Next Article in Journal
An Investigation of Parturient Ocular Appearance in Sows
Previous Article in Journal
Infrared Thermography as a Diagnostic Tool for the Assessment of Mastitis in Dairy Ruminants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Challenges of Vaccination in Fish Health Management

by
Avnish Kumar
1,
Sushil Kumar Middha
2,
Soumya Vettiyatil Menon
3,
Biswaranjan Paital
4,*,
Shyam Gokarn
3,
Meghana Nelli
3,
Rakshith Bangalore Rajanikanth
3,
Harish Mani Chandra
5,
Susithra Priyadarshni Mugunthan
5,†,
Sanwar Mal Kantwa
6,
Talambedu Usha
7,
Akshaya Kumar Hati
8,
Divyadharshini Venkatesan
5,
Abira Rajendran
3,
Tapas Ranjan Behera
9,
Swarupa Venkatesamurthy
3 and
Dipak Kumar Sahoo
10
1
Department of Biotechnology, School of Life Sciences, Dr. Bhimrao Ambedkar University, Agra 282004, India
2
Department of Biotechnology, Maharani Lakshmi Ammanni College for Women, 18th Cross, Malleswaram, Bangalore 560012, India
3
Department of Chemistry and Biochemistry, School of Sciences, Jain University, #34 JC Road, Bangalore 560027, India
4
Redox Regulation Laboratory, Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar 751003, India
5
Department of Biotechnology, Thiruvalluvar University, Serkkadu, Vellore 632115, India
6
Department of Zoology, B. S. Memorial P.G. College, NH 52, Ranoli, Sikar 332403, India
7
Department of Biochemistry, Maharani Lakshmi Ammanni College for Women, 18th Cross, Malleswaram, Bangalore 560012, India
8
Dr. Abhin Chandra Homoeopathic Medical College and Hospital, Homeopathic College Rd., Unit 3, Kharvela Nagar, Bhubaneswar 751001, India
9
Department of Community Medicine, Fakir Mohan Medical College and Hospital, Januganj Rd., Kalidaspur, Balia, Balasore 756019, India
10
Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
*
Author to whom correspondence should be addressed.
Present address: Amity Institute of Biotechnology, Amity University, Mumbai 410206, India.
Animals 2024, 14(18), 2692; https://doi.org/10.3390/ani14182692
Submission received: 27 April 2024 / Revised: 10 September 2024 / Accepted: 11 September 2024 / Published: 16 September 2024
(This article belongs to the Special Issue Current Advances in Vaccines against Fish Viruses)

Abstract

:

Simple Summary

About a 4% decline in fish production was recorded in 2020 in comparison to 2018 in inland aquaculture sectors. In 2014, about 10% of aquatic cultured animals were lost (costing global annual losses > USD 10 billion) due to infectious diseases. Contagious diseases generate several stresses, including oxidative stress in fishes that hampers their reproduction, growth, and production value. Aquaculture management needs to employ preventive measures against various contagious diseases for maintenance. Therefore, vaccination in fish, especially in broodstocks, is one of the most important measures to curb such fish losses. Here, we reviewed the current status of fish vaccines and the modern technology used to produce (alternative) fish vaccines, such as plant-derived vaccines for fish health management. Reliable aquaculture vaccines to increase economic productivity in aquaculture sectors are needed. Emerging contagious diseases in different fish species demand novel vaccines to be produced by adapting biotechnological approaches. Vaccination in broodstocks is important, and the fish vaccines are produced with whole-killed pathogens; subunits of a protein, peptide, or a recombinant protein; DNA vaccines; or live attenuated vaccines via different modes of administration. The use of adjuvants in fish vaccines needs to be researched.

Abstract

Vaccination is an essential method of immunological preventive care required for the health management of all animals, including fish. More particularly, immunization is necessary for in-land aquaculture to manage diseases in fish broodstocks and healthy seed production. According to the latest statistics in 2020, 90.3 million tons of capture fishery production was achieved from the aquaculture sector. Out of the above, 78.8 million tons were from marine water aquaculture sectors, and 11.5 million tons were from inland water aquaculture sectors. About a 4% decline in fish production was achieved in 2020 in comparison to 2018 from inland aquaculture sectors. On the other hand, the digestive protein content, healthy fats, and nutritional values of fish products are comparatively more affordable than in other meat sources. In 2014, about 10% of aquatic cultured animals were lost (costing global annual losses > USD 10 billion) due to infectious diseases. Therefore, vaccination in fish, especially in broodstocks, is one of the essential approaches to stop such losses in the aquaculture sector. Fish vaccines consist of whole-killed pathogens, protein subunits, recombinant proteins, DNA, or live-attenuated vaccines. Challenges persist in the adaption of vaccination in the aquaculture sector, the route of administration, the use of effective adjuvants, and, most importantly, the lack of effective results. The use of autogenous vaccines; vaccination via intramuscular, intraperitoneal, or oral routes; and, most importantly, adding vaccines in feed using top dressing methods or as a constituent in fish feed are now emerging. These methods will lower the risk of using antibiotics in cultured water by reducing environmental contamination.

1. Introduction

The rate of infectious diseases is higher in animals, which is very dangerous, as they may affect humans [1,2,3,4,5]. Some infectious agents and their causative diseases are explained in Table 1. To develop immunity in animals (including fish) and reduce the susceptibility of animals to communicable diseases, vaccination is crucial. Thus, vaccines have a significant role in managing live stocks (Figure 1). Vaccines have been applied as preventive measures to reduce the chance of diseases such as rabies and rinderpest, as well as other zoonotic ones that can be endemic [2,3,4,5,6,7,8,9,10,11,12,13,14].
Even though vaccines are available for animals based on the type of disease, prioritizing them is necessary. Irrespective of fish, seaweed, and crustaceans being vital food sources that provide protein, the aquaculture industry suffers from bacterial diseases, viruses, parasites, and fungi. Fish vaccines are limited in quantity and are rendered inactive by factors such as temperature, pH, and organic waste. Despite thorough investigation, only a limited number of authorized vaccines and antivirals demonstrate efficacy against viral infections [15]. Research on fish immunity mainly relies on vaccination, gene/protein sequencing, antigen screening, vaccine administration, and enhanced aquatic immunizations [16,17]. Numerous vaccines have been developed, such as live vaccines and recombinant vaccines, to prevent these infectious diseases, but still, the recovery rate from contracting the diseases is meager [15,18]. For example, commercial vaccines LF-89 and EM-90 of P. salmonis may not show protective efficacy [19]. TiLV (tilapia lake virus) vaccination can cause the virus to return to a virulent condition, retain some virulence, or cause virulence in fish with weakened immune systems [20,21]. Nevertheless, a wide range of commercial vaccines are available according to animal infectious disease types and infection rates (as given in Table 1). Still, the availability of such vaccines in fish may be able to curb the contagious diseases that affect their production and reproduction value (Figure 2).
Total fish weight is estimated to double in India, Mexico, and Brazil by 2050, as stated by the FAO’s food balance sheets, and the problems of total fish demand for the top 10 countries ranked in order of human consumption is expected to be solved by that time [21,22,23,24,25,26]. The controlled production of aquatic organisms such as fish, algae, mollusks, and other aquatic plants is known as aquaculture or aquafarming. In contrast to commercial fishing, which includes wild fish capturing, aquaculture involves raising populations of freshwater, brackish water, and saltwater species in controlled or semi-natural environments. However, their production is hampered severely by several (infectious) diseases.
Aquaculture ecosystems can naturally be home to many diseases, which have adverse economic effects on productivity. Diseases are also can be contagious in oceanic ecosystems. In hosts, different levels/percentages of infections occur; for example, 49% of fish, 21% of crustaceans, 28% of mollusks, and 1% of echinoderms are susceptible to various diseases [27,28]. Exotic fish may become more susceptible to diseases in new environmental conditions than local wild varieties. Opportunistic infections may impair the aquaculture sector due to high stocking densities of monoculture systems, leading to greater host contact with pathogenic organisms, which in turn cause increased stress for the fish and degrade the water quality [27,28]. The main objective is to prevent the occurrence of diseases in aquaculture, and this objective can be met by maintaining good water quality, moderating stocking density, producing disease-resistant, genetically modified stocks, using immuno-stimulants and therapeutics against fish pathogens, and utilizing vaccines against bacteria, viruses, and other pathogens in fish [27,28].
A vaccine can be a suspension of weakened or killed microorganisms, toxins, or any other biological preparation that consists of a protein or a polysaccharide antigen to induce a protective immunological response against the infecting pathogen [29]. Currently, three routes of administration of vaccines to fish, such as oral vaccines, are incorporated into the feed. Immersion vaccines are applied by dipping the fish for a short time in a concentrated solution of vaccine, and injection vaccines, as the name suggests, are injected into the fish. This current review attempts to update information on fish vaccines and their efficacy and prospects using a systematic literature survey [27,28,29]. It is to be noted that different commercial veterinary vaccines available at present, such as canine (Canileish Leishmania), avian (Trovac Flu), equine (Proteq Flu), salmonoid (Apex IHN), food (iPED+), and feline (PureVax rabies) vaccines, are available currently [30].

2. Literature Review Process

This study systematically reviews the literature on fish vaccines, utilizing a comprehensive search conducted across PubMed, AGRICOLA, Google Scholar, Scopus, Science Direct, and Web of Science. Search terms included “vaccines”, “fish vaccines”, and “fish”. The review encompassed both peer-reviewed and respected non-peer-reviewed English-language sources. The review examined English-language peer-reviewed publications, books, journals, and credible websites. Due to their enormous biomedical and scholarly collections, PubMed/MEDLINE and Google Scholar were used as primary sources. AGRICOLA, with 6 million records in agricultural and biological sciences, was especially important. The search technique adhered to current review procedures and included extra phrases such as “disease”, “immunity”, and “economic loss”. From AGRICOLA, 10,000 publications on “fish”, 1553 on “vaccine”, and 26 on “fish vaccines” were examined. PubMed returned 4018 hits, whereas Google Scholar returned nearly 489,000, all of which were filtered for relevance. The data were divided into subsections covering many areas of fish vaccines, such as the condition of aquaculture, present fish illnesses, and future possibilities [25,31,32]. Of the first findings, 187 articles were selected based on their direct relevance to fish vaccinations. The evaluation process examined the origins and development of animal and fish vaccines and their efficacy, commercialization, and influence on fish immunity, illness prevention, and economic value.
These publications were selected, categorized, and analyzed to offer a thorough overview, emphasizing the importance of fish vaccines and their future potential in aquaculture.

3. Need for Fish Vaccines

3.1. Status of Aquaculture Products

Fish are essential for ecosystem balance, economic contribution, water pollution prevention, disease control, and food sources [27]. Fish are also an essential source of byproducts like fish oil, liver oil, fish protein, fish flour, fish roes, fish silage, fish fertilizers, isinglass, etc. However, they face threats like climate change, habitat loss, overfishing, illegal fishing [33], ocean pollution, and disease. It is essential to protect them from these threats, and, in this concern, vaccination strategies must be developed to safeguard their existence.
According to the FAO, [25] world fisheries and aquaculture production increased from 110.7 metric tons to 178.9 metric tons in the years from 1990 to 2018 but slightly decreased the production of aquaculture and fisheries in the year 2019 (1.5 metric tons) and to 1.1 metric tons in 2020. Human consumption of aquaculture and fishery products increased from 86.6 metric tons to 158.1 metric tons from 1990 to 2019 but reduced in 2020 to 0.7 metric tons. The trade (export) of fisheries and aquaculture also increased from 1990 to 2018, from 39.6 metric tons to 66.8 metric tons, but decreased to 0.2 metric tons in 2019 and to 6.8 metric tons in 2020. The unusual COVID-19 outbreak caused lockdowns and market, port, and border closures worldwide. Commerce plummeted, disrupting aquatic food production and distribution and causing unemployment and livelihood losses. Furthermore, the “UN Decade of Ocean Science for Sustainable Development (2021–2030)” identifies three key challenges to reduce global biodiversity losses: involving diverse stakeholders, ensuring adequate resources, and developing a transparent monitoring system for progress [25,33].
India is the second largest fish-producing country after China. China produces 58.8 million metric tons (one-third of total world fish production), while India produces 9.46 million metric tons annually, although the aquaculture industry was established in the 19th century in India [34]. According to the Ministry of Fisheries, Animal Husbandry and Dairy, Government of India, fish production increased from 6,305,000 metric tons to 14,164,000 metric tons in just 16 years, i.e., from the year 2004 to 2020 [34].
Contagious and infectious diseases are still a significant threat to the development of the aquaculture industry in the world. Fish are vulnerable to many bacterial, viral, fungal, protozoan, and parasitic diseases like red pest, fungus, tuberculosis, argulus, velvet or rust, anchor worm, costia (Ichthyobodo), lymphocytes, etc. Some communicable diseases, along with their associated pathogens, symptoms, and treatments, are given in Table 2.

3.2. An Emergency Need for Fish Vaccines

Aquatic foods like sea plants, fish, and crustaceans are one of the largest sources of protein for humans. However, the aquaculture industry faces substantial economic losses due to infectious diseases. In aquaculture, bacteria account for more than 50% of infectious diseases, followed by viruses, parasites, and fungi [51]. These diseases impact fish internally and topically, including their gills, fins, and body surfaces. In general, fish farmers have used a variety of strategies such as biosecurity controls, better settings, and feeding the fish special diets to manage the diseases that affect fish. The use of disinfectants, the addition of antibiotics, and treatment with pharmaceuticals like therapeutic treatments are a few examples of synthetic methods [25]. Although antibiotics and chemotherapeutics have been used to treat diseases, there are some downsides, such as problems with drug resistance and consumer safety concerns [52,53,54,55]. Successful vaccination delivery and design are crucial, and these depend on aquatic species’ immunological responses, as well as proper dosing. Aquaculture vaccine research and deployment require particular vaccine-delivery methods with regard to vaccine technology, aquatic organism species and their reproductive stages, pathogen nature, infection pathways, and cost [56]. Nevertheless, a few fish vaccines are being studied and are available in the literature (Table 3).
In Table 3, we can see that the majority of vaccines are produced by inactivating bacterial and viral cultures, which are injected intra-peritoneally for emulsion-based injectable vaccines and intramuscularly for DNA plasmids [62], which produce higher protection rates against diseases for fish. However, using an injection to administer the vaccines in fish, even fingerlings, is tricky. Even with the automation of the vaccination process in fish, they still experience stress. However, economically important fish can be vaccinated [63]. Environmental conditions such as pH, the presence of organic matter, and temperature can impact a drug’s efficacy [64,65]. Viral infections are more challenging to control due to the lack of antivirals or efficient viral vaccinations [66]. Only a few viral vaccines have acquired licenses, even though companies and academic institutions have carried out numerous research investigations to create viable viral vaccines [67].
Vaccination is an effective method to improve the immune response and is cost-effective for aquaculture, but it should be developed to overcome the hurdle mentioned earlier. Aquatic vaccines will likely foresee advancement in the following years because of increased technological inventions like sequencing, antigen screening, the development of fish cell lines, the development of innovative vaccine protein expression and delivery systems, and increased fundamental knowledge about fish mucosal immunity.

4. Developments of Commercially Available Vaccines and Experimental Available Vaccines

A shortage of anti-viral medicines, efficient viral vaccinations, and illness resistance makes viral infections harder to treat. Unlike bacterial infections, viral illnesses in animals or fish have less effective treatment systems [60]. Therefore, the vaccination of fish, particularly broodstocks, is one of the most important ways to reduce losses in the aquaculture business. This is because the broodstocks of aquatic animals are used to produce and supply seeds from that species for most aquaculture systems [68]. Therefore, healthy and immunized seeds can be obtained from such immunized broodstocks.
This section provides current research on protein-based, live-attenuated, virus-like particles, bacterins, DNA fish vaccines, etc. These topics cover present knowledge of fish vaccine administration methodologies, effective adjuvants, and obstacles in adapting vaccines for the fishing industry, including studies on mucosal immunity. Novel techniques encompass intramuscular, intraperitoneal, oral, and feed-based autogenous vaccines. Furthermore, bioinformatics and in silico testing have accelerated the vaccine development process. Vaccination practices have been increasing in the aquaculture business.
Due to various legal guidelines, several farms have been routinely vaccinating their fish, which, when administered appropriately, provides a high level of protection. Approximately 30 vaccines were reported to offer protection for fish against significant bacterial and viral illnesses, meeting the need for continuous aquaculture despite the presence of disease-causing agents and environmental pressures [69]. With regard to enteric red mouth and vibriosis, the concept of immunizing fish on a commercial scale has finally been realized (Table 1, Figure 1 and Figure 2).

4.1. Early Works on Fish Vaccination

Fish were immunized for the first time successfully via an oral vaccine in carp and trout against Aeromonas punctata in 1938 and Aeromonas salmonicida in 1942, respectively [70]. The vaccinations against enteric red mouth disease (ERM), also known as yersiniosis and vibriosis (and sometimes only referred to as yersiniosis), were the first bacterial vaccines to be made available for commercial purchase in the United States in the late 1970s [71,72]. Bioveta, a Czechoslovak business, created the first viral immunization for fish in 1982 [70,71,72,73]. The vaccine was created by the fusion of two deactivated spring viremia of carp SVC virus strains in oil, followed by injection. The vaccination included no live virus. The only carp vaccine currently in the market for commercial sale in Asia is an inactivated injection that protects against grass carp bleeding disease caused by the reovirus. The manufacturer of a vaccine for koi herpesvirus for purchase in Israel uses an attenuated strain of CyHV-3 [70]. The history, types, and modes of administration of vaccines in fish are different for different vaccines [72]. Key researchers in this area have identified the application of adjuvants and immunostimulants in fish vaccines and their delivery methods. The data were obtained based on alternative methods (other than injection) for vaccine delivery and the protective efficacies of traditional and promising new-generation adjuvants [74,75].
Infectious illnesses have a negative effect on aquaculture practices all around the world. Pathogenic illnesses in aquaculture in India lead to considerable financial challenges and threaten the survival of fish populations. Antibiotic resistance patterns, residual effects, and environmental impact have all increased the need for establishing appropriate alternate preventive methods [76,77,78,79,80,81,82,83,84]. Vaccinations have been shown to be an effective technique for disease control, allowing for increased fish productivity. Aeromonas hydrophila is a widely spread pathogen in all farmed fish species in India. Most vaccine research has focused on bacterial illnesses, including Aeromoniasis and Edwardsiellosis. Despite significant efforts and potential pilot programs for developing vaccines against several parasitic and fungal infections, India’s aquaculture sector has no universally applicable immunizations. Indian researchers are attempting to develop a vaccine to protect the fish business, listing the problems associated with manufacturing and exploring the opportunities of earning money from such vaccines [53]. Tafalla et al. [75] defined and listed the evolution of commercially available fish. More than 26 licensed fish vaccines are available commercially and globally at present [17]. Vaccines have been approved for use by the United States Department of Agriculture (USDA) for diverse species of aquaculture, and most of them are produced by conventional methods that target pathogens via culture testing. This collection of vaccines has successfully protected fish against some severe fish diseases up to an extent.

4.2. Properties of Fish Vaccines and the Vaccination Process

Fish vaccinations provide protection against diseases and should be ecologically safe for both fish and their surrounding ecosystems, economically viable for large-scale production, easy to administer, and capable of eliciting a robust immune response in delicate species, with few adverse effects, if any [70,71,72,73,74,75]. Molecular research demonstrated that immunoprophylactic approaches in grass carp stimulate viral nucleic acid sensors, TLRs, HMGBs, RLRs, and PRRs, enhancing virus resistance [84]. Significantly, these approaches may be improved to provide the highest level of security. Toll-like receptors (TLRs) detect pathogen-associated molecular patterns (PAMPs) and initiate immunological signaling cascades that enhance innate immunity and adaptability [70,71,72,73,74,75]. TLR adjuvants and activators can enhance the immunization of fish and other aquatic animals [70].

4.3. Types of Fish Vaccines

Fish vaccines can be protein, live-attenuated vaccines, virus-like particles, bacterines, and DNA vaccines. Challenges remain in the fishing industry regarding the adaptation of vaccination, including the method of administration, the use of effective adjuvants, and, most importantly, the lack of data from more focused research on the mucosal immunity mechanism [15,85]. The use of autogenous vaccinations, which include intramuscular, intraperitoneal, and oral vaccinations and, most significantly, the incorporation of vaccines in feed via top dressing methods in feed or as a constituent in fish feed, are all examples of emerging practices [70,75].

4.3.1. Whole-Cell Vaccines

Whole-cell vaccines that have been killed or inactivated can be characterized as a suspension of pathogens that have been destroyed (either by heat or chemical treatment) but still provide protection when administered to the host. These vaccines are cheap to make. Pathogens such as Vibrio anguillarum, Vibrio salmonicida, Vibrio ordalli, Yersinia ruckeri, and Aeromonas salmonicida are combated by applying these vaccines to fish [86,87] (Table 4). These killed vaccinations are commercially available as formalin-inactivated whole-cell vaccines, which can be given with or without adjuvants. Vaccines against deadly fish viruses, including infectious pancreatic necrosis virus (IPNV), infectious hematopoietic necrosis virus (IHNV), viral hemorrhagic septicemia virus (VHSV), and spring viremia of carp virus (SVCV) have been produced. When the inactivated IPNV was injected into rainbow trout fry, the fish were well protected [88], and when it was given to brook trout with Freund’s complete adjuvant, the fish obtained a robust humoral response, but this did not stop them from spreading the virus [89,90]. Formaldehyde is used to sterilize fish. Although vaccination with inactivated vaccines effectively prevents the spread of diseases, this method is deemed unsuitable for some viruses because of the age at which symptoms first appear. The humoral immune response is induced by killed vaccines and, to maintain protection, booster dosages are required. The primary drawbacks of heat-killed vaccines are the high cost of cell culture manufacture, difficulty of purification, and limited administration options (Table 4).

4.3.2. Attenuated Vaccines

Attenuated vaccines are genetically or chemically weakened microbes that can produce short-lasting immune responses in the host [52]. They contain live bacteria and viruses that no longer cause illness.

4.3.3. Recombinant Vaccines

Recombinant vaccines are those that use only the immunogenic areas of the pathogen to be produced in a heterologous host and are then utilized as vaccines [17]. Additionally, recombinant vaccines can be genetically modified organisms (bacteria or virus) with reduced pathogenicity, thus being recombinant and attenuated to produce vaccines.

4.3.4. Synthetic Peptide Vaccines

Synthetic peptide vaccines can act as subunit vaccines, antigenic sites, or peptides that have been tested to induce antibodies against infectious pathogens such as nodavirus, rhabdovirus, birnavirus, IHNV, IPNV, and VHS [17].
Among various vaccines for fish, mRNA vaccines are in colossal demand compared to conventional vaccines. Live-attenuated vaccines (LAVs) successfully reduce diseases, but their safety and efficacy are still doubtful. The mRNA vaccine, on the other hand, is non-infectious; hence, it is safer.

4.3.5. DNA Vaccines

DNA vaccines containing plasmids that carry a pathogen’s antigen have garnered attention as a potential way to boost fish defense against illnesses [89]. Aquaculture researchers have boosted VHSV and IHNV resistance by injecting viral genes expressing surface glycoproteins intramuscularly (IM). VHSV glycoprotein and DNA immunization elicited an immune response in rainbow trout [17].

4.3.6. Mucosal Vaccinations

Mucosal vaccines against pathogenic infections are being developed because they can elicit protective responses at mucosal surfaces by preventing pathogen reproduction [90]. Oral, immersion, and nasal teleost fish mucosal vaccines stimulate B and T cells to induce systemic and mucosal responses. It has been shown that mucosal vaccines work if they imitate the natural route of infection, such as going through mucosal surfaces [74]. For example, rainbow trout (Oncorhynchus mykiss), a bony fish renowned for its specialized mucosal immune system, was studied by Magadan et al. [76]. The olfactory organ of this fish contains nasopharynx-associated lymphoid tissue (NALT), distinguished by a widespread network of myeloid and lymphoid cells. Myeloid and lymphoid cells activated by the nasal immunization showed distinctive dynamics of IgM and IgT repertoires at systemic and mucosal locales, demonstrating an unusual capacity to induce splenic Ig responses.

4.3.7. Plant-Based Edible Vaccines

Plants might provide an inexpensive foundation for creating cost-effective, edible, live-attenuated, pathogen-free vaccinations that can reduce several pathogenic fish illnesses to maintain a sustainable aquaculture. Plant-based vaccines may minimize the frequency of live-attenuated virus or bacterial booster doses. Modern plant researchers suggest that plant cells could express mammalian proteins after cloning [77]. Specific plant cell cultures or whole plants can be used as expression hosts. To express mammalian genes in plants, cloned complementary DNA must be spliced with a plant promoter, terminator, or regulatory region. Selectable markers help to identify recombinants [78]. Plant-based vaccines would be easy and convenient to consume orally. Follicle-associated epithelium M cells will grab the protein unit to activate mucosal immunity [74,78].

4.3.8. Nanoparticle-Based Vaccine

Nanoparticles provide tailored vaccination delivery, antigen stability, and adjuvant effectiveness. Vaccinating particular body parts with these systems might prevent disease transmission. Nanoparticles can increase weak antigen immunogenicity and release kinetics, stability, and targeted dispersion over traditional adjuvants. Oral immunization is preferred in aquaculture since each fish in a pond, especially small fish, cannot be injected with vaccines. Maintaining gastrointestinal stability is difficult with oral vaccines. Nanoparticles may solve this problem [79]. Alginate, chitosan, and PLGA have different chemicals mixed with antigens and immunostimulants to enhance delivery and immune responses.

4.4. Advancements in Vaccine Development

New biotechnology developments have enabled the quick production of vaccines against various pathogens. In bioinformatic approaches, software predicts the gene to produce recombinant proteins functional for vaccines [52]. Andreoni et al. [80] used this software-based approach to investigate vaccines against intracellular infections, such as Flavobacterium columnare and Edwardsiella tarda in Photobacterium damselae subsp. piscicida fishes. These cause the most common significant diseases of fish columnaris and Edwardsiellosis, respectively [90,91]. Both an in silico examination and a functional analysis could lead to the conclusion that an effective candidate gene exists, which could be prioritized to develop a vaccine to prevent a disease’s further spread. This would undoubtedly reduce the time required to develop a novel vaccine. There are approximately 30 vaccines available that have the potential to protect fish against the most lethal bacterial and viral diseases.

5. Contemporary Need for Vaccines in Fish, a Growing Field of Research

Aquaculture production was estimated to be 90.3 million tons worldwide, with a decline of 4% in 2020–2021 [25]. Etiological diseases in fish and their treatment methods are always challenging. The immunization of fish is necessary to combat typical diseases and maintain their renewable production for sustainability in aquaculture. The spreading of any infectious diseases in fish may be multiplied by environmental stress caused by pollutants. One of the species affected due to such stress is shrimp. A total loss of one billion US dollars was reported by Briggs et al. [91]. Infectious agents or parasites, such as bacteria, viruses, algae, etc., are generally present in water. Some of the primary etiological agents and infected species of fish are shown in Table 5 [92].
Reviews have focused on various vaccines for fish, among which mRNA vaccines are in huge demand compared to conventional vaccines [99,100]. Live-attenuated vaccines (LAVs), either commercially available or under experimental conditions, are noted to be successful in reducing diseases. Some examples are the Edwardsiella ictaluri vaccine (against enteric septicemia in catfish), arthrobacter vaccine (against bacterial kidney disease in salmonids), and Flavobacterium columnare vaccine (against columnaris infection in catfish). Nonetheless, the safety and efficacy of certain vaccines remain uncertain. The mRNA vaccine, on the other hand, is non-infectious; hence, it is safer [99,100]. Other types of vaccines used are nanoparticulate vaccines (nanogels, micelles) and liposomal vaccines (natural or synthetic lipids), which combat infectious diseases [101,102].
Some vaccines available for infectious diseases are inactivated (IPNV, ISAV), nucleic acid (DNA, RNA), and live-attenuated vaccines. DNA vaccines have been studied to be effective against viral infections and have been developed for a series of pathogens in water [103,104]. Even though numerous types of vaccines have been explored, tested, and controlled successfully for many diseases, enormous challenges persist in delivering effective immunization in fish. Disease outbreaks, mitigatory species of fish, anthropogenic activities, and morbidity rate increases are the challenges faced by farmed and traded aquatic species. Some activities, such as mining and construction works in nearby areas, lead to viral infections in marine ecosystems. Moreover, the consumption of infected fish, used as a protein source for farm fish, results in the infection of farm fish species. Another major drawback is the lack of disease diagnosis tools and surveillance measures, especially in underdeveloped countries [105,106,107]. Different management strategies and policies should be developed to manage aquaculture trading and diseases. A multifaceted approach must be designed to prevent further emerging etiological agents and the infectious diseases caused by them (Table 5).

6. Fish Vaccine Production against Various Pathogens

The discovery and subsequent production and commercialization of vaccines against infectious diseases in fish are commercially unavailable. These processes use multiple sources and steps. Each vaccine undergoes the steps of identification of the pathogen/disease, process development, production, validation with multiple individual fish, documentation, and vaccine licensing for commercialization (Figure 3). Various sources are used to achieve these steps.

6.1. Nocardiosis

Nocardiosis, caused by Nocardia seriolae, was first discovered in Japan’s central region [108]. It swiftly expanded to the country’s western areas, resulting in widespread Seriola fish mortality. Nocardiosis initially did not have a significant negative economic impact but is now considered a major cause of mortality in fish [109]. Although Seriola fish become easily infected by Lactococcus garvieae, nocardiosis has replaced it as the disease that causes the most economic harm to Japan’s aquaculture sector due to the development of a vaccine to control the bacteria [110]. Nocardiosis is distinguished by tubercles on the kidney and gills, as well as ulcers on the body’s exterior [111,112]. The creation of a vaccination against this illness is eagerly awaited. The administration of Freund’s incomplete adjuvant or formalin-killed cells of N. seriolae generated a humoral immune response, but the vaccine does not have a protective role against infectious diseases [113]. Numerous studies have documented that N. soli, N. fluminea, and N. uniformis could be vaccinated using live vaccines, such as in N. seriolae, a “codon-optimized antigen-85- like gene” [114]. Kato et al. [114] insisted on “DNA vaccines encoding the codon-optimized antigen-85-like” genotype of N. seriolae and malin-killed cell vaccine augmented with hybrid interleukin-12.

6.2. Bacterial Hemolytic Jaundice

It was documented that bacterial hemolytic jaundice in farmed yellowtails causes a 5–20% overall loss in cultured fish, like the Japanese amberjack population [115]. The hemolytic activity of the bacteria causes the skin and muscle tissue of the infected fish to turn yellow [116]. Similar infection may also occur in natural fish population [117]. The illness initially surfaced in the 1980s and has since spread rapidly over western Japan [16,118]. In addition to the losses caused by nocardiosis, a silent infection in yellowtail kingfish, the illness also results in considerable economic losses, as it affects a large number of fish before shipping. I. seriolicida exhibits low pathogenicity for greater amberjack and yellowtail kingfish [119].
Matsuyama et al. [117] reported that those fish that survive against naturally occurring contagions with Ichthyobacterium exhibit strong immunity and antibody-mediated death of the pathogen. Even though the bacterial serotype has not been identified, molecular epidemiological studies have shown that Ichthyobacterium populations are clonal populations with few variations. Furthermore, if low-cost growth techniques can be created and bacterial quality is steady, it would be possible to develop an effective vaccine. Takano et al. [119] sequenced Ichthyobacterium seriolicida isolated from Seriola quinqueradiata. This genome data of JBKA-6T sp. is being used in research to create a recombinant vaccine [119].

6.3. Bacterial Coldwater Disease

Fish that live in cold, fresh waters with temperatures of 16 °C or lower are susceptible to one of the bacterial diseases known as coldwater disease (CWD). Flavobacterium psychrophilum causes bacterial CWD [120] and rainbow trout fry disease [121]. In 1990 and 1993, Japanese researchers documented the presence of bacterial CWD in farmed coho salmon and natural sweetfish, respectively [122,123]. It has significantly harmed the abundance of sweetfish in numerous rivers. Genomics, serotypes, and lethal studies indicated different strains of F. psychrophilum in sweetfish and other species [124,125], and this has led to the indication of their immuno-compromised state under infection [126,127].
Antiserum obtained from bacterial CWD-infected fish used for passive immunization helps to battle illness and can be used to develop an efficient vaccine [128]. The use of an oil-based adjuvant, formalin-killed cell vaccine, has also demonstrated a protective effect [129]. This oil-based adjuvant takes at least two months to become completely washed off from the bodies of sweetfish. Since sweetfish are annual and all their organs are edible, this long-term residual adjuvant vaccine is unsuitable [130]. Further developments of a water-soluble adjuvant and its effectiveness with a short residence period have also been demonstrated [131,132,133].
Unfortunately, the adjuvant’s toxic properties prevented the vaccine from being marketed. No permanent impact has been attained despite studies on enteric micro-capsulated oral vaccinations and oral vaccines employing formalin-killed cells from logarithmic bacterial colonies [132]. Researchers have focused on recombinant vaccines of antigenic proteins against F. psychrophilum in sweetfish [133]. Although these antigenic proteins are promising candidates in the creation of bacterial CWD vaccines, further research is required to improve the efficacy of these vaccines [134].

6.4. Erythrocyte Inclusion Body Syndrome

The etiologic agent of EIBS in Oncorhynchus kisutch is porcine orthoreovirus 2 [135]. The illness strikes when the water temperature is below 10 °C, resulting in anemia and widespread deaths. PRV-1 is shown to be linked to PRV-2 [136,137] and PRV-3 [138], which are infections that cause HSMI in Salmo salar and resemble HSMI in rainbow trout [139]. Although there have been no serious attempts to culture porcine orthoreovirus 2, a DNA-based genetic approach may help to develop a vaccine. Haatveit et al. [140] studied the PRV-1 DNA vaccine in experimental fish, which showed a moderate protective effect. A recombinant vaccine might work for EIBS due to the correlation between PRV-1 and PRV-2.

6.5. Parasitosis

In Japan, farmed marine fishes are plagued by the “skin-parasitic capsalid monogeneans, Neobenedenia girellae and Benedenia seriolae”. The latter has strong host specificity for Seriola species, while the former parasitizes various fish species and exhibits moderate host specificity [141]. Even though praziquantel oral administration and bath treatments with freshwater or hydrogen peroxide solutions successfully control the parasite, they involve significant cost or work [142]. In numerous marine fish species, including the Japanese flounder (Paralichthys olivaceus), the ciliate Miamiensis avidus causes scuticociliatosis [143]. No effective treatment has made a vaccine compulsory for scuticociliatosis infection in several countries, like Malaysia and Japan [144]. The death rate was reduced after infections with ciliate to P. olivaceus immunized with formalin-killed peracetic M. avidus [143]. However, a vaccination study with Philasterides dicentrarchi or M. avidus in Spanish turbot indicated their effective protection when used in vaccine form [145,146]. Therefore, three different M. avidus or P. dicentrarchi serotypes are used in Malaysia and Japan [144].
Surface antigens implicated in M. avidus immobilization have been identified as highly antigenic, short, linear peptide fragments of proteins that could be associated with three serotypes [146]. As a result, it is believed that a combination of different immobilized M. avidus serotypes or polypeptides generated from different serotypes are potential antigens for vaccines.

6.6. Vaccines Other Than Inactivated Vaccines

Eel, Japanese flounder, and red sea bream aquaculture in Japan suffer commonly from Edwardsiellosis [147]. It was already established that cultured yellowtail and greater amberjack are negatively impacted by infectious diseases brought on by Mycobacterium spp. and N. seriolae [148,149,150].
Mochizuki et al. [151] specified that the nucleotide of IHNV isolates mutates quickly. It modulates its virulence capacity in rainbow trout farms, as observed in countries such as Japan and Malaysia. Various scientists worldwide are working to develop preventative measures against these intracellular infections. Edwardsiella, Mycobacterium, N. seriolae, and IHNV are infections occurring within a cell. To prevent these intracellular infections, a live-attenuated vaccine or a cytokine adjuvant-based vaccine has been established [113,152]. A DNA vaccine for IHNV infections has been approved against intracellular pathogens of fish in the USA and Canada. The rules for introducing additional vaccinations in fish have not yet been studied. In Japan, all known licensed fish vaccines are inactivated ones. Therefore, it is essential to build an administrative structure that can integrate live-attenuated or DNA-based vaccines into Japanese aquaculture and confirm their safety and efficacy [153]. Major disease prevention could be achieved using complete vaccination protocols in aquaculture. Major vaccination is used in the commercial aquaculture of species like barramundi, rainbow trout, Atlantic salmon, sea bass, sea bream, tilapia, turbot, and yellowtail. Currently, available vaccines for commercial aquaculture are listed in Table 6.
Additionally, experimental vaccinations are used to prevent illnesses, including Vibrio harveyi infection in barramundi and Photobacterium damsela subsp. damsela infection in salmonids, piscirickettsiosis, and bacterial kidney disease in salmonids, as well as Flexibacter maritimus infection in turbot. However, non-licensed vaccines remain utilized in industrial fish farms. Many approved bacterial vaccines are inactivated products, and only a small number of recombinant vaccine technologies have been applied thus far. Typically, intraperitoneal injections of multivalent vaccinations are used to immunize salmonid fish. Although booster vaccination is increasingly used predominantly in the Mediterranean province, immersion vaccination remains the standard immunization method for marine fish species [64,156,157].

6.7. Autogenous Vaccines

Japan forbids the use of autogenous vaccinations in aquaculture. Inactivated immunological VMPs made from pathogens and antigens taken from an animal or animals at a particular facility and used to treat that animal or those animals in the same area are known as autogenous vaccines [158]. A publicly funded facility working on a specific disease can produce an autogenous vaccine faster than a commercial vaccine, which can be created and granted a license [159] (Figure 2). Therefore, when there are no reports of effective commercial fish vaccines, these autogenous vaccines prepared from the pathogen strains in a precise fish farm may provide protection. Various diseases and their variations can typically arise when cultivating different fish species. The advancement of generating a fish vaccine may be appropriately aided by introducing autogenous vaccines in nations like Japan, where various fish species are cultured [160,161,162,163]. Therefore, more research on fish vaccines is now needed.

7. Plant-Derived Fish Vaccines—A New Perspective in Immunology

Plant molecular farming is an emerging expression system through which recombinant genes are expressed in plant cells. In this technique, foreign proteins are expressed in plants with the purpose of using only the protein rather than the plant [164]. In the late 1980s, attempts were made to express recombinant proteins in plants [165,166]. Initially, plants were used to express pharmaceutically essential proteins. In 2012, a plant-produced enzyme, Glucocerebrosidase, was the first plant-expressed product commercially available for human use [167]. Since then, numerous research has been conducted to produce human and animal recombinant proteins in plants. Three different platforms are used to express the recombinant protein in plants: stable nuclear expression, stable expression of transgenes in chloroplasts, and transient expression of transgenes [168] (Figure 3). Each approach has its benefits and drawbacks, and the choice of platform is mainly determined by the amount of required protein.
Currently, good manufacturing practices and regulatory concerns with plant-made recombinant proteins have also been thoroughly developed. Plant molecular farming may utilize a wide range of plant species. In lettuce, tomato, potato, cabbage, and other edible plants, nuclear and plastid genome engineering has been used to produce a variety of recombinant proteins. It has been suggested that a chloroplast expression system is a promising method of producing oral vaccinations. Vaccines against viruses, bacteria, and parasites have been made using transgenic plants [168].

7.1. Advantages of Plant-Derived Fish Vaccines

Plant-derived vaccines have been used for their multi-fold advantages such as the following: (1) the plant expression system is safe, and the vaccines do not have any toxic metabolites and hyperglycosylated proteins; (2) plants have a greater capacity for the biosynthesis of proteins on a large scale and perform complex assembly and folding of proteins; (3) plants are environmentally friendly, cost-effective, and sustainable when compared to existing expression systems; and (4) plant-derived vaccines can be effectively used for oral delivery without the requirement of sophisticated and time-consuming downstream processes.

7.2. Prospective Plant-Derived Fish Vaccines

The financial cost is crucial to consider while developing vaccines for the aquaculture industry. The plant expression system for vaccine production provides cost-effectiveness, safety, and efficacy. Plant-derived fish vaccines will provide increased and sustainable fish health in the flourishing global aquaculture industry. Research on plant-derived fish vaccines has been scarce, but the numbers of studies are now growing [169]. In aquaculture, there is much potential for oral vaccinations made from edible plants for fish. Additionally, a plant-generated recombinant vaccine can simultaneously deliver multiple antigenic proteins [170]. Table 7 shows some plant-derived vaccines in fish and other animals. Nevertheless, there is currently an absence of commercially available fish vaccines produced from plants (Figure 3). Therefore, it is necessary to conduct more studies on the development of fish vaccines employing plants as an important source [171].
Oral immunization offers a stress-free, time-saving means of delivery, with fewer labor costs, and minimizes the need for expensive downstream processing, purification, and cold storage while transporting [176]. Using a plant as an expression system, recombinant major capsid protein (rMCP) from iridovirus affecting Neoscorpis lithophilus was successfully expressed in rice callus. The rMCP was able to provide immune protection from iridovirus [62]. Likewise, nervous necrosis virus (NNV) coat protein was stably transformed into N. tabacum chloroplasts [58]. AcrV and VapA antigens from Aeromonas salmonicida affecting salmon were stably expressed in chloroplasts of Chlamydomonas reinhardtii, showing elevated immune production during infection [172]. White spot syndrome virus (WSSV) affecting crustaceans causes substantial economic loss, and the VP28 protein from WSSV was successfully expressed in Chlamydomonas reinhardtii and Dunaliella salina [173,174]. A successful transient expression of Atlantic cod nervous necrosis virus (ACNNV) VLPs in N. benthamiana was demonstrated, and an immunization study showed its effective defense against a virus challenge [175]. Plant-based virus-like particles (VLPs) against piscine myocarditis virus (PMCV), causing cardiomyopathy syndrome in wild Atlantic salmon, was transiently expressed in N. benthamiana. However, the plant-derived PMCV VLP vaccine induced limited immune protection against PMCV infection [170].
To elicit effective immune responses in fish, studies on plant-derived vaccines primarily focus on enhancing the quantity and purity of the produced antigens in the expressing plants (Figure 4). For the maximum amount and quality of antigens, targeting the most appropriate subcellular compartment in plant cells is crucial. Although there are numerous obstacles to overcome in the field of plant-derived vaccine production and application, the potential and appeal of enhanced plant-based immunizations are undeniable. Ultimately, plant biotechnology presents a promising option for developing future aquatic vaccines.

8. Updates on Strategies to Develop Fish Vaccines

A vaccine should be safe for a fish, and the mode of administration, if it is the oral mode, should be better and provide long lasting better protection. The main challenge to producing and developing vaccines remains in a sequence of events, from manufacturing to marketing. Another critical challenge is providing a cost-effective and readily available vaccine [176]. A significant restriction is the commercialization of oral vaccines and preservation of antigens. The licensing of vaccines is also not practically applicable for all fish antigens. Autogenous vaccines can be used as an alternative. Brooker et al. [177] studied an autogenous vaccine that was shown to be effective in controlling Aeromonas salomonicida when administered via injection in cleaner fish. Adjuvants are limited for mucosal vaccines, and adjuvant usage modulates the immunogenicity of an antigen [11]. Vaccines must be designed according to the type of fish species, production cycle, farming technology, environment, etc. Strategies must be developed based on the genomes of various fish, and the epidemiology with a multi-approach strategy needs to be taken care of for better production (Figure 4) [11].
Kole et al. [178] proposed that a trivalent oral vaccine consisting of attenuated viral hemorrhagic septicemia virus (VHSV), S. parauberis serotype I, and antigens of M. avidus, encapsulated in a chitosan–PLGA complex, could be an assuring strategy to prevent the outbreak of diseases in olive flounder. In addition, various other vaccination approaches are in their developmental phases, such as algal-enclosed oral vaccines, bacterial biofilm-based antigens, and exosome-derived vaccines [179,180,181]. Partial immunization in fish has led to the utilization of epitope-based vaccines, where one could, with the help of bioinformatics, formulate an antigen with multiple epitopes that would be potentially beneficial in creating effective vaccines [182]. Orally delivered vaccines have the disadvantage of disintegrating in the (acidic) gut of fish. As mentioned earlier, when encapsulated by a microalgal cell wall, these vaccines or recombinant proteins are protected from such harsh environments and are delivered to the system very easily via cell wall degradation [179].
Control and prevention strategies must be designed to appropriately treat infectious diseases in fish. Xu et al. [183] highlighted the necessity of employing multi-strategy approaches for fish production, emphasizing disease prevention, climate control, environmental management, and planned governance to meet the anticipated demand by 2050 for the national and global market distribution of fish products. This can be achieved with the geographical expansion of fish species, fishing capacity building, natural variability, and the employment of judicious culture techniques with newer science, policies, and interventions [183]. As per Xu et al.’s [183] report, although applicable measures have been identified, fewer vaccines are available for fish disease prevention and management. General prevention strategies are highlighted in Figure 5. Another major challenge in developing novel techniques is antibiotic resistance by microorganisms in fish.
The world’s need for fish is increasing every day, and the estimated demand for fish by 2050 is presented in Table 8. Therefore, loss due to infectious diseases in aquaculture must be controlled. Various vaccines proven to have a positive impact on reducing fish mortality have been outlined in this review. However, the most effective vaccines are necessary to address the increased demand for aquaculture, especially the need to manage large-scale fish farming.
Global capture-based aquaculture production has been recorded throughout various periods in different nations, revealing a slight decline in production in 2020 in the inland aquaculture system. Moreover, food security and sustainability concerns will increase as the world population approaches 9 billion by 2050 (Table 8) [184]. Therefore, economic expansion, job growth, nutritional profiles, and gender empowerment are a few variables that have contributed to the increased demand for aquaculture. The market expanded approximately 20-fold in 2020, compared to production in 1976. Billions of people depend on fishing and its allied sectors to meet their financial and health needs. The increase in fish consumption is primarily driven by technological developments in processing and distribution, directly affecting the increased demand for fish and fish products. Good management practices are crucial to reduce stress and minimize the prevalence of diseases (Figure 4 and Figure 5). Marine ecosystems are constantly bombarded with challenges that seriously threaten their sustainability. Several studies assert that climate change is the major player among all other factors [12]. Currently, there seem to be no climate-change measures that have been added to fish aquaculture management policies [185]. A shift in the regulatory guidelines of aquaculture has only occurred recently, with governments opting for a more ecosystem-based approach to aquaculture management (EAF). In simple terms, EAF considers all the species of a particular marine ecosystem rather than focusing on one species in isolation. Other components such as climate change, co-interactions with other species, and pollution also come into the picture (“Understanding Ecosystem-Based Fisheries Management,” National Oceanic and Atmospheric Administration). Understanding the role of each factor helps maximize the benefits of fish aquaculture and other marine ecosystems by avoiding over-exploitation of the available resources. It is vital to ensure that aquaculture is managed appropriately to attain the goals of food security, employment, and nutritional benefits. Apart from EAF, other existing strategies for managing aquaculture include rights-based management and management for resilience [186]. Therefore, fish vaccination is one of the significant tools for the management of aquaculture health. Hence, it is crucial to redirect attention away from traditional vaccines and instead prioritize novel fish immunization techniques. Vaccine development should employ advanced technologies like “Omics based” [187,188] and nano-carrier-based adjuvants [189,190] and environmentally friendly vaccines like plant-based vaccines [76,190], which are efficient and cost-effective, require small doses, and do not require the use of antibiotics. Such research on vaccine development is necessary in light of the increasing importance of aquaculture globally in recent years [16,191,192].

9. Conclusions

The demand for aquaculture products is growing faster than expected internationally. With the ever-growing demand for fish in world markets, their management in aquaculture by preventive measures against contagious diseases is a challenging area. Therefore, modern technology is presently employed in developing unique and alternative fish vaccines—for example, plant-derived vaccines—which help develop immune responses beneficial to fish health. The industry requires reliable aquaculture vaccines for economic success. The inefficient steps of conventional vaccine-developing methods must be modified to fight against emerging disease concerns. Research protocols for producing fish vaccines that employ newer technologies may be expensive but are essential. It will be crucial to leverage the abundance of biotechnology that is now available to address emerging disease concerns due to the continual demand for novel vaccines induced by the global development of aquaculture. In particular, vaccination in broodstocks is essential for fish seed production. The development of fish vaccines is known to use either whole-killed pathogens; subunits of a protein, peptide, or recombinant protein; DNA vaccines; or live-attenuated vaccines via different modes of administration. Further research is anticipated to explore strategies regarding the mode of administration and use of adjuvants.

Author Contributions

B.P.: Conceptualization; Literature survey and drafting outline, Data curation; Formal analysis; Funding acquisition; Investigation; Methodology; Project administration; Resources; Software; Supervision; Validation; Writing—original draft; editing, Coordination among all authors. A.K., S.K.M. and S.V.M. (Section 2, Section 3 and Section 4): Writing—original draft; Methodology, Writing—review and editing and Revision. A.R.: Drawing Figures (redrawn Figure 1, Figure 2 and Figure 4 in Biorender), writing and editing the original draft, editing reviews. S.G. (Section 1), M.N. (Section 6), R.B.R. (Section 6), H.M.C., S.P.M., and S.M.K. (Section 6.1, Section 8 and Section 9 with Table 5), T.U. (Section 6), A.K.H. (Section 7), D.V. (Section 2), T.R.B. (Section 5), S.V. (Section 7), and D.K.S. (Section 8): Writing—original draft; Methodology, Formal analysis; Writing—review and editing and revision. All authors have read and agreed to the published version of the manuscript.

Funding

The work was generously supported by the funding to BRP from the Science and Engineering Research Board, Department of Science and Technology, Govt. of India New Delhi, India (No. ECR/2016/001984) and Department of Science and Technology, Government of Odisha (Grant letter number 1188/ST, Bhubaneswar, dated 01.03.17, ST-(Bio)-02/2017). The work was partially supported by DBT Star College Scheme (12th Batch, Department of Biotechnology, Govt. of India, New Delhi) offered to BRP.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analyzed during this study are included in this published article.

Acknowledgments

The authors duly acknowledge using the Central Instrumentation Facility of the Odisha University of Agriculture and Technology. Language and grammatical corrections made by Aidan Mark Webster, United Kingdom, presently at the Department of Chemistry and Biochemistry, School of Sciences, Jain University, #34 JC Road, Bangalore, Karnataka, India, are acknowledged.

Conflicts of Interest

The authors declare that there are no conflicts of interest.

References

  1. WHO. Immunization, Vaccines and Biologicals. 2023. Available online: https://www.who.int/teams/immunization-vaccines-and-biologicals (accessed on 2 February 2023).
  2. Grace, D.; Gilbert, J.; Randolph, T.; Kang’ethe, E. The multiple burdens of zoonotic disease and an Ecohealth approach to their assessment. Trop. Anim. Health Prod. 2012, 44, 67–73. [Google Scholar] [CrossRef] [PubMed]
  3. Lurie, N.; Manolio, T.; Paterson, A.P.; Collins, F.; Frieden, T. Research as a part of public health emergency response. N. Eng. J. Med. 2013, 368, 1251–1255. [Google Scholar] [CrossRef] [PubMed]
  4. Monath, T.P. Vaccines against diseases transmitted from animals to humans: A one health paradigm. Vaccine 2013, 31, 5321–5338. [Google Scholar] [CrossRef] [PubMed]
  5. Taksdal, T.; Stangeland, K.; Dannevig, B.H. Induction of infectious pancreatic necrosis (IPN) in Atlantic salmon Salmo salar and brook trout Salvelinus fontinalis by bath challenge of fry with infectious pancreatic necrosis virus (IPNV) serotype Sp. Dis. Aquat. Organ. 1997, 28, 39–44. [Google Scholar] [CrossRef]
  6. Bebak, J.; McAllister, P.E.; Smith, G. Infectious pancreatic necrosis virus: Transmission from infectious to susceptible rainbow trout fry. J. Aquat. Anim. Health 1998, 10, 287–293. [Google Scholar] [CrossRef]
  7. Rahman, M.T.; Sobur, M.A.; Islam, M.S.; Ievy, S.; Hossain, M.J.; El Zowalaty, M.E.; Rahman, A.T.; Ashour, H.M. Zoonotic Diseases: Etiology, Impact, and Control. Microorganisms 2020, 8, 1405. [Google Scholar] [CrossRef]
  8. Ziarati, M.; Zorriehzahra, M.J.; Hassantabar, F.; Mehrabi, Z.; Dhawan, M.; Sharun, K.; Emran, T.B.; Dhama, K.; Chaicumpa, W.; Shamsi, S. Zoonotic diseases of fish and their prevention and control. Vet. Q. 2022, 42, 95–118. [Google Scholar] [CrossRef]
  9. Rupprecht, C.E.; Hanlon, C.A.; Blanton, J.; Manangan, J.; Morrill, P.; Murphy, S.; Niezgoda, M.; Orciari, L.A.; Schumacher, C.L.; Dietzschold, B. Oral vaccination of dogs with recombinant rabies virus vaccines. Virus Res. 2005, 111, 101–105. [Google Scholar] [CrossRef]
  10. Perkins, S.D.; Smither, S.J.; Atkins, H.S. Towards a Brucella vaccine for humans. FEMS Microbiol. Rev. 2010, 4, 379–394. [Google Scholar] [CrossRef]
  11. Adams, A. Progress, challenges and opportunities in fish vaccine development. Fish Shellfish Immunol. 2019, 90, 210–214. [Google Scholar] [CrossRef]
  12. Holsman, K.K.; Haynie, A.C.; Hollowed, A.B.; Reum, J.C.P.; Aydin, K.; Hermann, A.J.; Cheng, W.; Faig, A.; Ianelli, J.N.; Kearney, K.A. Ecosystem-Based Fisheries Management Forestalls Climate-Driven Collapse. Nat. Commun. 2020, 11, 4579. [Google Scholar] [CrossRef] [PubMed]
  13. Roth, J.A. Veterinary vaccines and their importance to animal health and public health. Procedia Vaccinol. 2011, 5, 127–136. [Google Scholar] [CrossRef] [PubMed]
  14. NAOH. National Office of Animal Health, Vaccination for Animals: An Overview. 2017. Available online: http://noah.co.uk (accessed on 2 February 2023).
  15. Du, Y.; Hu, X.; Miao, L.; Chen, J. Current status and development prospects of aquatic vaccines. Front. Immunol. 2022, 13, 1040336. [Google Scholar] [CrossRef] [PubMed]
  16. Qian, M.-B.; Keiser, J.; Utzinger, J.; Zhou, X.-N. Clonorchiasis and opisthorchiasis: Epidemiology, transmission, clinical features, morbidity, diagnosis, treatment, and control. Clin. Microbiol. Rev. 2024, 37, e0000923. [Google Scholar] [CrossRef]
  17. Ma, J.; Bruce, T.J.; Jones, E.M.; Cain, K.D. A Review of Fish Vaccine Development Strategies: Conventional Methods and Modern Biotechnological Approaches. Microorganisms 2019, 7, 569. [Google Scholar] [CrossRef] [PubMed]
  18. Lillehaug, A. Vaccination Strategies and Procedures. In Fish Vaccination, 1st ed.; Lillehaug, A.G.R., Evensen, Ø., Eds.; John Wiley and Sons Inc.: Oxford, UK, 2014; pp. 141–150. [Google Scholar]
  19. Figueroa, C.; Torrealba, D.; Morales-Lange, B.; Mercado, L.; Dixon, B.; Conejeros, P.; Silva, G.; Soto, C.; Gallardo, J.A. Commercial Vaccines Do Not Confer Protection against Two Genogroups of Piscirickettsia salmonis, LF-89 and EM-90, in Atlantic Salmon. Biology 2022, 11, 993. [Google Scholar] [CrossRef]
  20. He, T.; Zhang, Y.-Z.; Gao, L.-H.; Miao, B.; Zheng, J.-S.; Pu, D.-C.; Zhang, Q.-Q.; Zeng, W.-W.; Wang, D.-S.; Su, S.-Q.; et al. Identification and pathogenetic study of tilapia lake virus (TiLV) isolated from naturally diseased tilapia. Aquaculture 2023, 565, 739166. [Google Scholar] [CrossRef]
  21. Kembou-Ringert, J.E.; Steinhagen, D.; Readman, J.; Daly, J.M.; Adamek, M. Tilapia Lake Virus Vaccine Development: A Review on the Recent Advances. Vaccines 2023, 11, 251. [Google Scholar] [CrossRef]
  22. NSC. History of New England’s Ground Fish Fishery & Management. 2023. Available online: https://northeastseafoodcoalition.org/fishery-101/history/#:~:text=Before%20the%20Magnuson%20was%20implemented,New%20England%20Fishery%20Management%20Council (accessed on 2 February 2023).
  23. OWD. Seafood and fish production, World. 2023. Available online: https://ourworldindata.org/grapher/seafood-and-fish-production-thousand-tonnes (accessed on 2 February 2023).
  24. OECD. Food & Nations, A.O. of the U. OECD-FAO Agricultural Outlook 2020–2029. OECD iLibrary. 2020. Available online: https://www.oecd-ilibrary.org/agriculture-and-food/oecd-fao-agricultural-outlook-2020-2029_1112c23b-en (accessed on 2 February 2023). [CrossRef]
  25. FAO. The State of World Fisheries and Aquaculture 2022-Towards Blue Transformation; FAO: Rome, Italy, 2022; Available online: https://openknowledge.fao.org/items/11a4abd8-4e09-4bef-9c12-900fb4605a02 (accessed on 2 February 2023). [CrossRef]
  26. Naylor, R.L.; Kishore, A.; Sumaila, U.R.; Issifu, I.; Hunter, B.P.; Belton, B.; Bush, S.R.; Cao, L.; Gelcich, S.; Gephart, J.A. Blue food demand across geographic and temporal scales. Nat. Commun. 2021, 12, 5413. [Google Scholar] [CrossRef]
  27. Pollard, A.J.; Bijker, E.M. A guide to vaccinology: From basic principles to new developments. Nat. Rev. 2021, 21, 83–100. [Google Scholar] [CrossRef]
  28. Aida, V.; Pliasas, V.C.; Neasham, P.J.; North, J.F.; McWhorter, K.L.; Glover, S.R.; Kyriakis, C.S. Novel technologies in veterinary medicine: A Herald to human medicine vaccines. Front. Vet. Sci. 2021, 8, 654289. [Google Scholar] [CrossRef] [PubMed]
  29. Panda, F.; Pati, S.G.; Das, K.; Samanta, L.; Sahoo, D.K.; Paital, B. Biochemical and molecular responses of the freshwater snail Pila sp. To environmental pollutants, abiotic, and biotic stressors. Front. Environ. Sci. 2022, 10, 1033049. [Google Scholar] [CrossRef]
  30. Bal, A.; Panda, F.; Pati, S.G.; Anwar, T.A.; Das, K.; Paital, B. Influence of anthropogenic activities on redox regulation and oxidative stress responses in different phyla of animals in coastal water via change in salinity. Water 2022, 14, 4026. [Google Scholar] [CrossRef]
  31. Chhabria, S.; Mathur, S.; Vadakan, S.; Sahoo, D.K.; Mishra, P.; Paital, B. A review on Phytochemical and Pharmacological Facets of Tropical Ethnomedicinal Plants as Reformed DPP-IV Inhibitors to Regulate Incretin Activity. Front. Endocrinol. 2022, 13, 1027237. [Google Scholar] [CrossRef]
  32. Serkissian, M. Seven of the Biggest Problems Facing Fish in Our Oceans & Raquo; Marine Conservation Institute. Available online: https://marine-conservation.org/on-the-tide/seven-of-the-biggest-problems-facing-fish-in-our-oceans/ (accessed on 31 January 2023).
  33. Snieszko, S.F. Columnaris disease of fishes. USFWS Fish Leafl. 1958, 46, 1–3. [Google Scholar]
  34. Amend, D.F.; Ross, A.J. Experimental control of columnaris disease with a new nitrofuran drug, P-7138. Prog. Fish-Cult. 1970, 32, 19–25. [Google Scholar] [CrossRef]
  35. Ross, A.J. In vitro studies with nifurpirinol (P-7138) and bacterial fish pathogens. Prog. Fish-Cult. 1972, 34, 18–20. [Google Scholar] [CrossRef]
  36. Shiraki, K.; Miyamoto, F.; Sato, T.; Sonezaki, I.; Yano, K. Studies on a New Chemotherapautic Agent Nifurprazine (HB-115) against Fishinfectious Diseases-Part I. Fish Pathol. 1970, 4, 130–137. [Google Scholar] [CrossRef]
  37. Deufel, J. Prophylactic measures against bacterial diseases of salmonid fry (Prophylaktische Massnahmen gegen bakterielle Erkrankungen der Salmonidenbrut). Osterr Fisch 1974, 27, 1–5. [Google Scholar]
  38. Endo, T.; Ogishima, K.; Hayasaka, H.; Kaneko, S.; Ohshima, S. Application of oxolinic acid as a chemotherapeutic agent against infectious diseases in fishes-I. Antibacterial activity, chemotherapeutic effects and pharmacokinetics of oxolinic acid in fishes. Bull. Jpn. Soc. Sci. Fish. 1973, 39, 165–171. [Google Scholar] [CrossRef]
  39. Soltani, M.; Shanker, S.; Munday, B.L. Chemotherapy of Cytophaga/Flexibacter-like bacteria (CFLB) nfections in fish: Studies validating clinical efficacies of selected antimicrobials. J. Fish Dis. 1995, 18, 555–565. [Google Scholar] [CrossRef]
  40. Lilley, J.H.; Callinan, R.B.; Chinabut, S.; Kanchanakhan, S.; Macrae, I.H.; Phillips, M.J. Epizootic Ulcerative Syndrome (EUS) Technical Handbook; Aquatic Animal Health Research Institute: Bangkok, Thailand, 1998. [Google Scholar]
  41. Ahne, W.; Bjorklund, H.V.; Essbauer, S.; Fijan, N.; Kurath, G.; Winton, J.R. Spring viremia of carp (SVC). Dis. Aquat. Organ. 2002, 52, 261–272. [Google Scholar] [CrossRef] [PubMed]
  42. Dixon, P.F. Virus diseases of cyprinids. In Fish Diseases; Eiras, J.C., Segner, H., Wahli, T., Kapoor, B.G., Eds.; Science Publishers: Enfield, NH, USA, 2008; Volume 1, pp. 87–184. [Google Scholar]
  43. Emmenegger, E.J.; Kurath, G. DNA Vaccine Protects Ornamental Koi (Cyprinus carpio Koi) against North American Spring Viremia of Carp Virus. Vaccine 2008, 26, 6415–6421. [Google Scholar] [CrossRef] [PubMed]
  44. Noga, E.J. Lymphocystis in Fish Disease: Diagnosis and Treatment, 2nd ed.; Wiley-Blackwell: Ames, IA, USA, 2010; pp. 171–173. [Google Scholar]
  45. Wolf, K. Lymphocystis disease. In Fish Viruses and Fish Viral Diseases; Cornell University Press: Ithaca, NY, USA, 1988; pp. 268–291. [Google Scholar]
  46. Cusack, R.; Cone, D.K. A Review of Parasites as Vectors of Viral and Bacterial Diseases of Fish. J. Fish Dis. 1986, 9, 169–171. [Google Scholar] [CrossRef]
  47. GOI. Govt. of India Handbook on Fisheries Statistics, Ministry of Fisheries, Animal Husbandry and Dairy, Government of India. 2021. Available online: https://dof.gov.in/sites/default/files/2021-02/Final_Book.pdf (accessed on 2 February 2023).
  48. Masifundise. The Importance of Fish to Man: Not Just a Food Source 2016. Available online: https://www.masifundise.org/the-importance-of-fish-to-man-not-just-a-food-source/ (accessed on 31 January 2023).
  49. McLoughlin, M.F.; Graham, D.A. Alphavirus Infections in Salmonids? A Review. J. Fish Dis. 2007, 30, 511–531. [Google Scholar] [CrossRef]
  50. Sneeringer, S.; Bowman, M.; Clancy, M. The US and EU Animal Pharmaceutical Industries in the Age of Antibiotic Resistance. In USDA Economic Research Service Report Number 264; USDA: Washington, DC, USA, 2019. [Google Scholar]
  51. Horzinek, M.C.; Schijns, V.E.C.J.; Denis, M.; Desmettre, P.; Babiuk, L.A. General description of vaccines. In Veterinary Vaccinology; Pastoret, P.P., Blancou, J., Vannier, P., Verschueren, C., Eds.; Elsevier Press: Amsterdam, The Netherlands, 1997; pp. 132–152. [Google Scholar]
  52. Dadar, M.; Dhama, K.; Vakharia, V.N.; Hoseinifar, S.H.; Karthik, K.; Tiwari, R.; Khandia, R.; Munjal, A.; Salgado-Miranda, C.; Joshi, S.K. Advances in Aquaculture Vaccines Against Fish Pathogens: Global Status and Current Trends. Rev. Fish. Sci. Aquac. 2017, 25, 184–217. [Google Scholar] [CrossRef]
  53. DFO. Efficacy of the APEX Vaccine in Atlantic Salmon Subjected to an IHNV Exposure Simulating Natural and/or Elevated Field Challenges. Available online: https://www.dfo-mpo.gc.ca/aquaculture/rp-pr/acrdp-pcrda/projects-projets/P-07-04-010-eng.html (accessed on 2 February 2023).
  54. PHARMAQ Seguridad y Apoyo. Available online: https://www.pharmaq.com/es/pharmaq/ (accessed on 19 January 2023).
  55. HOME-MSD Animal Health Norge (Msd-Animal-Health.No). Available online: https://www.msd-animal-health.no/ (accessed on 21 January 2023).
  56. Tammas, I.; Bitchava, K.; Gelasakis, A.I. Transforming Aquaculture through Vaccination: A Review on Recent Developments and Milestones. Vaccines 2024, 12, 732. [Google Scholar] [CrossRef]
  57. Elanco South Africa. Available online: https://www.elanco.no/index (accessed on 24 January 2023).
  58. Aquavac-Vaccines Against Fish Diseases. Formerly Norvax. Available online: https://www.aquavac-vaccines.com/ (accessed on 16 January 2023).
  59. Leiva-Rebollo, R.; Labella, A.M.; Gémez-Mata, J.; Castro, D.; Borrego, J.J. Fish Iridoviridae: Infection, vaccination and immune response. Vet Res. 2024, 55, 88. [Google Scholar] [CrossRef]
  60. Dhar, A.K.; Manna, S.K.; Thomas Allnutt, F.C. Viral Vaccines for Farmed Finfish. Virus Dis. 2014, 25, 1–17. [Google Scholar] [CrossRef]
  61. Busch, R.A. Polyvalent vaccines in fish: The interactive effects of multiple antigens. Dev. Biol. Stand. 1997, 90, 245–256. [Google Scholar]
  62. Stuart, N. Treatment of Fish Disease. Veter. Rec. 1983, 112, 173–177. [Google Scholar] [CrossRef] [PubMed]
  63. Hegde, A.; Kabra, S.; Basawa, R.M.; Khile, D.A.; Abbu, R.U.F.; Thomas, N.A.; Manickam, N.B.; Raval, R. Bacterial diseases in marine fish species: Current trends and future prospects in disease management. World J. Microbiol. Biotechnol. 2023, 39, 317. [Google Scholar] [CrossRef] [PubMed]
  64. Sommerset, I.; Krossøy, B.; Biering, E.; Frost, P. Vaccines for Fish in Aquaculture. Expert Rev. Vaccines 2005, 4, 89–101. [Google Scholar] [CrossRef] [PubMed]
  65. Mondal, H.; Thomas, J. A Review on the Recent Advances and Application of Vaccines against Fish Pathogens in Aquaculture. Aquac. Int. 2022, 30, 1971–2000. [Google Scholar] [CrossRef] [PubMed]
  66. Nayak, S.K. Current Prospects and Challenges in Fish Vaccine Development in India with Special Reference to Aeromonas hydrophila Vaccine. Fish Shellfish Immunol. 2020, 100, 283–299. [Google Scholar] [CrossRef]
  67. Mai, T.T.; Kayansamruaj, P.; Soontara, C.; Kerddee, P.; Nguyen, H.; Senapin, S.; Costa, J.Z.; Thompson, K.D.; Rodkhum, C.; Dong, H.T. Immunization of Nile Tilapia (Oreochromis niloticus) Broodstock with Tilapia Lake Virus (TiLV) Inactivated Vaccines Elicits Protective Antibody and Passive Maternal Antibody Transfer. Vaccines 2022, 10, 167. [Google Scholar] [CrossRef]
  68. Shefat, S.H.T. Vaccines for infectious bacterial and viral diseases of fish. J. Bacteriol. Infect. Dis. 2018, 2, 1–5. [Google Scholar]
  69. Razali, S.A.; Shamsir, M.S.; Ishak, N.F.; Low, C.F.; Azemin, W.A. Riding the wave of innovation: Immunoinformatics in fish disease control. PeerJ 2023, 11, e16419. [Google Scholar] [CrossRef]
  70. Gudding, R.; Van Muiswinkel, W.B. A history of fish vaccination: Science-based disease prevention in aquaculture. Fish Shellfish Immunol. 2013, 35, 1683–1688. [Google Scholar] [CrossRef]
  71. Newman, S.G. Bacterial vaccines for fish. Annu. Rev. Fish Dis. 1992, 3, 145–185. [Google Scholar] [CrossRef]
  72. Plant, K.P.; LaPatra, S.E. Advances in fish vaccine delivery. Dev. Comp. Immunol. 2011, 35, 1256–1262. [Google Scholar] [CrossRef] [PubMed]
  73. Tafalla, C.; Bogwald, J.; Dalmo, R.A. Adjuvants and immunostimulants in fish vaccines: Current knowledge and future perspectives. Fish Shellfish Immunol. 2013, 35, 1740–1750. [Google Scholar] [CrossRef] [PubMed]
  74. Embregts, C.W.E.; Forlenza, M. Oral Vaccination of Fish: Lessons from Humans and Veterinary Species. Dev. Comp. Immunol. 2016, 64, 118–137. [Google Scholar] [CrossRef] [PubMed]
  75. Tafalla, C.; Bogwald, J.; Dalmo, R.A.; Munang’andu, H.M.; Evensen, O. Adjuvants in fish vaccines. In Fish Vaccination, 1st ed.; Gudding, R., Lillehaug, A., Evensen, Ø., Eds.; John. Wiley & Sons, Inc.: New York, NY, USA, 2014; pp. 68–84. [Google Scholar] [CrossRef]
  76. Magadan, S.; Jouneau, L.; Boudinot, P.; Salinas, I. Nasal Vaccination Drives Modifications of Nasal and Systemic Antibody Repertoires in Rainbow Trout. J. Immunol. 2019, 203, 1480–1492. [Google Scholar] [CrossRef]
  77. Milligan, S.B.; Skalla, D.; Lawton, K. Promoters for Regulation of Gene Expression in Plant Roots. 2023. Available online: https://patents.google.com/patent/US20050010974A1/en?oq=US+patent+No+US+2005%2f0010974A1 (accessed on 29 January 2023).
  78. Su, H.; Yakovlev, I.A.; van Eerde, A.; Su, J.; Clarke, J.L. Plant-Produced Vaccines: Future Applications in Aquaculture. Front. Plant Sci. 2021, 12, 718775. [Google Scholar] [CrossRef]
  79. Nagaraju, V.T. Nanovaccines in Aquaculture. Arch. Nanomed. Open Access J. 2019, 2, 153–159. [Google Scholar]
  80. Andreoni, F.; Amagliani, G.; Magnani, M. Selection of Vaccine Candidates for Fish Pasteurellosis Using Reverse Vaccinology and an In Vitro Screening Approach. In Vaccine Design, Book Series-Methods in Molecular Biology; Thomas, S., Ed.; Humana: New York, NY, USA, 2016; Volume 1404, pp. 181–192. [Google Scholar] [CrossRef]
  81. Rao, Y.; Su, J. Insights into the Antiviral Immunity against Grass Carp (Ctenopharyngodon idella) Reovirus (GCRV) in Grass Carp. J. Immunol. Res. 2015, 2015, 670437. [Google Scholar] [CrossRef]
  82. Miccoli, A.; Manni, M.; Picchietti, S.; Scapigliati, G. State-of-the-Art Vaccine Research for Aquaculture Use: The Case of Three Economically Relevant Fish Species. Vaccines 2021, 9, 140. [Google Scholar] [CrossRef]
  83. Irshath, A.A.; Rajan, A.P.; Vimal, S.; Prabhakaran, S.; Ganesan, R. Bacterial Pathogenesis in Various Fish Diseases: Recent Advances and Specific Challenges in Vaccine Development. Vaccines 2023, 11, 470. [Google Scholar] [CrossRef]
  84. Lan, N.G.T.; Dong, H.T.; Shinn, A.P.; Vinh, N.T.; Senapin, S.; Salin, K.R.; Rodkhum, C. Review of current perspectives and future outlook on bacterial disease prevention through vaccination in Asian seabass (Lates calcarifer). J. Fish Dis. 2024, 47, e13964. [Google Scholar] [CrossRef]
  85. Tamer, C.; Cavunt, A.; Durmaz, Y.; Ozan, E.; Kadi, H.; Kalayci, G.; Ozkan, B.; Isidan, H.; Albayrak, H. Inactivated infectious pancreatic necrosis virus (IPNV) vaccine and E.coli-expressed recombinant IPNV-VP2 subunit vaccine afford protection against IPNV challenge in rainbow trout. Fish Shellfish Immunol. 2021, 115, 205–211. [Google Scholar] [CrossRef] [PubMed]
  86. Bootland, L.M.; Dobos, P.; Stevenson, R.M.W. Immunization of adult brook trout, Salvelinus fontinalis, fails to prevent the infectious pancreatic necrosis virus (IPNV) carrier state. J. Fish Dis. 1995, 18, 449–458. [Google Scholar]
  87. Maret, J.J. Virulence Characteristics of Infectious Pancreatic Necrosis Virus (lPNV) in Brook Trout, Salvelinus fontinalis, Fry. Master’s Thesis, Oregon State University, Corvallis, OR, USA, 1997. [Google Scholar]
  88. Collins, C.; Lorenzen, N.; Collet, B. DNA vaccination for finfish aquaculture. Fish Shellfish Immunol. 2019, 85, 106–125. [Google Scholar] [CrossRef] [PubMed]
  89. Muñoz-Atienza EDiaz-Rosales, P.; Tafalla, C. Systemic and mucosal B and T cell responses upon mucosal vaccination of teleost fish. Front. Immunol. 2021, 11, 622377. [Google Scholar] [CrossRef]
  90. Mahendran, R.; Jeyabaskar, S.; Michael, D.; Vincent Paul, A.; Sitharaman, G. Computer-Aided Vaccine Designing Approach against Fish Pathogens Edwardsiella Tarda and Flavobacterium Columnare Using Bioinformatics Softwares. Drug Des. Dev. Ther. 2016, 1703–1714. [Google Scholar] [CrossRef]
  91. Briggs, M.; Funge-Smith, S.; Subasinghe, R.; Phillips, M. Introductions and Movement of Penaeus vannamei and Penaeus stylirostris in Asia and the Pacific; FAO: Bangkok, Thailand, 2004; RAP Publication 2004/10. [Google Scholar]
  92. Mishra, S.S.; Rakesh, D.; Dhiman, M.; Choudhary, P.; Debbarma, J.; Sahoo, S.N.; Barua, A.; Giri, B.S.; Ramesh, R.; Ananda, K.; et al. Present status of fish disease management in freshwater aquaculture in India: State-of-the-art-review. J. Aquac. Fish. 2017, 1, 003. [Google Scholar] [CrossRef]
  93. Zdanowicz, M.; Mudryk, Z.J.; Perlinski, P. Abundance and antibiotic resistance of Aeromonas isolated from the water of three carp ponds. Vet. Res. Commun. 2020, 44, 9–18. [Google Scholar] [CrossRef]
  94. Algammal, A.M.; Mabrok, M.; Sivaramasamy, E.; Youssef, F.M.; Atwa, M.H.; El-Kholy, A.W.; Hetta, H.F.; Hozzein, W.N. Emerging MDR-Pseudomonas aeruginosa in fish commonly harbor oprL and toxA virulence genes and blaTEM, blaCTX-M, and tetA antibiotic-resistance genes. Sci. Rep. 2020, 10, e15961. [Google Scholar] [CrossRef]
  95. Hashish, E.; Merwad, A.; Elgaml, S.; Amer, A.; Kamal, H.; Elsadek, A.; Marei, A.; Sitohy, M. Mycobacterium marinum infection in fish and man: Epidemiology, pathophysiology and management; a review. Vet. Q. 2018, 38, 35–46. [Google Scholar] [CrossRef]
  96. IASRI. Fish Diseases and Management. e-Krishi Shiksha. Available online: http://ecoursesonline.iasri.res.in (accessed on 2 February 2023).
  97. Kim, C.H.; Leong, J.A. Fish viruses. Encyclop. Virol. 2004, 558–568. [Google Scholar]
  98. Bajpai, V.; Pragyan, D.; Suman, K.; Mohanty, J.; Sahoo, P.K. Viral diseases in Indian freshwater and marine water pisciculture. Curr. Sci. 2022, 122, 267–280. [Google Scholar] [CrossRef]
  99. Nitika; Wei, J.; Hui, A.M. The Development of mRNA Vaccines for Infectious Diseases: Recent Updates. Infect. Drug Resist. 2021, 14, 5271–5285. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, Y.; Zhang, Z.; Luo, J.; Han, X.; Wei, Y.; Wei, X. mRNA vaccine: A potential therapeutic strategy. Mol. Cancer 2021, 16, 20–33. [Google Scholar] [CrossRef] [PubMed]
  101. Nisini, R.; Poerio, N.; Mariotti, S.; De Santis, F.; Fraziano, M. The Multirole of Liposomes in Therapy and Prevention of Infectious Diseases. Front. Immunol. 2018, 9, 155. [Google Scholar] [CrossRef] [PubMed]
  102. Bo, Y.; Wang, H. Materials based vaccines for infectious diseases. Wires Nanomedic. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnology 2022, 14, e1824. [Google Scholar] [CrossRef]
  103. Holvold, L.B.; Myhr, A.I.; Dalmo, R.A. Strategies and hurdles using DNA vaccines to fish. Vet. Res. 2014, 45, 21. [Google Scholar] [CrossRef]
  104. Chang, C.J. Immune sensing of DNA and strategies for fish DNA vaccine development. Fish Shellfish Immunol. 2020, 101, 252–260. [Google Scholar] [CrossRef]
  105. Coll, M.; Carreras, M.; Ciércoles, C.; Cornax, M.-J.; Gorelli, G.; Morote, E.; Saez, R. Assessing fishing and marine biodiversity changes using fishers’ perceptions: The Spanish Mediterranean and Gulf of Cadiz case study. PLoS ONE 2014, 9, e85670. [Google Scholar] [CrossRef]
  106. Mugimba, K.K.; Byarugaba, D.K.; Mutoloki, S.; Evensen, O.; Munang’andu, H.M. Challenges and Solutions to Viral Diseases of Finfish in Marine Aquaculture. Pathogens 2021, 10, 673. [Google Scholar] [CrossRef]
  107. Kageyama, A.; Yazawa, K.; Ishikawa, J.; Hotta, K.; Nishimura, K.; Mikami, Y. Nocardial Infections in Japan from 1992 to 2001, Including the First Report of Infection by Nocardia transvalensis. Eur. J. Epidemiol. 2004, 19, 383–389. [Google Scholar] [CrossRef]
  108. Singh, A.; Chhina, D.; Soni, R.K.; Kakkar, C.; Sidhu, U.S. Clinical spectrum and outcome of pulmonary nocardiosis: 5-year experience. Lung India: Official organ of Ind. Chest Soc. 2016, 33, 398–403. [Google Scholar]
  109. Vendrell, D.; Balcázar, J.L.; Ruiz-Zarzuela, I.; de Blas, I.; Gironés, O.; Múzquiz, J.L. Lactococcus garvieae in fish: A review. Comp. Immunol. Microbiol. Infect. Dis. 2006, 29, 177–198. [Google Scholar] [CrossRef] [PubMed]
  110. Shrestha, S.; Kanellis, J.; Korman, T.; Polkinghorne, K.R.; Brown, F.; Yii, M.; Kerr, P.G.; Mulley, W. Different faces of Nocardia infection in renal transplant recipients. Nephrology 2016, 21, 254–260. [Google Scholar] [CrossRef] [PubMed]
  111. Kariya, T.; Kubota, S.; Nakamura, Y.; Kira, K. Nocardial infection in cultured yellowtails (Seriola quinqueruiata and S. purpurascens)—I. Bacteriological study. Fish Pathol. 1968, 3, 16–23. (In Japanese) [Google Scholar] [CrossRef]
  112. Shimahara, Y.; Yasuda, H.; Nakamura, A.; Itami, T.; Yoshida, T. Detection of antibody response against Nocardia seriolae by enzyme-linked immunosorbent assay (ELISA) and a preliminary vaccine trial in yellowtail Seriola quinqueradiata. Bull. Eur. Assoc. Fish Pathol. 2005, 25, 270–275. [Google Scholar]
  113. Kato, G.; Yamashita, K.; Kondo, H.; Hirono, I. Protective efficacy and immune responses induced by a DNA vaccine encoding codonoptimized PPA1 against Photobacterium damselae subsp. Piscicida in Japanese flounder. Vaccine 2015, 33, 1040–1045. [Google Scholar]
  114. Nash, A.A.; Dalziel, R.G.; Fitzgerald, J.R. Chapter 8 Mechanisms of Cell and Tissue Damage. In Mims’ Pathogenesis of Infectious Disease, 6th ed.; Academic Press: Cambridge, MA, USA, 2015; pp. 171–231. ISBN 978-0-12-397188-3. [Google Scholar]
  115. Lampel, K.A.; Formal, S.B.; Maurelli, A.T. A Brief History of Shigella. EcoSal. Plus. 2018, 8, 10–1128. [Google Scholar] [CrossRef]
  116. Matsuyama, T.; Fukuda, Y.; Sakai, T.; Tanimoto, N.; Nakanishi, M.; Nakamura, Y.; Takano, T.; Nakayasu, C. Clonal structure in Ichthyobacterium seriolicida, the causative agent of bacterial haemolytic jaundice in yellowtail, Seriola quinqueradiata, inferred from molecular epidemiological analysis. J. Fish Dis. 2017, 40, 1065–1075. [Google Scholar] [CrossRef]
  117. Abdelsalam, M.; Asheg, A.; Eissa, A.E. Streptococcus dysgalactiae: An emerging pathogen of fishes and mammals. Int. J. Vet. Sci. Med. 2013, 1, 1–6. [Google Scholar] [CrossRef]
  118. Takano, T.; Nakamura, Y.; Matsuyama, T.; Sakai, T.; Shigenobu, Y.; Sugaya, T.; Yasuike, M.; Fujiwara, A.; Kondo, H.; Hirono, I.; et al. Complete Genome Sequence of Ichthyobacterium seriolicida JBKA-6T, Isolated from Yellowtail (Seriola quinqueradiata) Affected by Bacterial Hemolytic Jaundice. Genome Announc. 2017, 5, e01574-16. [Google Scholar] [CrossRef]
  119. Castillo, D.; Higuera, G.; Villa, M.; Middelboe, M.; Dalsgaard, I.; Madsen, L.; Espejo, R.T. Diversity of Flavobacterium psychrophilum and the potential use of its phages for protection against bacterial cold water disease in salmonids. J. Fish Dis. 2012, 35, 193–201. [Google Scholar] [CrossRef] [PubMed]
  120. Garcia, C.; Pozet, F.; Michel, C. Standardization of experimental infection with Flavobacterium psychrophilum, the agent of rainbow trout Oncorhynchus mykiss fry syndrome. Dis. Aquatic. Org. 2000, 42, 191–197. [Google Scholar] [CrossRef] [PubMed]
  121. Wakabayashi, H.; Horiuchi, M.; Bunya, T.; Hoshiai, G. Outbreaks of cold-water disease in coho salmon in Japan. Fish Pathol. 1991, 26, 211–212. (In Japanese) [Google Scholar] [CrossRef]
  122. Iida, Y.; Mizokami, A. Outbreaks of coldwater disease in wild ayu and pale chub. Fish Pathol. 1996, 31, 157–164. [Google Scholar] [CrossRef]
  123. Rochat, T.; Calvez, S.; Dalsgaard, I.; Madsen, L.; Calteau, A.; Lunazzi, A.; Nicolas, P.; Wiklund, T.; Bernardet, J.; Duchaud, E. Genomic Characterization of Flavobacterium psychrophilum Serotypes and Development of a Multiplex PCR-Based Serotyping Scheme. Front. Microbiol. 2017, 8, 1752. [Google Scholar] [CrossRef]
  124. Duchaud, E.; Rochat, T.; Habib, C.; Barbier, P.; Loux, V.; Guérin, C.; Dalsgaard, I.; Madsen, L.; Nilsen, H.; Sundell, K.; et al. Genomic Diversity and Evolution of the Fish Pathogen Flavobacterium psychrophilum. Front. Microbiol. 2018, 9, 138. [Google Scholar] [CrossRef]
  125. Keller, M.A.; Stiehm, E.R. Passive Immunity in Prevention and Treatment of Infectious Diseases. Clin. Microbiol. Rev. 2000, 13, 602–614. [Google Scholar] [CrossRef]
  126. Gravningen, K.; Sakai, M.; Mishiba, T.; Fujimoto, T. The efficacy and safety of an oil-based vaccine against Photobacterium damsela subsp. Piscicida in yellowtail (Seriola quinqueradiata): A field study. Fish Shellfish Immunol. 2008, 24, 523–529. [Google Scholar]
  127. Awate, S.; Babiuk, L.A.; Mutwiri, G. Mechanisms of Action of Adjuvants. Front. Immunol. 2013, 4, 114. [Google Scholar] [CrossRef]
  128. Sarkar, I.; Garg, R.; van Drunen Littel-van den Hurk, S. Selection of adjuvants for vaccines targeting specific pathogens. Expert Rev. Vac. 2019, 18, 505–521. [Google Scholar] [CrossRef]
  129. Rahman, M.; Ototake, M.; Nakanishi, T. Water-soluble adjuvants enhance the protective effect of Flavobacterium psychrophilum vaccines in ayu Plecoglossus altivelis. Fish Pathol. 2003, 38, 171–176. [Google Scholar] [CrossRef]
  130. Nagai, T.; Lida, Y.; Yoneji, T. Field trials of a vaccine with water-soluble adjuvant for bacterial coldwater disease in ayu Plecoglossus altivelis. Fish Pathol. 2003, 38, 63–65. (In Japanese) [Google Scholar] [CrossRef]
  131. Rhee, J.H. Current and New Approaches for Mucosal Vaccine Delivery. Mucos. Vac. 2020, 325–356. [Google Scholar] [CrossRef]
  132. Gómez, E.; Méndez, J.; Cascales, D.; Guijarro, J.A. Flavobacterium psychrophilum vaccine development: A difficult task. Microbial. Biotechnol. 2014, 7, 414–423. [Google Scholar] [CrossRef]
  133. Dumetz, F.; Duchaud, E.; LaPatra, S.E.; Marrec, C.L.; Claverol, S.; Urdaci, C.; Hénaff, M.L. A Protective Immune Response Is Generated in Rainbow Trout by an OmpH-Like Surface Antigen (P18) of Flavobacterium psychrophilum. Appl. Environ. Microbiol. 2006, 72, 4845–4852. [Google Scholar]
  134. Fux, R.; Arndt, D.; Langenmayer, M.C.; Schwaiger, J.; Ferling, H.; Fischer, N.; Indenbirken, D.; Grundhoff, A.; Dölken, L.; Adamek, M.; et al. Piscine Orthoreovirus 3 Is Not the Causative Pathogen of Proliferative Darkening Syndrome (PDS) of Brown Trout (Salmo trutta fario). Viruses 2019, 11, 112. [Google Scholar] [CrossRef]
  135. Takano, T.; Nawata, A.; Sakai, T.; Matsuyama, T.; Ito, T.; Kurita, J.; Terashima, S.; Yasuike, M.; Nakamura, Y.; Fujiwara, A.; et al. Full-Genome Sequencing and Confirmation of the Causative Agent of Erythrocytic Inclusion Body Syndrome in Coho Salmon Identifies a New Type of Piscine Orthoreovirus. PLoS ONE 2016, 11, e0165424. [Google Scholar] [CrossRef]
  136. Wessel, O.; Braaen, S.; Alarcon, M.; Haatveit, H.; Roos, N.; Markussen, T.; Tengs, T.; Dahle, M.; Rimstad, E. Infection with purified Piscine orthoreovirus demonstrates a causal relationship with heart and skeletal muscle inflammation in Atlantic salmon. PLoS ONE 2017, 12, e0183781. [Google Scholar] [CrossRef]
  137. Olsen, A.; Hjortaas, M.; Tengs, T.; Hellberg, H.; Johansen, R. First description of a new disease in rainbow trout (Oncorhynchus mykiss (Walbaum)) similar to heart and skeletal muscle inflammation (HSMI) and detection of a gene sequence related to piscine orthoreovirus (PRV). PLoS ONE 2015, 10, e0131638. [Google Scholar] [CrossRef]
  138. Marcotte, H.; Hammarström, L. Passive Immunization: Toward Magic Bullets. Mucos. Immunol. 2015, 2, 1403–1434. [Google Scholar]
  139. Haatveit, H.; Hodneland, K.; Braaen, S.; Hansen, E.; Nyman, I.; Dahle, M.; Frost, P.; Rimstad, E. DNA vaccine expressing the non-structural proteins of Piscine orthoreovirus delay the kinetics of PRV infection and induces moderate protection against heart-and skeletal muscle inflammation in Atlantic salmon (Salmo salar). Vaccine 2018, 36, 7599–7608. [Google Scholar] [CrossRef] [PubMed]
  140. Ogawa, K.; Bondad-Reantaso, M.; Fukudome, M.; Wakabayashi, H. Neobenedenia girellae (Hargis, 1955) Yamaguti, 1963 (Monogenea: Capsalidae) from cultured marine fishes of Japan. J. Parasitol. 1995, 81, 223–227. [Google Scholar] [CrossRef] [PubMed]
  141. Norbury, L.J.; Shirakashi, S.; Power, C.; Nowak, B.F.; Bott, N.J. Praziquantel use in aquaculture–Current status and emerging issues. Int. J. Parasitol. Drug Res. 2022, 18, 87–102. [Google Scholar] [CrossRef] [PubMed]
  142. Jung, S.-J.; Kitamura, S.-I.; Aoyana, M.; Song, J.-Y.; Kim, B.-K.; Oh, M.-J. Immune response of olive flounder, Paralichthys olivaceus against Miamiensis avidus (Ciliophora: Scuticociliatida). J. Fish Pathol. 2006, 19, 173–181. [Google Scholar]
  143. Song, J.Y.; Sasaki, K.; Okada, T.; Sakashita, M.; Kawakami, H.; Matsuoka, S.; Kang, H.S.; Nakayama, K.; Jung, S.J.; Oh, M.J. Antigenic differences of the scuticociliate Miamiensis avidus from Japan. J. Fish Dis. 2009, 12, 1027–1034. [Google Scholar] [CrossRef] [PubMed]
  144. Kole, S.; Dar, S.A.; Shin, M.; Jeong, J.; Jung, J. Potential Efficacy of Chitosan-Poly (Lactide-Co-Glycolide)-Encapsulated Trivalent Immersion Vaccine in Olive Flounder (Paralichthys olivaceus) Against Viral Hemorrhagic Septicemia Virus, Streptococcus parauberis Serotype I, and Miamiensis avidus (Scuticociliate). Front. Immunol. 2021, 12, 761130. [Google Scholar]
  145. Motokawa, S.; Narasaki, Y.; Song, J.Y.; Yokoyama, Y.; Hirose, E.; Murakami, S.; Jung, S.-J.; Oh, M.J.; Nakayama, K.; Kitamura, S.I. Analysis of genes encoding high antigenicity polypeptides in three serotypes of Miamiensis avidus. Parasitol. Int. 2018, 67, 196–202. [Google Scholar] [CrossRef]
  146. Nakamura, Y.; Takano, T.; Yasuike, M.; Sakai, T.; Matsuyama, T.; Sano, M. Comparative genomics reveals that a fish pathogenic bacterium Edwardsiella tarda has acquired the locus of enterocyte effacement (LEE) through horizontal gene transfer. BMC Genom. 2013, 14, 642. [Google Scholar] [CrossRef]
  147. Kusuda, R.; Inoue, M.; Sugiura, H.; Kawai, K. Characteristics of a pathogenic Mycobacterium sp. isolated from cultured striped jack, Pseudocaranx dentex. Aquac. Sci. 1993, 41, 125–131. [Google Scholar]
  148. Kusuda, R.; Kawakami, K.; Kawai, K. A fish-pathogenic Mycobacterium sp. isolated from an epizootic of cultured yellowtail. Nippon. Suisan Gakkaishi 1987, 53, 1797–1804. [Google Scholar] [CrossRef]
  149. Maekawa, S.; Yoshida, T.; Wang, P.; Chen, S. Current knowledge of nocardiosis in teleost fish. J. Fish Dis. 2018, 41, 413–419. [Google Scholar] [CrossRef] [PubMed]
  150. Mochizuki, M.; Kim, H.J.; Kasai, H.; Nishizawa, T.; Yoshimizu, M. Virulence Change of Infectious Hematopoietic Necrosis Virus against Rainbow trout Oncorhynchus mykiss with Viral Molecular Evolution. Fish Pathol. 2009, 44, 159–165. [Google Scholar] [CrossRef]
  151. Watt, J.; Liu, J. Preclinical Progress of Subunit and Live Attenuated Mycobacterium tuberculosis Vaccines: A Review following the First in Human Efficacy Trial. Pharmaceutics 2020, 12, 848. [Google Scholar] [CrossRef] [PubMed]
  152. Redding, L.; Werner, D.B. DNA vaccines in veterinary use. Expert Rev. Vaccines 2009, 8, 1251. [Google Scholar] [CrossRef] [PubMed]
  153. Håstein, T.; Gudding, R.; Evensen, Ø. Bacterial vaccines for fish–an update of the current situation worldwide. Dev. Biol. 2005, 121, 55–74. [Google Scholar]
  154. Torres-Corral, Y.; Girons, A.; González-Barreiro, O.; Seoane, R.; Riaza, A.; Santos, Y. Effect of Bivalent Vaccines against Vibrio anguillarum and Aeromonas salmonicida Subspecie Achromogenes on Health and Survival of Turbot. Vaccines 2021, 9, 906. [Google Scholar] [CrossRef]
  155. Silvaraj, S.; Md Yasin, I.S.; A. Karim, M.M.; Saad, M.Z. Elucidating the Efficacy of Vaccination against Vibriosis in Lates calcarifer Using Two Recombinant Protein Vaccines Containing the Outer Membrane Protein K (r-OmpK) of Vibrio alginolyticus and the DNA Chaperone J (r-DnaJ) of Vibrio harveyi. Vaccines 2020, 8, 660. [Google Scholar]
  156. Soler, E.; Houdebine, M. Preparation of recombinant vaccines. Biotechnol. Ann. Rev. 2007, 13, 65–94. [Google Scholar]
  157. Lee, H.; Lee, A.; Park, Y.; Song, S.; Choi, S.; Lee, B. A review of vaccine development and research for industry animals in Korea. Clin. Exp. Vaccine Res. 2012, 1, 18–34. [Google Scholar] [CrossRef]
  158. Chambers, M.A.; Graham, S.P.; La Ragione, R.M. Challenges in Veterinary Vaccine Development and Immunization. Vaccines Vet. Dis. 2016, 2, 3–35. [Google Scholar]
  159. Ogawa, K.; Yokoyama, H. Parasitic diseases of cultured marine fish in Japan. Fish Pathol. 1998, 33, 303–309. [Google Scholar] [CrossRef]
  160. Piacentini, S.C.; Rohovec, J.S.; Fryer, J.L. Epizootiology of erythrocytic inclusion body syndrome. J. Aquat. Anim. Health 1989, 1, 173–179. [Google Scholar] [CrossRef]
  161. Starliper, C.E. Bacterial coldwater disease of fishes caused by Flavobacterium psychrophilum. J. Adv. Res. 2011, 2, 97–108. [Google Scholar] [CrossRef]
  162. Castells-Graells, R.; Lomonossoff, G.P. Plant-based Production Can Result in Covalent Cross-linking of Proteins. Plant Biotechnol. J. 2021, 19, 1095–1097. [Google Scholar] [CrossRef] [PubMed]
  163. Barta, A.; Sommergruber, K.; Thompson, D.; Hartmuth, K.; Matzke, M.A.; Matzke, A.J.M. The Expression of a Nopaline Synthase? Human Growth Hormone Chimaeric Gene in Transformed Tobacco and Sunflower Callus Tissue. Plant Mol. Biol. 1986, 6, 347–357. [Google Scholar] [CrossRef]
  164. Hiatt, A.; Caffferkey, R.; Bowdish, K. Production of Antibodies in Transgenic Plants. Nature 1989, 342, 76–78. [Google Scholar] [CrossRef]
  165. Malik, M.S.; Elahi, I.; Sameeullah, M.; Ijaz, F.; Batool, N.; Khalid, F.; Gurel, E.; Saba, K.; Waheed, M.T. In silico designing and characterization of outer membrane protein K (OmpK) from Vibrio anguillarum and its expression in Nicotiana tabacum for the development of a plant-based vaccine against fish vibriosis. J Biotechnol. 2024, 380, 51–63. [Google Scholar] [CrossRef]
  166. Shaaltiel, Y.; Gingis-Velitski, S.; Tzaban, S.; Fiks, N.; Tekoah, Y.; Aviezer, D. Plant-Based Oral Delivery of β-Glucocerebrosidase as an Enzyme Replacement Therapy for Gaucher’s Disease. Plant Biotechnol. J. 2015, 13, 1033–1040. [Google Scholar] [CrossRef]
  167. Guan, Z.; Guo, B.; Huo, Y.; Guan, Z.; Dai, J.; Wei, Y. Recent Advances and Safety Issues of Transgenic Plant-Derived Vaccines. Appl. Microbiol. Biotechnol. 2013, 97, 2817–2840. [Google Scholar] [CrossRef]
  168. Su, H.; van Eerde, A.; Steen, H.S.; Heldal, I.; Haugslien, S.; Ørpetveit, I.; Wüstner, S.C.; Inami, M.; Løvoll, M.; Rimstad, E.; et al. Establishment of a Piscine Myocarditis Virus (PMCV) Challenge Model and Testing of a Plant-Produced Subunit Vaccine Candidate against Cardiomyopathy Syndrome (CMS) in Atlantic Salmon Salmo Salar. Aquaculture 2021, 541, 736806. [Google Scholar] [CrossRef]
  169. Buyel, J.F. Plant Molecular Farming–Integration and Exploitation of Side Streams to Achieve Sustainable Biomanufacturing. Front. Plant Sci. 2019, 9, 1893. [Google Scholar] [CrossRef] [PubMed]
  170. Shin, Y.J.; Kwon, T.H.; Seo, J.Y.; Kim, T.J. Oral Immunization of Fish against Iridovirus Infection Using Recombinant Antigen Produced from Rice Callus. Vaccine 2013, 31, 5210–5215. [Google Scholar] [CrossRef] [PubMed]
  171. Michelet, L.; Lefebvre-Legendre, L.; Burr, S.E.; Rochaix, J.-D.; Goldschmidt-Clermont, M. Enhanced Chloroplast Transgene Expression in a Nuclear Mutant of Chlamydomonas: Enhanced Chloroplast Transgene Expression. Plant Biotechnol. J. 2011, 9, 565–574. [Google Scholar] [CrossRef] [PubMed]
  172. Xue, R.; Liu, L.; Cao, G.; Xu, S.; Li, J.; Zou, Y.; Chen, H.; Gong, C. Oral Vaccination of BacFish-Vp6 against Grass Carp Reovirus Evoking Antibody Response in Grass Carp. Fish Shellfish Immunol. 2013, 34, 348–355. [Google Scholar] [CrossRef] [PubMed]
  173. Marsian, J.; Hurdiss, D.L.; Ranson, N.A.; Ritala, A.; Paley, R.; Cano, I.; Lomonossoff, G.P. Plant-Made Nervous Necrosis Virus-Like Particles Protect Fish Against Disease. Front. Plant Sci. 2019, 10, 880. [Google Scholar] [CrossRef]
  174. Clarke, J.L.; Paruch, L.; Dobrica, M.-O.; Caras, I.; Tucureanu, C.; Onu, A.; Ciulean, S.; Stavaru, C.; Eerde, A.; Wang, Y.; et al. Lettuce-Produced Hepatitis C Virus E1E2 Heterodimer Triggers Immune Responses in Mice and Antibody Production after Oral Vaccination. Plant Biotechnol. J. 2017, 15, 1611–1621. [Google Scholar] [CrossRef]
  175. Grisez, L.; Tan, Z. Vaccine development for Asian Aquaculture. Diseases in Asian Aquaculture V Fish health section. In Proceedings of the Fifth Symposium in Asian Aquaculture; Walker, P., Lester, R., Bondad-Reantaso, M.G., Eds.; Asian Fisheries Society: Goldcoast, Australia, 2005; pp. 483–494. [Google Scholar]
  176. Brooker, A.J.; Papadopoulou, A.; Gutierrez, C.; Rey, S.; Davie, A.; Migaud, H. Sustainable production and use of cleaner fish for the biological control of sea lice: Recent advances and current challenges. Vet. Rec. 2018, 183, 383. [Google Scholar] [CrossRef]
  177. Kole, S.; Shin, S.M.; Kwak, I.S.; Cho, S.H.; Jung, S.J. Efficacy of Chitosan-PLGA encapsulated trivalent oral vaccine against viral haemorrhagic septicemia virus, Streptococcus parauberis, and Miamiensis avidus in olive flounder (Paralichthys olivaceus). Fish Shellfish Immunol. 2022, 127, 843–854. [Google Scholar] [CrossRef]
  178. Kwon, K.C.; Lamb, A.; Fox, D.; Jegathese, P.S.J. An evaluation of microalgae as a recombinant protein oral delivery platform for fish using green fluorescent protein (GFP). Fish Shellfish Immunol. 2019, 87, 414–420. [Google Scholar] [CrossRef]
  179. Loera-Muro, A.; Guerrero-Barrera, A.; Tremblay, D.N.Y.; Hathroubi, S.; Angulo, C. Bacterial biofilm-derived antigens: A new strategy for vaccine development against infectious diseases. Expert Rev. Vaccines 2021, 20, 385–396. [Google Scholar] [CrossRef]
  180. Montaner-Tarbes, S.; Fraile, L.; Montoya, M.; Del Portillo, H. Exosome-Based Vaccines: Pros and Cons in the World of Animal Health. Viruses 2021, 13, 1499. [Google Scholar] [CrossRef] [PubMed]
  181. Hajissa, K.; Zakaria, R.; Suppian, R.; Mohamed, Z. Epitope-based vaccine as a universal vaccination strategy against Toxoplasma gondii infection: A mini-review. J. Adv. Vet. Anim. Res. 2019, 6, 174. [Google Scholar] [CrossRef] [PubMed]
  182. Xu, K.; Wang, Y.; Yang, W.; Cai, W.; Zhang, Y.; Huang, L. Strategies for prevention and control of vibriosis in Asian fish culture. Vaccines 2023, 11, 98. [Google Scholar] [CrossRef] [PubMed]
  183. Garcia, S.M.; Rosenberg, A.A. Food security and marine capture fisheries: Characteristics, trends, drivers and future perspectives. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2010, 365, 2869–2880. [Google Scholar] [CrossRef] [PubMed]
  184. Karp, M.A.; Peterson, J.O.; Lynch, P.D.; Griffis, R.B.; Adams, C.F.; Arnold, W.S.; Barnett, L.A.K.; deReynier, Y.; DiCosimo, J.; Fenske, K.H.; et al. Accounting for Shifting Distributions and Changing Productivity in the Development of Scientific Advice for Fishery Management. ICES J. Mar. Sci. 2019, 76, 1305–1315. [Google Scholar] [CrossRef]
  185. Andrew, N.L.; Evans, L. Approaches and frameworks for management and research in small-scale fisheries. In Smallscale Fisheries Management: Frameworks and Approaches for the Developing World; CAB International: Oxfordshire, UK, 2011; pp. 16–34. [Google Scholar]
  186. Natnan, M.E.; Mayalvanan, Y.; Jazamuddin, F.M.; Aizat, W.M.; Low, C.F.; Goh, H.H.; Azizan, K.A.; Bunawan, H.; Baharum, S.N. Omics strategies in current advancements of infectious fish disease management. Biology 2021, 10, 1086. [Google Scholar] [CrossRef]
  187. Shivam, S.; El-Matbouli, M.; Kumar, G. Development of Fish Parasite Vaccines in the OMICs Era: Progress and Opportunities. Vaccines 2021, 9, 179. [Google Scholar] [CrossRef]
  188. Vinay, T.N.; Bhat, S.; Gon Choudhury, T.; Paria, A.; Jung, M.-H.; Shivani Kallappa, G.; Jung, S.-J. Recent advances in application of nanoparticles in fish vaccine delivery. Rev. Fish. Sci. Aquac. 2018, 26, 29–41. [Google Scholar] [CrossRef]
  189. Giri, S.S.; Kim, S.G.; Kang, J.W.; Kwon, J.; Bin Lee, S.; Jung, W.J.; Park, S.C. Applications of carbon nanotubes and polymeric micro-/nanoparticles in fish vaccine delivery: Progress and future perspectives. Rev. Aquac. 2021, 13, 1844–1863. [Google Scholar] [CrossRef]
  190. Clarke, J.L.; Waheed, M.T.; Lössl, A.G.; Martinussen, I.; Daniell, H. How can plant genetic engineering contribute to cost-effective fish vaccine development for promoting sustainable aquaculture? Plant Mol. Biol. 2013, 83, 33–40. [Google Scholar] [CrossRef]
  191. Assefa, A.; Abunna, F. Maintenance of Fish Health in Aquaculture: Review of Epidemiological Approaches for Prevention and Control of Infectious Disease of Fish. Vet. Med. Int. 2018, 2018, 5432497. [Google Scholar] [CrossRef] [PubMed]
  192. Snow, M. The contribution of molecular epidemiology to the understanding and control of viral diseases of salmonid aquaculture. Vet. Res. 2011, 42, 56. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Management of fish health with multiple approaches. This panel depicts ecosystem-level management, which includes disease and environmental control measures.
Figure 1. Management of fish health with multiple approaches. This panel depicts ecosystem-level management, which includes disease and environmental control measures.
Animals 14 02692 g001
Figure 2. Vaccine classification based on their mode of action and their sources. This Figure is modified after Assefa and Abunna [21].
Figure 2. Vaccine classification based on their mode of action and their sources. This Figure is modified after Assefa and Abunna [21].
Animals 14 02692 g002
Figure 3. Schematic diagram for fish vaccine regulatory approval.
Figure 3. Schematic diagram for fish vaccine regulatory approval.
Animals 14 02692 g003
Figure 4. Schematic representation of the steps involved in plant-based fish vaccine development.
Figure 4. Schematic representation of the steps involved in plant-based fish vaccine development.
Animals 14 02692 g004
Figure 5. Control and prevention strategies for infectious diseases in fish [25,183]. Different approaches are used to maintain fish health, such as nano-based drugs/vaccines, injectable vaccines, probiotics, plant-based medicines, or edible vaccines.
Figure 5. Control and prevention strategies for infectious diseases in fish [25,183]. Different approaches are used to maintain fish health, such as nano-based drugs/vaccines, injectable vaccines, probiotics, plant-based medicines, or edible vaccines.
Animals 14 02692 g005
Table 1. Different infectious agents, diseases they cause, and commercial vaccines against them in animals.
Table 1. Different infectious agents, diseases they cause, and commercial vaccines against them in animals.
S. No.Infectious Agent/Etiological AgentDiseasesAnimals InfectedVaccines Used to Prevent the Disease (Either Used in Humans or Animals)
1.Avian Influenzae virusAvian InfluenzaPoultry: chickens, ducks, turkeys, geeseAfluria Quadrivalent, Fluarix Quadrivalent, FluLaval Quadrivalent, and Fluzone Quadrivalent
2.Herpes virus, Gallid alphaherpesvirus 2Marek’s disease
(fowl paralysis)
Poultry: chickensHSV vaccine candidate (mRNA-1608), herpes zoster
3.Salmonella sp.SalmenollosisAquatic animals: fishes; tortoises, birds, cattle, pigs, horsesTy21a, Vivotif (Typhoid Vaccine Live Oral Ty21a), Typbar TCV®, Typhim Vi, Vivotif
4. Rabies lyssavirusRabiesDogsHDCV or PCEC, human rabies immune globulin (HRIG)
5.BacillusanthracisAnthraxCattle, sheep, goats, camelsAnthrax Vaccine Adsorbed (AVA) or BioThraxTM
6.Brucella bacteriaBrucellosisCattle, sheep, goats, swine, equinesLive-attenuated Brucella abortus strain 19 (S19 vaccine), Brucella abortus S19
7. ListeriamonocytogenesListeriosisBirds, crustaceansUnder clinical trials
8.SARS-CoVSevere acute respiratory syndrome Bats, birds, cattleCOMIRNATY®, COMIRNATY® Original/Omicron BA.1, COMIRNATY® Original/Omicron BA.4-5, VAXZEVRIA, COVISHIELD™, COVID-19 Vaccine, SPIKEVAX, Inactivated COVID-19 Vaccine (Vero Cell), CoronaVac, COVAXIN®, COVOVAX™, NUVAXOVID™, CONVIDECIA
9.Nipah virus (NiV)NipahBats, pigs, horses, goats, sheepNipah Virus Vaccine (PHV02)
10.Monkey pox virusMonkey poxRope squirrels, dormice, non-human primates, etc. ACAM2000®, JYNNEOS™ (Imvamune or Imvanex or MVA-BN)
11.ClostridiumbotulinumBotulismFishes, mainly troutFabrizio Anniballi,
Alfonsina Fiore,
Charlotta Löfström,
Viveca Båverud.
12.Mycobacterium sp.Mycobacterial infections All fish Bacille Calmette-Guérin (BCG)
13.E. coliAvian bacterial infections (AVECs)All avian sp.Poulvac E. coli
A list of different causatives and infectious or etiological agents is given in this Table. Susceptible animals indicate that the adaptation of vaccination is recommended in animals; data adapted from The World Health Organization [1].
Table 2. Communicable diseases, associated pathogens, symptoms, and treatments in different fish.
Table 2. Communicable diseases, associated pathogens, symptoms, and treatments in different fish.
S. No. DiseasePathogenSymptomsTreatmentReferences
1.ColumnarisDiseaseFlavobacterium columnareLesions in skin, fin erosion, necrosis in gillsAmphenicol, Nifurpirinol,
Nifurprazine, Oxolinic acid
[35,36,37,38,39,40,41]
2. Epizootic ulcerative syndrome (EUS), or “red spot disease”AphanomycesinvadensRed lesions (sores) or deep ulcersNo effective treatment but can be treated with different parts of Azadirachta indica[42]
3.Spring viremia of carpRhabdovirus, spring viremia of carp virusDestruction of kidney, spleen and liver tissuesDNA
vaccination may be protectable in fish
[43,44,45]
4.LymphocystisLymphocysti virus or Lymphocystis disease virusPebble- or wart-like nodules mostly on the fins, skin, gills, etc.No effective treatment [46,47]
5.Carp poxCyprinid herpesvirus-1 (CyHV-1)Milky skin lesions, thickening of finsNo effective treatment [48,49,50,51,52,53,54,55,56,57,58,59,60]
Table 3. Commercial names of a few fish vaccines and their mode of administration.
Table 3. Commercial names of a few fish vaccines and their mode of administration.
SpeciesDiseaseOrganismName of the VaccineMode of AdministrationType of VaccineReference
SalmonInfectious hematopoietic necrosisInfectious hematopoietic necrosis virusAPEX-IHN®IMIHNV plasmid vaccine[57]
Enteric red mouth disease, yersiniosisYersinia ruckeri serotype O1bAlpha ERM SalarIPInactivated bacterial vaccine[58]
Aquavac YER knowsIPInactivated bacterial vaccine[59]
Pancreatic diseaseSalmon pancreas disease virusALPHA JECT micro 1 PDIPInactivated viral vaccine[58]
Salmonid alphavirusClynavIM.DNA plasmid[60]
Salmon alphaviruses (SAV)PD Norvax® Compact PDIPInactivated viral vaccine[59]
Infectious salmon anemiaInfectious salmon anemia virus (ISAV)ALPHA JECT® micro 1 ISAIPInactivated viral vaccine[58]
TilapiaStreptococcosisS. agalactiae serotype lbAQUAVAC® Strep SaIPInactivated viral vaccine[61]
S. agalactiae serotype Ia and serotype IIIAQUAVAC® Strep Sa1IPInactivated viral vaccine[61]
Tilapia, seabassS. iniaeAQUAVAC® Strep SiDip immersion/IPInactivated viral vaccine[61]
Streptococcus agalactiae IbALPHA JECT® micro 1 TilaIPInactivated viral vaccine[58]
KoiKoi herpes virus diseaseKoi herpes virus (KHV)KV-3Immersion/InjectionAttenuated viral vaccine (not used because of its safety issues)[61]
Sea bassViral nervous necrosisNodavirusALPHA JECT micro® 1 NodaIPInactivated viral vaccine[58]
Aeromonas veronii infectionAeromonas veroniiAutogenous Aeromonas veronii vaccineIPInactivated bacterial culture[58]
VibriosisListonella anguillarumALPHA DIP® VibDip vaccineInactivated bacterial vaccine[58]
Asian seabassEpizootic hematopoietic necrosisIridovirusAQUAVAC® IridoVIPInactivated viral vaccine[61]
Seabass, rainbow troutInfectious pancreatic necrosisInfectious pancreatic necrosis virus (IPNV)AquaVac® IPN OralOralInactivated viral vaccine[59]
Alpha Jects® 1000IPInactivated viral vaccine[58]
PangasiusEnteric septicemia disease,
motile aeromonad septicemia
Aeromonas hydrophila and Edwardsiella icataluriALPHA JECT® Panga 2IPInactivated bacterial vaccine[58]
Gray mullet (Mugil cephalus), Nile tilapiaLactococcosisLactococcus garvieaeIchtiovac-LgIPInactivated vaccine
Table 4. List of vaccines proved to be preventing infections in fish.
Table 4. List of vaccines proved to be preventing infections in fish.
Types of VaccineName of VaccineFishInfectionRemark
Inactivated or heat-killed whole-cell vaccineApha Ject® 1000, NorwaySalmonInfectious pancreatic necrosis virusMonovalent
Inactivated SVCVCarpSpring viremia of carp virusSVCV emulsified in oil
Formalin-inactivated IHNVRainbow troutInfectious hematopoietic necrosis virus
Killed VHSVRainbow troutViral hemorrhagic septicemia virus--
Attenuated vaccinesAttenuated KHV IsraelCarpKoi herpes virus
Attenuated
Flavobacterium columnare
All freshwater finfishFlavobacterium columnare--
Septicemia due to enterococciCatfishEdwardsiella ictaluri--
Kidney infection in fish due to bacteriaPacific salmon and Atlantic salmonRenibacterium salmoninarum--
Recombinant vaccinesRecombinant G proteinCarpSVCV--
CIBA-Nodavac-RAll types of fishes infected with nervous necrosis virus (NNV)Red-spotted grouper NNVFirst vaccine indigenously developed in India
Synthetic peptide vaccinesSubunit vaccine IPNV aquabirnavirusRainbow trout and Atlantic salmonInfectious pancreatic necrosis virusesTarget: VP2, VP3 and Capsid proteins
DNA vaccinesApex IHN, CanadaSalmonViruses having G antigen
DNASalmonidsInfectious hematopoietic necrosis rhabdovirusTarget: G glycoprotein
Mucosal vaccinesMicroMatrix™ delivery system (Piscirickettsia salmonis, ISAV and IPNV, Centrovet)Atlantic salmonY. ruckeri V. anguillarum, P. salmonis and IPNV or other similar mucosal infection.Pathogen killed by heat or inactivated by formalin
Plant-based edible vaccinesUnder development in plant Nicotiana benthamianaSalmonidsPMCV and cardiomyopathy syndrome--
Nanoparticle-based vaccinesChitosan-NPs based vaccine formulation NPrgpG, pICrgpG, CSrgpG, NpiVZebrafishViral hemorrhagic septicemia virus.Under experimental trials
OCMCS-hyaluronic acid, OCMCS/aerA-NPs, OCMCS-HA/aerA-NPsEuropean carpAeromonas hydrophilaUnder experimental trials
Monovalent and polyvalent vaccinesME-VAC Aqua StreptNile tilapia fish, Nile tilapiaStreptococcus infectionsEffective against bacterial strains, Streptococcoci, Enterococcoi, and Lactococci
Table 4 shows the “available” list of fish vaccines. Data are collected from Mondal and Thomas [65] and Dhar et al. [60].
Table 5. Some examples of infectious diseases affecting fish that cause economic losses in the aquaculture sector.
Table 5. Some examples of infectious diseases affecting fish that cause economic losses in the aquaculture sector.
Sr. No.Causative AgentDiseasesType of Fish InfectedLoss in Production/Economic Loss (%)References
1.Aeromonas bacteriaAeromonas infectionsCarps, freshwater fish80–100% [93]
2.Pseudomonas sp.Strawberry diseaseCarps, rainbow trout, tench 50%[94]
3.ShewanellaputrefaciensShewanellosisCarps, rainbow trout, zebra fish-[93,94]
4. Mycobacterium sp.MycobacteriosisAll freshwater and marine fish 50%[95]
5.FlavobacteriumflavobacterBacterial gill diseasesAll fish 60–70%[96]
6. BirnavirusNecrosisFreshwater fish like salmonids50%[25]
7.RetrovirusAnemiaWalley pike50%[97]
8.RanavirusAnemiaCarp50%[98]
9.MegalocytivirusAnemiaCarp and other freshwater fish60–70%[98]
Due to the severity and varied symptoms of diseases, the total production and diversity of fish are hugely affected, especially in India. Disease outbreaks are mainly due to malpractices in aquaculture. The mentioned causative agents have the ability to cause diseases in a variety of animals and birds as well.
Table 6. Major vaccinations used in commercial aquaculture systems.
Table 6. Major vaccinations used in commercial aquaculture systems.
S. No.SpeciesVaccination againstReference
1Cold-water vibriosis, Classical vibriosis, Listonella anguillarum, Vibrio ordalii[154]
2FurunculosisAeromonas salmonicida subspecies achromogenes[155]
3VibriosisVibrio salmonicida
Vibrio anguillarum
[155]
4Yersiniosis Yersinia ruckeri[154]
5PasteurellosisPhotobacterium damselae subspecie piscicida[154]
6EdwardsiellosisEdwardsiella ictaluri[154]
7Winter ulcer Moritella viscosa[154]
8Streptococcosis/LactococcosisStreptococcus iniae, Lactococcus garviae[154]
Table 7. List of plant-derived vaccines in fish and other animals under development.
Table 7. List of plant-derived vaccines in fish and other animals under development.
S. NoProtein ExpressedExpression SystemTreated AnimalsReference
1.Recombinant major capsid protein (rMCP) of iridovirusRice callusNeoscorpis lithophilus[160]
2.Nervous necrosis virus (NNV) coat proteinTobacco chloroplast Grouper fish[58]
3.AcrV and VapA antigens from Aeromonas salmonicidaChloroplasts of Chlamydomonas reinhardtiiSalmon[172]
4.VP28 from white spot syndrome virusChlamydomonas reinhardtiiPenaeus monodon[173]
5.VP28 from white spot syndrome virusDunaliella salinaCrayfish[174]
6.Virus-like-particle from Atlantic cod nervous necrosis virus (ACNNV)Nicotiana benthamianaSalmonids[175]
7. ORF1 from cardiomyopathy syndrome (PMCV)Nicotiana benthamianaSalmonids[170]
Table 8. Prediction for fish demand for human consumption by 2050 in the top ten fish-producing countries.
Table 8. Prediction for fish demand for human consumption by 2050 in the top ten fish-producing countries.
World Position NumberCountryTotal Consumption (Million Tons)
1China99,875
2India24,601
3United States10,423
4Mexico6061
5Brazil5460
6Nigeria5359
7France3494
8Spain2529
9Peru23,331
10Ghana9121
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kumar, A.; Middha, S.K.; Menon, S.V.; Paital, B.; Gokarn, S.; Nelli, M.; Rajanikanth, R.B.; Chandra, H.M.; Mugunthan, S.P.; Kantwa, S.M.; et al. Current Challenges of Vaccination in Fish Health Management. Animals 2024, 14, 2692. https://doi.org/10.3390/ani14182692

AMA Style

Kumar A, Middha SK, Menon SV, Paital B, Gokarn S, Nelli M, Rajanikanth RB, Chandra HM, Mugunthan SP, Kantwa SM, et al. Current Challenges of Vaccination in Fish Health Management. Animals. 2024; 14(18):2692. https://doi.org/10.3390/ani14182692

Chicago/Turabian Style

Kumar, Avnish, Sushil Kumar Middha, Soumya Vettiyatil Menon, Biswaranjan Paital, Shyam Gokarn, Meghana Nelli, Rakshith Bangalore Rajanikanth, Harish Mani Chandra, Susithra Priyadarshni Mugunthan, Sanwar Mal Kantwa, and et al. 2024. "Current Challenges of Vaccination in Fish Health Management" Animals 14, no. 18: 2692. https://doi.org/10.3390/ani14182692

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

Article Metrics

Back to TopTop