species utilise lymphocyte receptors (VLRs) for the recognition and response to pathogens [44]. The VLRs are structurally different from antibodies while fulfilling a comparable role in antigen recognition [45]. This system exemplifies the diversity of immune strategies present in vertebrates.

Nutritional immunity is defined as the host's active sequestration of essential trace metals to inhibit pathogen growth during infection [[46], [47], [48], [49], [50]]. Nutritional immunity serves as a crucial adaptive mechanism in aquatic ecosystems, linking environmental factors such as salinity, temperature, pollution, and pH with immune responses, all of which influence water chemistry and metal bioavailability. Aquatic vertebrates, including fish, employ iron-binding proteins such as transferrin and hepcidin to diminish extracellular iron availability during infections [51,52]. This mechanism deprives pathogens of an essential resource while also regulating systemic iron levels. The expression of hepcidin is modulated by inflammatory signals and environmental stressors, highlighting its dual role in immunity and metal homeostasis [53]. Additionally, certain pathogens in aquatic environments have developed siderophore-mediated strategies to extract iron from the host or surroundings, underscoring the evolutionary conflict between host defences and microbial acquisition methods [54]. The results of this competition are frequently influenced by the availability of abiotic nutrients in the water column or sediment, positioning nutritional immunity as both an internal immune strategy and a response that is sensitive to external environmental conditions. Nutritional immunity acts as a functional connection between abiotic stressors, such as nutrient limitation and water chemistry, and biotic challenges, including infections. This illustrates the evolutionary adaptations of aquatic species in defending themselves through the regulation of metabolic resources in varied and dynamic environments.

Biotic factors influencing immune responses

Biotic variables influence immunological responses in fish. Thus, it is crucial to understand how fish protect themselves from illnesses and other risk factors in the wild and farmed environments. The biotic variables influencing immune responses in aquatic organisms include pathogens (bacteria, viruses, fungi), parasites (protozoans and helminths), and the internal microbiome.

Pathogens

Pathogens, including bacteria, viruses, and fungi, represent a major biotic stressor to fish's immune system (Table 1). Pathogenic threats compromise the immune system and increase the fish’s susceptibility to infections, which may result in significant mortality and economic losses in aquaculture [55]. Aquatic animals depend significantly on their innate immune systems to combat bacterial infections [56]. According to Wangkahart et al. [57], the innate immune system identifies bacterial components, including lipopolysaccharides (LPS) and flagellin, to initiate an immune response. Some bacteria, such as Vibrio spp., Aeromonas spp., and Edwardsiella spp., can evade or suppress host immune responses to facilitate their survival within the host. These microbes secrete toxins that harm immune cells or inhibit phagocytosis. Pathogens can also affect the fish’s adaptive immune response by disrupting antibody production responsible for neutralising bacterial toxins, leading to chronic infections that are more difficult to manage and treat [[58], [59], [60]]. Therefore, understanding the influence of pathogens on immune responses in aquatic animals is essential for managing fish health in aquaculture, improving infection resilience, and conserving wild populations.

Table 1.

Pathogen effects on the immune system of aquatic species.

Pathogen Aquatic species Effects on the immune system References
Aeromonas veronii Freshwater fish Immunosuppression and acute infections in aquatic animals decrease mucosal defences De Silva and Heo [61]
Microsporidia Fish (general) Induces immunosuppression in aquatic animals, impacting specific and non-specific immune defences Bojko and Stentiford [62]
Ichthyophthirius multifiliis Freshwater fish (tilapia, catfish, carp) Reduced immune reaction and caused skin sores on fish Teixeira Alves and Taylor [63]
Probiotic bacteria Shrimp, fish (general) Improved non-specific defences and bacterial resistance in aquaculture species Jamal et al. [64]
Vibriosis Marine fish Impacted innate defences and compromised the immune system, resulting in elevated mortality rates among aquaculture species Ina-Salwany et al. [65]
Vibrio vulnificus Marine fish, shellfish Induced immunosuppression and tissue necrosis, compromising the immune system in marine organisms Allam and Raftos [66]
Listeria monocytogenes Fish (general) Disruption of the mucosal barriers affected the fish's immune system, reducing its innate defence Morley [67]
Spring viremia of carp virus (SVCV) Cyprinus carpio (Common carp) Infects carp species by suppressing their innate and adaptive immune responses Morley [67]
Saprolegnia spp. Freshwater fish (salmon, trout, catfish) Affected immune responses through tissue damage and the suppression of protective barriers in aquatic organisms Duan et al. [68]
Aeromonas salmonicida Salmonids (salmon, trout) Impaired immune responses in salmonids led to significant tissue necrosis and impaired specific and nonspecific immunity Rauta et al. [59]
Aeromonas hydrophila Freshwater fish (carp, catfish, tilapia) Inhibited innate immune responses and diminished the host's defence mechanisms Ben Hamed et al. [69]
Flavobacterium columnare Freshwater fish (tilapia, catfish, carp) Infected multiple fish species, compromising their immune defences and causing skin lesions that further diminish immune responses Teixeira Alves and Taylor [63]
Edwardsiella tarda Eels, catfish, and other freshwater fish Impaired immune responses in eels hindered the production of immune cells and elevated their vulnerability to bacterial infections Shoemaker et al. [70]
Vibrio spp. Marine fish and shellfish Immunosuppression in fish and shellfish weakened their immune responses and increased susceptibility to diseases Derome et al. [71]
Yersinia ruckeri Salmonids (salmon, trout) Redmouth disease in fish impaired immune responses and heightened vulnerability to secondary infections Mydlarz et al. [72]
Aeromonas hydrophila Zebrafish (Danio rerio) The bacterial infection induces a pro-inflammatory response, increases oxidative stress, and results in tissue damage in essential organs such as the kidney. This immune activation constitutes a component of the fish's defense mechanism against bacterial infection. Rodríguez et al. [73]
A. hydrophila, A. salmonicida, L. garvieae, S. agalactiae, and V. parahaemolyticus Goldfish (Carassius auratus) The pathogens (A. hydrophila, A. salmonicida, L. garvieae, S. agalactiae, and V. parahaemolyticus) adversely impact the immune system, as these bacteria are recognized for inducing illnesses in fish. Yi et al. [74]
Aphanomyces invadans Cirrhina mrigala Aphanomyces invadans compromises the immune system by inducing infections that elevate mortality rates in untreated fish. Harikrishnan et al. [75]
Aphanomyces invadans Zebrafish (Danio rerio) Aphanomyces invadans compromises the immune system by inducing infections that elevate mortality rates in untreated fish. Yang et al. [76]
species of Vibrio Wild eel Vibrio species influence the immune system of wild eels by colonizing and potentially compromising the mucosal barrier, which serves as the primary defense mechanism in fish immunity. Carda-Diéguez et al. [77]
Vibrio spp., Aeromonas hydrophila, Edwardsiella tarda Orange-spotted grouper (Epinephelus coioides) The pathogens may impair the immune system by inducing infections that result in tissue damage and systemic disease. Kuo et al. [78]

Bacterial pathogens

Bacterial infections are prevalent in freshwater and marine fish, particularly in aquaculture settings, due to the high fish density [79]. Therefore, it is important to manage bacterial diseases to prevent morbidity and mortality and maintain fish health. Vibrio spp., for instance, is responsible for a significant disease affecting marine fish and shrimp known as vibriosis. As a result, numerous studies have investigated the effects of Vibrio spp. on infections, immune modulation, and preventive strategies across various fish species [[65], [80], [81], [82], [83], [84], [85], [86], [87]]. It was found that these bacteria triggered immune responses by activating the innate immune system to produce antimicrobial peptides (AMPs) and activate phagocytic cells. Moreover, Vibrio infections upregulated immune-related genes in fish, such as the cytokine IL-1β, which is crucial for inflammation and immune cell recruitment [88].

Aeromonas spp., particularly Aeromonas hydrophila, is recognised for its substantial impact on the immunological response of numerous fish species. The pathogen infiltrates the host by disrupting the integumentary or gastrointestinal barriers, regulating cytokines and triggering inflammation. In Zhejiang province, China, A. hydrophila outbreaks caused substantial economic losses by inducing biofilm formation and mass mortalities in aquaculture species [89,90]. Aeromonas spp. regulated the immunological response of fish via many pathways, affected by bacterial virulence mechanisms and extrinsic factors such as nutrition and environmental conditions. In red crucian carp, A. hydrophila infection affects the gut-liver axis, modifying immune signalling pathways that regulate pro- and anti-inflammatory cytokines [89]. Recent research indicated that the removal of virulence factors, such as orf02889 from A. hydrophila, increased the expression of crucial immune-related genes in zebrafish, including interleukin-1β (il-1β) and antibacterial peptides such as lysozyme (lyz-c). These changes suggest the immune response may be activated under specific conditions, potentially facilitating vaccine development [90]. Nutritional therapies such as food supplementation with immunostimulants have also shown promising results in managing infections in aquaculture species. For example, administering algal extracts to Labeo rohita (rohu) enhanced their resistance to A. hydrophila due to increased neutrophil and lysozyme activities. Furthermore, the algal diet improved their survival rates compared to the control group after bacterial challenge [91]. This finding illustrates that dietary and environmental variables can influence the fish’s immune response to Aeromonas infections.

Edwardsiella tarda significantly influences aquatic animals' immune responses by activating innate and adaptive defences while employing evasion mechanisms that facilitate its persistence and pathogenicity. Comprehending these immune interactions is essential for formulating effective vaccines and treatment strategies to safeguard aquatic species, especially in aquaculture settings where E. tarda represents a considerable risk. Edwardsiella tarda is another pathogenic bacterium that significantly affects the immune response of aquatic animals [92,93]. When exposed to E. tarda infection, aquatic animals initiate an oxidative burst that produces reactive oxygen species (ROS) to eliminate pathogens [94,95]. However, E. tarda can neutralise reactive oxygen species (ROS) and continue to evade the host's immune mechanisms by producing antioxidant enzymes, such as iron-cofactored superoxide dismutase (FeSOD) [96,97]. Meanwhile, infected zebrafish and flounder generated pro-inflammatory cytokines to attract immune cells to the infection site as a strategy against E. tarda attacks. Nonetheless, E. tarda has developed mechanisms to evade detection and partially circumvent the immune response [98], such as inhibiting macrophage activity and reducing phagocytosis to reduce the innate immune response [99].

The adaptive immune response, which encompasses humoral and cell-mediated immunity, is essential for defence against E. tarda [100]. Infected flounder and tilapia were found to increase their antibody production, particularly IgM, to neutralise E. tarda [101]. Meanwhile, cellular immunity in aquaculture species helps protect them against this bacterium by activating T-cells and other immune cells to eradicate infected cells. Vaccinating flounder and carp with inactivated E. tarda has been proven to increase antibody production and cellular immune responses [102,103], serving as critical measures for safeguarding aquatic species against E. tarda infections [102]. Aquatic species infected with E. tarda also demonstrated notable metabolic and transcriptomic alterations. For instance, exposure to E. tarda triggered metabolic changes in Chinese soft-shelled turtles that enhanced their immune functions by elevating energy and protein metabolism [104]. Transcriptomic analyses in flounder and tilapia exhibited the upregulation of immune-related genes associated with inflammation and pathogen recognition as a response to E. tarda infection [105].

Synergistic effects of temperature and bacterial infection

The interaction between abiotic and biotic stressors significantly influences immune responses in aquatic species. Temperature fluctuations can exacerbate bacterial infections, leading to a synergistic effect that undermines host immunity [[49], [106]]. Increased water temperatures frequently enhance pathogen virulence, growth rates, and transmission dynamics, while concurrently compromising the host's immune function [107]. Vibrio spp. and Aeromonas hydrophila demonstrate increased proliferation in warmer conditions, leading to more severe infections and immune suppression in fish species, including Nile tilapia and common carp [108].

High temperatures induce oxidative stress and immune dysregulation in aquatic animals, weakening essential immune components, including phagocytic activity, lysozyme levels, and cytokine signalling [109,110]. The modifications diminish the host's ability to effectively eliminate pathogens. The molecular synergy is evident, as temperature stress influences the expression of immune genes such as IL-1β, TNF-α, and heat shock proteins, which collectively modulate responses to bacterial challenges [111]. The compounding effect is notably significant in aquaculture, where temperature regulation is essential for controlling disease outbreaks. Recognizing and managing the synergistic impacts of thermal and bacterial stressors is essential for sustaining immune resilience in aquatic organisms, especially in the context of global warming and intensified aquaculture practices.

Virus

Viral infections pose significant challenges in aquaculture due to their high transmissibility and the complexities associated with treating viral diseases after they are transmitted [112]. Numerous viruses directly inhibit or circumvent the immune system, facilitating their rapid replication within host cells. Infectious hematopoietic necrosis virus (IHNV) is a major viral pathogen that predominantly impacts salmonid fish species, especially in freshwater ecosystems [[113], [114], [115]]. The immune response involves the activation of interferons, signalling proteins that impede viral replication. Chaves-Pozo et al. [116] demonstrated that interferon-stimulated genes (ISGs) are essential for fish protection against IHNV, as they enhance the antiviral state of the immune cells. Viral hemorrhagic septicemia virus (VHSV) is a viral pathogen that impacts various fish species, resulting in internal haemorrhaging. Fish activate innate immune responses via the secretion of cytokines such as TNF-α and IFN-γ. Infected fish display a robust antiviral response alongside suppression of adaptive immunity against VHSV, highlighting the virus's capacity to circumvent more specific immune defences [117].

Aquaculture increasingly use antiviral agents to manage viral outbreaks that threaten fish and shellfish populations. Antiviral drug development for aquaculture remains in its early stages; however, several promising examples demonstrate the potential effectiveness of this therapy in managing viral infections. Mondal et al. [118] revealed the application of polyvinylpyrrolidone-iodine (PVP-I) serves as an example of a broad-spectrum antiseptic utilized to inhibit viral replication and decrease viral loads in aquaculture systems. PVP-I effectively inhibits IHNV in salmonids by preventing viral adhesion to epithelial cells, thereby restricting infection spread. Another example involves the utilization of coumarin derivatives, which exhibit antiviral properties against IHNV. Research indicates that coumarin compounds can inhibit viral entry and replication, resulting in a reduced mortality rate among infected fish populations [114]. Antiviral activities against the spring viraemia of carp virus have been identified in extracts of medicinal plants such as Schisandra chinensis, thereby confirming their potential application as natural antivirals in aquaculture [119]. The general application of antiviral drugs in aquaculture is limited by stringent regulatory approvals and the necessity for extensive field trials to demonstrate their efficacy and safety. The control of viral diseases in aquaculture relies significantly on biosecurity protocols, vaccination initiatives, and selective breeding for enhanced disease resistance.

Fungus

Fungal infections frequently take advantage of weakened immune systems or are promoted by environmental factors like inadequate water quality that support fungal proliferation., elevating mortality rates within affected populations [[120], [121], [122]]. Saprolegnia spp. is recognised as the most prevalent fungal infection, a water mould that induces saprolegniosis in freshwater fish or infections of the skin and gills [123]. The fish’s innate immune system prevents the spreading of infection by activating reactive oxygen species (ROS) and the release of pro-inflammatory cytokines. Parra-Laca et al. [124] demonstrated enhanced melanin production in fish during Saprolegnia infections to encapsulate and isolate the pathogen.

Scientists have developed and implemented various preventive measures to mitigate fungal infections in aquatic species, focusing on environmental management and direct interventions for fish health. Maintaining optimal water quality is essential, as suboptimal conditions promote fungal proliferation [123]. Regular monitoring and control of parameters, including temperature, pH, and dissolved oxygen levels, resulted in decreasing the growth of fungal. According to Hu et al. [125], hydrogen peroxide and potassium permanganate are significant antifungal agents employed in the management of Saprolegnia outbreaks in aquaculture. Additionally, significant research efforts are directed towards immunostimulants and probiotics that enhance the immune response to fungal infections. Barde et al. [126] indicate that biological interventions enhance innate immunity, thereby protecting fish from environmental stressors. Another promising strategy involves selective breeding for resistance to Saprolegnia infections. Research in genetics and breeding initiatives aims to create fish strains that exhibit improved resistance to fungal pathogens [127]. Vaccine development against Saprolegnia remains in the experimental phase and is being investigated as a potential long-term preventive measure [128].

The application of aquatic antifungal agents is governed by considerations of food safety, environmental sustainability, and the welfare of aquatic organisms. Various regulatory frameworks govern the approval, application, and monitoring of antifungal agents used in the aquaculture industry. Antifungal agents require rigorous evaluation by regulatory bodies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA). Rodgers and Furones [129] examine the safety, efficacy, and potential environmental impacts of the substances to reduce risks to consumers and ecosystems. Some antifungal agents, including malachite green, are restricted or prohibited because of their carcinogenic characteristics and detrimental environmental impacts [130,131]. Regulatory bodies determine allowable antifungal agents and set maximum residue limits (MRLs) for aquaculture products meant for human consumption [132]. According to Patil et al. [133], producers must comply with Good Aquaculture Practices (GAP), which focus on disease prevention and the judicious use of antifungal agents to mitigate the risk of drug resistance and environmental contamination.

Monitoring programs, such as the National Residue Program (NRP) in the United States, are essential and perform periodic assessments and residue testing to ensure compliance with established safety thresholds. These initiatives are essential for monitoring antifungal usage to ensure that aquaculture products adhere to acceptable safety limits [134]. Training and education programs enhance the appropriate use of antifungal agents, emphasizing correct dosages, withdrawal periods, and waste management to mitigate environmental discharge and protect aquatic ecosystems. Comprehensive regulatory measures will enable the aquaculture industry to sustainably manage fungal infections, reduce risks associated with antifungal agents, and ensure the production of safe, healthy aquatic products for global consumption. The integrated management strategies that combine water quality control, targeted antifungal treatments, genetic enhancement, and immunoprophylaxis effectively protect aquaculture from fungal infections, thereby ensuring healthier fish populations and enhancing sustainability within the industry.

Parasites

Parasites, such as protozoans and helminths, are notable biotic stressors in fish, often evading or manipulating the host immune system to establish chronic infections [135]. Protozoan parasites, including Ichthyophthirius multifiliis (Ich) and Cryptocaryon irritans, significantly impact aquaculture by affecting all aquatic species [136,137]. Ich is responsible for white spot disease in freshwater fish, inducing immune responses such as heightened mucus production to encapsulate the parasite and activate complement proteins to lyse its cells. In fish with adaptive immunity, their stress response to parasitic infection is characterised by the rise of antibody levels and heat shock proteins are upregulated [138]. Cryptocaryon irritans induce analogous infections in the gills and skin of marine fish, triggering a protective mechanism to thwart additional parasitic invasion known as gill hyperplasia [139]. Digenetic trematodes are multicellular parasitic helminths that complicate immune responses because of their intricate life cycles, frequently resulting in chronic infections. Wu et al. [102] reported that trematode infections in fish lead to granuloma formation, where immune cells encapsulate the parasites to isolate and neutralise them.

In summary, parasitic infections pose a major concern in aquaculture, as protozoans and helminths can evade or manipulate the immune system. Understanding the fish’s immune responses, including mucus production, expression of heat shock proteins, gill hyperplasia, and granuloma formation. Future studies should focus on developing effective treatments and preventive strategies to reduce the impact of parasitic diseases in aquaculture, such as vaccines and improved biosecurity measures.

Microbiome and symbiotic interactions

The fish microbiome is crucial for regulating immune responses and sustaining overall health [140]. Microbial communities act as a primary defence mechanism by inhibiting the colonisation of harmful pathogens through competitive exclusion and the synthesis of antimicrobial compounds (Zhang et al. [141]. Furthermore, the microbiome is crucial in maintaining immune homeostasis through its interactions with the host's immune system, affecting innate and adaptive immune responses. Beneficial bacteria in the gut, skin, and gills enhance immune function by strengthening physical barriers, including the mucosal layer, and facilitating the production of immune effectors [[142], [143], [144], [145]].

The microbiome significantly contributes to enhancing immune resilience in fish, primarily through its interactions with beneficial probiotic bacteria. These mechanisms are critical for managing fish health in aquaculture and facilitate the development of targeted probiotic therapies to enhance disease resistance in fish populations. Probiotics improve fish immunity in aquaculture, primarily influencing the mucosal immune system in the gastrointestinal tract [146]. These live beneficial bacteria regulate immune responses by stimulating IgM to neutralise pathogens and activate the complement system [147,148]. In addition, probiotics enhance AMP synthesis, which is the synthesis of small molecules that directly target and eliminate invading pathogens. Recent studies have reported that probiotic supplementation significantly promoted Toll-like receptor (TLRs) expression in fish [[149], [150], [151]]. These pattern-recognition receptors are crucial to the innate immune system, identifying pathogen-associated molecular patterns (PAMPs), including bacterial lipopolysaccharides and viral RNA. Upon recognition of these PAMPs, TLRs activate immune signalling pathways, resulting in downstream immune responses such as producing pro-inflammatory cytokines and recruiting immune cells to infection sites [152].

Abiotic factors affecting immune responses

Aquatic animals live in environments characterized by frequent fluctuations in physical and chemical conditions, which can substantially affect their immune responses. Factors including temperature, salinity, pH, oxygen levels, and pollutants significantly influence the function and efficacy of both innate and acquired immune systems. These stressors can either inhibit or stimulate immune pathways, influencing disease susceptibility, resistance, and overall health. Fig. 2 illustrates the primary abiotic factors and their impacts on the immune system components in aquatic species, facilitating a deeper understanding of these interactions.

Fig. 2.

Fig 2:

Abiotic factors influencing immune systems in aquatic animals.

Temperature

Temperature is a vital abiotic factor that has substantial impacts on the innate and adaptive immune responses of aquatic animals. Aquatic animals are ectothermic and, thus, rely on external temperatures to regulate their physiological processes, such as immune function [153]. Water temperature fluctuation can either strengthen or diminish the fish’s immune responses, influencing their susceptibility to pathogens. As the primary defence mechanism, innate immunity is more sensitive to temperature variation. When the temperature decreases, immune cells' (macrophages and phagocytes) activities are reduced, thereby impairing their capacity to recognise and eliminate pathogens [154]. Furthermore, cold conditions lower enzymatic activities critical for immune defence, thus reducing the efficacy of lysozyme and complement proteins. Meanwhile, elevated temperatures can initially enhance immune function. However, prolonged exposure may lead to thermal stress, resulting in decreased immune efficiency and heightened disease risk.

Table 2 demonstrates that the adaptive immune responses and innate immunity of aquatic animals are influenced by temperature, particularly lymphocyte proliferation, antibody production, and expression of major histocompatibility complex (MHC) molecules [155,156]. Cold conditions frequently result in delayed immune responses, characterised by reduced antibody production and poorer capacity to fight infections [157]. In contrast, higher temperatures can accelerate immune responses. Nonetheless, the accuracy of immune recognition may be compromised under this condition, leading to decreased efficiency in pathogen clearance. Temperature also influences pathogen-host interactions by affecting pathogen virulence and replication rates. Pathogens often flourish within temperature ranges, and sudden temperature fluctuations can establish conditions conducive to pathogen proliferation and compromise the host's immune defences. This dynamic presents a considerable challenge in aquaculture, where maintaining optimal water temperatures is essential for disease prevention in fish and their overall health.

Table 2.

Impact of temperature on the immune responses of various aquatic species.

Species Optimal temperature Effects on the immune system Reference
Grass carp (Ctenopharyngodon idellus) High temperatures (varied) Elevated temperatures adversely impact the expression of immune-related genes [158]
Pacific oyster, Mediterranean mussel Varied temperature stress Temperature fluctuation influenced the immune system Rahman et al. [159]
Nile Tilapia (Oreochromis niloticus) Varied (low/high) Increased temperatures enhanced immune responses Qiang et al. [160]
Japanese flounder (Paralichthys olivaceus) Varied (against LCDV virus) Reduced temperatures diminished immune efficacy in combating viral infections Xu et al. [161]
Antarctic fish (Harpagifer species) 2 °C - 10 °C Temperature increase enhanced immune responses Saravia et al. [162]
Abalone (Haliotis rubra) Elevated temperatures Elevated temperatures diminish antibacterial and antiviral defences Dang et al. [163]
Surf clams (Mactra veneriformis) Immediate temperature changes Immediate temperature fluctuations adversely impact immune responses Yu et al. [164]
Turbot (Scophthalmus maximus) High temperatures The skin's immune system suffered when water temperature increase Huang et al. [165]
Freshwater crayfish (Astacus leptodactylus) High temperatures Immune response diminished, increasing the risk of white spot syndrome virus (WSSV). Jiravanichpaisal et al. [166]
Channel catfish (Ictalurus punctatus) 15–20 °C Reduced temperatures inhibited immune responses, while a moderate increase promoted immune activity Martins et al. [167]
Tambaqui (Colossoma macropomum) Varied temperatures The immune response was influenced by temperature and environmental pollutants Salazar-Lugo et al. [168]
Arctic charr (Salvelinus alpinus) 10 °C The immune response of Arctic charr to vaccination is favorable at temperatures up to 10 °C; however, higher temperatures do not provide further advantages. Pylkkàet al. [169]
Common carp (Cyprinus carpio L.) Minimizing temperature fluctuations The study indicates that reducing temperature fluctuations is essential for preserving immune competence in carp. Engelsma et al. [170]
Sablefish (Anoplopoma fimbria) 12-14 °C At 18 °C, immune responses, including lysozyme activity and phagocytosis, were elevated, suggesting an activation of the immune system at increased temperatures. Kim et al. [171]
Shore crab (Carcinus maenas) 20 °C A gradual temperature increase from 10 °C to 20 °C significantly elevated circulating haemocyte (immune cell) counts, suggesting enhanced immune activity. Truscott and White [172]
Sockeye salmon (Oncorhynchus nerka) 8-12 °C Temperature at 8-12 °C enhances the non-specific immune responses of fish Alcorn et al. [173]
Surf clams (Mactra veneriformis) 20 °C Decreased haemocyte proliferation and lysozyme activity may impair the clam's capacity to defend against infections. Yu et al. [164]
Farrer's scallop Chlamys farreri < 29 °C A temperature of 29 °C negatively impacts immune function and energy balance. Wang et al. [174]
Blue mussels (Mytilus spp.) 5-10 °C This study demonstrated that temperature influences immune function, with low temperatures enhancing hemocyte health and phagocytosis, whereas high temperatures may induce inflammatory responses and metabolic stress in blue mussels. Beaudry et al. [175]
Rainbow trout (Oncorhynchus mykiss) 15 °C Higher temperatures (23 °C) induce immune and metabolic stress, thereby increasing susceptibility to pollutants and parasites. (2019)
Mediterranean finfish aquaculture N/A Temperature influences fish immune function and vulnerability to diseases in Mediterranean aquaculture. Cascarano et al. [176]
Walbaum (Oncorhynchus mykiss) 10 °C Temperature significantly influences immune responses and pathogen development, as elevated conditions promote parasite growth and host decline. Kocan et al. [177]
Catla (Catla catla) 28 °C Maintaining Catla catla within the temperature range of 25–28 °C is essential for ensuring immune competence, optimal growth, and survival. It is critical to avoid temperatures below 20 °C or above 35 °C to prevent stress and mortality. Sharma et al. [178]
Mozambique tilapia (Oreochromis mossambicus) 27 °C Temperature significantly influences immune responses in tilapia, with both low and high temperatures impairing disease resistance. Maintaining the fish at 27 °C reduces physiological stress, thereby enhancing survival rates in both cultured and natural environments. Ndong et al. [110]
Pufferfish (Takifugu obscurus) 25 °C Reduced temperature resulted in Immunosuppression diminishes the capacity of fish to combat infections and recuperate from diseases. Cheng et al. [179]
Pufferfish (Takifugu obscurus) High temperatures The immune system is influenced by temperature, as elevated temperatures can induce oxidative stress and lead to potential tissue damage. Cheng et al. [180]
Manila clam (Ruditapes philippinarum) 18 °C High temperatures can lead to immune suppression or overactivation, compromising the clam's ability to defend against pathogens and increasing susceptibility to disease and mortality. Menike et al. [181]
Mytilus coruscus < 31 °C Temperatures exceeding 31 °C are associated with increased mortality and impact the immune system. Li et al. [182]
Polar cod (Boreogadus saida) 4 °C Increased temperature suggests that increased thermal conditions enhance stress and immune responses. Kim et al. [183]
Nile tilapia (Oreochromis niloticus) 27.9 °C - 29.4 °C Temperature around 20.0 °C resulted in higher mortality and reduced the immune system of fish Qiang et al. [160]
Pacific herring (Clupea pallasii) 12.6 °C The immune system of Pacific herring is significantly influenced by temperature, which modulates the speed and efficacy of vaccine-induced responses. Hart et al. [184]
Atlantic salmon (Salmo salar) 16.5 - 17.5 °C Temperature influences immune function by affecting stress levels, metabolic demand, and susceptibility to pathogens. Stehfest et al. [185]
Olive flounder (Paralichthys olivaceus) > 16.5 °C Temperature influences the immune system by inhibiting growth and immune function; however, taurine supplementation can alleviate these effects, thereby improving the flounder's resistance to cold stress. Kim et al. [186]
Shell mussel (Mytilus coruscus) 25 °C Temperature significantly influences the immune system, as extended exposure to high temperatures diminishes immune efficiency and enhances cellular damage. Wu et al. [187]
Orange-spotted grouper (Epinephelus coioides) 28 °C Temperature has a significant impact on the immune system, where cold stress induces immunosuppression, inflammation, and apoptosis. Sun et al. [111]
Hard clams (Mercenaria mercenaria) 21 °C Temperature has a significant impact on the immune system through the modulation of gene expression associated with immune recognition, pathogen defense, and oxidative stress. Wang et al. [188]
Diamond-backed terrapins (Malaclemys terrapin) 34 °C Elevated temperatures, approaching 34 °C, are likely to improve immune competence by facilitating accelerated metabolism and more effective physiological processes. Tamplin et al. [189]
Thick-shelled river mussel (Unio crassus) 17 °C Increased temperatures can lead to either immune suppression or hyperactivation, potentially impacting the host's capacity to endure the parasitic phase. Taeubert et al. [190]
Sea cucumber (Holothuria scabra) 27 °C At elevated temperatures, low levels of reactive oxygen species (ROS) and minimal oxidative damage were noted, indicating that the immune system efficiently mitigated stress during the experiment. Kamyab et al. [191]
Pacific white shrimp (Litopenaeus vannamei) 28 °C The immune system is influenced by temperature through modifications in the expression of heat shock proteins and genes associated with apoptosis. Peng et al. [192]
Oscar (Astronotus ocellatus) 22–27 °C High temperatures induce elevated stress in fish, which may lead to potential liver damage and impaired immune function, thereby increasing their vulnerability to disease. Esmaeili et al. [193]

<: Below; >: Higher.

One notable consequence of temperature-driven immune modulation in fish is the phenomenon known as Winter Syndrome, first described by [194]. This condition occurs with exposure to low water temperatures, impairing both innate and adaptive immune functions in aquatic species [195]. Reduced phagocytic activity, diminished lysozyme levels, and suppressed cytokine expression are frequently observed. The immunosuppressive condition heightens the susceptibility of fish to opportunistic bacterial and viral infections, including Aeromonas hydrophila and Vibrio spp., particularly in aquaculture systems without thermal regulation. Winter Syndrome illustrates the manner in which seasonal abiotic stressors can exacerbate biotic challenges, underscoring the necessity for temperature regulation and immunostimulatory strategies in colder periods.

Understanding the relationship between temperature and immune responses is essential in aquaculture and conservation. Global climate change modifies temperature patterns in aquatic ecosystems, potentially intensifying disease dynamics and elevating the risk of pathogen outbreaks in wild populations, thereby jeopardising biodiversity. Regulating temperature in controlled environments (aquaculture systems) can boost immune resilience and minimise disease occurrence. Temperature significantly influences the immune function of aquatic animals, and its effects are increasingly concerning, given the changing environmental conditions.

Salinity

Salinity is a critical abiotic factor in aquatic environments that significantly influences the immune responses of aquatic animals. Fish and invertebrates frequently experience variations in salinity, which can disturb their physiological homeostasis and subsequently affect their immune function [[196], [197], [198]]. The impact of salinity on immune function is primarily mediated by mechanisms associated with osmotic balance, ion regulation, and cellular stress (Fig. 3). Elevated salinity (hyperosmotic conditions) often results in cellular dehydration, impairing immune cells' proliferation and function, including leukocytes. Lu et al. [199] found that elevated salinity levels diminished leukocyte phagocytic activity, reduced cytokine production, and compromised the overall immune response in fish. Conversely, low salinity (hypoosmotic conditions) may lead to cellular swelling and stress, thereby initiating an acute immune response. Prolonged exposure to these conditions can lead to immune suppression. For instance, euryhaline fish exhibited elevated levels of stress-related proteins in response to hypoosmotic stress, including heat shock proteins and pro-inflammatory cytokines [200].

Fig. 3.

Fig 3:

Salinity effects on fish immune system [[199], [201], [202], [203], [204]].

The relationship between salinity and immunity is often mediated by ionic imbalance, oxidative stress, and endocrine responses. Salinity stress alters ionic balance, forcing aquatic animals to invest more energy towards osmoregulation compared to immune defence [205]. Increased salinity also causes oxidative stress that is harmful to immune cells and their functions [206]. Stress hormones such as cortisol are activated by salinity changes, suppressing immune responses, diminishing leukocyte activity, and decreasing the production of immune proteins [207].

Table 3 presents how salinity significantly influences the immune responses of aquatic animals. Euryhaline species that can withstand a wide range of salinities have developed adaptive immune mechanisms. Atlantic salmon (Salmo salar) demonstrated an upregulation of immune-related genes during the transition from freshwater to seawater, notably increasing AMPs [208]. In contrast, stenohaline species, which flourish within a limited salinity range, exhibit increased susceptibility to immune suppression when subjected to salinity stress. Wang et al. [209] reported that Pacific white shrimp (Litopenaeus vannamei) exhibit reduced hemocyte activity and immune-related gene expression under hyper- and hypo-osmotic conditions, suggesting impairments in the shrimp's infection resistance due to salinity fluctuations.

Table 3.

Effects of salinity on immune responses across various aquatic species.

Species Best salinity Effects on the immune system/Findings related to salinity and immunity Reference
Scatophagus argus 25 ppt Elevated immune responses were observed, characterized by increased expression of cytokine genes, enhanced leukocyte proliferation, and more robust immune parameters during infection. Lu et al. [199]
Syngnathus typhle Ambient salinity Enhances immune health through increased activity and proliferation of immune cells. Birrer et al. [196]
Mytilus edulis 15 ppt Reduced salinity immune responses are significantly suppressed, leading to a reduction in both functional and molecular immune characteristics. Wu et al. [210]
Nibea albiflora 6-30 ppt High salinity (42 ppt) induces stress, adversely affecting immune enzyme activity, growth, and gut health. Tian et al. [211]
Oreochromis niloticus 16 ppt At higher ppt levels, the immune systems of the fish experienced stress, resulting in an increased susceptibility to illness. El-Leithy et al. [212]
Pangasianodon hypophthalmus 10 ppt Salinity affects immune ability and may decrease the resilience of catfish to infections, including Edwardsiella ictalurid. Schmitz et al. [213]
Alosa sapidissima 14-21 ppt 14 to 21 ppt enhanced immune respond, better growth, improved enzyme activity and optimal fatty acid composition. Liu et al. [214]
Oreochromis niloticus 0-10 ppt Higher ppt inhibited immune relayed genes of IgM, IL-1β, and IFN-γ Wang et al. [215]
Takifugu fasciatus 10 ppt Low salinity supporting the immune system and reducing the physiological stress of fish Wen et al. [216]
Acanthopagrus latus and Lates calcarifer 6-12 ppt Rising salinity influenced the humoral immune responses of fish. Mozanzadeh et al. [217]
Procambarus clarkii 0-2 ppt Higher salinity effected the immune system of fish Xiao et al. [218]
Portunus trituberculatus 31 ppt Lower salinity induced temporary immune supression Wang et al. [219]
Anguilla japonica <0.5 ppt Freshwater condition improve immune-related gene expression Gu et al. [220]
Cyprinus carpio 0-10 ppt High salinity effects the immune and physiological stress of fish Dawood et al. [221]
Scophthalmus maximus 24-30 ppt High salinity effect the immune system, particularly IgM expression in the kidney. Huang et al. [222]
Anoplopoma fimbria 31.5-35 ppt Higher salinity improves immune system of fish Kim et al. [171]
Echinometra lucunter 35 ppt Reduced salinity may trigger oxidative stress, while elevated salinity can result in mitochondrial dysfunction. Honorato et al. [223]
Eriocheir sinensis 8 ppt High salinity effect immune system of the crab Yang et al. [224]
Gadus morhua 10 ppt The density did not affect the immune system of fish; however, 10 ppt is the optimal value for growth. Árnason et al. [207]
Litopenaeus vannamei 1-5 ppt Low salt-tolerant hybrid shrimp demonstrate enhanced immune performance and antioxidant capacity. Ye et al. [225]
Notopterus chitala 0-3 ppt Salinity exceeding 6 ppt can result in considerable oxidative damage and immune suppression. Moniruzzaman et al. [226]
Litopenaeus vannamei 25-30 ppt Salinity at 25-30 ppt increase the immune respond and disease resistance Wang and Chen [227]
Penaeus monodon 20 ppt Salinity at 20 ppt produced optimal growth, survival, and a stable immune response. Rahi et al. [228]
Acanthopagrus schlegelii 22 ppt At a salinity of 22 ppt, fish experience reduced physiological stress and enhanced immune function, allowing them to survive even at lower salinities of 4-5 ppt. Li et al., [229]
Haliotis diversicolor supertexta 30 ppt Salinity at 30 ppt enhances immune system of Haliotis diversicolor supertexta Cheng et al. [230]
Dicentrarchus labrax 6-12 ppt Results indicate that Dicentrarchus labrax acclimatized at intermediate salinities (6 and 12 ppt) exhibit superior performance during exposure to extreme cold conditions (8 °C). Jakiul Islam et al. [231]
Aquarana catesbeiana 2-4 ppt Higher salinity can induce oxidative stress and potential immune suppression Zheng et al. [232]
Litopenaeus vannamei 36 ppt Salinities under 36 ppt are recommended for L. vannamei aquaculture to enhance immune health and metabolic balance. Long et al. [233]
Haliotis discus discus 25 ppt Keeping salinity levels below this threshold (25 ppt) may enhance immune function and overall health in disk abalone. De Zoysa et al. [234]
Procambarus clarkii 6 ppt High salinity up to 18 ppt cause immune disruption and metabolic stress Luo et al. [235]
Ctenopharyngodon idella 2 ppt Higher salinity (6 ppt) lead to immune suppression and decline in growth performance Liu et al. [236]
Larimichthys polyactis 22.1 ppt The fish can adapt at low salinity, but salinity at 22.1 ppt improve immune system and physiological function Mengjie et al. [237]
Scapharca subcrenata 22 ppt Salinity changes effect the immune system, growth performance and physiological balance of Scapharca subcrenata Mo et al. [238]
Oreochromis spp. 5-10 ppt Moderate salinity improve immune responses and health of fish Ulkhaq et al. [239]
Pseudosciaena crocea 5-10 ppt High salinity (15-20ppt) is not suitable for growth and immune system Wang et al. [240]
Litopenaeus vannamei 56 ppt A salinity level of 56 ppt is recommended to ensure immune stability and minimize physiological stress. Shen et al. [241]
Cherax quadricarinatus 5 ppt Salinity at 5 ppt improve the immune function, antioxidant activity and gut health Liu et al. [242]
Litopenaeus vannamei 0-4 ppt Rearing fish with multispecies of probiotic is recommend at low salinity (0-4 ppt) Zannat et al. [243]
Oreochromis niloticus 0-10 ppt High salinity levels (15-20 ppt) impact the health and growth of fish when dietary supplementation with Aspergillus oryzae is administered. Shukry et al. [244]
Macrobrachium rosenbergii 13 ppt Salinity of 13 ppt is recognized as optimal for enhancing growth, immunity, enzyme activity, and successful larval development in Macrobrachium rosenbergii Wei et al. [245]
Sinonovacula constricta 30 ppt Exceeding 35 ppt has a detrimental impact on health and growth performance. Cao et al. [246]
Ruditapes philippinarum 23.3-31.1 ppt Low salinity (15 ppt) enhances susceptibility to metabolic and oxidative disruptions. Wu et al. [247]
Oreochromis niloticus 4-8 ppt Salinity levels of 4 to 8 ppt may partially reduce ammonia toxicity and enhance antioxidant defense mechanisms. Motamedi-Tehrani et al. [248]
Eriocheir sinensis ≤ 6 ppt Higher salinity (≥ 12 ppt) lead to physiological stress and increase mortality rate Zhang et al. [249]
Penaeus vannamei 40-47 g/L High salinity affected the amino acid composition in the body, which directly influences the immune system of fish. Li et al. [250]
Selenotoca multifasciata 5 ppt High salinity increased physiological strain and greater stress response Liu et al. [251]
Oryzias melastigma 15 ppt Improve growth and health performance Li et al. [252]
Giant clams (Bivalvia: Tridacnidae) 34 ppt Reduced salinities adversely affected growth and long-term health conditions. Lee et al. [253]

It is clear that environmental challenges such as climate change and human activities may increase salinity variability in aquatic ecosystems. Therefore, evaluating the relationship between salinity and immune function is crucial for effectively managing aquaculture systems. Future research should focus on examining species-specific adaptations to salinity stress and exploring strategies to improve immune resilience in aquaculture via environmental management or selective breeding.

Oxygen levels (Hypoxia)

Hypoxia is characterised by oxygen levels falling below the normal threshold (< 2 mg/L), profoundly affecting aquatic organisms' physiological and immune responses [254]. Aquatic animals under hypoxia are pressured to focus on physiological responses aimed at maintaining oxygen homeostasis, compromising their immune function and increasing their susceptibility to infections. Moreover, hypoxia modifies immune-related gene expression, diminishing the function of critical immune cells (macrophages, neutrophils) and decreasing the synthesis of reactive oxygen species (ROS) vital for pathogen eradication [255,256]. Aquatic animals exposed to hypoxic conditions frequently exhibit diminished immune defences, rendering them more vulnerable to bacterial, viral, and parasitic infections, as illustrated in Table 4.

Table 4.

Oxygen levels and immune responses in aquatic species under hypoxic conditions.

Species Optimal oxygen level Critical hypoxia threshold Immune response under hypoxia Reference
Salmo salar > 95 % saturation (∼8.3 mg/L) 70 % saturation (∼5.9 mg/L) Decreased expression of immune-related genes in low oxygen conditions, compromised stress responses, and inflammation associated with prolonged hypoxia Beemelmanns et al. [257]
Cyprinus carpio 6–8 mg/L ∼3 mg/L Improved hypoxia tolerance is associated with a slight decrease in immune function Cerra et al. [258]
Danio rerio 6–7 mg/L < 3 mg/L Enhanced survival rates in chronic hypoxia and improved immune response adaptation during juvenile stages following prolonged exposure Chun and Kim [259]
Pseudobagrus ussuriensis Above 6 mg/L 1.5 mg/L Acute hypoxia results in immune suppression, skin damage, and heightened susceptibility to infections following reoxygenation Liu et al. [260]
Sander lucioperca 8.3 mg/L ∼3.2 mg/L Chronic hypoxia affects leukocyte count, impairs acute inflammatory responses, and increases susceptibility to infections Huo et al. [261]
Apostichopus japonicus 6–8 mg/L 1.5–2 mg/L Reduced activities of antioxidant enzymes, diminished immune defences, lowered oxygen uptake, and increased oxidative stress Huo et al. [261]
Ictalurus punctatus ∼6 mg/L < 2 mg/L Elevated heart rate and enhanced oxygen extraction support immune function; however, these mechanisms are impaired under prolonged hypoxia Cerra et al. [258]
Carassius auratus > 5 mg/L < 1 mg/L Acute hypoxia promotes cardiovascular adaptation; however, it may lead to a suppression of immune function over an extended period Cerra et al. [258]
Hypophthalmichthys molitrix > 6 mg/L < 0.76 mg/L Elevated oxidative stress and immune suppression, particularly in hepatic and brain tissues, lead to compromised physiological function Wang et al. [262]
Scylla paramamosain > 6 mg/L 1 mg/L Reduced antioxidant capacity and altered glutaredoxin activity in gills diminished immune defence against pathogens. Jie et al. [263]
Ruffe and Flounder > 6 mg/L (normoxia) < 1.5 mg/L Tissue damage and downregulation of immune genes in the gills and heart, alongside diminished metabolic and immune functions, occur under conditions of severe hypoxia Tiedke et al. [264]
Karpowicz et al. [265]
Benthic organisms (Coastal) > 4–5 mg/L < 2 mg/L Reduction in biodiversity, immune system failure, and behaviour changes when animals relocate or die from oxygen loss Rabalais et al. [266]
Zooplankton (Lake ecosystems) > 5 mg/L < 2 mg/L Reduced immunological responses and physiological stress, severe hypoxia collapses communities Karpowicz et al. [265]

<: Less than; >: More than

The innate immune system is susceptible to hypoxic stress. Oxygen-dependent mechanisms are impaired in low-oxygen environments. For instance, reduced ROS production decreased the efficacy of macrophages and neutrophils in eliminating pathogens. Hypoxia-induced immune suppression has been documented in O. niloticus (Nile tilapia) [267]. Wang et al. [268] revealed that prolonged hypoxia stress diminished phagocytic activity and heightened vulnerability to Streptococcus iniae infections. Likewise, the adaptive immune system, which includes lymphocytes such as T and B cells, is also compromised during hypoxia. Hypoxia hampers lymphocyte proliferation, antibody production, and dendritic cell maturation, essential for antigen presentation and immunological memory development [269]. This disruption diminishes the capacity of aquatic animals to generate a robust adaptive immune response to pathogens.

The hypoxia-inducible factor (HIF) pathway is crucial in regulating immune changes at the molecular level during hypoxia. Stabilising HIF-1α in hypoxic conditions activates genes that regulate oxygen homeostasis and facilitate metabolic adaptation. This shift frequently compromises immune function, as metabolic resources are redirected towards survival instead of maintaining immune competence [270]. In fish and other aquatic animals, sustained activation of the HIF pathway during chronic hypoxia results in immune suppression, diminishing the organism's overall fitness and capacity to respond to pathogenic threats. Consequently, overcrowding and poor water quality can worsen hypoxia and result in disease outbreaks in natural and aquaculture populations [271].

The implications of hypoxia-induced immune suppression in aquatic environments are substantial, particularly regarding climate change and anthropogenic oxygen depletion. Increasing hypoxic conditions in aquatic habitats, driven by eutrophication and elevated water temperatures, may impair the disease resistance of aquatic organisms, potentially resulting in population declines and disruptions to ecosystem equilibrium. Investigating the molecular and physiological mechanisms through which hypoxia influences immune function in aquatic animals is essential for formulating mitigation strategies to enhance these organisms' resilience to environmental stressors.

Pollutants and chemicals

Aquatic environments are increasingly affected by various pollutants, such as heavy metals, pesticides, and pharmaceuticals, which can notably impair the immune systems of aquatic organisms. A summary of the effects of pollutants and chemicals on the immune systems of aquatic animals can be found in Table 5. Heavy metals, including cadmium (Cd), lead (Pb), mercury (Hg), and copper (Cu), are immunotoxic, causing oxidative stress and dysfunction in immune cells. Cadmium exposure in O. niloticus (Nile tilapia) diminished phagocytic activity and ROS generation, which is essential for innate immune defence [272]. Similarly, Mytilus galloprovincialis (marine mussels) exposed to cadmium exposure demonstrated impaired hemocyte function in their immune responses [273,274]. Mercury, especially in its methylmercury (MeHg) form, inhibited immune responses in Danio rerio (zebrafish) by influencing T-cell proliferation and antibody production [275,276]. Meanwhile, lead exposure decreased cytokine production critical for initiating immune responses, including interleukin-1β (IL-1β) and tumour necrosis factor-alpha (TNF-α) [277,278].

Table 5.

Effects of pollutants and chemicals on the immune systems of aquatic animals.

Species Pollutant/Chemical Effects on the immune system Reference
Common carp (Cyprinus carpio) Chlorpyrifos Inhibition of macrophage function and decreased lysozyme synthesis, compromising innate immune responses Taheri Mirghaed et al. [279]
Rainbow trout (Oncorhynchus mykiss) Pyrethroids Impairment of humoral immunity and a decrease in antibody production Major and Brander [280]
African clawed frog (Xenopus laevis) Atrazine Endocrine disruption, diminished immune cell proliferation, and modified cytokine profiles Galbiati et al. [281]; Zhang et al. [282]
Atlantic salmon (Salmo salar) Antibiotics A decrease in gut microbial diversity leads to compromised immune defences Wang et al. [283]; Wang et al. [284]
Grass shrimp (Palaemonetes pugio) Fluoxetine Reduced antimicrobial peptide release and impaired hemocyte activity Coates and Costa-Paiva [285]; Alboni et al. [286]
European sea bass (Dicentrarchus labrax) Diclofenac (NSAID) Inhibition of prostaglandin production leads to decreased inflammatory and phagocytic activities Courant et al. [287]
Rainbow trout (Oncorhynchus mykiss) Cadmium Oxidative stress leads to damage in immune cells, resulting in impaired immune function Eskander and Saleh [288]
Atlantic cod (Gadus morhua) Cortisol (stress hormone, induced by pollutants) Increased cortisol concentrations result in immunosuppression. Campbell et al. [289]
Mediterranean mussel (Mytilus galloprovincialis) Various NSAIDs (e.g., diclofenac) Decreased immune responses, encompassing gill integrity and phagocytic activity Chaabani et al. [290]; Courant et al. [287]
European plaice (Pleuronectes platessa) Heavy metals (cadmium) Impaired innate immunity encompasses alterations in lysozyme levels and phagocytic activity Eskander and Saleh [288]
Mummichog (Fundulus heteroclitus) Polycyclic aromatic hydrocarbons (PAHs) Impairment of immune cell function and reduced susceptibility to infections Pijanowski et al. [291]

NSAID: Non-steroidal anti-inflammatory drugs

Pesticides, such as organophosphates and pyrethroids, are also immunotoxic to aquatic organisms. Chlorpyrifos, an extensively utilised organophosphate insecticide, adversely impacted the innate immunity of Cyprinus carpio (common carp) by inhibiting macrophage function and diminishing lysozyme synthesis [279]. Meanwhile, pyrethroid exposure impaired the humoral immunity in Oncorhynchus mykiss (rainbow trout), reducing their antibody production [280]. In addition, herbicides, such as Atrazine, disrupted endocrine functions in amphibians, particularly Xenopus laevis, leading to impaired immune cell proliferation and altered cytokine profiles [281]. One of the most recent risks of immunotoxic aquatic pollutants is pharmaceuticals. Antibiotics, including tetracyclines and fluoroquinolones, can alter the microbiome of aquatic animals, which is crucial for immune regulation. Antibiotic exposure in Salmo salar (Atlantic salmon) reduces gut microbial diversity, impairing their immune defences [[283], [284]]. Non-steroidal anti-inflammatory drugs (NSAIDs), such as diclofenac, inhibit prostaglandin production, impair inflammatory responses and reduce phagocytic activity in fish [[287], [290]]. Meanwhile, antidepressants such as fluoxetine have been shown to impair hemocyte activity in Palaemonetes pugio (grass shrimp), reducing AMP release and increasing susceptibility to pathogens [285].

The mechanisms responsible for these immunotoxic effects typically include the induction of oxidative stress, disruption of endocrine pathways, and interference with immune signalling processes. Oxidative stress, often induced by heavy metals and pesticides, disrupts immune cell function and diminishes its ability to combat infections [288,292]. Pollutants can activate the hypothalamic-pituitary-interrenal (HPI) axis, increasing cortisol levels linked to immunosuppression [[289], [293]]. Endocrine-disrupting chemicals (EDCs), including atrazine, exacerbate immune dysfunction by modifying cytokine signalling pathways, compromising innate and adaptive immune responses. Therefore, it is important to understand pollutants' impact on aquatic organisms' immune health, which potentially influences population dynamics and ecosystem stability.

pH levels

pH levels significantly affect the immune responses of aquatic animals due to ocean acidification due to elevated atmospheric carbon dioxide (CO₂) (Table 6). The dissolution of CO₂ in seawater results in carbonic acid formation, which decreases pH levels and can adversely impact the physiological and immunological health of numerous species. Research indicates that Crassostrea gigas (Pacific oyster) exhibited reduced hemocyte activity and impaired phagocytic responses under acidic conditions, which compromised their innate immune function and heightened their susceptibility to pathogens [[294], [295], [296]]. Mytilus edulis (blue mussel) susceptibility to bacterial infections and mortality rates increased significantly in acidified waters, illustrating the correlation between compromised immune responses and increased vulnerability [[297], [298], [299]].

Table 6.

Impact of pH levels on the immune responses of various aquatic species.

Species Optimal pH Stressful pH Effect on the immune system Reference
Pacific white shrimp (Litopenaeus vannamei) 7.5–8.5 < 7.0 Decreased hemocyte activity and dysregulation of the Na+/H+ exchanger under acidic stress negatively affect immune response Li et al. [300]; Townhill et al. [301]
Blue mussel (Mytilus edulis) 7.8–8.3 < 7.6 Decreased phagocytic ability of hemocytes increased mussel’s vulnerability to infections in acidic environments Guo et al. [298]; Mackenzie et al. [299]
Pacific oyster (Crassostrea gigas) 7.8–8.4 < 7.6 Reduced expression of immune genes and compromised hemocyte function hinder pathogen defence Dineshram et al. [302]; Gazeau et al. [297]
Arctic copepod (Calanus glacialis) ∼8.0 < 7.6 Decreased metabolic rates and diminished immune function adversely affected survival in acidic environments Thor et al. [303]
Channel catfish (Ictalurus punctatus) 7.5–8.5 < 7.0 Decreased white blood cell and phagocytic activity Sánchez-Martínez et al. [304]; Zhao et al. [305]
European sea bass (Dicentrarchus labrax) 7.8–8.3 < 7.4 Disruption of skin mucosal immunity increased susceptibility to infections, particularly Vibrio harveyi Kataoka and Kashiwada [306]
Pufferfish (Takifugu rubripes) 7.6–8.2 < 7.0 A decrease in haematological parameters (RBC, haemoglobin) was attributed to oxidative stress due to low pH exposure Xu et al. [307]
Thick shell mussel (Mytilus coruscus) 7.8–8.3 < 7.3 Decreased haemocyte count, impaired phagocytosis, diminished esterase activity at low pH, and elevated reactive oxygen species levels Wu et al. [308]
Common carp (Cyprinus carpio) 7.0–8.5 < 6.5 Lower pH levels of ammonia elevate oxidative stress and induce immune suppression Xu et al. [307]
Pipefish (Syngnathus typhle) 7.5–8.0 < 7.3 Immunosuppression and heightened vulnerability to pathogens due to ocean acidification Tort and Balasch [309]
Amur minnow (Phoxinus lagowskii) N/A Higher Ph The pH, especially in the context of saline-alkaline stress, influences the immune system of fish. Zhou et al. [198]
Spotted babylon (Babylonia areolata) 7.5–8.5 N/A Ph influence the immune system of spotted babylon Ding et al. [310]
Chinese mitten crab (Eriocheir sinensis) 7.5–8.5 N/A Low pH impairs the Toll-like receptor (TLR) pathway, thereby diminishing the immune defense of crabs. Zhang et al. [311]
Common carp (Cyprinus carpio); Mrigal (Cirrhinus mrigala); Rohu (Labeo rohita) 8.0 9.0 The pH level impacts the immune system of fish by modulating the activity of non-specific immune enzymes present in skin mucus. Sridhar et al. [312]
White shrimp (Litopenaeus vannamei) 8.0–8.1 > 8.1 Shrimp raised at pH 8.1 demonstrated significantly enhanced immune activation and immune parameters relative to those raised at pH 6.8. Chen et al. [313]
White shrimp (Litopenaeus vannamei) 7.8–8.2 >8.2 Shrimp subjected to pH 6.5 (low) and pH 10.1 (high) demonstrated reduced immune responses relative to those maintained at pH 8.2 (control). Li and Chen [314]
Nile tilapia (Oreochromis niloticus) 7.0–8.0 N/A pH influences the immune system of Nile tilapia (Oreochromis niloticus) Dominguez et al. [315]
Rohu (Labeo rohita) 6.5–7.5 < 6.5 Fish reared at pH 7.0 demonstrated superior immune and growth performance, indicating that this pH level is optimal for Labeo rohita. Singh et al. [316]
Indian white shrimp (Fenneropenaeus indicus) 7.8–8.5 < 7.0 Low pH exposure significantly compromised immune functions in F. indicus. Sharma et al. [317]

N/A: Not available.

Acidification also influences the development of immune systems during the early life stages of aquatic species. For instance, Ostrea edulis (European flat oyster) larvae cultivated in low pH environments demonstrated modified immune gene expression, which may affect their health and survival in the long term [318,319]. Furthermore, pH alterations directly impact immunity and disrupt metabolic processes, leading to physiological stress [320,321]. These findings indicated the cumulative impact of compromised immune responses due to fluctuating pH levels on aquatic species, manifesting as increased disease susceptibility and altered metabolic processes. Investigating these dynamics could aid stakeholders in predicting and overcoming the effects of climate change on marine ecosystems and their resident organisms.

Ultraviolet radiation

Ultraviolet (UV) radiation can significantly impact the immune responses of aquatic animals, as shown in Table 7. The UV-B (280–320 nm), in particular, can penetrate aquatic ecosystems at various levels depending on water clarity and depth. High UV exposure in aquatic animals causes oxidative stress, DNA damage, and compromised immune responses essential to combat pathogens and environmental stressors [[322], [323], [324]]. The UV-induced oxidative stress produces ROS that is harmful to cellular components, including DNA, proteins, and lipids, impairing cellular functions [325]. In fish and marine invertebrates, oxidative damage hampered immune parameters, such as phagocytic cell activity, AMP production, and immune competence. For instance, O. mykiss (rainbow trout) exposed to UV-B exhibited reduced lymphocyte proliferation and poor macrophage function, thereby diminishing their ability to combat pathogens [[323], [326]].

Table 7.

Effects of ultraviolet radiation on the immune responses of various aquatic species.

Aquatic species Type of UV radiation Effects on the immune system and other biological responses References
Rainbow trout (Oncorhynchus mykiss) UV-B Reduced lymphocyte proliferation; compromised macrophage activity; heightened vulnerability to pathogens Fajardo et al. [326]
Pacific oyster (Crassostrea gigas) UV-B Reduced lymphocyte proliferation; compromised macrophage activity; heightened vulnerability to pathogens Mello et al. [327]
Water flea (Daphnia magna) UV-B Increased oxidative stress; modified immune response resulting in increased vulnerability to pathogens; diminished reproductive success Ivanina et al. [328]
Seahorse (Hippocampus erectus) UV-A and UV-B Modified immune responses result in decreased resistance to viral infections and may cause endocrine disruption Song et al. [329]
Rohu fish (Labeo rohita) UV-B Ultraviolet B radiation adversely impacts immune responses, diminishing defence mechanisms in aquatic larvae Singh et al. [330]
High-altitude fish species UV-B Inhibition of primary and secondary immune responses due to UV-B exposure Subramani et al. [331]
Fish (species unspecified) UV-A and UV-B UV-B exposure modified immune system function in fish, resulting in immunosuppression Salo et al. [332]
Zebrafish (Danio rerio) UV-B UV-B radiation, in conjunction with varying temperatures, influenced stress and innate immune responses. Icoglu Aksakal and Ciltas [333]
Various fish species UV-B UV-B irradiation impaired non-specific and specific immune responses in fish Sharma and Chakrabarti [334]
Teleost fish UV-A and UV-B Ultraviolet radiation negatively affected immune function and heightened disease susceptibility in fish Lawrence et al. [335]
Three-spined stickleback (Gasterosteus aculeatus) UV-B UV-B exposure reduced innate and adaptive immune responses in sticklebacks Vitt et al. [336]
Common carp (Cyprinus carpio) and Rainbow trout (Oncorhynchus mykiss) UV-B UV-B influenced specific and nonspecific immune responses in fish Markkula et al. [337]
Marine zooplankton and ichthyoplankton UV-A and UV-B Ultraviolet radiation negatively affected immune function during the early developmental stages of marine organisms Browman et al. [338]
Three-spined sticklebacks (Gasterosteus aculeatus) UV-B Ultraviolet-B (UVB) radiation influences the immune system of fish by lower splenosomatic index and higher granulocyte to lymphocyte ration Vitt et al. [336]
European seabass (Dicentrarchus labrax) UV-B Exposure of European seabass (Dicentrarchus labrax) to UVB radiation resulted in alterations in humoral immune parameters, indicating immune system modulation across all UVB doses (low, moderate, and high). Alves et al. [339]
Gilthead Seabream (Sparus aurata) UV-B UVB exposure resulted in decreased total anti-protease and total peroxidase activities, indicating a modulation of the innate immune system. Alves et al. [340]
Atlantic Salmon (Salmo salar) UV-B Effect the plasma lgM levels, lysozyme activity and complement bacteriolytic activity Jokinen et al. [341]
Indian major carp (Catla catla) UV-B UVB radiation resulted in a marked reduction of lysozyme levels in the larvae of Catla catla. Sharma et al. [342]
African catfish (Clarias gariepinus) UV-A Exposure to UV-A resulted in diminished immune responses in African catfish (Clarias gariepinus), as indicated by: 1. A reduction in lysozyme (LYZ) activity, which serves as a marker for non-specific immune function; 2. A decrease in phagocytic activity (PhA), essential for pathogen defense. Hamed et al. [343]
Rutilus roach (Rutilus rutilus) UV-B UVB irradiation resulted in decreased activity of neutrophils and macrophages in Rutilus rutilus Salo et al. [344]
Atlantic Salmon (Salmo salar) UV-B Increased UVB radiation influenced plasma immunoglobulin levels, which are essential for the adaptive immune response. Jokinen et al. [345]
Senegalese sole (Solea senegalensis) UVR Increase oxidative stress resulted effects the immune response of fish by damaging the cells and tissues. Araújo et al. [346]
African catfish (Clarias gariepinus) UV-A UV-A radiation adversely affects the immune system of Clarias gariepinus through the induction of oxidative stress, hormonal imbalance, and tissue damage. Ibrahim [347]
Rainbow trout (Oncorhynchus mykiss) UVR Ultraviolet radiation influences the immune response of rainbow trout by diminishing phagocytic activity while not significantly affecting T lymphocyte proliferation. Hébert et al. [348]
Rainbow Trout (Oncorhynchus mykiss) UV-B UVB radiation influences the immune system of rainbow trout, leading to a gradual decrease in their resistance to parasitic and bacterial infections. Markkula et al. [349]
Shrimp (Litopenaeus vannamei) UV-A/ UV-B Ultraviolet radiation, specifically UVA, has a beneficial effect on immune responses and growth in Litopenaeus vannamei through the enhancement of antioxidant enzyme activity and the reduction of apoptosis-related gene expression. Fei et al. [350]
European sea bass (Dicentrarchus labrax) UVR Ultraviolet (UV) radiation affects the immune system, particularly in the context of developing a UV-inactivated viral vaccine. Valero et al. [351]
Pacific White Shrimp (Penaeus vannamei) UV-A Increased concentrations of acid phosphatase (ACP), phenol oxidase (PO), and lysozyme (LZM) serve as significant indicators of innate immunity. Wang et al. [352]
Atlantic Salmon (Salmo salar) UV-B The modifications in fatty acid composition resulting from UVB exposure and temperature stress may indirectly affect immune function and overall health in Atlantic salmon. Arts et al. [353]
Common carp (Cyprinus carpio) UVR Ultraviolet (UV) radiation exerts an indirect influence on the immune system of fish through the induction of oxidative stress. Britto et al. [354]
Rainbow Trout (Oncorhynchus mykiss) UVR The detrimental impacts of UV light on the skin of rainbow trout can indirectly impair immune function by compromising the skin, an essential barrier against infection. Bullock and Coutts [355]
Caspian Sea Salmon (Salmo trutta caspius) UVR Ultraviolet (UV) radiation indirectly affects the immune system by damaging the skin, which is essential for the defense mechanisms in fish. Ghanizadeh Kazerouni† and Khodabandeh [356]
Intertidal fish UVR The physiological effects may indirectly influence the immune system by undermining overall health and energy distribution, thereby increasing the fish's vulnerability to disease. García-Huidobro et al. [357]
Rainbow trout (Oncorhynchus mykiss) UV-B Ultraviolet-B (UVB) radiation affects the immune system of fish indirectly by causing damage to skin and tissues essential for defense mechanisms. Abedi et al. [358]
African Catfish (Clarias gariepinus) UV-A Ultraviolet-A (UVA) radiation affects the immune system of fish, primarily characterized by physiological and biochemical alterations. Sayed et al. [359]
Atlantic Cod Larvae (Gadus morhua) UVR Behavioural and survival impairments in larval cod resulting from UV exposure may indirectly influence overall health and resilience, potentially impacting immune function. Fukunishi et al. [360]
Freshwater Carp (Labeo rohita) UV-B Ultraviolet-B (UV-B) radiation affects the immune system of fish, demonstrated by alterations in biochemical and physiological markers following UV-B exposure. Singh et al. [361]

UV-A: Ultraviolet-A (320-400 nm); UV-B: Ultraviolet-B (280-320 nm); UVR: Ultraviolet radiation

Bivalves are marine invertebrates that are susceptible to UV-induced immunosuppression. Studies on C. gigas (Pacific oyster) indicated that UV-B radiation exposure adversely impacted hemocyte activity, reducing phagocytosis and pathogen elimination [328]. Moreover, UV radiation can alter the immune-related gene expression equilibrium, reducing the ability to respond to bacterial infections. Reports have also revealed that UV radiation directly and indirectly influences immune function by modifying microbial communities in aquatic ecosystems [362]. These alterations increased pathogen diversity, which further strained the immune systems of aquatic animals. Stressors and UV-induced immunosuppression may increase the vulnerability of aquatic species to diseases, particularly in areas where ozone depletion enhances UV-B penetration into the water column. Therefore, it is crucial for researchers to find a solution for long-term ecological consequences of UV-induced immune suppression.

Interaction between biotic and abiotic factors

The interaction between biotic and abiotic factors influences the immune function of aquatic species, leading to synergistic or antagonistic effects. Biotic factors, such as pathogens, parasites, and interspecies interactions, directly affect immune responses by triggering various defence mechanisms, including inflammation and the synthesis of antimicrobial peptides. Meanwhile, abiotic factors (temperature, salinity, pH, and pollutants) impact the efficacy of their immune responses. At higher temperatures, pathogen replication rates increase and intensify the immunological challenges on the host. This situation creates a synergistic effect through the simultaneous escalation of pathogen pressure and immune stress [363]. Furthermore, increased temperatures can accelerate the host's metabolic processes, which improves their immune response in the initial stages. Nevertheless, prolonged thermal stress may result in immune suppression, mainly when the pathogen load is also high. This occurrence is primarily attributed to climate change.

Pollutants such as heavy metals and organic pollutants are abiotic factors that can disrupt immune function by interfering with signalling pathways or inducing oxidative stress, diminishing the organism's ability to respond to biotic threats. Under specific circumstances, an antagonistic effect may occur. For example, certain pollutants can reduce pathogen viability, thereby reducing disease transmission and alleviating immune pressure on the host [[364], [365], [366]]. Salinity fluctuations can also affect immune function and pathogen dynamics. Increased salinity can lead to osmoregulatory stress, which may impair immune function. Conversely, this condition can also create an unfavourable environment for pathogens and decrease the overall biotic challenge. The connection among these factors is dependent on species and ecological context. In summary, the equilibrium between synergistic and antagonistic interactions results from the cumulative and context-dependent effects of environmental and biological stressors on the immune systems of aquatic organisms.

Adaptation and immunological plasticity

Immune plasticity refers to the ability of aquatic animals to utilise diverse immune strategies to adapt to extreme environments and fluctuating conditions. This plasticity enables dynamic modifications to immune function in reaction to environmental stressors, including temperature variations, salinity alterations, and pathogen presence. Changes in aquatic species subjected to pollution and temperature variations have been noted, which enhance their immune responses to environmental stressors [367,368]. In extreme environments, such as polar regions or deep-sea habitats, immune systems must adapt to low pathogen diversity while retaining the capacity to respond effectively to pathogenic threats [369]. Antarctic fish have adapted to freezing temperatures by enhancing the production of heat shock proteins, which support immune function during thermal stress [[162], [370]]. Deep-sea organisms possess distinct immune receptor proteins to identify and react to specific microbial threats in their environments [371].

Short-term acclimatisation is a vital mechanism through which aquatic animals enhance immune function temporarily in reaction to environmental changes. The process sometimes involves immune priming, equipping the organism for more significant challenges. Exposure to minor elevations in temperature or salinity promotes their immune defences. An upregulation in pro-inflammatory cytokines and immune-regulating molecules was evident in O. niloticus (Nile tilapia) after short-term exposure to elevated salinity, which enhanced its immune response [372,373]. In addition, organisms can experience metabolic reprogramming to reallocate energy resources to enhance immune functions in response to environmental stress. Studies have detected this process in zebrafish during temporary temperature elevations [374,375].

Epigenetic mechanisms also significantly contribute to immune plasticity in response to long-term environmental changes such as ocean warming or acidification. For instance, DNA methylation and histone modification enable organisms to rapidly modify gene expression in reaction to environmental challenges without changing the fundamental genetic code. Evolution has allowed epigenetic changes to be established within populations, manifested as adaptations to improve survival in extreme environments. In summary, aquatic animals depend on short-term acclimatisation mechanisms and long-term evolutionary adaptations to survive and thrive under extreme and fluctuating conditions. Adaptations are frequently driven by immune plasticity, supported by epigenetic modifications allowing swift and reversible reactions to environmental changes.

Implications for aquaculture and conservation

Immune responses to biotic and abiotic factors are essential for the health and survival of aquatic animals, significantly impacting aquaculture practices and the conservation of wild populations. In fish and shrimp farming, bacteria, viruses, and parasites are biotic factors that can significantly impair the immune systems of cultured species, leading to increased mortality rates and considerable economic losses. Vibrio species are known to cause diseases in shrimp, highlighting the importance of understanding pathogen-host dynamics for developing effective disease management strategies [376]. Additionally, abiotic factors, including environmental conditions like temperature, salinity, pH, and pollutant presence, significantly influence immune function. Variations in water temperature can intensify stress responses, leading to compromised immune function and heightened vulnerability to infectious diseases [377]. Therefore, aquaculture operations must identify and address these environmental stressors to ensure the optimal health of cultured species.

Various strategies can be implemented to improve immune health in aquaculture. The added benefit of probiotics has demonstrated the potential to enhance gut microbiota, improve digestion, and augment immune responses in aquatic animals [378]. Probiotics such as Lactobacillus and Bacillus can help prevent disease outbreaks in tilapia and shrimp by competing against harmful pathogens and enhancing their health [379]. Beta-glucans and mannan oligosaccharides are immune stimulants incorporated in the diets of cultured species to activate and improve the immune response and disease resistance [380]. Maintaining optimal environmental conditions is also crucial in enhancing the immune health of cultured organisms, which involves the consistent monitoring of water quality parameters, such as temperature, salinity, and oxygen level and prompt corrective actions when necessary. Biosecurity measures such as access control to farms and prevention of cross-contamination can significantly reduce the risk of disease outbreaks [[380], [381], [382]].

Environmental changes significantly threaten immune functions in wild populations, resulting in biodiversity decline. Climate change, pollution, habitat degradation, rising ocean temperatures and altered salinity levels negatively impact the immune systems of wild fish and invertebrates, increasing their susceptibility to diseases. Coral reef fish also become immunocompromised when exposed to higher temperatures, increasing their mortality rates during disease outbreaks [[383], [384], [385], [386]]. Furthermore, pollution from heavy metals and microplastics impaired immune functions in multiple species, including salmon, thus contributing to population declines [387,388].

These challenges highlight the necessity of immunological adjustment strategies to conserve endangered aquatic species. Immunocompromised populations in the wild may be more vulnerable and at risk of biodiversity losses. Prioritising habitat preservation and restoration can help maintain the immune health of wild aquatic species and survive environmental changes [389]. Understanding the complex interactions between immune responses and environmental factors is crucial for advancing aquaculture sustainability and strengthening conservation efforts for wild aquatic populations.

Future research directions

Climate change, habitat degradation, and rising pollution levels have contributed to the emergence of novel diseases in aquatic ecosystems. Increasing global temperatures modified the immune responses of coral reef fish, increasing their susceptibility to pathogens such as Vibrio spp. that thrive in warmer waters [390]. Coastal development and pollution are examples of habitat degradation that disrupt the ecosystem balance and offer an optimal environment for opportunistic pathogens [391]. Heavy metals and microplastics pollution also induce immunotoxicity in aquatic species that prevent them from responding effectively to infections. These challenges underscore the necessity for additional research to investigate the interactions between novel pathogens and extreme environmental conditions and their effects on aquatic immune systems. Comprehending these intricate dynamics is crucial for formulating effective management strategies to reduce the risks associated with emerging diseases and safeguard the health of wild and farmed aquatic species.

Genomics, proteomics, and immunologic advancements offer essential tools for improving our comprehension of aquatic immune responses. The integration of these technologies may facilitate the development of biomarkers that indicate immune health, offering insights into population resilience in varying environments. Genomic technologies help identify genetic markers linked to disease resistance and immune function in aquaculture, enabling selective breeding of resilient strains [392,393]. Proteomic analyses aid in the detection od specific proteins associated with immune responses, elucidating how aquatic organisms adapt to environmental stressors [393]. In addition, biosensors and non-invasive monitoring systems are promising innovations for monitoring immune health in wild and cultured species. Wearable biosensors can transform aquaculture management by facilitating real-time fish health assessments by observing physiological parameters indicative of immune status [394]. Utilizing these technological advancements enables researchers and aquaculturists to understand the complexities of aquatic immune systems and improve their resilience against emerging threats.

Conclusion

Biotic and abiotic factors interact and influence the immune responses of aquatic animals. Under environmental pressures and elevated pathogen loads, immune suppression may occur in various species. Pollution, temperature variations, and salinity changes also reduce the organism’s capacity to defend themselves against pathogens. As climate change continues to modify aquatic environments, understanding these interactions is critical for managing farmed and wild aquatic populations and providing optimal environments to improve their immune resilience. Therefore, it is recommended that future research investigate species-specific responses to environmental stressors and develop strategies to alleviate the impacts of pollution, temperature fluctuations, and pathogen pressure on the immune health of aquatic species in their respective ecosystems.

Data availability statement

The data that support the findings of this study are available on request from the corresponding author

Funding statement

The primary source of funding for this research project was provided by the Ministry of Education Malaysia under the Fundamental Research Grant Scheme (FRGS) (FRGS/1/2022/STG03/UMK/03/1).

Ethics approval statement

Not applicable.

Patient consent statement

Not applicable.

Permission to reproduce material from other sources

Not applicable.

CRediT authorship contribution statement

Zulhisyam Abdul Kari: Writing – review & editing, Writing – original draft, Supervision.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  • 1.Abdul Razak S., Griffin M.J., Mischke C.C., Bosworth B.G., Waldbieser G.C., Wise D.J., Marsh T.L., Scribner K.T. Biotic and abiotic factors influencing channel catfish egg and gut microbiome dynamics during early life stages. Aquaculture. 2019;498:556–567. doi: 10.1016/j.aquaculture.2018.08.073. [DOI] [Google Scholar]
  • 2.Kasamatsu J. Evolution of innate and adaptive immune systems in jawless vertebrates. Microbiol. Immunol. 2013;57(1):1–12. doi: 10.1111/j.1348-0421.2012.00500.x. [DOI] [PubMed] [Google Scholar]
  • 3.Smith N.C., Rise M.L., Christian S.L. A comparison of the innate and adaptive immune systems in cartilaginous fish, ray-finned fish, and lobe-finned fish. Front. Immunol. 2019;10 doi: 10.3389/fimmu.2019.02292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zhu L.-y., Nie L., Zhu G., Xiang L.-x., Shao J.-z. Advances in research of fish immune-relevant genes: a comparative overview of innate and adaptive immunity in teleosts. Dev. Comp. Immunol. 2013;39(1):39–62. doi: 10.1016/j.dci.2012.04.001. [DOI] [PubMed] [Google Scholar]
  • 5.Bacheler N.M., Paramore L.M., Buckel J.A., Hightower J.E. Abiotic and biotic factors influence the habitat use of an estuarine fish. Mar. Ecol. Prog. Ser. 2009;377:263–277. https://www.int-res.com/abstracts/meps/v377/p263-277/ [Google Scholar]
  • 6.Burns M.D., Knouft J.H., Dillman C.B. The role of abiotic and biotic factors in the unequal body shape diversification of a Gondwanan fish radiation (Otophysi: Characiformes) Evolution. 2023;78(2):253–266. doi: 10.1093/evolut/qpad203. [DOI] [PubMed] [Google Scholar]
  • 7.Claramunt R.M., Wahl D.H. The effects of abiotic and biotic factors in determining larval fish growth rates: a comparison across species and reservoirs. Trans. Am. Fish Soc. 2000;129(3):835–851. doi: 10.1577/1548-8659(2000)129&#x0003c;0835:TEOAAB&#x0003e;2.3.CO;2. [DOI] [Google Scholar]
  • 8.Echevarría G., Lujan N.K., Montoya J., Granda-Albuja M.G., Valdiviezo-Rivera J., Sánchez F., Cuesta F., Ríos-Touma B. Abiotic and biotic factors influencing heavy metals pollution in fisheries of the Western Amazon. Sci. Total Environ. 2024;908 doi: 10.1016/j.scitotenv.2023.168506. [DOI] [PubMed] [Google Scholar]
  • 9.Magoulick D.D. Spatial and temporal variation in fish assemblages of drying stream pools: the role of abiotic and biotic factors. Aquat. Ecol. 2000;34(1):29–41. doi: 10.1023/A:1009914619061. [DOI] [Google Scholar]
  • 10.Solovyev M.M., Izvekova G.I., Kashinskaya E.N., Gisbert E. Dependence of pH values in the digestive tract of freshwater fishes on some abiotic and biotic factors. Hydrobiologia. 2018;807(1):67–85. doi: 10.1007/s10750-017-3383-0. [DOI] [Google Scholar]
  • 11.Trujillo-Jiménez P., López-López E., Díaz-Pardo E., Camargo J.A. Patterns in the distribution of fish assemblages in Río Amacuzac, Mexico: influence of abiotic factors and biotic factors. Rev. Fish Biol. Fish. 2010;20(4):457–469. doi: 10.1007/s11160-009-9153-y. [DOI] [Google Scholar]
  • 12.Lafferty K.D., Porter J.W., Ford S.E. Are diseases increasing in the ocean?*. Annu Rev. Ecol. Evol. Syst. 2004;35:31–54. doi: 10.1146/annurev.ecolsys.35.021103.105704. Volume 35, 2004. [DOI] [Google Scholar]
  • 13.Lafferty K.D., Kuris A.M. How environmental stress affects the impacts of parasites. Limnol. Ocean. 1999;44(3part2):925–931. doi: 10.4319/lo.1999.44.3_part_2.0925. [DOI] [Google Scholar]
  • 14.Bornette G., Puijalon S. Response of aquatic plants to abiotic factors: a review. Aquat. Sci. 2011;73(1):1–14. doi: 10.1007/s00027-010-0162-7. [DOI] [Google Scholar]
  • 15.Kansara K., Bolan S., Radhakrishnan D., Palanisami T., Al-Muhtaseb AaH, Bolan N., Vinu A., Kumar A., Karakoti A. A critical review on the role of abiotic factors on the transformation, environmental identity and toxicity of engineered nanomaterials in aquatic environment. Environ. Pollut. 2022;296 doi: 10.1016/j.envpol.2021.118726. [DOI] [PubMed] [Google Scholar]
  • 16.Tejerina-Garro F.L., De Mérona B. Spatial variability of biotic and abiotic factors of the aquatic habitat in French Guiana. Regul. Rivers: Res. Manag. 2001;17(2):157–169. doi: 10.1002/rrr.619. [DOI] [Google Scholar]
  • 17.Raeeszadeh M., Khoei A.J., Parhizkar S., Rad F.T., Salimi B. Assessment of some heavy metals and their relationship with oxidative stress and immunological parameters in aquatic animal species. Biol. Trace Elem. Res. 2023;201(9):4547–4557. doi: 10.1007/s12011-022-03507-w. [DOI] [PubMed] [Google Scholar]
  • 18.Gomez D., Sunyer J.O., Salinas I. The mucosal immune system of fish: the evolution of tolerating commensals while fighting pathogens. Fish Shellfish. Immunol. 2013;35(6):1729–1739. doi: 10.1016/j.fsi.2013.09.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kiron V. Fish immune system and its nutritional modulation for preventive health care. Anim. Feed Sci. Technol. 2012;173(1):111–133. doi: 10.1016/j.anifeedsci.2011.12.015. [DOI] [Google Scholar]
  • 20.Muiswinkel Wv, Vervoorn-Van Der Wal B. CABI; 2006. The Immune System Of Fish; pp. 678–701. [DOI] [Google Scholar]
  • 21.Bols N.C., Brubacher J.L., Ganassin R.C., Lee L.E.J. Ecotoxicology and innate immunity in fish. Dev. Comp. Immunol. 2001;25(8):853–873. doi: 10.1016/S0145-305X(01)00040-4. [DOI] [PubMed] [Google Scholar]
  • 22.Boehm T. Design principles of adaptive immune systems. Nat. Rev. Immunol. 2011;11(5):307–317. doi: 10.1038/nri2944. [DOI] [PubMed] [Google Scholar]
  • 23.Matsunaga T., Rahman A. What brought the adaptive immune system to vertebrates? - the jaw hypothesis and the seahorse. Immunol. Rev. 1998;166(1):177–186. doi: 10.1111/j.1600-065X.1998.tb01262.x. [DOI] [PubMed] [Google Scholar]
  • 24.Sutoh Y., Kasahara M. The immune system of jawless vertebrates: insights into the prototype of the adaptive immune system. Immunogenetics. 2021;73(1):5–16. doi: 10.1007/s00251-020-01182-6. [DOI] [PubMed] [Google Scholar]
  • 25.Rathinam R.B., Acharya A., Robina A.J., Banu H., Tripathi G. The immune system of marine invertebrates: earliest adaptation of animals. Comp. Immunol. Rep. 2024;7 doi: 10.1016/j.cirep.2024.200163. [DOI] [Google Scholar]
  • 26.Guryanova S.V., Ovchinnikova T.V. Innate immunity mechanisms in marine multicellular organisms. Mar. Drugs. 2022;20(9):549. doi: 10.3390/md20090549. https://www.mdpi.com/1660-3397/20/9/549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Esteban M.Á., Cuesta A., Chaves-Pozo E., Meseguer J. Phagocytosis in teleosts. Implications of the new cells involved. Biology. 2015;4(4):907–922. doi: 10.3390/biology4040907. https://www.mdpi.com/2079-7737/4/4/907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.MacArthur J.I., Fletcher T.C. In: Fish Immunology. Manning M.J., Tatner M.F., editors. Academic Press; 1985. Phagocytosis in Fish; pp. 29–46. [DOI] [Google Scholar]
  • 29.Fischer U., Koppang E.O., Nakanishi T. Teleost T and NK cell immunity. Fish Shellfish. Immunol. 2013;35(2):197–206. doi: 10.1016/j.fsi.2013.04.018. [DOI] [PubMed] [Google Scholar]
  • 30.Azumi K., Yoshimizu M., Suzuki S., Ezura Y., Yokosawa H. Inhibitory effect of halocyamine, an antimicrobial substance from ascidian hemocytes, on the growth of fish viruses and marine bacteria. Experientia. 1990;46(10):1066–1068. doi: 10.1007/BF01940675. [DOI] [PubMed] [Google Scholar]
  • 31.Li F., Chang X., Xu L., Yang F. Different roles of crayfish hemocytes in the uptake of foreign particles. Fish Shellfish. Immunol. 2018;77:112–119. doi: 10.1016/j.fsi.2018.03.029. [DOI] [PubMed] [Google Scholar]
  • 32.Li L., Cardoso J.C.R., Félix R.C., Mateus A.P., Canário A.V.M., Power D.M. Fish lysozyme gene family evolution and divergent function in early development. Dev. Comp. Immunol. 2021;114 doi: 10.1016/j.dci.2020.103772. [DOI] [PubMed] [Google Scholar]
  • 33.Saurabh S., Sahoo P.K. Lysozyme: an important defence molecule of fish innate immune system. Aquac. Res. 2008;39(3):223–239. doi: 10.1111/j.1365-2109.2007.01883.x. [DOI] [Google Scholar]
  • 34.Song Q., Xiao Y., Xiao Z., Liu T., Li J., Li P., Han F. Lysozymes in fish. J. Agric. Food Chem. 2021;69(50):15039–15051. doi: 10.1021/acs.jafc.1c06676. [DOI] [PubMed] [Google Scholar]
  • 35.Boshra H., Li J., Sunyer J.O. Recent advances on the complement system of teleost fish. Fish Shellfish. Immunol. 2006;20(2):239–262. doi: 10.1016/j.fsi.2005.04.004. [DOI] [PubMed] [Google Scholar]
  • 36.Masso-Silva J.A., Diamond G. Antimicrobial peptides from fish. Pharmaceuticals. 2014;7(3):265–310. doi: 10.3390/ph7030265. https://www.mdpi.com/1424-8247/7/3/265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rajanbabu V., Chen J.-Y. Applications of antimicrobial peptides from fish and perspectives for the future. Peptides. 2011;32(2):415–420. doi: 10.1016/j.peptides.2010.11.005. [DOI] [PubMed] [Google Scholar]
  • 38.Valero Y., Saraiva-Fraga M., Costas B., Guardiola F.A. Antimicrobial peptides from fish: beyond the fight against pathogens. Rev. Aquac. 2020;12(1):224–253. doi: 10.1111/raq.12314. [DOI] [Google Scholar]
  • 39.Álvarez C.A., Santana P.A., Salinas-Parra N., Beltrán D., Guzmán F., Vega B., Acosta F., Mercado L. Immune modulation ability of hepcidin from teleost fish. Animals. 2022;12(12):1586. doi: 10.3390/ani12121586. https://www.mdpi.com/2076-2615/12/12/1586 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Brinchmann M.F., Patel D.M., Pinto N., Iversen M.H. Functional aspects of fish mucosal lectins—Interaction with non-self. Molecules. 2018;23(5):1119. doi: 10.3390/molecules23051119. https://www.mdpi.com/1420-3049/23/5/1119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Elumalai P., Rubeena A.S., Arockiaraj J., Wongpanya R., Cammarata M., Ringø E., Vaseeharan B. The role of lectins in Finfish: a review. Rev. Fish Sci. Aquac. 2019;27(2):152–169. doi: 10.1080/23308249.2018.1520191. [DOI] [Google Scholar]
  • 42.Thompson K.D. In: Fish Diseases. Jeney G., editor. Academic Press; 2017. Chapter 1 - immunology: improvement of innate and adaptive immunity; pp. 1–17. [DOI] [Google Scholar]
  • 43.Dawood M.A.O. Nutritional immunity of fish intestines: important insights for sustainable aquaculture. Rev. Aquac. 2021;13(1):642–663. doi: 10.1111/raq.12492. [DOI] [Google Scholar]
  • 44.Xu Y., Zheng J., Yang Y., Cui Z. New insight of variable lymphocyte receptor-likes in anti-bacteria activity from Eriocheir sinensis. Fish Shellfish. Immunol. 2023;134 doi: 10.1016/j.fsi.2023.108592. [DOI] [PubMed] [Google Scholar]
  • 45.Bela-ong D.B., Kim J., Thompson K.D., Jung T.S. Leveraging the biotechnological promise of the hagfish variable lymphocyte receptors: tools for aquatic microbial diseases. Fish Shellfish. Immunol. 2024;150 doi: 10.1016/j.fsi.2024.109565. [DOI] [PubMed] [Google Scholar]
  • 46.Martínez D., Garrido M., Ponce C., Zumelzu Y., Coronado J., Santibañez N., Quilapi A.M., Vargas-Lagos C., Pontigo J.P., Oyarzún-Salazar R., Godoy M., Enríquez R., Muñoz J.L., Vargas-Chacoff L., Romero A. Comparative analysis of the stress and immune responses in Atlantic salmon (Salmo salar) inoculated with live and inactivated Piscirickettsia salmonis. Fish Shellfish. Immunol. 2025;157 doi: 10.1016/j.fsi.2024.110111. [DOI] [PubMed] [Google Scholar]
  • 47.Martínez D., Oyarzún R., Pontigo J.P., Romero A., Yáñez A.J., Vargas-Chacoff L. Nutritional immunity triggers the modulation of iron metabolism genes in the Sub-antarctic notothenioid Eleginops maclovinus in response to Piscirickettsia salmonis. Front. Immunol. 2017;8 doi: 10.3389/fimmu.2017.01153. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Martínez D., Oyarzún R., Vargas-Lagos C., Pontigo J.P., Soto-Dávila M., Saravia J., Romero A., Núñez J.J., Yáñez A.J., Vargas-Chacoff L. Identification, characterization and modulation of ferritin-H in the sub-antarctic notothenioid Eleginops maclovinus challenged with Piscirickettsia salmonis. Dev. Comp. Immunol. 2017;73:88–96. doi: 10.1016/j.dci.2017.03.015. [DOI] [PubMed] [Google Scholar]
  • 49.Martínez D., Vargas-Lagos C., Oyarzún R., Loncoman C.A., Pontigo J.P., Yáñez A.J., Vargas-Chacoff L. Temperature modulates the immunological response of the sub-antarctic notothenioid fish Eleginops maclovinus injected with Piscirickettsia salmonis. Fish Shellfish. Immunol. 2018;82:492–503. doi: 10.1016/j.fsi.2018.08.042. [DOI] [PubMed] [Google Scholar]
  • 50.Murdoch C.C., Skaar E.P. Nutritional immunity: the battle for nutrient metals at the host–pathogen interface. Nat. Rev. Microbiol. 2022;20(11):657–670. doi: 10.1038/s41579-022-00745-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.De Domenico I., Ward D.M., Kaplan J. Hepcidin and Ferroportin: the new players in iron metabolism. Semin. Liver. Dis. 2011;31(03):272–279. doi: 10.1055/s-0031-1286058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rodrigues P.N.S., Vázquez-Dorado S., Neves J.V., Wilson J.M. Dual function of fish hepcidin: response to experimental iron overload and bacterial infection in sea bass (Dicentrarchus labrax) Dev. Comp. Immunol. 2006;30(12):1156–1167. doi: 10.1016/j.dci.2006.02.005. [DOI] [PubMed] [Google Scholar]
  • 53.Chen J., Shi Y.H., Li M.Y. Changes in transferrin and hepcidin genes expression in the liver of the fish Pseudosciaena crocea following exposure to cadmium. Arch. Toxicol. 2008;82(8):525–530. doi: 10.1007/s00204-008-0297-7. [DOI] [PubMed] [Google Scholar]
  • 54.Gomes A.F.R., Almeida M.C., Sousa E., Resende D. Siderophores and metallophores: metal complexation weapons to fight environmental pollution. Sci. Total Environ. 2024;932 doi: 10.1016/j.scitotenv.2024.173044. [DOI] [PubMed] [Google Scholar]
  • 55.Maldonado-Miranda J.J., Castillo-Pérez L.J., Ponce-Hernández A., Carranza-Álvarez C. In: Bacterial Fish Diseases. Dar G.H., Bhat R.A., Qadri H., Al-Ghamdy K.M., Hakeem K.R., editors. Academic Press; 2022. Chapter 19 - summary of economic losses due to bacterial pathogens in aquaculture industry; pp. 399–417. [DOI] [Google Scholar]
  • 56.Magnadóttir B. Innate immunity of fish (overview) Fish Shellfish. Immunol. 2006;20(2):137–151. doi: 10.1016/j.fsi.2004.09.006. [DOI] [PubMed] [Google Scholar]
  • 57.Wangkahart E., Secombes C.J., Wang T. Vol. 9. 2019. Studies on the use of flagellin as an immunostimulant and vaccine adjuvant in fish aquaculture. (Front. Immunol.). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Harikrishnan R., Balasundaram C., Heo M.-S. Impact of plant products on innate and adaptive immune system of cultured finfish and shellfish. Aquaculture. 2011;317(1):1–15. doi: 10.1016/j.aquaculture.2011.03.039. [DOI] [Google Scholar]
  • 59.Rauta P.R., Nayak B., Das S. Immune system and immune responses in fish and their role in comparative immunity study: a model for higher organisms. Immunol. Lett. 2012;148(1):23–33. doi: 10.1016/j.imlet.2012.08.003. [DOI] [PubMed] [Google Scholar]
  • 60.Rauta P.R., Samanta M., Dash H.R., Nayak B., Das S. Toll-like receptors (TLRs) in aquatic animals: signaling pathways, expressions and immune responses. Immunol. Lett. 2014;158(1):14–24. doi: 10.1016/j.imlet.2013.11.013. [DOI] [PubMed] [Google Scholar]
  • 61.De Silva L., Heo G.-J. Biofilm formation of pathogenic bacteria isolated from aquatic animals. Arch. Microbiol. 2022;205(1):36. doi: 10.1007/s00203-022-03332-8. [DOI] [PubMed] [Google Scholar]
  • 62.Bojko J., Stentiford G.D. In: Microsporidia: Current Advances in Biology. Weiss L.M., Reinke A.W., editors. Springer International Publishing; 2022. Microsporidian pathogens of aquatic animals; pp. 247–283. [DOI] [Google Scholar]
  • 63.Teixeira Alves M., Taylor N.G.H. Models suggest pathogen risks to wild fish can be mitigated by acquired immunity in freshwater aquaculture systems. Sci. Rep. 2020;10(1):7513. doi: 10.1038/s41598-020-64023-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jamal M.T., Abdulrahman I.A., Al Harbi M., Chithambaran S. Probiotics as alternative control measures in shrimp aquaculture: a review. J. Appl. Biol. Biotechnol. 2019;7(3):69–77. [Google Scholar]
  • 65.Ina-Salwany M.Y., Al-saari N., Mohamad A., Mursidi F.-A., Mohd-Aris A., Amal M.N.A., Kasai H., Mino S., Sawabe T., Zamri-Saad M. Vibriosis in fish: a review on disease development and prevention. J. Aquat. Anim. Health. 2019;31(1):3–22. doi: 10.1002/aah.10045. [DOI] [PubMed] [Google Scholar]
  • 66.Allam B., Raftos D. Immune responses to infectious diseases in bivalves. J. Invertebr. Pathol. 2015;131:121–136. doi: 10.1016/j.jip.2015.05.005. [DOI] [PubMed] [Google Scholar]
  • 67.Morley N.J. Interactive effects of infectious diseases and pollution in aquatic molluscs. Aquat. Toxicol. 2010;96(1):27–36. doi: 10.1016/j.aquatox.2009.09.017. [DOI] [PubMed] [Google Scholar]
  • 68.Duan, J., Feng, Y.H., Wang, K., Geng, Y., Deng, Y., Ou, Y., Chen, D..Yang, S. 2018. A review: factors affecting outbreaks of saprolegniosis on aquatic animals.
  • 69.Ben Hamed S., Tavares Ranzani-Paiva M.J., Tachibana L., de Carla Dias D., Ishikawa C.M., Esteban M.A. Fish pathogen bacteria: adhesion, parameters influencing virulence and interaction with host cells. Fish Shellfish. Immunol. 2018;80:550–562. doi: 10.1016/j.fsi.2018.06.053. [DOI] [PubMed] [Google Scholar]
  • 70.Shoemaker C., Xu D.-H., LaFrentz B., LaPatra S. Dietary Nutrients, Additives, and Fish Health. 2015. Overview of Fish immune system and infectious diseases; pp. 1–24. [DOI] [Google Scholar]
  • 71.Derome N., Gauthier J., Boutin S., Llewellyn M. In: The Rasputin Effect: When Commensals and Symbionts Become Parasitic. Hurst C.J., editor. Springer International Publishing; 2016. Bacterial opportunistic pathogens of fish; pp. 81–108. [DOI] [Google Scholar]
  • 72.Mydlarz L.D., Jones L.E., Harvell C.D. Innate immunity, environmental drivers, and disease ecology of marine and freshwater invertebrates. Annu. Rev. Ecol. Evol. Syst. 2006;37:251–288. doi: 10.1146/annurev.ecolsys.37.091305.110103. (Volume 37, 2006) [DOI] [Google Scholar]
  • 73.Rodríguez I., Novoa B., Figueras A. Immune response of zebrafish (Danio rerio) against a newly isolated bacterial pathogen Aeromonas hydrophila. Fish Shellfish. Immunol. 2008;25(3):239–249. doi: 10.1016/j.fsi.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 74.Yi Y., Zhang Z., Zhao F., Liu H., Yu L., Zha J., Wang G. Probiotic potential of Bacillus velezensis JW: antimicrobial activity against fish pathogenic bacteria and immune enhancement effects on Carassius auratus. Fish Shellfish. Immunol. 2018;78:322–330. doi: 10.1016/j.fsi.2018.04.055. [DOI] [PubMed] [Google Scholar]
  • 75.Harikrishnan R., Balasundaram C., Dharaneedharan S., Moon Y.-G., Kim M.-C., Kim J.-S., Heo M.-S. Effect of plant active compounds on immune response and disease resistance in Cirrhina mrigala infected with fungal fish pathogen, Aphanomyces invadans. Aquac. Res. 2009;40(10):1170–1181. doi: 10.1111/j.1365-2109.2009.02213.x. [DOI] [Google Scholar]
  • 76.Yang H.-T., Zou S.-S., Zhai L.-J., Wang Y., Zhang F.-M., An L.-G., Yang G.-W. Pathogen invasion changes the intestinal microbiota composition and induces innate immune responses in the zebrafish intestine. Fish Shellfish. Immunol. 2017;71:35–42. doi: 10.1016/j.fsi.2017.09.075. [DOI] [PubMed] [Google Scholar]
  • 77.Carda-Diéguez M., Ghai R., Rodríguez-Valera F., Amaro C. Wild eel microbiome reveals that skin mucus of fish could be a natural niche for aquatic mucosal pathogen evolution. Microbiome. 2017;5(1):162. doi: 10.1186/s40168-017-0376-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kuo I.P., Lee P.-T., Nan F.-H. Rheum officinale extract promotes the innate immunity of orange-spotted grouper (Epinephelus coioides) and exerts strong bactericidal activity against six aquatic pathogens. Fish Shellfish. Immunol. 2020;102:117–124. doi: 10.1016/j.fsi.2020.04.024. [DOI] [PubMed] [Google Scholar]
  • 79.Maekawa S., Wang P.-C., Chen S.-C. Comparative study of immune reaction against bacterial infection from transcriptome analysis. Front. Immunol. 2019;10 doi: 10.3389/fimmu.2019.00153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Biswas G., Korenaga H., Nagamine R., Kawahara S., Takeda S., Kikuchi Y., Dashnyam B., Yoshida T., Kono T., Sakai M. Elevated cytokine responses to Vibrio harveyi infection in the Japanese pufferfish (Takifugu rubripes) treated with Lactobacillus paracasei spp. Paracasei (06TCa22) isolated from the Mongolian dairy product. Fish Shellfish. Immunol. 2013;35(3):756–765. doi: 10.1016/j.fsi.2013.06.004. [DOI] [PubMed] [Google Scholar]
  • 81.Cámara-Ruiz M., Cerezo I.M., Guardiola F.A., García-Beltrán J.M., Balebona M.C., Moriñigo M.Á., Esteban M.Á. Alteration of the immune response and the microbiota of the skin during a natural infection by Vibrio harveyi in European Seabass (Dicentrarchus labrax) Microorganisms. 2021;9(5):964. doi: 10.3390/microorganisms9050964. https://www.mdpi.com/2076-2607/9/5/964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Cheng A.-C., Cheng S.-A., Chen Y.-Y., Chen J.-C. Effects of temperature change on the innate cellular and humoral immune responses of orange-spotted grouper Epinephelus coioides and its susceptibility to Vibrio alginolyticus. Fish Shellfish. Immunol. 2009;26(5):768–772. doi: 10.1016/j.fsi.2009.03.011. [DOI] [PubMed] [Google Scholar]
  • 83.Do Huu H., Sang H.M., Thanh Thuy N.T. Dietary β-glucan improved growth performance, vibrio counts, haematological parameters and stress resistance of pompano fish, Trachinotus ovatus Linnaeus 1758. Fish Shellfish. Immunol. 2016;54:402–410. doi: 10.1016/j.fsi.2016.03.161. [DOI] [PubMed] [Google Scholar]
  • 84.Kewcharoen W., Srisapoome P. Probiotic effects of Bacillus spp. From Pacific white shrimp (Litopenaeus vannamei) on water quality and shrimp growth, immune responses, and resistance to vibrio parahaemolyticus (AHPND strains) Fish Shellfish. Immunol. 2019;94:175–189. doi: 10.1016/j.fsi.2019.09.013. [DOI] [PubMed] [Google Scholar]
  • 85.Liu C.-H., Chen J.-C. Effect of ammonia on the immune response of white shrimpLitopenaeus vannamei and its susceptibility to Vibrio alginolyticus. Fish Shellfish. Immunol. 2004;16(3):321–334. doi: 10.1016/S1050-4648(03)00113-X. [DOI] [PubMed] [Google Scholar]
  • 86.Manchanayake T., Salleh A., Amal M.N.A., Yasin I.S.M., Zamri-Saad M. Pathology and pathogenesis of Vibrio infection in fish: a review. Aquac. Rep. 2023;28 doi: 10.1016/j.aqrep.2022.101459. [DOI] [Google Scholar]
  • 87.Reyes-Becerril M., Guluarte C., Ceballos-Francisco D., Angulo C., Esteban M.Á. Enhancing gilthead seabream immune status and protection against bacterial challenge by means of antigens derived from Vibrio parahaemolyticus. Fish Shellfish. Immunol. 2017;60:205–218. doi: 10.1016/j.fsi.2016.11.053. [DOI] [PubMed] [Google Scholar]
  • 88.Yina S., Zhongjie C., Kaiyu C., Chenghua L., Xiaodong Z. Target of rapamycin signaling inhibits autophagy in sea cucumber apostichopus japonicus. Fish Shellfish. Immunol. 2020;102:480–488. doi: 10.1016/j.fsi.2020.05.013. [DOI] [PubMed] [Google Scholar]
  • 89.Wang F., Qin Z.-L., Luo W.-S., Xiong N.-X., Luo S.-W. Aeromonas hydrophila can modulate synchronization of immune response in gut-liver axis of red crucian carp via the breach of gut barrier. Aquac. Int. 2024;32(1):871–885. doi: 10.1007/s10499-023-01185-6. [DOI] [Google Scholar]
  • 90.Zhang L., Sun L., Srinivasan R., Lin M., Gong L., Lin X. Unveiling a virulence-regulating mechanism in Aeromonas hydrophila: a quantitative exoproteomic analysis of an AraC-like protein. Front. Immunol. 2023;14 doi: 10.3389/fimmu.2023.1191209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Sattanathan G., Liu W.-C., Padmapriya S., Pushparaj K., Sureshkumar S., Lee J.-W., Balasubramanian B., Kim I.H. Effects of dietary blend of algae extract supplementation on growth, biochemical, haemato-immunological response, and immune gene expression in Labeo rohita with Aeromonas hydrophila post-challenges. Fishes. 2023;8(1):7. https://www.mdpi.com/2410-3888/8/1/7 [Google Scholar]
  • 92.Muratori M.C.S., De Oliveira A.L., Ribeiro L.P., Leite R.C., Costa A.P.R., Da Silva M.C.C. Edwardsiella tarda isolated in integrated fish farming. Aquac. Res. 2000;31(6):481–483. doi: 10.1046/j.1365-2109.2000.00448.x. [DOI] [Google Scholar]
  • 93.Park S.B., Aoki T., Jung T.S. Pathogenesis of and strategies for preventing edwardsiella tarda infection in fish. Vet. Res. 2012;43(1):67. doi: 10.1186/1297-9716-43-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ye J., Su Y., Peng X., Li H. Reactive oxygen species-related ceftazidime resistance is caused by the Pyruvate cycle perturbation and reverted by Fe3 + in Edwardsiella tarda. Front. Microbiol. 2021;12 doi: 10.3389/fmicb.2021.654783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yin K., Wang Q., Xiao J., Zhang Y. Comparative proteomic analysis unravels a role for EsrB in the regulation of reactive oxygen species stress responses in Edwardsiella piscicida. FEMS Microbiol. Lett. 2017;364(1):fnw269. doi: 10.1093/femsle/fnw269. [DOI] [PubMed] [Google Scholar]
  • 96.Cheng S., Zhang M., Sun L. The iron-cofactored superoxide dismutase of Edwardsiella tarda inhibits macrophage-mediated innate immune response. Fish Shellfish. Immunol. 2010;29(6):972–978. doi: 10.1016/j.fsi.2010.08.004. [DOI] [PubMed] [Google Scholar]
  • 97.Zhang M., Qin Y., Huang L., Yan Q., Mao L., Xu X., Wang S., Zhang M., Chen L. The role of sodA and sodB in Aeromonas hydrophila resisting oxidative damage to survive in fish macrophages and escape for further infection. Fish Shellfish. Immunol. 2019;88:489–495. doi: 10.1016/j.fsi.2019.03.021. [DOI] [PubMed] [Google Scholar]
  • 98.Pressley M.E., Phelan P.E., Eckhard Witten P., Mellon M.T., Kim C.H. Pathogenesis and inflammatory response to Edwardsiella tarda infection in the zebrafish. Dev. Comp. Immunol. 2005;29(6):501–513. doi: 10.1016/j.dci.2004.10.007. [DOI] [PubMed] [Google Scholar]
  • 99.Grayfer L., Hodgkinson J.W., Belosevic M. Antimicrobial responses of teleost phagocytes and innate immune evasion strategies of intracellular bacteria. Dev. Comp. Immunol. 2014;43(2):223–242. doi: 10.1016/j.dci.2013.08.003. [DOI] [PubMed] [Google Scholar]
  • 100.Yamasaki M., Araki K., Nakanishi T., Nakayasu C., Yoshiura Y., Iida T., Yamamoto A. Adaptive immune response to Edwardsiella tarda infection in ginbuna crucian carp, Carassius auratus langsdorfii. Vet. Immunol. Immunopathol. 2013;153(1):83–90. doi: 10.1016/j.vetimm.2013.02.004. [DOI] [PubMed] [Google Scholar]
  • 101.Velázquez J., Rodríguez A., Aragón H., Haidar A., González M., Valdés R., Garay H.E., Abreu D.D., Ramos Y., Cabrales A., Morales A., González O., Herrera F., Estrada M.P., Carpio Y. Monoclonal antibody against Nile tilapia (Oreochromis niloticus) IgM heavy chain: a valuable tool for detection and quantification of IgM and IgM+ cells. Fish Shellfish. Immunol. 2021;110:44–54. doi: 10.1016/j.fsi.2020.12.007. [DOI] [PubMed] [Google Scholar]
  • 102.Wu X., Xing J., Tang X., Sheng X., Chi H., Zhan W. Protective cellular and humoral immune responses to edwardsiella tarda in flounder (Paralichthys olivaceus) immunized by an inactivated vaccine. Mol. Immunol. 2022;149:77–86. doi: 10.1016/j.molimm.2022.06.008. [DOI] [PubMed] [Google Scholar]
  • 103.Yamasaki M., Araki K., Maruyoshi K., Matsumoto M., Nakayasu C., Moritomo T., Nakanishi T., Yamamoto A. Comparative analysis of adaptive immune response after vaccine trials using live attenuated and formalin-killed cells of Edwardsiella tarda in ginbuna crucian carp (Carassius auratus langsdorfii) Fish Shellfish. Immunol. 2015;45(2):437–442. doi: 10.1016/j.fsi.2015.04.038. [DOI] [PubMed] [Google Scholar]
  • 104.Liang Q., Zhu N., Zheng X., Ding X., He R., Xu H., Cao F., Xue H., Zhou F., Zheng T. Transcriptome analysis of immune responses and metabolic regulations of Chinese soft-shelled turtle (Pelodiscus sinensis) against Edwardsiella tarda infection. Fishes. 2022;7(2):79. https://www.mdpi.com/2410-3888/7/2/79 [Google Scholar]
  • 105.Zhu J., Li C., Ao Q., Tan Y., Luo Y., Guo Y., Lan G., Jiang H., Gan X. Trancriptomic profiling revealed the signatures of acute immune response in tilapia (Oreochromis niloticus) following Streptococcus iniae challenge. Fish Shellfish. Immunol. 2015;46(2):346–353. doi: 10.1016/j. [DOI] [PubMed] [Google Scholar]
  • 106.Jacobsen K.L., Griffin M., Phinney B.S., Salemi M., Yazdi Z., Balami S., Older C.E., Soto E. Temperature-dependent alterations in the proteome of the emergent fish pathogen edwardsiella piscicida. J. Fish Dis. 2024 doi: 10.1111/jfd.14017. n/a(n/a) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Okon E.M., Oyesiji A.A., Okeleye E.D., Kanonuhwa M., Khalifa N.E., Eissa E.-S.H., Mathew R.T., Eissa M.E.H., Alqahtani M.A., Abdelnour S.A. The escalating threat of climate change-driven diseases in fish: evidence from a global perspective – A literature review. Env. Res. 2024;263 doi: 10.1016/j.envres.2024.120184. [DOI] [PubMed] [Google Scholar]
  • 108.Jinagool P., Wipassa V., Chaiyasing R., Chukanhom K., Aengwanich W. Vol. 589. 2024. Effect of increasing ambient temperature on physiological changes, oxidative stress, nitric oxide, total antioxidant power, and mitochondrial activity of Nile tilapia (Oreochromis niloticus Linn.) (Aquaculture). [DOI] [Google Scholar]
  • 109.Cheng C.-H., Guo Z.-X., Luo S.-W., Wang A.-L. Effects of high temperature on biochemical parameters, oxidative stress, DNA damage and apoptosis of pufferfish (Takifugu obscurus) Ecotoxicol. Env. Saf. 2018;150:190–198. doi: 10.1016/j.ecoenv.2017.12.045. [DOI] [PubMed] [Google Scholar]
  • 110.Ndong D., Chen Y.-Y., Lin Y.-H., Vaseeharan B., Chen J.-C. The immune response of tilapia oreochromis mossambicus and its susceptibility to Streptococcus iniae under stress in low and high temperatures. Fish Shellfish. Immunol. 2007;22(6):686–694. doi: 10.1016/j.fsi.2006.08.015. [DOI] [PubMed] [Google Scholar]
  • 111.Sun Z., Tan X., Liu Q., Ye H., Zou C., Xu M., Zhang Y., Ye C. Physiological, immune responses and liver lipid metabolism of orange-spotted grouper (Epinephelus coioides) under cold stress. Aquaculture. 2019;498:545–555. doi: 10.1016/j.aquaculture.2018.08.051. [DOI] [Google Scholar]
  • 112.Kibenge, F.S.B. 2019. Emerging viruses in aquaculture. Curr. Opin. Virol.. 34: 97-103. 10.1016/j.coviro.2018.12.008. [DOI] [PubMed]
  • 113.Jia P., Breyta R.B., Li Q., Qian X., Wu B., Zheng W., Wen Z., Liu Y., Kurath G., Hua Q., Jin N., Liu H. Insight into infectious hematopoietic necrosis virus (IHNV) in Chinese rainbow trout aquaculture from virus isolated from 7 provinces in 2010–2014. Aquaculture. 2018;496:239–246. doi: 10.1016/j.aquaculture.2018.06.062. [DOI] [Google Scholar]
  • 114.Liu L., Shan L.-P., Xue M.-Y., Lu J.-F., Hu Y., Liu G.-L., Chen J. Potential application of antiviral coumarin in aquaculture against IHNV infection by reducing viral adhesion to the epithelial cell surface. Antivir. Res. 2021;195 doi: 10.1016/j.antiviral.2021.105192. [DOI] [PubMed] [Google Scholar]
  • 115.Xu L., Zhao J., Ren G., Ying D., Lin J., Cao Y., Yin J., Liu H., Lu T., Zhang Q. Co-infection of infectious hematopoietic necrosis virus (IHNV) and infectious pancreatic necrosis virus (IPNV) caused high mortality in farmed rainbow trout (Oncorhynchus mykiss) in China. Aquaculture. 2019;512 doi: 10.1016/j.aquaculture.2019.734286. [DOI] [Google Scholar]
  • 116.Chaves-Pozo E., Guardiola F.A., Meseguer J., Esteban M.A., Cuesta A. Nodavirus infection induces a great innate cell-mediated cytotoxic activity in resistant, gilthead seabream, and susceptible, European sea bass, teleost fish. Fish Shellfish. Immunol. 2012;33(5):1159–1166. doi: 10.1016/j.fsi.2012.09.002. [DOI] [PubMed] [Google Scholar]
  • 117.Lin Y., Ren G., Zhao J., Shao Y., He B., Tang X., Sha O., Zhao W., Liu Q., Xu L., Lu T. Long-term protection elicited by an inactivated vaccine supplemented with a water-based adjuvant against infectious hematopoietic necrosis virus in rainbow trout (Oncorhynchus mykiss) Microbiol. Spectr. 2022;10(6) doi: 10.1128/spectrum.03245-22. e03245-03222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Mondal H., Chandrasekaran N., Mukherjee A., Thomas J. Viral infections in cultured fish and shrimps: current status and treatment methods. Aquac. Int. 2022;30(1):227–262. doi: 10.1007/s10499-021-00795-2. [DOI] [Google Scholar]
  • 119.Qiu T.-X., Liu L., Wang H., Hu Y., Chen J. Schisandrin A: a sustainable antiviral and immunomodulatory agent against spring viraemia of carp virus in aquaculture. Fish Shellfish. Immunol. 2024;154 doi: 10.1016/j.fsi.2024.109914. [DOI] [PubMed] [Google Scholar]
  • 120.El-Hissy F.T., Khallil A.-R.M., El-Nagdy M.A. Aquatic fungi associated with seven species of nile fishes (Egypt) Zentralbl. Mikrobiol. 1989;144(5):305–314. doi: 10.1016/S0232-4393(89)80002-2. [DOI] [PubMed] [Google Scholar]
  • 121.Grossart H.-P., Van den Wyngaert S., Kagami M., Wurzbacher C., Cunliffe M., Rojas-Jimenez K. Fungi in aquatic ecosystems. Nat. Rev. Microbiol. 2019;17(6):339–354. doi: 10.1038/s41579-019-0175-8. [DOI] [PubMed] [Google Scholar]
  • 122.Kaydu V., Yardımcı B. Fungus infections in fish. Acta Aquat. Turc. 2024;20(3):256–266. doi: 10.22392/actaquatr.1363489. [DOI] [Google Scholar]
  • 123.van den Berg A.H., McLaggan D., Diéguez-Uribeondo J., van West P. The impact of the water moulds saprolegnia diclina and saprolegnia parasitica on natural ecosystems and the aquaculture industry. Fungal. Biol. Rev. 2013;27(2):33–42. doi: 10.1016/j.fbr.2013.05.001. [DOI] [Google Scholar]
  • 124.Parra-Laca R., Hernández-Hernández F., Lanz-Mendoza H., Borrego Enríquez L., García Gil F. Isolation and identification of saprolegnia sp from fresh water aquarium fishes and the hemolymph immune response of dactylopus coccus Costa de 1835 (Homoptera: Coccoidea: Dactylopidae) against this oomycete. Entomol. Ornithol. Herpetol. 2015;4(149) 2161-0983.10001. [Google Scholar]
  • 125.Hu X.-G., Liu L., Hu K., Yang X.-L., Wang G.-X. In vitro screening of fungicidal chemicals for antifungal activity against saprolegnia. J. World Aquac. Soc. 2013;44(4):528–535. doi: 10.1111/jwas.12052. [DOI] [Google Scholar]
  • 126.Barde R.D., Deshpande M., Nagthane N., Darak O., Baig M. A review of saprolegnia infection in freshwater fishes and control of the saprolegniosis. Sustain. Humanosph. 2020;16(1):702–711. [Google Scholar]
  • 127.Lindholm-Lehto P.C., Pylkkö P. Saprolegniosis in aquaculture and how to control it? Aquac. Fish Fish. 2024;4(4):e2200. doi: 10.1002/aff2.200. [DOI] [Google Scholar]
  • 128.Earle G., Hintz W. New approaches for controlling saprolegnia parasitica, the causal agent of a devastating fish disease. Trop. Life Sci. Res. 2014;25(2):101–109. [PMC free article] [PubMed] [Google Scholar]
  • 129.Rodgers C., Furones M. Antimicrobial agents in aquaculture: practice, needs and issues. Options Méditerr. 2009;86:41–59. [Google Scholar]
  • 130.Hashimoto J.C., Paschoal J.A.R., de Queiroz J.F., Reyes F.G.R. Considerations on the use of malachite green in aquaculture and analytical aspects of determining the residues in fish: a review. J. Aquat. Food Prod. Technol. 2011;20(3):273–294. doi: 10.1080/10498850.2011.569643. [DOI] [Google Scholar]
  • 131.Scherpenisse P., Bergwerff A.A. Determination of residues of malachite green in finfish by liquid chromatography tandem mass spectrometry. Anal. Chim. Acta. 2005;529(1):173–177. doi: 10.1016/j.aca.2004.08.009. [DOI] [PubMed] [Google Scholar]
  • 132.Henriksson P.J.G., Rico A., Troell M., Klinger D.H., Buschmann A.H., Saksida S., Chadag M.V., Zhang W. Unpacking factors influencing antimicrobial use in global aquaculture and their implication for management: a review from a systems perspective. Sustain. Sci. 2018;13(4):1105–1120. doi: 10.1007/s11625-017-0511-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Patil P.K., Mishra S.S., Pradhan P.K., Manna S.K., Abraham J.T., Solanki H.G., Shahi N., Swain P., Sahoo S.N., Avunje S., Sharma K.S.R., Geetha R., Priyadharshini R., Nagaraju V.T., Paniprasad K., Kumar A., Debnath D., Panikkar P., Raja R.A.…Jena J. Usage pattern of chemicals, biologicals and veterinary medicinal products in Indian aquaculture. Rev. Aquac. 2022;14(4):2038–2063. doi: 10.1111/raq.12688. [DOI] [Google Scholar]
  • 134.Sedyaaw P., Bhatkar V. A review on application of aquaculture drugs for sustainable aquaculture. J. Dev. Res. 2024;14(09):66685–66690. [Google Scholar]
  • 135.O’Dwyer K., Dargent F., Forbes M.R., Koprivnikar J. Parasite infection leads to widespread glucocorticoid hormone increases in vertebrate hosts: a meta-analysis. J. Anim. Ecol. 2020;89(2):519–529. doi: 10.1111/1365-2656.13123. [DOI] [PubMed] [Google Scholar]
  • 136.Li Y., Jiang B., Mo Z., Li A., Dan X. Cryptocaryon irritans (Brown, 1951) is a serious threat to aquaculture of marine fish. Rev. Aquac. 2022;14(1):218–236. doi: 10.1111/raq.12594. [DOI] [Google Scholar]
  • 137.Wang Q., Yu Y., Zhang X., Xu Z. Immune responses of fish to Ichthyophthirius multifiliis (Ich): a model for understanding immunity against protozoan parasites. Dev. Comp. Immunol. 2019;93:93–102. doi: 10.1016/j.dci.2019.01.002. [DOI] [PubMed] [Google Scholar]
  • 138.Zhou C.-Q., Ka W., Yuan W.-K., Wang J.-L. The effect of acute heat stress on the innate immune function of rainbow trout based on the transcriptome. J. Therm. Biol. 2021;96 doi: 10.1016/j.jtherbio.2021.102834. [DOI] [PubMed] [Google Scholar]
  • 139.Xia Y., Yu X., Yuan Z., Yang Y., Liu Y. Whole-transcriptome analysis reveals potential CeRNA regulatory mechanism in Takifugu rubripes against cryptocaryon irritans infection. Biology. 2024;13(10):788. doi: 10.3390/biology13100788. https://www.mdpi.com/2079-7737/13/10/788 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Sehnal L., Brammer-Robbins E., Wormington A.M., Blaha L., Bisesi J., Larkin I., Martyniuk C.J., Simonin M., Adamovsky O. Microbiome composition and function in aquatic vertebrates: small organisms making big impacts on aquatic animal health. Front. Microbiol. 2021;12 doi: 10.3389/fmicb.2021.567408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Zhang Z., Yang Q., Liu H., Jin J., Yang Y., Zhu X., Han D., Zhou Z., Xie S. Potential functions of the gut microbiome and modulation strategies for improving aquatic animal growth. Rev. Aquac. 2024 doi: 10.1111/raq.12959. n/a(n/a) [DOI] [Google Scholar]
  • 142.Abdul Kari Z., Kabir M.A., Mat K., Rusli N.D., Razab M., Ariff N., Edinur H.A., Rahim M.Z.A., Pati S., Dawood M.A.O., Wei L.S. The possibility of replacing fish meal with fermented soy pulp on the growth performance, blood biochemistry, liver, and intestinal morphology of African catfish (Clarias gariepinus) Aquac. Rep. 2021;21 doi: 10.1016/j.aqrep.2021.100815. [DOI] [Google Scholar]
  • 143.Kari Z.A., Kabir M.A., Dawood M.A.O., Razab M., Ariff N., Sarkar T., Pati S., Edinur H.A., Mat K., Ismail T.A., Wei L.S. Effect of fish meal substitution with fermented soy pulp on growth performance, digestive enzyme, amino acid profile, and immune-related gene expression of African catfish (Clarias gariepinus) Aquaculture. 2022;546 doi: 10.1016/j.aquaculture.2021.737418. [DOI] [Google Scholar]
  • 144.Kari Z.A., Wee W., Hamid N.K.A., Mat K., Rusli N.D., Khalid H.N.M., Sukri S.A.M., Harun H.C., Dawood M.A., Hakim A.H. Recent advances of phytobiotic utilization in carp farming: a review. Aquac. Nutr. 2022;2022 [Google Scholar]
  • 145.Zakaria M.K., Kari Z.A., Van Doan H., Kabir M.A., Che Harun H., Mohamad Sukri S.A., Goh K.W., Wee W., Khoo M.I., Wei L.S. Fermented soybean meal (FSBM) in African catfish (Clarias gariepinus) diets: effects on growth performance, fish gut microbiota analysis, blood haematology, and liver morphology. Life. 2022;12(11):1851. doi: 10.3390/life12111851. https://www.mdpi.com/2075-1729/12/11/1851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Dawood M.A.O., Koshio S., Abdel-Daim M.M., Van Doan H. Probiotic application for sustainable aquaculture. Rev. Aquac. 2019;11(3):907–924. doi: 10.1111/raq.12272. [DOI] [Google Scholar]
  • 147.Ashraf R., Shah N.P. Immune system stimulation by probiotic microorganisms. Crit. Rev. Food Sci. Nutr. 2014;54(7):938–956. doi: 10.1080/10408398.2011.619671. [DOI] [PubMed] [Google Scholar]
  • 148.Guimarães M.C., Cerezo I.M., Fernandez-Alarcon M.F., Natori M.M., Sato L.Y., Kato C.A.T., Moriñigo M.A., Tapia-Paniagua S., Dias DdC, Ishikawa C.M., Ranzani-Paiva M.J.T., Cassiano L.L., Bach E.E., Clissa P.B., Orefice D.P., Tachibana L. Oral administration of probiotics (Bacillus subtilis and Lactobacillus plantarum) in Nile Tilapia (Oreochromis niloticus) vaccinated and challenged with Streptococcus agalactiae. Fishes. 2022;7(4):211. https://www.mdpi.com/2410-3888/7/4/211 [Google Scholar]
  • 149.Dang Y., Sun Y., Zhou Y., Men X., Wang B., Li B., Ren Y. Effects of probiotics on growth, the toll-like receptor mediated immune response and susceptibility to Aeromonas salmonicida infection in rainbow trout Oncorhynchus mykiss. Aquaculture. 2022;561 doi: 10.1016/j.aquaculture.2022.738668. [DOI] [Google Scholar]
  • 150.Liu Z.-Y., Yang H.-L., Wei C.-Y., Cai G.-H., Ye J.-D., Zhang C.-X., Sun Y.-Z. Commensal Bacillus siamensis LF4 induces antimicrobial peptides expression via TLRs and NLRs signaling pathways in intestinal epithelial cells of Lateolabrax maculatus. Fish Shellfish. Immunol. 2023;134 doi: 10.1016/j.fsi.2023.108634. [DOI] [PubMed] [Google Scholar]
  • 151.Xu S., Wang Q., Wang F., Li X., Wang B., Zhou Y., Zou P., Tang L., Yu D., Li W. Improved immune function of Chinese soft-shelled turtles (Pelodiscus sinensis) through oral probiotics via the TLR signaling pathway. Aquaculture. 2022;555 doi: 10.1016/j.aquaculture.2022.738126. [DOI] [Google Scholar]
  • 152.Zhang B., Yang H., Cai G., Nie Q., Sun Y. The interactions between the host immunity and intestinal microorganisms in fish. Appl. Microbiol. Biotechnol. 2024;108(1):30. doi: 10.1007/s00253-023-12934-1. [DOI] [PubMed] [Google Scholar]
  • 153.Golovanova I.L. Effects of abiotic factors (temperature, pH, heavy metals) on activities of glycosidases in invertebrate animals. J. Evol. Biochem. Physiol. 2011;47(1):15–20. doi: 10.1134/S0022093011010026. [DOI] [PubMed] [Google Scholar]
  • 154.Makrinos D.L., Bowden T.J. Natural environmental impacts on teleost immune function. Fish Shellfish. Immunol. 2016;53:50–57. doi: 10.1016/j.fsi.2016.03.008. [DOI] [PubMed] [Google Scholar]
  • 155.Khasani I., Febrianti R., Sularto, Pamungkas W., Rosada K.K. Identification of potential molecular markers for disease resistance in giant gourami through major histocompatibility complex (MHC) II gene analysis. Fish Aquat. Sci. 2024;27(3):159–170. doi: 10.47853/FAS.2024.e16. [DOI] [Google Scholar]
  • 156.Yamaguchi T., Dijkstra J.M. Major histocompatibility complex (MHC) genes and disease resistance in fish. Cells. 2019;8(4):378. doi: 10.3390/cells8040378. https://www.mdpi.com/2073-4409/8/4/378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Abram Q.H., Dixon B., Katzenback B.A. Impacts of low temperature on the teleost immune system. Biology. 2017;6(4):39. doi: 10.3390/biology6040039. https://www.mdpi.com/2079-7737/6/4/39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Yang Y., Yu H., Li H., Wang A., Yu H.-y. Effect of high temperature on immune response of grass carp (Ctenopharyngodon idellus) by transcriptome analysis. Fish Shellfish. Immunol. 2016;58:89–95. doi: 10.1016/j.fsi.2016.09.014. [DOI] [PubMed] [Google Scholar]
  • 159.Rahman M.A., Henderson S., Miller-Ezzy P., Li X.X., Qin J.G. Immune response to temperature stress in three bivalve species: pacific oyster Crassostrea gigas, Mediterranean mussel Mytilus galloprovincialis and mud cockle Katelysia rhytiphora. Fish Shellfish. Immunol. 2019;86:868–874. doi: 10.1016/j.fsi.2018.12.017. [DOI] [PubMed] [Google Scholar]
  • 160.Qiang J., Yang H., Wang H., Kpundeh M.D., Xu P. Interacting effects of water temperature and dietary protein level on hematological parameters in Nile tilapia juveniles, oreochromis niloticus (L.) and mortality under Streptococcus iniae infection. Fish Shellfish. Immunol. 2013;34(1):8–16. doi: 10.1016/j.fsi.2012.09.003. [DOI] [PubMed] [Google Scholar]
  • 161.Xu G., Sheng X., Xing J., Zhan W. Effect of temperature on immune response of Japanese flounder (Paralichthys olivaceus) to inactivated lymphocystis disease virus (LCDV) Fish Shellfish. Immunol. 2011;30(2):525–531. doi: 10.1016/j.fsi.2010.11.026. [DOI] [PubMed] [Google Scholar]
  • 162.Saravia J., Paschke K., Pontigo J.P., Nualart D., Navarro J.M., Vargas-Chacoff L. Effects of temperature on the innate immune response on antarctic and sub-antarctic fish harpagifer antarcticus and Harpagifer bispinis challenged with two immunostimulants, LPS and Poly I:C: in vivo and in vitro approach. Fish Shellfish. Immunol. 2022;130:391–408. doi: 10.1016/j.fsi.2022.09.025. [DOI] [PubMed] [Google Scholar]
  • 163.Dang V.T., Speck P., Benkendorff K. Influence of elevated temperatures on the immune response of abalone, Haliotis rubra. Fish Shellfish. Immunol. 2012;32(5):732–740. doi: 10.1016/j.fsi.2012.01.022. [DOI] [PubMed] [Google Scholar]
  • 164.Yu J.H., Song J.H., Choi M.C., Park S.W. Effects of water temperature change on immune function in surf clams, Mactra veneriformis (Bivalvia: Mactridae) J. Invertebr. Pathol. 2009;102(1):30–35. doi: 10.1016/j.jip.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 165.Huang Z.H., Ma A.J., Wang X.A. The immune response of turbot, scophthalmus maximus (L.), skin to high water temperature. J. Fish Dis. 2011;34(8):619–627. doi: 10.1111/j.1365-2761.2011.01275.x. [DOI] [PubMed] [Google Scholar]
  • 166.Jiravanichpaisal P., Söderhäll K., Söderhäll I. Effect of water temperature on the immune response and infectivity pattern of white spot syndrome virus (WSSV) in freshwater crayfish. Fish Shellfish. Immunol. 2004;17(3):265–275. doi: 10.1016/j.fsi.2004.03.010. [DOI] [PubMed] [Google Scholar]
  • 167.Martins M.L., Xu D.H., Shoemaker C.A., Klesius P.H. Temperature effects on immune response and hematological parameters of channel catfish Ictalurus punctatus vaccinated with live theronts of Ichthyophthirius multifiliis. Fish Shellfish. Immunol. 2011;31(6):774–780. doi: 10.1016/j.fsi.2011.07.015. [DOI] [PubMed] [Google Scholar]
  • 168.Salazar-Lugo R., Estrella A., Oliveros A., Rojas-Villarroel E., de Villalobos L.B., Lemus M. Paraquat and temperature affect nonspecific immune response of Colossoma macropomum. Env. Toxicol. Pharmacol. 2009;27(3):321–326. doi: 10.1016/j.etap.2008.11.010. [DOI] [PubMed] [Google Scholar]
  • 169.PylkkàP., LyytikÃÂinen T., Ritola O., Pelkonen S., Valtonen E.T. Temperature effect on the immune defense functions of Arctic charr Salvelinus alpinus. Dis. Aquat. Org. 2002;52(1):47–55. doi: 10.3354/dao052047. https://www.int-res.com/abstracts/dao/v52/n1/p47-55/ [DOI] [PubMed] [Google Scholar]
  • 170.Engelsma M.Y., Hougee S., Nap D., Hofenk M., Rombout J., van Muiswinkel W.B., Lidy Verburg-van Kemenade B.M. Multiple acute temperature stress affects leucocyte populations and antibody responses in common carp, cyprinus carpio L. Fish Shellfish. Immunol. 2003;15(5):397–410. doi: 10.1016/S1050-4648(03)00006-8. [DOI] [PubMed] [Google Scholar]
  • 171.Kim J.-H., Park H.-J., Kim K.-W., Hwang I.-K., Kim D.-H., Oh C.W., Lee J.S., Kang J.-C. Growth performance, oxidative stress, and non-specific immune responses in juvenile sablefish, Anoplopoma fimbria, by changes of water temperature and salinity. Fish Physiol. Biochem. 2017;43(5):1421–1431. doi: 10.1007/s10695-017-0382-z. [DOI] [PubMed] [Google Scholar]
  • 172.Truscott R., White K.N. The influence of metal and temperature stress on the immune system of crabs. Funct. Ecol. 1990;4(3):455–461. doi: 10.2307/2389609. [DOI] [Google Scholar]
  • 173.Alcorn S.W., Murray A.L., Pascho R.J. Effects of rearing temperature on immune functions in sockeye salmon (Oncorhynchus nerka) Fish Shellfish. Immunol. 2002;12(4):303–334. doi: 10.1006/fsim.2001.0373. [DOI] [PubMed] [Google Scholar]
  • 174.Wang X., Wang L., Zhang H., Ji Q., Song L., Qiu L., Zhou Z., Wang M., Wang L. Immune response and energy metabolism of Chlamys farreri under Vibrio anguillarum challenge and high temperature exposure. Fish Shellfish. Immunol. 2012;33(4):1016–1026. doi: 10.1016/j.fsi.2012.08.026. [DOI] [PubMed] [Google Scholar]
  • 175.Beaudry A., Fortier M., Masson S., Auffret M., Brousseau P., Fournier M. Effect of temperature on immunocompetence of the blue mussel (Mytilus edulis) J. Xenobiot. 2016;6(1):5889. doi: 10.4081/xeno.2016.5889. https://www.mdpi.com/2039-4713/6/1/5889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Cascarano M.C., Stavrakidis-Zachou O., Mladineo I., Thompson K.D., Papandroulakis N., Katharios P. Mediterranean aquaculture in a changing climate: temperature effects on pathogens and diseases of three farmed fish species. Pathogens. 2021;10(9):1205. doi: 10.3390/pathogens10091205. https://www.mdpi.com/2076-0817/10/9/1205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Kocan R., Hershberger P., Sanders G., Winton J. Effects of temperature on disease progression and swimming stamina in Ichthyophonus-infected rainbow trout, Oncorhynchus mykiss (Walbaum) J. Fish Dis. 2009;32(10):835–843. doi: 10.1111/j.1365-2761.2009.01059.x. [DOI] [PubMed] [Google Scholar]
  • 178.Sharma J., Singh S.P., Chakrabarti R. Effect of temperature on digestive physiology, immune-modulatory parameters, and expression level of hsp and LDH genes in Catla catla (Hamilton, 1822) Aquaculture. 2017;479:134–141. doi: 10.1016/j.aquaculture.2017.05.031. [DOI] [Google Scholar]
  • 179.Cheng C.-H., Ye C.-X., Guo Z.-X., Wang A.-L. Immune and physiological responses of pufferfish (Takifugu obscurus) under cold stress. Fish Shellfish. Immunol. 2017;64:137–145. doi: 10.1016/j.fsi.2017.03.003. [DOI] [PubMed] [Google Scholar]
  • 180.Cheng C.-H., Guo Z.-X., Ye C.-X., Wang A.-L. Effect of dietary astaxanthin on the growth performance, non-specific immunity, and antioxidant capacity of pufferfish (Takifugu obscurus) under high temperature stress. Fish Physiol. Biochem. 2018;44(1):209–218. doi: 10.1007/s10695-017-0425-5. [DOI] [PubMed] [Google Scholar]
  • 181.Menike U., Lee Y., Oh C., Wickramaarachchi W.D.N., Premachandra H.K.A., Park S.C., Lee J., De Zoysa M. Oligo-microarray analysis and identification of stress-immune response genes from manila clam (Ruditapes philippinarum) exposure to heat and cold stresses. Mol. Biol. Rep. 2014;41(10):6457–6473. doi: 10.1007/s11033-014-3529-3. [DOI] [PubMed] [Google Scholar]
  • 182.Li Y.-F., Yang N., Liang X., Yoshida A., Osatomi K., Power D., Batista F.M., Yang J.-L. Elevated seawater temperatures decrease microbial diversity in the gut of mytilus coruscus. Front. Physiol. 2018;9 doi: 10.3389/fphys.2018.00839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Kim S.M., Lee D.W., Kim Y.J., Jun L.J., Park H.K., Kim Y.J., Jeong Y.Y., Lee S.H., Kwon M.G., Jeong J.B. Field experiment on effect of butaphosphan and cyanocobalamin complex on the immunity and stress of olive flounder at low temperature. Fish Aquat. Sci. 2021;24(4):153–162. doi: 10.47853/FAS.2021.e15. [DOI] [Google Scholar]
  • 184.Hart L.M., Lorenzen N., Einer-Jensen K., Purcell M.K., Hershberger P.K. Influence of temperature on the efficacy of homologous and heterologous DNA vaccines against viral hemorrhagic septicemia in Pacific Herring. J. Aquat. Anim. Health. 2017;29(3):121–128. doi: 10.1080/08997659.2017.1307287. [DOI] [PubMed] [Google Scholar]
  • 185.Stehfest K.M., Carter C.G., McAllister J.D., Ross J.D., Semmens J.M. Response of Atlantic salmon Salmo salar to temperature and dissolved oxygen extremes established using animal-borne environmental sensors. Sci. Rep. 2017;7(1):4545. doi: 10.1038/s41598-017-04806-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Kim J.-M., Malintha G.H.T., Gunathilaka G., Lee C., Kim M.-G., Lee B.-J., Kim J.-D., Lee K.-J. Taurine supplementation in diet for olive flounder at low water temperature. Fish Aquat. Sci. 2017;20(1):20. doi: 10.1186/s41240-017-0065-9. [DOI] [Google Scholar]
  • 187.Wu F., Lu W., Shang Y., Kong H., Li L., Sui Y., Hu M., Wang Y. Combined effects of seawater acidification and high temperature on hemocyte parameters in the thick shell mussel mytilus coruscus. Fish Shellfish. Immunol. 2016;56:554–562. doi: 10.1016/j.fsi.2016.08.012. [DOI] [PubMed] [Google Scholar]
  • 188.Wang K., Pales Espinosa E., Tanguy A., Allam B. Alterations of the immune transcriptome in resistant and susceptible hard clams (Mercenaria mercenaria) in response to Quahog Parasite unknown (QPX) and temperature. Fish Shellfish. Immunol. 2016;49:163–176. doi: 10.1016/j.fsi.2015.12.006. [DOI] [PubMed] [Google Scholar]
  • 189.Tamplin J.W., Moran V.F., Riesberg E.J. Response of juvenile diamond-backed terrapins (Malaclemys terrapin) to an aquatic thermal gradient. J. Therm. Biol. 2013;38(7):434–439. doi: 10.1016/j.jtherbio.2013.06.005. [DOI] [Google Scholar]
  • 190.Taeubert J.-E., El-Nobi G., Geist J. Effects of water temperature on the larval parasitic stage of the thick-shelled river mussel (Unio crassus) Aquat. Conserv.: Mar. Freshw. Ecosyst. 2014;24(2):231–237. doi: 10.1002/aqc.2385. [DOI] [Google Scholar]
  • 191.Kamyab E., Kühnhold H., Novais S.C., Alves L.M.F., Indriana L., Kunzmann A., Slater M., Lemos M.F.L. Effects of thermal stress on the immune and oxidative stress responses of juvenile sea cucumber Holothuria scabra. J. Comp. Physiol. B. 2017;187(1):51–61. doi: 10.1007/s00360-016-1015-z. [DOI] [PubMed] [Google Scholar]
  • 192.Peng M., Wang H., Wen S., Liang Z., Huang Z., Zhang B., Chen T., Liu Q., Li Q., Meng Y., Huang Y., Yang C., Zeng D., Li M., Zhu W., Zhao Y. Identification and expression analysis of the heat shock proteins Hsp70, Hsp90, and Hsp90b in Litopenaeus vannamei under low-temperature stress. Aquac. Rep. 2025;40 doi: 10.1016/j.aqrep.2024.102591. [DOI] [Google Scholar]
  • 193.Esmaeili N., Zare M., Choupani S.M.H., Kazempour M., Hosseini H., Akhavan S.R., Salini M. Responses of oscar (Astronotus ocellatus) to different temperatures and its interaction with early mild stress. Aquaculture. 2025;594 doi: 10.1016/j.aquaculture.2024.741454. [DOI] [Google Scholar]
  • 194.Tort L., PadrÓS F., Rotllant J., Crespo S. Winter syndrome in the gilthead sea breamSparus aurata. Immunological and histopathological features. Fish Shellfish. Immunol. 1998;8(1):37–47. doi: 10.1006/fsim.1997.0120. [DOI] [Google Scholar]
  • 195.Ibarz A., Padrós F., Gallardo M.Á., Fernández-Borràs J., Blasco J., Tort L. Low-temperature challenges to gilthead sea bream culture: review of cold-induced alterations and ‘Winter Syndrome. Rev. Fish Biol. Fish. 2010;20(4):539–556. doi: 10.1007/s11160-010-9159-5. [DOI] [Google Scholar]
  • 196.Birrer S.C., Reusch T.B.H., Roth O. Salinity change impairs pipefish immune defence. Fish Shellfish. Immunol. 2012;33(6):1238–1248. doi: 10.1016/j.fsi.2012.08.028. [DOI] [PubMed] [Google Scholar]
  • 197.Velasco J., Gutiérrez-Cánovas C., Botella-Cruz M., Sánchez-Fernández D., Arribas P., Carbonell J.A., Millán A., Pallarés S. Effects of salinity changes on aquatic organisms in a multiple stressor context. Philos. Trans. R. Soc. B: Biol. Sci. 2019;374(1764) doi: 10.1098/rstb.2018.0011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Zhou H., Yao T., Zhang T., Sun M., Ning Z., Chen Y., Mu W. Effects of chronic saline-alkaline stress on gill, liver and intestinal histology, biochemical, and immune indexes in Amur minnow (Phoxinus lagowskii) Aquaculture. 2024;579 doi: 10.1016/j.aquaculture.2023.740153. [DOI] [Google Scholar]
  • 199.Lu M., Su M., Liu N., Zhang J. Effects of environmental salinity on the immune response of the coastal fish scatophagus argus during bacterial infection. Fish Shellfish. Immunol. 2022;124:401–410. doi: 10.1016/j.fsi.2022.04.029. [DOI] [PubMed] [Google Scholar]
  • 200.Li R., Liu J., Leung C.T., Lin X., Chan T.F., Tse W.K.F., Lai K.P. Transcriptomic analysis in marine Medaka Gill reveals that the hypo-osmotic stress could alter the immune response via the IL17 signaling pathway. Int. J. Mol. Sci. 2022;23(20) doi: 10.3390/ijms232012417. https://www.mdpi.com/1422-0067/23/20/12417 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Bado-Nilles A., Tebby C., Pinet A., Turiès C., Lignot J.-H., Porcher J.-M. How short-term change in temperature or salinity affect cellular immune parameters of three-spined stickleback, Gasterosteus aculeatus? Mar. Env. Res. 2025;204 doi: 10.1016/j.marenvres.2025.106972. [DOI] [PubMed] [Google Scholar]
  • 202.Leprêtre M., Hamar J., Urias M.B., Kültz D. Comparative proteomics of salinity stress responses in fish and aquatic invertebrates. Proteomics. 2025 doi: 10.1002/pmic.202400255. n/a(n/a) [DOI] [PubMed] [Google Scholar]
  • 203.Siddika A., Akram W., Mridul M.M.I., Zeehad M.S.K., Islam M.R., Salin K.R., Hurwood D.A., Rahi M.L. Effects of elevated salinity levels on the biological alterations of rohu (Labeo rohita): initiative for developing salinity tolerant line. Aquac. Int. 2024;33(1):21. doi: 10.1007/s10499-024-01708-9. [DOI] [Google Scholar]
  • 204.Sun S., Gong C., Deng C., Yu H., Zheng D., Wang L., Sun J., Song F., Luo J. Effects of salinity stress on the growth performance, health status, and intestinal microbiota of juvenile Micropterus salmoides. Aquaculture. 2023;576 doi: 10.1016/j.aquaculture.2023.739888. [DOI] [Google Scholar]
  • 205.Zimmer A.M., Goss G.G., Glover C.N. Reductionist approaches to the study of ionoregulation in fishes. Comp. Biochem. Physiol. B: Biochem. Mol. Biol. 2021;255 doi: 10.1016/j.cbpb.2021.110597. [DOI] [PubMed] [Google Scholar]
  • 206.Bal A., Panda F., Pati S.G., Das K., Agrawal P.K., Paital B. Modulation of physiological oxidative stress and antioxidant status by abiotic factors especially salinity in aquatic organisms. Comp. Biochem. Physiol. C: Toxicol. Pharmacol. 2021;241 doi: 10.1016/j.cbpc.2020.108971. [DOI] [PubMed] [Google Scholar]
  • 207.Árnason T., Magnadóttir B., Björnsson B., Steinarsson A., Björnsson B.T. Effects of salinity and temperature on growth, plasma ions, cortisol and immune parameters of juvenile Atlantic cod (Gadus morhua) Aquaculture. 2013;380-383:70–79. doi: 10.1016/j.aquaculture.2012.11.036. [DOI] [Google Scholar]
  • 208.van Muilekom D.R., Mueller J., Lindemeyer J., Schultheiß T., Maser E., Seibel H., Rebl A., Schulz C., Goldammer T. Salinity change evokes stress and immune responses in Atlantic salmon with microalgae showing limited potential for dietary mitigation. Front. Physiol. 2024;15 doi: 10.3389/fphys.2024.1338858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Wang Z., Zhou J., Li J., Zou J., Fan L. The immune defense response of Pacific white shrimp (Litopenaeus vannamei) to temperature fluctuation. Fish Shellfish. Immunol. 2020;103:103–110. doi: 10.1016/j.fsi.2020.04.053. [DOI] [PubMed] [Google Scholar]
  • 210.Wu F., Falfushynska H., Dellwig O., Piontkivska H., Sokolova I.M. Interactive effects of salinity variation and exposure to ZnO nanoparticles on the innate immune system of a sentinel marine bivalve, Mytilus edulis. Sci. Total Environ. 2020;712 doi: 10.1016/j.scitotenv.2019.136473. [DOI] [PubMed] [Google Scholar]
  • 211.Tian L., Tan P., Yang L., Zhu W., Xu D. Effects of salinity on the growth, plasma ion concentrations, osmoregulation, non-specific immunity, and intestinal microbiota of the yellow drum (Nibea albiflora) Aquaculture. 2020;528 doi: 10.1016/j.aquaculture.2020.735470. [DOI] [Google Scholar]
  • 212.El-Leithy A.A.A., Hemeda S.A., El Naby W., El Nahas A.F., Hassan S.A.H., Awad S.T., El-Deeb S.I., Helmy Z.A. Optimum salinity for Nile tilapia (Oreochromis niloticus) growth and mRNA transcripts of ion-regulation, inflammatory, stress- and immune-related genes. Fish Physiol. Biochem. 2019;45(4):1217–1232. doi: 10.1007/s10695-019-00640-7. [DOI] [PubMed] [Google Scholar]
  • 213.Schmitz M., Ziv T., Admon A., Baekelandt S., Mandiki S.N.M., L'Hoir M., Kestemont P. Salinity stress, enhancing basal and induced immune responses in striped catfish Pangasianodon hypophthalmus (Sauvage) J. Proteom. 2017;167:12–24. doi: 10.1016/j.jprot.2017.08.005. [DOI] [PubMed] [Google Scholar]
  • 214.Liu Z.-F., Gao X.-Q., Yu J.-X., Qian X.-M., Xue G.-P., Zhang Q.-Y., Liu B.-L., Hong L. Effects of different salinities on growth performance, survival, digestive enzyme activity, immune response, and muscle fatty acid composition in juvenile American shad (Alosa sapidissima) Fish Physiol. Biochem. 2017;43(3):761–773. doi: 10.1007/s10695-016-0330-3. [DOI] [PubMed] [Google Scholar]
  • 215.Wang J., He R.-Z., Lu G.-L., Luo H.-L., Lu D.-Q., Li A.-X. Vaccine-induced antibody level as the parameter of the influence of environmental salinity on vaccine efficacy in Nile tilapia. Fish Shellfish. Immunol. 2018;82:522–530. doi: 10.1016/j.fsi.2018.08.025. [DOI] [PubMed] [Google Scholar]
  • 216.Wen X., Chu P., Xu J., Wei X., Fu D., Wang T., Yin S. Combined effects of low temperature and salinity on the immune response, antioxidant capacity and lipid metabolism in the pufferfish (Takifugu fasciatus) Aquaculture. 2021;531 doi: 10.1016/j.aquaculture.2020.735866. [DOI] [Google Scholar]
  • 217.Mozanzadeh M.T., Safari O., Oosooli R., Mehrjooyan S., Najafabadi M.Z., Hoseini S.J., Saghavi H., Monem J. The effect of salinity on growth performance, digestive and antioxidant enzymes, humoral immunity and stress indices in two euryhaline fish species: yellowfin seabream (Acanthopagrus latus) and asian seabass (Lates calcarifer) Aquaculture. 2021;534 doi: 10.1016/j.aquaculture.2020.736329. [DOI] [Google Scholar]
  • 218.Xiao Y., Zhang Y.-M., Xu W.-B., Chen D.-Y., Li B.-W., Cheng Y.-X., Guo X.-L., Dong W.-R., Shu M.-A. The effects of salinities stress on histopathological changes, serum biochemical index, non-specific immune and transcriptome analysis in red swamp crayfish Procambarus clarkii. Sci. Total Environ. 2022;840 doi: 10.1016/j.scitotenv.2022.156502. [DOI] [PubMed] [Google Scholar]
  • 219.Wang L., Pan L., Ding Y., Ren X. Effects of low salinity stress on immune response and evaluating indicators of the swimming crab Portunus trituberculatus. Aquac. Res. 2018;49(2):659–667. doi: 10.1111/are.13495. [DOI] [Google Scholar]
  • 220.Gu J., Dai S., Liu H., Cao Q., Yin S., Lai K.P., Tse W.K.F., Wong C.K.C., Shi H. Identification of immune-related genes in gill cells of Japanese eels (Anguilla japonica) in adaptation to water salinity changes. Fish Shellfish. Immunol. 2018;73:288–296. doi: 10.1016/j.fsi.2017.12.026. [DOI] [PubMed] [Google Scholar]
  • 221.Dawood M.A.O., Alkafafy M., Sewilam H. The antioxidant responses of gills, intestines and livers and blood immunity of common carp (Cyprinus carpio) exposed to salinity and temperature stressors. Fish Physiol. Biochem. 2022;48(2):397–408. doi: 10.1007/s10695-022-01052-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Huang Z., Ma A., Wang Xa, Lei J., Li W., Wang T., Yang Z., Qu J. Interaction of temperature and salinity on the expression of immunity factors in different tissues of juvenile turbot Scophthalmus maximus based on response surface methodology. Chin. J. Oceanol. Limnol. 2015;33(1):28–36. doi: 10.1007/s00343-015-4016-y. [DOI] [Google Scholar]
  • 223.Honorato T.B.M., Boni R., da Silva P.M., Marques-Santos L.F. Effects of salinity on the immune system cells of the tropical sea urchin echinometra lucunter. J. Exp. Mar. Biol. Ecol. 2017;486:22–31. doi: 10.1016/j.jembe.2016.09.012. [DOI] [Google Scholar]
  • 224.Yang Z., Zhu L., Zhao X., Cheng Y. Effects of salinity stress on osmotic pressure, free amino acids, and immune-associated parameters of the juvenile Chinese mitten crab, Eriocheir sinensis. Aquaculture. 2022;549 doi: 10.1016/j.aquaculture.2021.737776. [DOI] [Google Scholar]
  • 225.Ye Y., Zhu B., Yun J., Yang Y., Tian J., Xu W., Du X., Zhao Y., Li Y. Comparison of antioxidant capacity and immune response between low salinity tolerant hybrid and normal variety of Pacific white shrimp (Litopenaeus vannamei) Aquac. Int. 2024;32(2):1879–1894. doi: 10.1007/s10499-023-01248-8. [DOI] [Google Scholar]
  • 226.Moniruzzaman M., Mukherjee M., Kumar S., Chakraborty S.B. Effects of salinity stress on antioxidant status and inflammatory responses in females of a “Near threatened” economically important fish species notopterus chitala: a mechanistic approach. Environ. Sci. Pollut. Res. 2022;29(49):75031–75042. doi: 10.1007/s11356-022-21142-9. [DOI] [PubMed] [Google Scholar]
  • 227.Wang L.-U., Chen J.-C. The immune response of white shrimp Litopenaeus vannamei and its susceptibility to Vibrio alginolyticus at different salinity levels. Fish Shellfish. Immunol. 2005;18(4):269–278. doi: 10.1016/j.fsi.2004.07.008. [DOI] [PubMed] [Google Scholar]
  • 228.Rahi M.L., Azad K.N., Tabassum M., Irin H.H., Hossain K.S., Aziz D., Moshtaghi A., Hurwood D.A. Effects of salinity on physiological, biochemical and gene expression parameters of black tiger shrimp (Penaeus monodon): potential for farming in low-salinity environments. Biology. 2021;10(12):1220. doi: 10.3390/biology10121220. https://www.mdpi.com/2079-7737/10/12/1220 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Li X., Shen Y., Bao Y., Wu Z., Yang B., Jiao L., Zhang C., Tocher D.R., Zhou Q., Jin M. Physiological responses and adaptive strategies to acute low-salinity environmental stress of the euryhaline marine fish black seabream (Acanthopagrus schlegelii) Aquaculture. 2022;554 doi: 10.1016/j.aquaculture.2022.738117. [DOI] [Google Scholar]
  • 230.Cheng W., Juang F.-M., Chen J.-C. The immune response of Taiwan abalone Haliotis diversicolor supertexta and its susceptibility to Vibrio parahaemolyticus at different salinity levels. Fish Shellfish. Immunol. 2004;16(3):295–306. doi: 10.1016/S1050-4648(03)00111-6. [DOI] [PubMed] [Google Scholar]
  • 231.Jakiul Islam M., James Slater M., Thiele R., Kunzmann A. Influence of extreme ambient cold stress on growth, hematological, antioxidants, and immune responses in European seabass, Dicentrarchus labrax acclimatized at different salinities. Ecol. Indic. 2021;122 doi: 10.1016/j.ecolind.2020.107280. [DOI] [Google Scholar]
  • 232.Zheng X., Liang X., Chen Q., Xie J., Dong H., Yang J., Zhang J. Physiological responses of juvenile bullfrogs (Aquarana catesbeiana) to salinity stress. Animals. 2024;14(23):3454. doi: 10.3390/ani14233454. https://www.mdpi.com/2076-2615/14/23/3454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Long J., Cui Y., Wang R., Chen Y., Zhao N., Wang C., Wang Z., Li Y. Combined effects of high salinity and ammonia-N exposure on the energy metabolism, immune response, oxidative resistance and ammonia metabolism of the Pacific white shrimp Litopenaeus vannamei. Aquac. Rep. 2021;20 doi: 10.1016/j.aqrep.2021.100648. [DOI] [Google Scholar]
  • 234.De Zoysa M., Whang I., Lee Y., Lee S., Lee J.-S., Lee J. Transcriptional analysis of antioxidant and immune defense genes in disk abalone (Haliotis discus discus) during thermal, low-salinity and hypoxic stress. Comp. Biochem. Physiol. B: Biochem. Mol. Biol. 2009;154(4):387–395. doi: 10.1016/j.cbpb.2009.08.002. [DOI] [PubMed] [Google Scholar]
  • 235.Luo L., Yang L.-S., Huang J.-H., Jiang S.-G., Zhou F.-L., Li Y.-D., Jiang S., Yang Q.-B. Effects of different salinity stress on the transcriptomic responses of freshwater crayfish (Procambarus clarkii, Girard, 1852) Biology. 2024;13(7):530. doi: 10.3390/biology13070530. https://www.mdpi.com/2079-7737/13/7/530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Liu D., Zhang Z., Song Y., Yang J., Lu Y., Lai W., Wu Z., Zhao D., Lin H., Zhang Y., Zhang J., Li S. Effects of salinity on growth, physiology, biochemistry and gut microbiota of juvenile grass carp (Ctenopharyngodon idella) Aquat. Toxicol. 2023;258 doi: 10.1016/j.aquatox.2023.106482. [DOI] [PubMed] [Google Scholar]
  • 237.Mengjie W., Tianqi C., Feng L., Wei Z., Bao L., Wantu X. Effect of salinity stress on the antioxidant enzymes, non-specific immune enzymes, and Na+/K+ ATPase activities in Larimichthys polyactis. Haiyang Xuebao. 2021;43(2):59. doi: 10.12284/hyxb2021038. [DOI] [Google Scholar]
  • 238.Mo Z., Li L., Ying L., Xiaolong G. Effects of sudden drop in salinity on osmotic pressure regulation and antioxidant defense mechanism of Scapharca subcrenata. Front. Physiol. 2020;11 doi: 10.3389/fphys.2020.00884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Ulkhaq M.F., Safitri K.R., Asrin D.I., Sulmartiwi L., LOH J.-Y. Effects of salinity changes on hematological blood parameters and stress responses in red Tilapia (Oreochromis spp.) infected with Vibrio harveyi. Borneo. J. Resour. Sci. Technol. 2024;14(2):54–67. [Google Scholar]
  • 240.Wang Y., Li W., Li L., Zhang W., Lu W. Effects of salinity on the physiological responses of the large yellow croaker seudosciaena crocea under indoor culture conditions. Aquac. Res. 2016;47(11):3410–3420. doi: 10.1111/are.12788. [DOI] [Google Scholar]
  • 241.Shen M., Cui Y., Wang R., Dong T., Ye H., Wang S., Fu R., Li Y. Acute response of Pacific white shrimp Litopenaeus vannamei to high-salinity reductions in osmosis-, metabolism-, and immune-related enzyme activities. Aquac. Int. 2020;28(1):31–39. doi: 10.1007/s10499-019-00441-y. [DOI] [Google Scholar]
  • 242.Liu S., Qi C., Jia Y., Gu Z., Li E. Growth and intestinal health of the red claw crayfish, Cherax quadricarinatus, reared under different salinities. Aquaculture. 2020;524 doi: 10.1016/j.aquaculture.2020.735256. [DOI] [Google Scholar]
  • 243.Zannat M.M., Rohani M.F., Jeba R.-O.Z., Shahjahan M. Multi-species probiotics ameliorate salinity-induced growth retardation In striped catfish pangasianodon hypophthalmus. Int. J. Environ. Res. 2024;18(5):89. doi: 10.1007/s41742-024-00636-z. [DOI] [Google Scholar]
  • 244.Shukry M., Abd El-Kader M.F., Hendam B.M., Dawood M.A.O., Farrag F.A., Aboelenin S.M., Soliman M.M., Abdel-Latif H.M.R. Dietary Aspergillus oryzae modulates serum biochemical indices, immune responses, oxidative stress, and transcription of HSP70 and cytokine genes in Nile Tilapia exposed to salinity stress. Animals. 2021;11(6):1621. doi: 10.3390/ani11061621. https://www.mdpi.com/2076-2615/11/6/1621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Wei J., Tian L., Wang Y., Yu L., Zhu X. Effects of salinity, photoperiod, and light spectrum on larval survival, growth, and related enzyme activities in the giant freshwater prawn, Macrobrachium rosenbergii. Aquaculture. 2021;530 doi: 10.1016/j.aquaculture.2020.735794. [DOI] [Google Scholar]
  • 246.Cao W., Bi S., Chi C., Dong Y., Xia S., Liu Z., Zhou L., Sun X., Geng Y., Wu B. Effects of high salinity stress on the survival, gill tissue, enzyme activity and free amino acid content in razor clam sinonovacula constricta. Front. Mar. Sci. 2022;9 doi: 10.3389/fmars.2022.839614. [DOI] [Google Scholar]
  • 247.Wu H., Liu X., Zhang X., Ji C., Zhao J., Yu J. Proteomic and metabolomic responses of clam Ruditapes philippinarum to arsenic exposure under different salinities. Aquat. Toxicol. 2013;136-137:91–100. doi: 10.1016/j.aquatox.2013.03.020. [DOI] [PubMed] [Google Scholar]
  • 248.Motamedi-Tehrani J., Peyghan R., Shahriari A., Razijalali M., Ebrahimi E. The influence of ammonia-N and salinity levels on oxidative stress markers, hepatic enzymes, and acid phosphatase activity in Nile tilapia (Oreochromis niloticus) Sci. Rep. 2025;15(1):559. doi: 10.1038/s41598-024-84136-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Zhang H., Dong S., Shan H., Yang C., Wang F. Application of the DEB-TKTD model with multi-omics data: prediction of life history traits of Chinese mitten crab (Eriocheir sinensis) under different salinities. Ecotoxicol. Env. Saf. 2025;290 doi: 10.1016/j.ecoenv.2024.117635. [DOI] [PubMed] [Google Scholar]
  • 250.Li C., Wu X., Lei K., Tian X., Liu Y., Shan H. Characterization of the quality and physiological status of Penaeus vannamei in salt pans at various salinities. Aquac. Int. 2024;32(6):8131–8150. doi: 10.1007/s10499-024-01559-4. [DOI] [Google Scholar]
  • 251.Liu J., Ai T., Yang J., Shang M., Jiang K., Yin Y., Gao L., Jiang W., Zhao N., Ju J., Qin B. Effects of salinity on growth, digestive enzyme activity, and antioxidant capacity of spotbanded scat (Selenotoca multifasciata) juveniles. Fishes. 2024;9(8):309. https://www.mdpi.com/2410-3888/9/8/309 [Google Scholar]
  • 252.Li T.-Z., Chen C.-Z., Xing S.-Y., Liu L., Li P., Li Z.-H. The influence of triphenyltin exposure on the osmoregulatory capacity of Marine medaka (Oryzias melastigma) at different salinities. Water. 2024;16(7):921. https://www.mdpi.com/2073-4441/16/7/921 [Google Scholar]
  • 253.Lee R.P.-T., Lin Y.-R., Huang C.-Y., Nan F.-H. Effects of nutrient source, temperature, and salinity on the growth and survival of three giant clam species (Tridacnidae) Animals. 2024;14(7):1054. doi: 10.3390/ani14071054. https://www.mdpi.com/2076-2615/14/7/1054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Abdel-Tawwab M., Monier M.N., Hoseinifar S.H., Faggio C. Fish response to hypoxia stress: growth, physiological, and immunological biomarkers. Fish Physiol. Biochem. 2019;45(3):997–1013. doi: 10.1007/s10695-019-00614-9. [DOI] [PubMed] [Google Scholar]
  • 255.Bickler P.E., Buck L.T. Hypoxia tolerance in reptiles, amphibians, and fishes: life with variable oxygen availability. Annu. Rev. Physiol. 2007;69:145–170. doi: 10.1146/annurev.physiol.69.031905.162529. (Volume 69, 2007) [DOI] [PubMed] [Google Scholar]
  • 256.Lushchak V.I. Environmentally induced oxidative stress in aquatic animals. Aquat. Toxicol. 2011;101(1):13–30. doi: 10.1016/j.aquatox.2010.10.006. [DOI] [PubMed] [Google Scholar]
  • 257.Beemelmanns, A., Zanuzzo, F.S., Sandrelli, R.M., Rise, M.L. Gamperl, A.K. 2021. The Atlantic salmon’s stress-and immune-related transcriptional responses to moderate hypoxia, an incremental temperature increase, and these challenges combined. G3. 11(7): jkab102. [DOI] [PMC free article] [PubMed]
  • 258.Cerra M.C., Filice M., Caferro A., Mazza R., Gattuso A., Imbrogno S. Cardiac hypoxia tolerance in fish: from functional responses to cell signals. Int. J. Mol. Sci. 2023;24(2):1460. doi: 10.3390/ijms24021460. https://www.mdpi.com/1422-0067/24/2/1460 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Chun Y., Kim J. AMPK–mTOR signaling and cellular adaptations in hypoxia. Int. J. Mol. Sci. 2021;22(18):9765. doi: 10.3390/ijms22189765. https://www.mdpi.com/1422-0067/22/18/9765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Liu Q., Li Y., Cao Y., Gu L., Li T., Liu Y., Song J., Wang W., Wang X., Li B., Liu S. Transcriptome analysis of brain and skin reveals immune responses to acute hypoxia and reoxygenation in Pseudobagrus ussuriensis. Animals. 2024;14(2):246. doi: 10.3390/ani14020246. https://www.mdpi.com/2076-2615/14/2/246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Huo D., Sun L., Ru X., Zhang L., Lin C., Liu S., Xin X., Yang H. Impact of hypoxia stress on the physiological responses of sea cucumber Apostichopus japonicus: respiration, digestion, immunity and oxidative damage. PeerJ. 2018;6:e4651. doi: 10.7717/peerj.4651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Wang Q., Li X., Sha H., Luo X., Zou G., Liang H. Identification of microRNAs in Silver carp (Hypophthalmichthys molitrix) response to hypoxia stress. Animals. 2021;11(10):2917. doi: 10.3390/ani11102917. https://www.mdpi.com/2076-2615/11/10/2917 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Jie Y.-K., Cheng C.-H., Ma H.-L., Liu G.-X., Fan S.-G., Jiang J.-J., Guo Z.-X. Hypoxia affects the antioxidant activity of glutaredoxin 3 in Scylla paramamosain through Hypoxia Response elements. Antioxidants. 2023;12(1):76. doi: 10.3390/antiox12010076. https://www.mdpi.com/2076-3921/12/1/76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Tiedke J., Thiel R., Burmester T. Molecular response of estuarine fish to hypoxia: a comparative study with Ruffe and flounder from field and laboratory. PLoS One. 2014;9(3) doi: 10.1371/journal.pone.0090778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Karpowicz M., Ejsmont-Karabin J., Kozłowska J., Feniova I., Dzialowski A.R. Zooplankton community responses to oxygen stress. Water. 2020;12(3):706. https://www.mdpi.com/2073-4441/12/3/706 [Google Scholar]
  • 266.Rabalais N., Díaz R., Levin L., Turner R., Gilbert D., Zhang J. Dynamics and distribution of natural and human-caused coastal hypoxia. Biogeosci. Dis. 2009;6(5) [Google Scholar]
  • 267.Li D., Yu J., Zhu J., Xiao W., Zou Z., Chen B., Wei C., Zhu J., Yang H. Identification of the effects of hypoxia on the liver tissues of Nile tilapia Oreochromis niloticus. BMC Genom. 2024;25(1):946. doi: 10.1186/s12864-024-10700-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Wang J., Lu D.-Q., Jiang B., Luo H.-L., Lu G.-L., Li A.-X. The effect of intermittent hypoxia under different temperature on the immunomodulation in Streptococcus agalactiae vaccinated Nile tilapia (Oreochromis niloticus) Fish Shellfish. Immunol. 2018;79:181–192. doi: 10.1016/j.fsi.2018.04.040. [DOI] [PubMed] [Google Scholar]
  • 269.Kiani A.A., Elyasi H., Ghoreyshi S., Nouri N., Safarzadeh A., Nafari A. Study on hypoxia-inducible factor and its roles in immune system. Immunol. Med. 2021;44(4):223–236. doi: 10.1080/25785826.2021.1910187. [DOI] [PubMed] [Google Scholar]
  • 270.Semenza G.L. Hypoxia-inducible factors in physiology and medicine. Cell. 2012;148(3):399–408. doi: 10.1016/j.cell.2012.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Dagoudo M., Qiang J., Bao J.-W., Tao Y.-F., Zhu H.-J., Tumukunde E.M., Ngoepe T.K., Xu P. Effects of acute hypoxia stress on hemato-biochemical parameters, oxidative resistance ability, and immune responses of hybrid yellow catfish (Pelteobagrus fulvidraco × P. vachelli) juveniles. Aquac. Int. 2021;29(5):2181–2196. doi: 10.1007/s10499-021-00742-1. [DOI] [PubMed] [Google Scholar]
  • 272.Hu F., Yin L., Dong F., Zheng M., Zhao Y., Fu S., Zhang W., Chen X. Effects of long-term cadmium exposure on growth, antioxidant defense and DNA methylation in juvenile Nile tilapia (Oreochromis niloticus) Aquat. Toxicol. 2021;241 doi: 10.1016/j.aquatox.2021.106014. [DOI] [PubMed] [Google Scholar]
  • 273.Nardi A., Benedetti M., Gorbi S., Regoli F. Interactive immunomodulation in the Mediterranean mussel mytilus galloprovincialis under thermal stress and cadmium exposure. Front. Mar. Sci. 2021;8 doi: 10.3389/fmars.2021.751983. [DOI] [Google Scholar]
  • 274.Parisi M.G., Pirrera J., La Corte C., Dara M., Parrinello D., Cammarata M. Effects of organic mercury on mytilus galloprovincialis hemocyte function and morphology. J. Comp. Physiol. B. 2021;191(1):143–158. doi: 10.1007/s00360-020-01306-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Kang B., Wang J., Guo S., Yang L. Mercury-induced toxicity: mechanisms, molecular pathways, and gene regulation. Sci. Total Environ. 2024;943 doi: 10.1016/j.scitotenv.2024.173577. [DOI] [PubMed] [Google Scholar]
  • 276.Lu X., Xiang Y., Yang G., Zhang L., Wang H., Zhong S. Transcriptomic characterization of zebrafish larvae in response to mercury exposure. Comp. Biochem. Physiol. C: Toxicol. Pharmacol. 2017;192:40–49. doi: 10.1016/j.cbpc.2016.12.006. [DOI] [PubMed] [Google Scholar]
  • 277.Sarkar O., Dey K.K., Islam S., Chattopadhyay A. In: Biomarkers in Toxicology. Patel V.B., Preedy V.R., Rajendram R., editors. Springer International Publishing; 2023. Lead and aquatic ecosystems, biomarkers, and implications for humankind; pp. 961–988. [DOI] [Google Scholar]
  • 278.Zhang J., Ren Z., Chen M. Immunotoxicity and transcriptome analyses of zebrafish (Danio rerio) embryos exposed to 6:2 FTSA. Toxics. 2023;11(5):459. doi: 10.3390/toxics11050459. https://www.mdpi.com/2305-6304/11/5/459 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Taheri Mirghaed A., Baes M., Hoseini S.M. Humoral immune responses and gill antioxidant-related gene expression of common carp (Cyprinus carpio) exposed to lufenuron and flonicamide. Fish Physiol. Biochem. 2020;46(2):739–746. doi: 10.1007/s10695-019-00747-x. [DOI] [PubMed] [Google Scholar]
  • 280.Major K.M., Brander S.M. In: Pyrethroid Insecticides. Eljarrat E., editor. Springer International Publishing; 2020. The ecological and evolutionary implications of pyrethroid exposure: a new perspective on aquatic ecotoxicity; pp. 109–148. [DOI] [Google Scholar]
  • 281.Galbiati V., Buoso E., d'Emmanuele di Villa Bianca R., Paola R.D., Morroni F., Nocentini G., Racchi M., Viviani B., Corsini E. Immune and nervous systems interaction in endocrine disruptors toxicity: the case of Atrazine. Front. Toxicol. 2021;3 doi: 10.3389/ftox.2021.649024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Zhang X., Wang M., Gao S., Ren R., Zheng J., Zhang Y. Atrazine-induced apoptosis of splenocytes in BALB/C mice. BMC Med. 2011;9(1):117. doi: 10.1186/1741-7015-9-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Wang J., Li Y., Jaramillo-Torres A., Einen O., Jakobsen J.V., Krogdahl Å., Kortner T.M. Exploring gut microbiota in adult Atlantic salmon (Salmo salar L.): associations with gut health and dietary prebiotics. Anim. Microbiome. 2023;5(1):47. doi: 10.1186/s42523-023-00269-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Wang C., Sun G., Li S., Li X., Liu Y. Intestinal microbiota of healthy and unhealthy Atlantic salmon Salmo salar L. in a recirculating aquaculture system. J. Oceanol. Limnol. 2018;36(2):414–426. doi: 10.1007/s00343-017-6203-5. [DOI] [Google Scholar]
  • 285.Coates C.J., Costa-Paiva E.M. In: Vertebrate and Invertebrate Respiratory Proteins, Lipoproteins and other Body Fluid Proteins. Hoeger U., Harris J.R., editors. Springer International Publishing; 2020. Multifunctional roles of hemocyanins; pp. 233–250. [DOI] [Google Scholar]
  • 286.Alboni S., van Dijk R.M., Poggini S., Milior G., Perrotta M., Drenth T., Brunello N., Wolfer D.P., Limatola C., Amrein I., Cirulli F., Maggi L., Branchi I. Fluoxetine effects on molecular, cellular and behavioral endophenotypes of depression are driven by the living environment. Mol. Psychiatry. 2017;22(4):552–561. doi: 10.1038/mp.2015.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Courant F., Arpin-Pont L., Bonnefille B., Vacher S., Picot-Groz M., Gomez E., Fenet H. Exposure of marine mussels to diclofenac: modulation of prostaglandin biosynthesis. Environ. Sci. Pollut. Res. 2018;25(7):6087–6094. doi: 10.1007/s11356-017-9228-6. [DOI] [PubMed] [Google Scholar]
  • 288.Eskander S.B., Saleh H.M. In: Cellular and Molecular Phytotoxicity of Heavy Metals. Faisal M., Saquib Q., Alatar A.A., Al-Khedhairy A.A., editors. Springer International Publishing; 2020. Heavy metal-induced oxidative stress and related cellular process; pp. 99–123. [DOI] [Google Scholar]
  • 289.Campbell J.H., Dixon B., Whitehouse L.M. The intersection of stress, sex and immunity in fishes. Immunogenetics. 2021;73(1):111–129. doi: 10.1007/s00251-020-01194-2. [DOI] [PubMed] [Google Scholar]
  • 290.Chaabani R., Bejaoui M., Ben Jeddou I., Zaouali M.A., Haouas Z., Belgacem S., Peralta C., Ben Abdennebi H. Effect of the non-steroidal anti-inflammatory drug diclofenac on ischemia–Reperfusion injury in rat liver: a nitric oxide-dependent mechanism. Inflammation. 2023;46(4):1221–1235. doi: 10.1007/s10753-023-01802-9. [DOI] [PubMed] [Google Scholar]
  • 291.Pijanowski L., Jurecka P., Irnazarow I., Kepka M., Szwejser E., Verburg-van Kemenade B.M.L., Chadzinska M. Activity of the hypothalamus–pituitary–interrenal axis (HPI axis) and immune response in carp lines with different susceptibility to disease. Fish Physiol. Biochem. 2015;41(5):1261–1278. doi: 10.1007/s10695-015-0084-3. [DOI] [PubMed] [Google Scholar]
  • 292.Sun X., Deng Y., Fang L., Ni M., Wang X., Zhang T., Chen Y., Cai G., Pan F. Association of exposure to heavy metal mixtures with systemic immune-inflammation Index among US adults in NHANES 2011–2016. Biol. Trace Elem. Res. 2024;202(7):3005–3017. doi: 10.1007/s12011-023-03901-y. [DOI] [PubMed] [Google Scholar]
  • 293.Anne Brown J. In: Fish Ecophysiology. Rankin J.C., Jensen F.B., editors. Springer; Netherlands: 1993. Endocrine responses to environmental pollutants; pp. 276–296. [DOI] [Google Scholar]
  • 294.García-Lagunas N., Romero-Geraldo R., Kao-Godinez A.K., Hernández-Saavedra N.Y. Differential expression of immune response genes in Pacific oyster, Crassostrea gigas spat, fed with dinoflagellates Gymnodinium catenatum and Prorocentrum lima. Lat. Am. J. Aquat. Res. 2019;47(4):699–705. [Google Scholar]
  • 295.Ginger K.W.K., Vera C.B.S., R D., Dennis C.K.S., Adela L.J., Yu Z., Thiyagarajan V. Larval and post-Larval stages of Pacific Oyster (Crassostrea gigas) are resistant to elevated CO2. PLoS One. 2013;8(5) doi: 10.1371/journal.pone.0064147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Wang Q., Cao R., Ning X., You L., Mu C., Wang C., Wei L., Cong M., Wu H., Zhao J. Effects of ocean acidification on immune responses of the Pacific oyster crassostrea gigas. Fish Shellfish. Immunol. 2016;49:24–33. doi: 10.1016/j.fsi.2015.12.025. [DOI] [PubMed] [Google Scholar]
  • 297.Gazeau F., Gattuso J.-P., Dawber C., Pronker A., Peene F., Peene J., Heip C., Middelburg J. Effect of ocean acidification on the early life stages of the blue mussel Mytilus edulis. Biogeosciences. 2010;7(7):2051–2060. [Google Scholar]
  • 298.Guo F., Ye Y., Zhu K., Lin S., Wang Y., Dong Z., Yao R., Li H., Wang W., Liao Z., Guo B., Yan X. Genetic diversity, population structure, and environmental adaptation signatures of Chinese coastal hard-shell mussel mytilus coruscus revealed by whole-genome sequencing. Int. J. Mol. Sci. 2023;24(17) doi: 10.3390/ijms241713641. https://www.mdpi.com/1422-0067/24/17/13641 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Mackenzie C.L., Lynch S.A., Culloty S.C., Malham S.K. Future oceanic warming and acidification alter immune response and disease status in a commercial shellfish species, Mytilus edulis L. Plos One. 2014;9(6) doi: 10.1371/journal.pone.0099712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Li H., Ren C., Jiang X., Cheng C., Ruan Y., Zhang X., Huang W., Chen T., Hu C. Na+/H+ exchanger (NHE) in Pacific white shrimp (Litopenaeus vannamei): molecular cloning, transcriptional response to acidity stress, and physiological roles in pH homeostasis. PLoS One. 2019;14(2) doi: 10.1371/journal.pone.0212887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Townhill B., Artioli Y., Pinnegar J., Birchenough S. Exposure of commercially exploited shellfish to changing pH levels: how to scale-up experimental evidence to regional impacts. ICES J. Mar. Sci. 2022;79(9):2362–2372. [Google Scholar]
  • 302.Dineshram R., Xiao S., Ko G.W.K., Li J., Smrithi K., Thiyagarajan V., Zhang Y., Yu Z. Ocean acidification triggers cell signaling, suppress immune and calcification in the Pacific oyster larvae. Front. Mar. Sci. 2021;8 doi: 10.3389/fmars.2021.782583. [DOI] [Google Scholar]
  • 303.Thor P., Bailey A., Halsband C., Guscelli E., Gorokhova E., Fransson A. Seawater pH predicted for the year 2100 affects the metabolic response to feeding in copepodites of the Arctic Copepod Calanus glacialis. PLoS One. 2016;11(12) doi: 10.1371/journal.pone.0168735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Sánchez-Martínez J.G., Rábago-Castro J.L., Vázquez-Sauceda M.L., Pérez-Castañeda R., Blanco-Martínez Z., Benavides-González F. Effect of β-glucan dietary levels on immune response and hematology of channel catfish Ictalurus punctatus juveniles. Lat. Am. J. Aquat. Res. 2017;45(4):690–698. [Google Scholar]
  • 305.Zhao S., Shi X., Sun B., Liu Y., Tian Z., Huotari J. Effects of pH on phosphorus form transformation in lake sediments. Water Supply. 2021;22(2):1231–1243. doi: 10.2166/ws.2021.356. [DOI] [Google Scholar]
  • 306.Kataoka C., Kashiwada S. Ecological risks due to immunotoxicological effects on aquatic organisms. Int. J. Mol. Sci. 2021;22(15):8305. doi: 10.3390/ijms22158305. https://www.mdpi.com/1422-0067/22/15/8305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Xu Z., Cao J., Qin X., Qiu W., Mei J., Xie J. Toxic effects on bioaccumulation, hematological parameters, oxidative stress, immune responses and tissue structure in fish exposed to Ammonia nitrogen: a review. Animals. 2021;11(11):3304. doi: 10.3390/ani11113304. https://www.mdpi.com/2076-2615/11/11/3304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Wu F., Xie Z., Lan Y., Dupont S., Sun M., Cui S., Huang X., Huang W., Liu L., Hu M., Lu W., Wang Y. Short-term exposure of mytilus coruscus to decreased pH and salinity change impacts immune parameters of their haemocytes. Front. Physiol. 2018;9 doi: 10.3389/fphys.2018.00166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Tort L., Balasch J.C. In: Principles of Fish Immunology : From Cells and Molecules to Host Protection. Buchmann K., Secombes C.J., editors. Springer International Publishing; 2022. Stress and immunity in fish; pp. 609–655. [DOI] [Google Scholar]
  • 310.Ding R., Yang R., Fu Z., Zhao W., Li M., Yu G., Ma Z., Bai Z. Response of antioxidation and immunity to combined influences of pH and ammonia nitrogen in the spotted babylon (<em>Babylonia areolata</em>) Heliyon. 2024;10(8) doi: 10.1016/j.heliyon.2024.e29205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Zhang Y., Zhang X., Xu J., Zheng J., Cui Z. Combined effects of low pH stress and bacterial infection on the transcriptional changes of hemocytes in Chinese mitten crab Eriocheir sinensis. Fish Shellfish. Immunol. 2024;155 doi: 10.1016/j.fsi.2024.109995. [DOI] [PubMed] [Google Scholar]
  • 312.Sridhar A., Krishnasamy Sekar R., Manikandan D.B., Arumugam M., Veeran S., Ramasamy T. Activity profile of innate immune-related enzymes and bactericidal of freshwater fish epidermal mucus extract at different pH. Environ. Sci. Pollut. Res. 2021;28(26):33914–33926. doi: 10.1007/s11356-020-11173-5. [DOI] [PubMed] [Google Scholar]
  • 313.Chen Y.-Y., Chen J.-C., Tseng K.-C., Lin Y.-C., Huang C.-L. Activation of immunity, immune response, antioxidant ability, and resistance against vibrio alginolyticus in white shrimp Litopenaeus vannamei decrease under long-term culture at low pH. Fish Shellfish. Immunol. 2015;46(2):192–199. doi: 10.1016/j.fsi.2015.05.055. [DOI] [PubMed] [Google Scholar]
  • 314.Li C.-C., Chen J.-C. The immune response of white shrimp Litopenaeus vannamei and its susceptibility to Vibrio alginolyticus under low and high pH stress. Fish Shellfish. Immunol. 2008;25(6):701–709. doi: 10.1016/j.fsi.2008.01.007. [DOI] [PubMed] [Google Scholar]
  • 315.Dominguez M., Takemura A., Tsuchiya M. Effects of changes in environmental factors on the non-specific immune response of Nile tilapia, oreochromis Niloticus L . Aquac. Res. 2005;36(4):391–397. doi: 10.1111/j.1365-2109.2005.01220.x. [DOI] [Google Scholar]
  • 316.Singh S.K., Tiwari V.K., Chadha N.K., Munilkumar S., Prakash C., Pawar N.A. Effect of dietary synbiotic supplementation on growth, immune and physiological status of Labeo rohita juveniles exposed to low pH stress. Fish Shellfish. Immunol. 2019;91:358–368. doi: 10.1016/j.fsi.2019.05.023. [DOI] [PubMed] [Google Scholar]
  • 317.Sharma S., Jayaprakash S., Philipose K., Radhakrishnan E. Effect of salinity and pH on selected immune functions of the Indian white shrimp, Fenneropenaeus indicus (H. Milne Edwards, 1837) Indian J. Fish. 2009;56(3):183–187. [Google Scholar]
  • 318.Cocci P., Roncarati A., Capriotti M., Mosconi G., Palermo F.A. Transcriptional alteration of gene biomarkers in hemocytes of wild ostrea edulis with molecular evidence of infections with Bonamia spp. and/or marteilia refringens parasites. Pathogens. 2020;9(5):323. doi: 10.3390/pathogens9050323. https://www.mdpi.com/2076-0817/9/5/323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Kamermans P., Saurel C. Interacting climate change effects on mussels (Mytilus edulis and M. galloprovincialis) and oysters (Crassostrea gigas and Ostrea edulis): experiments for bivalve individual growth models. Aquat. Living Resour. 2022;35:1. doi: 10.1051/alr/2022001. [DOI] [Google Scholar]
  • 320.Strilbytska O.M., Semaniuk U.V., Storey K.B., Yurkevych I.S., Lushchak O. Insulin signaling in intestinal stem and progenitor cells as an important determinant of physiological and metabolic traits in drosophila. Cells. 2020;9(4):803. doi: 10.3390/cells9040803. https://www.mdpi.com/2073-4409/9/4/803 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Xu Z., Zhang H., Guo M., Fang D., Mei J., Xie J. Analysis of acute nitrite exposure on physiological stress response, oxidative stress, Gill tissue morphology and immune response of large yellow croaker (Larimichthys crocea) Animals. 2022;12(14):1791. doi: 10.3390/ani12141791. https://www.mdpi.com/2076-2615/12/14/1791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Häder D.-P., Williamson C.E., Wängberg S.-Å., Rautio M., Rose K.C., Gao K., Helbling E.W., Sinha R.P., Worrest R. Effects of UV radiation on aquatic ecosystems and interactions with other environmental factors. Photochem. Photobiol. Sci. 2015;14(1):108–126. doi: 10.1039/c4pp90035a. [DOI] [PubMed] [Google Scholar]
  • 323.Mitchell D.L., Adams-Deutsch T., Olson M.H. Dose dependence of DNA repair in rainbow trout (Oncorhynchus mykiss) larvae exposed to UV-B radiation. Photochem. Photobiol. Sci. 2009;8(1):75–81. doi: 10.1039/b807469k. [DOI] [PubMed] [Google Scholar]
  • 324.Neale P.J., Williamson C.E., Banaszak A.T., Häder D.P., Hylander S., Ossola R., Rose K.C., Wängberg S.Å., Zepp R. The response of aquatic ecosystems to the interactive effects of stratospheric ozone depletion, UV radiation, and climate change. Photochem. Photobiol. Sci. 2023;22(5):1093–1127. doi: 10.1007/s43630-023-00370-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Xue S., Zang Y., Chen J., Shang S., Gao L., Tang X. Ultraviolet-B radiation stress triggers reactive oxygen species and regulates the antioxidant defense and photosynthesis systems of intertidal red algae Neoporphyra haitanensis. Front. Mar. Sci. 2022;9 doi: 10.3389/fmars.2022.1043462. [DOI] [Google Scholar]
  • 326.Fajardo C., Santos P., Passos R., Vaz M., Azeredo R., Machado M., Fernández-Boo S., Baptista T., Costas B. Functional and molecular immune response of rainbow trout (Oncorhynchus mykiss) following challenge with Yersinia ruckeri. Int. J. Mol. Sci. 2022;23(6):3096. doi: 10.3390/ijms23063096. https://www.mdpi.com/1422-0067/23/6/3096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Mello D.F., De Oliveira E.S., Vieira R.C., Simoes E., Trevisan R., Dafre A.L., Barracco M.A. Cellular and transcriptional responses of crassostrea gigas hemocytes exposed in vitro to brevetoxin (PbTx-2) Mar. Drugs. 2012;10(3):583–597. doi: 10.3390/md10030583. https://www.mdpi.com/1660-3397/10/3/583 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Ivanina A.V., Falfushynska H.I., Beniash E., Piontkivska H., Sokolova I.M. Biomineralization-related specialization of hemocytes and mantle tissues of the Pacific oyster Crassostrea gigas. J. Exp. Biol. 2017;220(18):3209–3221. doi: 10.1242/jeb.160861. [DOI] [PubMed] [Google Scholar]
  • 329.Song H., Dong M., Xu W., Xie C., Zhang Y., Huang H., Zhang K., Han Y., Liu Y., Wei L., Wang X. Regulation of biomineralization and autophagy by the stress-sensing transcription factor CgRunx1 in crassostrea gigas under daylight ultraviolet B radiation. Mar. Biotechnol. 2024 doi: 10.1007/s10126-024-10370-4. [DOI] [PubMed] [Google Scholar]
  • 330.Singh M.K., Sharma J.G., Chakrabarti R. Effect of UV-B radiation on the defence system of Labeo rohita (Actinopterygii: Cypriniformes: Cyprinidae) larvae and its modulation by seed of devil’s horsewhip, Achyranthes aspera. Acta Ichthyol. Piscat. 2013;43:119–126. doi: 10.3750/AIP2013.43.2.04. [DOI] [Google Scholar]
  • 331.Subramani P.A., Hameed B., Michael R.D. Effect of UV-B radiation on the antibody response of fish – Implication on high altitude fish culture. J. Photochem. Photobiol. B: Biol. 2015;143:1–4. doi: 10.1016/j.jphotobiol.2014.12.021. [DOI] [PubMed] [Google Scholar]
  • 332.Salo H.M., Jokinen E.I., Markkula S.E., Aaltonen T.M., Penttilä H.T. Comparative effects of UVA and UVB irradiation on the immune system of fish. J. Photochem. Photobiol. B: Biol. 2000;56(2):154–162. doi: 10.1016/S1011-1344(00)00072-5. [DOI] [PubMed] [Google Scholar]
  • 333.Icoglu Aksakal F., Ciltas A. The impact of ultraviolet B (UV-B) radiation in combination with different temperatures in the early life stage of zebrafish (Danio rerio) Photochem. Photobiol. Sci. 2018;17(1):35–41. doi: 10.1039/c7pp00236j. [DOI] [PubMed] [Google Scholar]
  • 334.Sharma J.G., Chakrabarti R. In: Frontiers in Aquaculture Biotechnology. Lakra W.S., Goswami M., Trudeau V.L., editors. Academic Press; 2023. Chapter 12 - ultraviolet-B radiation, a potential threat to the aquatic organisms, and its mitigation; pp. 155–162. [DOI] [Google Scholar]
  • 335.Lawrence K.P., Young A.R., Diffey B.L., Norval M. The impact of solar ultraviolet radiation on fish: immunomodulation and photoprotective strategies. Fish Fish. 2020;21(1):104–119. doi: 10.1111/faf.12420. [DOI] [Google Scholar]
  • 336.Vitt S., Rahn A.K., Drolshagen L., Bakker T.C.M., Scharsack J.P., Rick I.P. Enhanced ambient UVB light affects growth, body condition and the investment in innate and adaptive immunity in three-spined sticklebacks (Gasterosteus aculeatus) Aquat. Ecol. 2017;51(4):499–509. doi: 10.1007/s10452-017-9632-5. [DOI] [Google Scholar]
  • 337.Markkula E., Salo H.M., Rikalainen K., Jokinen I.E. Long-term UVB irradiation affects the immune functions of carp (Cyprinus carpio) and rainbow trout (Oncorhynchus mykiss) Photochem. Photobiol. 2009;85(1):347–352. doi: 10.1111/j.1751-1097.2008.00446.x. [DOI] [PubMed] [Google Scholar]
  • 338.Browman H.I., Rodriguez C.A., Béland F., Cullen J.J., Davis R.F., Kouwenberg J.H., Kuhn P.S., McArthur B., Runge J.A., St-Pierre J.-F. Impact of ultraviolet radiation on marine crustacean zooplankton and ichthyoplankton: a synthesis of results from the estuary and Gulf of St. Lawrence. Can. Mar. Ecol. Prog. Ser. 2000;199:293–311. [Google Scholar]
  • 339.Alves R.N., Justo M.S.S., Laranja J.L.Q., Alarcon J.F., Al Suwailem A., Agustí S. Exposure to natural ultraviolet B radiation levels has adverse effects on growth, behavior, physiology, and innate immune response in juvenile European seabass (Dicentrarchus labrax) Aquaculture. 2021;533 doi: 10.1016/j.aquaculture.2020.736215. [DOI] [Google Scholar]
  • 340.Alves R.N., Mahamed A.H., Alarcon J.F., Al Suwailem A., Agustí S. Adverse effects of ultraviolet radiation on growth, behavior, skin condition, physiology, and immune function in Gilthead Seabream (Sparus aurata) Front. Mar. Sci. 2020;7 doi: 10.3389/fmars.2020.00306. [DOI] [Google Scholar]
  • 341.Jokinen I.E., Salo H.M., Markkula E., Rikalainen K., Arts M.T., Browman H.I. Additive effects of enhanced ambient ultraviolet B radiation and increased temperature on immune function, growth and physiological condition of juvenile (parr) Atlantic Salmon, Salmo salar. Fish Shellfish. Immunol. 2011;30(1):102–108. doi: 10.1016/j.fsi.2010.09.017. [DOI] [PubMed] [Google Scholar]
  • 342.Sharma J., Vasudeva Rao Y., Kumar S., Chakrabarti R. Impact of UV-B radiation on the digestive enzymes and immune system of larvae of Indian major carp Catla catla. Int. J. Radiat. Biol. 2010;86(3):181–186. doi: 10.3109/09553000903419312. [DOI] [PubMed] [Google Scholar]
  • 343.Hamed M., Monteiro C.E., Said R.E.M., Abdel-Maksoud M.A., Almanaa T.N., Naguib M., Abdel-Tawab H.S., Osman A., Lee J.-S., Sayed A.E.-D.H. UV-A-induced oxidative stress and immunotoxicity in African catfish (Clarias gariepinus): the bioremediation potential of Spirulina platensis for aquaculture. Aquac. Int. 2024;33(1):45. doi: 10.1007/s10499-024-01724-9. [DOI] [Google Scholar]
  • 344.Salo H.M., Aaltonen T.M., Markkula S.E., Jokinen E.I. Ultraviolet B irradiation modulates the Immune system of fish (Rutilus rutilus, Cyprinidae) I. Phagocytes. Photochem. Photobiol. 1998;67(4):433–437. doi: 10.1111/j.1751-1097.1998.tb05223.x. [DOI] [PubMed] [Google Scholar]
  • 345.Jokinen I.E., Markkula E.S., Salo H.M., Kuhn P., Nikoskelainen S., Arts M.T., Browman H.I. Exposure to increased ambient ultraviolet B radiation has negative effects on growth, condition and immune function of juvenile Atlantic salmon (Salmo salar) Photochem. Photobiol. 2008;84(5):1265–1271. doi: 10.1111/j.1751-1097.2008.00358.x. [DOI] [PubMed] [Google Scholar]
  • 346.Araújo M.J., Quintaneiro C., Soares A., Monteiro M.S. Effects of ultraviolet radiation to Solea senegalensis during early development. Sci. Total Environ. 2021;764 doi: 10.1016/j.scitotenv.2020.142899. [DOI] [PubMed] [Google Scholar]
  • 347.Ibrahim A.T.A. Negative impacts of ultraviolet-A radiation on antioxidant and oxidative stress biomarkers of African catfish Clarias gariepinus. Photochem. Photobiol. Sci. 2015;14(7):1337–1345. doi: 10.1039/c5pp00112a. [DOI] [PubMed] [Google Scholar]
  • 348.Hébert N., Gagné F., Cejka P., Bouchard B., Hausler R., Cyr D.G., Blaise C., Fournier M. Effects of ozone, ultraviolet and peracetic acid disinfection of a primary-treated municipal effluent on the Immune system of rainbow trout (Oncorhynchus mykiss) Comp. Biochem. Physiol. C: Toxicol. Pharmacol. 2008;148(2):122–127. doi: 10.1016/j.cbpc.2008.04.007. [DOI] [PubMed] [Google Scholar]
  • 349.Markkula S.E., Karvonen A., Salo H., Tellervo Valtonen E., Ilmari Jokinen E. Ultraviolet B irradiation affects resistance of rainbow trout (Oncorhynchus mykiss) against bacterium Yersinia ruckeri and trematode diplostomum spathaceum. Photochem. Photobiol. 2007;83(5):1263–1269. doi: 10.1111/j.1751-1097.2007.00165.x. [DOI] [PubMed] [Google Scholar]
  • 350.Fei F., Liu B., Gao X., Wang X., Liu Y., Bin H. Effects of supplemental ultraviolet light on growth, oxidative stress responses, and apoptosis-related gene expression of the shrimp Litopenaeus vannamei. Aquaculture. 2020;520 doi: 10.1016/j.aquaculture.2020.735013. [DOI] [Google Scholar]
  • 351.Valero Y., Mokrani D., Chaves-Pozo E., Arizcun M., Oumouna M., Meseguer J., Esteban M.Á., Cuesta A. Vaccination with UV-inactivated nodavirus partly protects European sea bass against infection, while inducing few changes in immunity. Dev. Comp. Immunol. 2018;86:171–179. doi: 10.1016/j.dci.2018.05.013. [DOI] [PubMed] [Google Scholar]
  • 352.Wang X., Liu B., Gao X., Wang X., Li H., Xu L., Wang G., Zhao K., Huang B. The effects of different UVA photoperiods on the growth performance, immune responses, antioxidant status and apoptosis-related gene expression of the Pacific white shrimp (Penaeus vannamei) Antibiotics. 2022;11(1):37. doi: 10.3390/antibiotics11010037. https://www.mdpi.com/2079-6382/11/1/37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Arts M.T., Palmer M.E., Skiftesvik A.B., Jokinen I.E., Browman H.I. UVB radiation variably affects n-3 fatty acids but elevated temperature reduces n-3 fatty acids in juvenile Atlantic Salmon (Salmo salar) Lipids. 2012;47(12):1181–1192. doi: 10.1007/s11745-012-3719-5. [DOI] [PubMed] [Google Scholar]
  • 354.Britto R.S., Garcia M.L., Martins da Rocha A., Flores J.A., Pinheiro M.V.B., Monserrat J.M., Ferreira J.L.R. Effects of carbon nanomaterials fullerene C60 and fullerol C60(OH)18–22 on gills of fish Cyprinus carpio (Cyprinidae) exposed to ultraviolet radiation. Aquat. Toxicol. 2012;114-115:80–87. doi: 10.1016/j.aquatox.2012.02.018. [DOI] [PubMed] [Google Scholar]
  • 355.Bullock A.M., Coutts R.R. The impact of solar ultraviolet radiation upon the skin of rainbow trout, Salmo gairdneri Richardson, farmed at high altitude in Bolivia. J. Fish Dis. 1985;8(3):263–272. doi: 10.1111/j.1365-2761.1985.tb00942.x. [DOI] [Google Scholar]
  • 356.Ghanizadeh Kazerouni† E., Khodabandeh S. Effects of ultraviolet radiation on skin structure and ultrastructure in Caspian Sea Salmon, Salmo trutta caspius, during alevin stage. Toxicol. Environ. Chem. 2010;92(5):903–914. doi: 10.1080/02772240903127284. [DOI] [Google Scholar]
  • 357.García-Huidobro M.R., Aldana M., Duarte C., Galbán-Malagón C., Pulgar J. Seawater-temperature and UV-radiation interaction modifies oxygen consumption, digestive process and growth of an intertidal fish. Mar. Env. Res. 2017;129:408–412. doi: 10.1016/j.marenvres.2017.06.013. [DOI] [PubMed] [Google Scholar]
  • 358.Abedi S., Sharifpour I., Mozanzadeh M.T., Zorriehzahra J., Khodabandeh S., Gisbert E. A histological and ultrastructural study of the skin of rainbow trout (Oncorhynchus mykiss) alevins exposed to different levels of ultraviolet B radiation. J. Photochem. Photobiol. B: Biol. 2015;147:56–62. doi: 10.1016/j.jphotobiol.2015.02.021. [DOI] [PubMed] [Google Scholar]
  • 359.Sayed A.E.-D.H., Ibrahim A.T., Mekkawy I.A.A., Mahmoud U.M. Acute effects of Ultraviolet-A radiation on African Catfish Clarias gariepinus (Burchell, 1822) J. Photochem. Photobiol. B: Biol. 2007;89(2):170–174. doi: 10.1016/j.jphotobiol.2007.09.010. [DOI] [PubMed] [Google Scholar]
  • 360.Fukunishi Y., Browman H.I., Durif C.M.F., Bjelland R.M., Skiftesvik A.B. Effect of sub-lethal exposure to ultraviolet radiation on the escape performance of Atlantic cod larvae (Gadus morhua) PLoS One. 2012;7(4) doi: 10.1371/journal.pone.0035554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.Singh M.K., Sharma J., Chakrabarti R. Impact of UV-B radiation on the physiology of freshwater carp labeo rohita larvae and evaluation of UV-B protective properties of seeds of achyranthes aspera and vitamin C. Agric. Res. 2013;2(2):166–171. doi: 10.1007/s40003-013-0060-z. [DOI] [Google Scholar]
  • 362.Häder D.P., Helbling E.W., Williamson C.E., Worrest R.C. Effects of UV radiation on aquatic ecosystems and interactions with climate change. Photochem. Photobiol. Sci. 2011;10(2):242–260. doi: 10.1039/C0PP90036B. [DOI] [PubMed] [Google Scholar]
  • 363.Mao R., Zeng D.-H., Zhang X.-H., Song C.-C. Responses of plant nutrient resorption to phosphorus addition in freshwater marsh of Northeast China. Sci. Rep. 2015;5(1):8097. doi: 10.1038/srep08097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Chong C.M., Lee P.-T., Rakus K., Wangkahart E. Editorial: the epidemiologic triads in aquaculture: host, pathogen and environment. Front. Immunol. 2023;14 doi: 10.3389/fimmu.2023.1305784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Ivshina I.B., Kuyukina M.S., Krivoruchko A.V., Tyumina E.A. Responses to ecopollutants and pathogenization risks of saprotrophic rhodococcus species. Pathogens. 2021;10(8):974. doi: 10.3390/pathogens10080974. https://www.mdpi.com/2076-0817/10/8/974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Ray S., Mukherjee S., Bhunia N.S., Bhunia A.S., Ray M. Emerging Pollutants in the Environment—Current and Further Implications. InTech; 2015. Immunotoxicological threats of pollutants in aquatic invertebrates; pp. 149–167. [Google Scholar]
  • 367.Cavalieri V., Spinelli G. Environmental epigenetics in zebrafish. Epigenet. Chromatin. 2017;10(1):46. doi: 10.1186/s13072-017-0154-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Marchione A.D., Thompson Z., Kathrein K.L. DNA methylation and histone modifications are essential for regulation of stem cell formation and differentiation in zebrafish development. Br. Funct. Genom. 2021;20(6):378–393. doi: 10.1093/bfgp/elab022. [DOI] [PubMed] [Google Scholar]
  • 369.Wang Y., Guo B. Adaption to extreme environments: a perspective from fish genomics. Rev. Fish Biol. Fish. 2019;29(4):735–747. doi: 10.1007/s11160-019-09577-9. [DOI] [Google Scholar]
  • 370.Roberts R.J., Agius C., Saliba C., Bossier P., Sung Y.Y. Heat shock proteins (chaperones) in fish and shellfish and their potential role in relation to fish health: a review. J. Fish Dis. 2010;33(10):789–801. doi: 10.1111/j.1365-2761.2010.01183.x. [DOI] [PubMed] [Google Scholar]
  • 371.Beinart R.A., Gartman A., Sanders J.G., Luther G.W., Girguis P.R. The uptake and excretion of partially oxidized sulfur expands the repertoire of energy resources metabolized by hydrothermal vent symbioses. Proc. R. Soc. B: Biol. Sci. 2015;282(1806) doi: 10.1098/rspb.2014.2811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Awad S.T., Hemeda S.A., El Nahas A.F., Abbas E.M., Abdel-Razek M.A.S., Ismail M., Mamoon A., Ali F.S. Gender-specific responses in gene expression of Nile tilapia (Oreochromis niloticus) to heavy metal pollution in different aquatic habitats. Sci. Rep. 2024;14(1) doi: 10.1038/s41598-024-64300-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 373.Sherif A.H., Farag E.A.H., Mahmoud A.E. Temperature fluctuation alters immuno-antioxidant response and enhances the susceptibility of Oreochromis niloticus to Aeromonas hydrophila challenge. Aquac. Int. 2024;32(2):2171–2184. doi: 10.1007/s10499-023-01263-9. [DOI] [Google Scholar]
  • 374.Lim M.Y.-T., Bernier N.J. Intergenerational plasticity to cycling high temperature and hypoxia affects offspring stress responsiveness and tolerance in zebrafish. J. Exp. Biol. 2023;226(16) doi: 10.1242/jeb.245583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Long Y., Li L., Li Q., He X., Cui Z. Transcriptomic characterization of temperature stress responses in larval zebrafish. PLoS One. 2012;7(5) doi: 10.1371/journal.pone.0037209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Lightner D. Special publication World Aquacult. Soc.; Baton Rouge LA: 1996. A Handbook of Pathology and Diagnostic Procedures for Diseases of Shrimps. [Google Scholar]
  • 377.Lupes S.C., Davis M.W., Olla B.L., Schreck C.B. Capture-related stressors impair immune system function in sablefish. Trans. Am. Fish Soc. 2006;135(1):129–138. doi: 10.1577/T04-198.1. [DOI] [Google Scholar]
  • 378.Kari Z.A., Kabir M.A., Dawood M.A., Razab M., Ariff N., Sarkar T., Pati S., Edinur H.A., Mat K., Ismail T.A. Effect of fish meal substitution with fermented soy pulp on growth performance, digestive enzyme, amino acid profile, and immune-related gene expression of African catfish (Clarias gariepinus) Aquaculture. 2022;546 [Google Scholar]
  • 379.Nya E.J., Austin B. Use of garlic, Allium sativum, to control Aeromonas hydrophila infection in rainbow trout, oncorhynchus mykiss (Walbaum) J. Fish Dis. 2009;32(11):963–970. doi: 10.1111/j.1365-2761.2009.01100.x. [DOI] [PubMed] [Google Scholar]
  • 380.Habte-Tsion H.-M. A review on fish immuno-nutritional response to indispensable amino acids in relation to TOR, NF-κb and Nrf2 signaling pathways: trends and prospects. Comp. Biochem. Physiol. B: Biochem. Mol. Biol. 2020;241 doi: 10.1016/j.cbpb.2019.110389. [DOI] [PubMed] [Google Scholar]
  • 381.Boyd C.E., Tucker C.S. Springer Science & Business Media; 2012. Pond Aquaculture Water Quality Management. [Google Scholar]
  • 382.Owusu Kwateng K., Darko J.E. Total quality management practices in aquaculture companies: a case from Ghana. TQM J. 2017;29(4):624–647. [Google Scholar]
  • 383.Bruno J.F., Selig E.R., Casey K.S., Page C.A., Willis B.L., Harvell C.D., Sweatman H., Melendy A.M. Thermal stress and coral cover as drivers of coral disease outbreaks. PLoS Biol. 2007;5(6):e124. doi: 10.1371/journal.pbio.0050124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Chiswell S.M. Colonisation and connectivity by intertidal limpets among New Zealand, Chatham and Sub-Antarctic Islands. II. Oceanographic connections. Mar. Ecol. Prog. Ser. 2009;388:121–135. [Google Scholar]
  • 385.Mugwanya M., Dawood M.A.O., Kimera F., Sewilam H. Anthropogenic temperature fluctuations and their effect on aquaculture: a comprehensive review. Aquac. Fish. 2022;7(3):223–243. doi: 10.1016/j.aaf.2021.12.005. [DOI] [Google Scholar]
  • 386.Sokolow S. Effects of a changing climate on the dynamics of coral infectious disease: a review of the evidence. Dis. Aquat. Org. 2009;87(1-2):5–18. doi: 10.3354/dao02099. https://www.int-res.com/abstracts/dao/v87/n1-2/p5-18/ [DOI] [PubMed] [Google Scholar]
  • 387.Masud N., Davies-Jones A., Griffin B., Cable J. Differential effects of two prevalent environmental pollutants on host-pathogen dynamics. Chemosphere. 2022;295 doi: 10.1016/j.chemosphere.2022.133879. [DOI] [PubMed] [Google Scholar]
  • 388.Wu H., Hou J., Wang X. A review of microplastic pollution in aquaculture: sources, effects, removal strategies and prospects. Ecotoxicol. Env. Saf. 2023;252 doi: 10.1016/j.ecoenv.2023.114567. [DOI] [PubMed] [Google Scholar]
  • 389.Descombes P., Wisz M.S., Leprieur F., Parravicini V., Heine C., Olsen S.M., Swingedouw D., Kulbicki M., Mouillot D., Pellissier L. Forecasted coral reef decline in marine biodiversity hotspots under climate change. Glob. Chang. Biol. 2015;21(7):2479–2487. doi: 10.1111/gcb.12868. [DOI] [PubMed] [Google Scholar]
  • 390.Harvell C.D., Mitchell C.E., Ward J.R., Altizer S., Dobson A.P., Ostfeld R.S., Samuel M.D. Climate warming and disease risks for terrestrial and marine biota. Science. 2002;296(5576):2158–2162. doi: 10.1126/science.1063699. [DOI] [PubMed] [Google Scholar]
  • 391.Misund A., Thorvaldsen T., Strand A.V., Oftebro T.L., Dahle S.W. Opportunities and challenges in new production systems for salmon farming in Norway—Industry perspective. Mar. Policy. 2024;170 doi: 10.1016/j.marpol.2024.106394. [DOI] [Google Scholar]
  • 392.Ahmed F., Kumar G., Soliman F.M., Adly M.A., Soliman H.A.M., El-Matbouli M., Saleh M. Proteomics for understanding pathogenesis, immune modulation and host pathogen interactions in aquaculture. Comp. Biochem. Physiol. D: Genom. Proteom. 2019;32 doi: 10.1016/j.cbd.2019.100625. [DOI] [PubMed] [Google Scholar]
  • 393.Ye H., Lin Q., Luo H. Applications of transcriptomics and proteomics in understanding fish immunity. Fish Shellfish. Immunol. 2018;77:319–327. doi: 10.1016/j.fsi.2018.03.046. [DOI] [PubMed] [Google Scholar]
  • 394.Bohara K., Joshi P., Acharya K.P., Ramena G. Emerging technologies revolutionising disease diagnosis and monitoring in aquatic animal health. Rev. Aquac. 2024;16(2):836–854. doi: 10.1111/raq.12870. [DOI] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available on request from the corresponding author


Articles from Comparative Immunology Reports are provided here courtesy of Elsevier

RESOURCES