Open access peer-reviewed chapter

Antibiotics in Chilean Aquaculture: A Review

Written By

Ivonne Lozano, Nelson F. Díaz, Susana Muñoz and Carlos Riquelme

Submitted: 11 April 2017 Reviewed: 19 October 2017 Published: 20 December 2017

DOI: 10.5772/intechopen.71780

From the Edited Volume

Antibiotic Use in Animals

Edited by Sara Savic

Chapter metrics overview

2,252 Chapter Downloads

View Full Metrics

Abstract

Aquaculture in Chile has been practiced since the 1920s; however, it was not until the 1990s that aquaculture became an important sector here. Important species in Chilean aquaculture include salmonids, algae, mollusks, and turbot. Salmonids are the dominant species in Chilean aquaculture for both harvest volume and export value, their production reaching greater than 800-thousand tons in 2015. However, this growth has been accompanied by an increase in disease presence, requiring greater drug use to control. This increase in drug use is an environmental and public health concern for the authorities, the salmon industry itself, and the destination markets. In this chapter, we review the literature on drug use, antibiotic resistance, regulatory framework, and alternatives, with focus on Chile.

Keywords

  • aquaculture
  • Salmon
  • antibiotics
  • food safety

1. Introduction: brief history of antibiotics use in Chilean aquaculture

Antibiotics have been used to treat animals since the 1940s, which was soon followed by the appearance of resistant bacteria [1, 2]. In 1969, the House of Lords in the United Kingdom published the “Swann report,” highlighting the excessive use of antibiotics in animals and its potential risks to human and animal health. The Swan report suggested that antibiotic use should be restricted and regulated. The accumulated evidence from Europe and North America supports the notion that antibiotic use should be regulated and restricted to specific clinical situations [2].

In Chile, between 1973 and 1976, the first commercial fish farming of salmonids in the Region of the Lakes was consolidated and has grown ever since [3]. In the subsequent decades, four species of salmonids of commercial importance have been cultivated in Chile: rainbow trout (Oncorhynchus mykiss), Chinook (Oncorhynchus tshawytscha), coho (Oncorhynchus kisutch), and Atlantic salmon (Salmo salar). In the period between 1987 and 2010, four diseases appeared in the salmon farming industry in Chile, coinciding with the beginning of antibiotic use by the industry in 1989. The diseases reported in these years were primarily due to the ectoparasite Caligus and bacterial kidney disease in 1987 [4, 5, 6]; the presence of Piscirickettsia salmonis in 1989 [4]; presenting later high mortalities due to IPN in 1997; and by the outbreaks of the infectious salmon anemia disease in the years 2002, 2007, and 2008 [5].

Sixteen antibiotics are used in animal treatments in Chile, compared to three in the United States (US) and four in Norway [7]. In the case of aquaculture in Chile, antibiotics have been mainly used in sea water Atlantic salmon farming, which accounted for 80% of the total use of antibiotics used for 2015, followed by 11% for coho salmon, 9% for rainbow trout, and 0% for Chinook salmon [8].

The consumption of antibiotics in the salmon industry in Chile has increased by 56% from 2005 to 2015, with a production increase for those years of 23.48%. The highest consumption was recorded in 2014, with a total use of 563.2 tons of antibiotics with 955,179 tons of salmonids produced. In 2016, there was a 30.66% decrease in antibiotic use compared to 2015, using a total of 382.5 tons of antibiotics to produce 727,812 tons of fish ( Table 1 ) [8, 9, 10, 11].

Year Antibiotics use annual quantity (ton) Annual production of salmonids (ton)
2005 239.1 614.435
2006 343.8 647.302
2007 385.6 600.862
2008 325.6 630.647
2009 184.4 474.174
2010 142.2 466.857
2011 206.8 649.492
2012 337.9 836.949
2013 450.7 786.091
2014 563.2 955.179
2015 557.2 846.163
2016 382.5 727.812

Table 1.

Historical consumption of antibiotics in salmon farming industry in Chile [8, 9, 10, 11].

The most used antibiotics in the salmon farming industry in Chile are florfenicol and oxytetracycline. Florfenicol use has increased steadily since 2013 and accounted for 87% and 82.50% of the total antibiotics used in 2015 and 2016, respectively [8, 9, 10, 11]. Florfenicol is mainly used in the seawater stage to control piscirickettsiosis (SRS) caused by the Gram-negative facultative intracellular pathogen P. salmonis. In salmonids, this epizootic disease has high mortality rates (78.9% for Atlantic salmon, 82.9% for rainbow trout, and 59.3% for coho salmon) [9].

In 2005, SRS was the most diagnosed pathology, accounting for 77.04% of the antimicrobials used in the year 2005 by the industry [9]. This trend has been maintained; 89.3% of all antibiotics used for the year 2016 have been for the control of SRS at the seawater stage followed by 6.8% for the control of renibacterosis [10].

Advertisement

2. Antibiotic resistance

2.1. Mechanisms of P. salmonis infection

In the last decade, there have been significant advances in the knowledge of P. salmonis, including aspects of its survival behaviors under stress conditions and genomic data. One of the first achievements has been the culture of this pathogen in the cell-free medium [12, 13]; previously, it had been necessary to develop and maintain cultures of fish cell lines. This progress has allowed the study of the physiology and behavior of this bacterium. An interesting recent finding is that P. salmonis can form biofilms. The development of P. salmonis biofilms occurs under stress conditions and salt concentrations similar to those of seawater. The biofilm matrix of P. salmonis is composed of exopolysaccharides and is disaggregated when treated with cellulases, which are relevant since biofilm formation might be a survival mechanism in the marine environment of this bacterium [14].

It is known that within biofilms, microorganisms are more resistant to the action of chemotherapeutics and have better survival rates under adverse conditions [15]. Recent findings have identified genes that have a role in the formation of P. salmonis biofilms such as the cheA gene [16]. This gene plays a key role in modulating the initiation of bacterial chemotaxis in other bacteria, such as Pseudomonas pseudoalcaligenes KF707 [17]. Using real-time PCR, it has been shown that cheA expression is increased during P. salmonis biofilm development. The results obtained in this research also suggest interaction between the formation of biofilms and the genes involved in the chemotaxis of this pathogen. Biofilm production has been reported as a potential mechanism of pathogenicity in several aquatic bacteria [18, 19]. It is very likely that the first contact with fish for the development of biofilm is produced by chemotactic responses. Chemotaxis to fish mucus has been previously reported as the first step in the development of pathogenic activity [20].

Some authors have suggested that P. salmonis infections begin when bacteria overgrow the skin barrier or gills [21]. In this regard, experimental infections were performed in juveniles of Oncorhynchus mykiss obtained from areas where the presence of SRS has never been reported, infecting them at six different entry sites. These authors found that the main entrance routes are through skin and gills and that the oral route is not used to initiate P. salmonis infection of salmonids [22]. Later, this same research group performed experimental infections in coho salmon (Oncorhynchus kisutch). The results of cumulative mortality and survival analyses showed that the most effective entry portal was the skin, followed by intestinal intubation and finally by gill infection [23]. These findings show that P. salmonis can penetrate and then systemically invade the Coho salmon through the skin and mucous membranes, which appear intact at the macroscopic level, and that the skin is probably the most important site of entry of this bacterium into salmonids. These findings support the notion that biofilm formation initiates colonization of the fish, thereby activating other virulence factors such as proteases to initiate ulcerations and invasion of the organism. P. salmonis is not a motile bacteria and a chemotaxis process could not be activated toward the fish mucus as it happens in other fish pathogens [20]. Studies have shown that other nonmotile pathogenic bacteria can adhere to their host through their net electrostatic charges [24]. Also, nonmotile pathogens infect a host using proteases, and the genes encoding these proteases can be transmitted to nonprotease mutant strains [24].

Recent research carried out on coding and noncoding transcript during an in vivo infection process of Atlantic salmon with P. salmonis identified a common response associated with oxidation-reduction processes, endocytosis, and ion responses. In the different types of analyzed tissues, the clathrin protein, which plays a major role in the formation of coated vesicles, was significantly upregulated in infected individuals, suggesting the importance of clathrin-mediated endocytosis for the bacterial internalization. Moreover, several endocytosis receptors were repressed during the challenge [25].

2.2. Piscirickettsia salmonis resistance to antibiotics

As mentioned above, the use of oxytetracycline and florfenicol are mostly used to control P. salmonis. Florfenicol use almost doubled between 2013 and 2016, suggesting that the battle against this pathogen has been unsuccessful. The evolution of resistance of P. salmonis to antibiotics has been demonstrated. Recent isolates of P. salmonis (SLGO94 and SLGO95) present a higher level of resistance to antibiotics than earlier isolates (LF-89 and EM-90), suggestive of antibacterial resistance [26]. Subsequently, a large-scale study conducted to evaluate the susceptibility profiles for quinolones, florfenicol, and oxytetracycline from 292 field isolates obtained from different farm sites over a 5-year period revealed a high incidence of resistance to quinolones and early resistance to oxytetracyclines and florfenicol [27, 28].

P. salmonis genes encoding membrane-carrying proteins are upregulated in the presence of antibiotics [29]. The P. salmonis genome encodes efflux pumps that enable this bacterium to survive at critical concentrations of florfenicol [30]. Thus, despite the use of antibiotics, there are antibiotic-resistant (especially quinolone) bacteria in sediments near farming areas [31]. These bacteria carry plasmids that confer resistance to quinolones in marine bacteria [32]. Figure 1 shows a proposed model for P. salmonis infection during the seawater stage in salmonids.

Figure 1.

Model of in situ infection of P. salmonis. (A) Biofilms of P. salmonis in culture net; (B) biofilms in microaggregates; (C) transference of resistance genes in sediments; (D) colonization of P. salmonis on fish surface in captivity and onset of pathogenesis by contact with microaggregates or net. Elaborated by: Carlos Riquelme and Victor Sanchez.

Advertisement

3. Analysis of the regulatory framework

In Chile, in its Article 86, the Fisheries and Aquaculture law prohibits the preventive application of antimicrobials in a preventive way in aquaculture, as well as any use harmful to human health. Subsequently, there have also been the supreme decree N°319/2001, Regulation on Protection Measures for the Control and Eradication of High-Risk Diseases for Hydrobiological Species; the exempt resolution N°8228/2015 [33], the Manual of Good Practices in the Use of Antimicrobials and Antiparasitics in Chilean Salmon Farming [34]; the exempt resolution No 5.125/ 2016, Manual on Food Safety and Certification [35]; and the Quality Assurance Program (PAC) for Fisheries and Factories Vessels [36]. This regulatory framework is jurisdiction of SENAPESCA (National Fisheries and Aquaculture Service) institution that is part of and depends on the Ministry of Economy, Development and Tourism.

According to the current legislation, only pharmaceuticals for exclusive veterinary use registered or authorized for application in hydrobiological species can be used [37]. The pharmaceutical products authorized by Servicio Agrícola Ganadero (SAG) are shown in Table 2 [34]. The studies of effectiveness, adequate dosage, animal safety, and human food safety (toxicology) of these authorized pharmaceutical products are not available to the public or were not found.

Generic name Trade name and registry no. Presentation/route of administration Registered by Withdrawal period (degree days) Dose mg/kg lw * /day Tolerance level in muscle tissue (μg kg−1)
Oxolinic acid 80% Reg. N°441 Powder/oral FAV S.A. 450 20 per 10 days 100
Litoflox Reg N°648 Powder/oral Centrovet LTDA. 450 10–30 per 10 days
Bandrol Reg N°481 Powder/oral Veterquímica S.A. 450 10 per 10 days
Amoxicillin 50% Amox-Feed Reg N°121 Powder/oral Veterquímica S.A. 300 70.4 per 10 days 50
Erythromycin 50% Vetromic Reg. N°1402-B Powder/oral Centrovet LTDA. 500 75–100 per 21 days 200
Erythromycin 80% Eritofeed Reg. N°616-B Powder/oral Veterquímica S.A. 500 92.5 per 21 days
Vetromic Reg N°1803-B Powder/oral Centrovet LTDA 500 75–100 per 14–21 days
Flumequine 10% Flumepren Reg. N°79 Powder/oral Centrovet LTDA 300 (−) *** 500
**** 600
Flumequine 50% Reg. N°484 Powder/oral FAV S.A. 300 12–25 per 10–12 days
Reg. N°646 Powder/oral Centrovet LTDA 300 12–30 per 10 days
Flumequine 80% Reg. N°442 Powder/oral FAV S.A. 300 20 per 10 days
Flox-Feed Reg. N°478 Powder/oral Veterquímica S.A. 300 10 per 10 days
Flumepren Reg. N°645 Powder/oral Centrovet LTDA. 600 12–30 per 10–15 days
Florfenicol 50% Florfenox Reg. N°1537 Powder/oral Bayer S.A. 300 10 per 10 days 1000
Veterin Reg. N°1556 Powder/oral Centrovet LTDA. 300 10 per 10 days
Duflosan Reg. N°1769 Powder/oral Veterquímica S.A. 300 10 per 10 days
Florfenicol 50% Duflosan L Reg. N°2264 Solution/oral Veterquímica S.A. 100 10 per 10 days 1000
Aquafen Reg. N°1193 Powder/oral Intervet Chile LTDA. 200 10 per 10 days
Reg. N°1598 Powder/oral FAV S.A. 300 10 per 10 days
Oxytetracycline hydrochloride 20% Terrivet F200 Reg. N°2252 Suspension for injection Veterquímica S.A. 1060 ** 20 200 (−) Tetracyclines
Oxytetracycline 50% Terrivet Reg. N°149 Powder/oral Veterquímica S.A. 600 75 per 15 days
Oxytetracycline 80% Terrivet Reg. N°485 Powder/oral Veterquímica S.A. 600 75 per 15 days
Reg. N°1595 Powder/oral FAV S.A. 600 55–82 per 10 days
Zanil Reg. N°1380 Powder/oral Centrovet LTDA. 600 75 per 10 days
Oxytetracycline 40% Reg. N°309 Powder/oral Laboratorio Veterinario Quimagro S.A. 600 13.57–20.75 per 10 days

Table 2.

Antimicrobials for salmonids authorized by the Veterinary Medicines Registry (SAG) [34].

lw = live weight.


mg/kg/lw.


Trout.


Other salmonids.


The Chilean authority supervises the use of pharmaceutical products in hydrobiological species, and in accordance with the provisions of the general and specific health programs, therapeutic treatments applied to populations of hydrobiological species should be prescribed by a veterinarian and the application of antimicrobials for prophylactic purposes is prohibited. Before the application of the antimicrobials, fish samples should be obtained for subsequent confirmation of the diagnosis by laboratory analysis [37]. Farming facilities should keep records of antimicrobial treatments performed and antimicrobial treatments should be reported monthly through the Aquaculture Inspection System (SIFA) [34].

Extralabel antimicrobials can be prescribed by a veterinarian when the health of an animal is at risk, there is danger of death, or there is suffering of the animal; or when one of the following is fulfilled: dosage, timing, duration of treatment or route of administration for a registered product does not obtain the expected response; the product is temporarily unavailable on the market; or there is no registered product to treat a diagnosed condition [34].

The Manual on Food Safety and Certification (resolution No 5.125/ 2016) describes the norms and procedures that allow to guarantee the sanitary quality of the fishery and aquaculture products destined for international markets along the whole productive chain. Regarding the procedures for the control of residues of pharmaceutical products, each farm facility must demonstrate (and issue a declaration of guarantee) that the concentrations of residues of pharmaceutical products in fish do not exceed the limits established by the Chilean authority [35].

The analysis must be carried out in authorized laboratories, according to the Methods of Analysis of Residues of Pharmaceutical Products and Contaminants for Export Fishery Products. If the maximum allowable limits are not met, the withdrawal period should be extended and a new sampling should be carried out [35].

The government of Chile also maintains a Program of Surveillance and Control of Piscirickettsiosis, in which monitoring system and the application of control measures are established for this disease [38]. Upon request, the Chilean authority issues fish farming centers a certificate stating that the fish are free of antimicrobial and/or antiparasitic treatments [39]. The Quality Assurance Program (PAC) is a voluntary certification program, based on the concept of hazard analysis and critical control point (HACCP), which applies only to fishing plants and factory vessels. This program, however, is mandatory for all companies that are authorized to export to the European Union and the United States. The Chilean authority must approve the quality assurance plan for the industry and supervise its subsequent operation [36]. The administrative procedures, work guides, and specific requirements of this program (PAC) are not publicly available.

The main Chilean salmon markets are the US and Japan. In the case of the US, the Food and Drug Administration (FDA) is in charge of regulating the use of antibiotics in fish, primarily through its regulation 21 Code of Federal Regulations (CFR) 123 ‟Procedures for the Safe and Sanitary Processing and Importing of Fish,” which aims to ensure the safe and sanitary processing of fish and fishery products (seafood), including imported seafood [40]. The regulation mandates the application of HACCP principles to the processing of seafood as a preventive system of hazard control that can be used by processors to ensure the safety of their products to consumers. For the control of drugs for use in food of animal origin, direct medication or for addition to feed must be approved, conditionally approved, or index listed by the FDA (Federal Food, Drug, and Cosmetic Act Section 512) [41].

Under certain conditions authorized by FDA, unapproved new animal drugs may be used in conformance with the terms of an Investigational New Animal Drug (INAD) application (21 CFR 511) [42] and FDA’s Center of Veterinary Medicine (CVM) guide 1240.3025. When a drug is approved by CVM, the condition of the approval is listed on its label or in the labeling (21 CFR 514.1) [43]; this condition specifies the species for which the drug is approved for use, indications for use, dosage regimen, and other limitations such as route of administration and withdrawal time. Labeled withdrawal times must be followed to ensure that no harmful drug residues are present in the edible tissue of the animal when harvested for human consumption; tolerances for some drug residues in the edible tissue have been established [44].

Relatively few drugs have been approved for aquaculture in the US ( Table 3 ). This has led to the inappropriate use of unapproved drugs, general-purpose chemicals, or approved drugs in a manner that deviates from the labeled instructions [44]. Studies establishing the effectiveness, adequate dosage, animal safety, and human food safety (toxicology) of these approved drugs are available to the public [45].

Antibiotic Dose Approved for: Route of administration Tolerance level in muscle tissue Withdrawal period
Terramycin 200 (oxytetracycline dehydrate) 2.5–3.75 g/100 lb. of fish/day
Maximum dose = 8.33 mg/kg/day/10 days
Salmonids: For control of ulcer disease caused by Haemophilus piscium, furunculosis caused by Aeromonas salmonicida, bacterial hemorrhagic septicemia caused by A. liquefaciens, and pseudomonas disease, for control of mortality due to cold-water disease associated with Flavobacterium psychrophilum. Freshwater-reared Oncorhynchus mykiss: For control of mortality due to columnaris disease associated with Flavobacterium columnare Administer in mixed ration for 10 days; do not liberate fish or slaughter fish for food for 21 days following the last administration of medicated feed. 2 ppm (As the sum of tetracycline residues) 21 days for disease control in salmonids
7 days for marking skeletal tissue in Pacific salmon
OxyMarine Oxytetracycline HCL soluble powder-343, TETROXY Aquatic 200–700 mg oxytetracycline/L of water for 2–6 h For marking of skeletal tissues in finfish fry and fingerlings as an aid identification Immersion 2 ppm (as the sum of tetracycline residues) 21CFR 556.500
Sulfadimethoxine/ormetoprim combination Romet-30 For control of furunculosis in salmonids (trout and salmon) caused by Aeromonas salmonicida Medicated feed 0.1 ppm for each drug (21 CFR 556.490) 42 days (21 CFR 558.575)
Florfenicol Aquaflor 10 mg/kg fish per day for 10 consecutive days For the control of mortality in freshwater-reared salmonids due to cold-water disease associated with Flavobacterium psychrophilum Medicated feed 2 ppm in salmon muscle/skin 15 days

Table 3.

FDA aquaculture approved drugs, route of administration, and tolerance levels [40, 44].

In the case of Japan, fishery products are regulated by the Food Sanitation Act and the Food Safety Basic Act. The authorities involved with in the Food Sanitation Act are as follows: the Office of Import Food Safety; Inspection and Safety Division; Pharmaceutical and Food Safety Bureau; Ministry of Health, Welfare and Labor; and the Department of Food Safety. The purpose of the Food Sanitation Act is to prevent the occurrence of health hazard arising from human food so as to contribute to the protection of people health by conducting regulations and measures deemed necessary, from the view point of public health and for securing food safety [46].

The purpose of the Food Safety Basic Act is to promote comprehensive measures to secure food safety by laying down the basic principles of safety for food, defining the responsibility of the government, local authorities, and food-related businesses, clarifying the role of consumers, and establishing basic policies for developing measures. The authority concerned is the Consumer Affair Agency [46].

Antibiotic residue concentrations for edible products from food-producing animals are determined based on jurisdictional-specific regulations that result in the determination of a tolerance or maximum residue level (MRL) for specific drugs in a specific tissue for specific animal species and based on toxicological assessments. This index estimates the amount of substance in food that can be ingested over a lifetime by humans without significant risk to health [47]. There are notable differences among MRLs or tolerances set by the different agencies regarding the two antibiotics most used in Chilean salmon farming ( Table 4 ).

Antibiotic Europe (μg kg−1) MRL e Chile (μg kg−1) f USA tolerance (ppm) g Japan (ppm)
Oxytetracycline a 100 200 c 2 d 0.2
Florfenicol b 1000 1000 1 0.2

Table 4.

Antibiotics used in Chilean salmon farming and their maximum residue limits in salmonids (MRLs).

508/1999/EC.


1322/2001/EC.


As a sum of tetracycline residues [44].


Calculated as oxytetracycline.


[34].


[44].


[48].


Many methods have been developed for analysis of antibiotics in fish. HPLC and mass spectrometry (HPLC-MS/MS) is the most sensitive method for the detection of these antibiotics and is currently regarded as the tool of choice for analysis of antibiotic residues in animal-derived food [47, 49], having a limit of detection (LOD) in fish of 10.3 ng/l [50] and a limit of quantification of 20 ng/l for tetracyclines [51].

Advertisement

4. Alternative antibiotic treatment in salmon farming

Fish are considered as the earliest class of vertebrates to have both innate and adaptive immunity, though the latter defense mechanism is not as elaborate as in higher vertebrates. Unlike in mammals, the alternative complement pathway in teleosts is relatively high and can mediate the lysis of target erythrocytes from several species. These features, along with their potential to function at varying temperatures, suggest that the complement system is a powerful defense mechanism in fish [52, 53, 54], and they are in constant interaction with their surroundings and therefore could easily encounter potential pathogens. In the wild, fish can protect themselves using innate defense mechanisms (either constitutive or responsive) [52].

The various alternatives to the use of antibiotics can be classified according to the action toward the pathogen or host. Pathogen-directed strategies include inhibitors of growth and virulence genes, antibacterial compounds, and the phage therapy. Host-directed strategies include the improvement of health, stress prevention, stimulation of the defense system, and selective breeding for disease resistance [55].

One of the first lines of defense against bacterial infection is the withholding of nutrients, termed nutritional immunity. The most significant form of nutritional immunity is the sequestration of iron [56]. Recent studies have detected a relationship between iron transporter glycoproteins and Salmo salar susceptibility to pathogens [57, 58]. In salmonids, an iron transporter glycoprotein has been identified as a vaccine enhancer [59]. In vertebrates, it has been shown that iron transporter glycoproteins exert antibiofilm therapeutic [60, 61] and antimicrobial activity by binding to iron, thereby preventing its use by bacteria [61, 62, 63, 64] and thus causing alterations in the bacterial wall and, ultimately, death. Because of its cationic nature, this glycoprotein binds to the lipopolysaccharides of Gram-negative bacteria, thereby attenuating those proinflammatory processes induced by bacterial lipopolysaccharides [63]. Among alternative sources of bioactive compounds, ingredients or products derived from marine algae show great potential for use in aquaculture [65]. Rainbow trout–supplemented diets with phytopharmaceutical of herbal and macroalgal origin have improved resistance against P. salmonis [66].

Several bacteriophages have been isolated against the following pathogenic bacteria, Edwardsiella tarda, Edwardsiella ictaluri, Lactococcus garvieae, Pseudomonas plecoglossicida, Streptococcus iniae, Flavobacterium columnare, Flavobacterium psychrophilum, Aeromonas salmonicida, Aeromonas hydrophila, Vibrio anguillarum, Vibrio harveyi, and Vibrio parahaemolyticus, and their potential to be used as a therapeutic agent has been studied by several researchers [67]. In salmonids, Flavobacterium psychrophilum phages have shown protection against bacterial cold water disease in in vitro conditions. Each phage isolate rarely infected F. psychrophilum strains other than the strain used for its enrichment and isolation. Some bacteriophages decrease mortality from intraperitoneal injection of their host strain when added together with the bacteria at a ratio of 10 plaque-forming units per colony-forming unit [68].

Promising results have been obtained in laboratory studies. However, high concentrations of bacteriophages in seawater can induce bacterial genetic variation. This occurs through mutation and bacteriophage-mediated horizontal transmission of genetic material between different bacteria mediated by bacteriophages [5, 69]. The use of phages can also influence bacterial community dynamics and ecosystem biogeochemistry. These influences differ depending on whether phages establish lytic, chronic, or lysogenic infections. The impacts of lysogeny are well studied at the cellular level, but ecosystem-level consequences remain underexplored [70].

Probiotics have been credited for producing improved nutrition, health benefits, reduced disease incidence, improving growth, health status, immunity, feed conversion, microbial balance, and water quality, as well as food production in an environmental-friendly way [71, 72, 73]. Probiotics in aquaculture can be live or dead preparations, including cellular/extracellular components of the microorganism(s), administered either as a feed supplement or to the rearing water. Probiotics can be used to control a range of bacterial pathogens in various fish species [69]. For example, rainbow trout (Oncorhynchus mykiss) were protected against Aeromonas salmonicida and Yersinia ruckeri when administered with dietary Carnobacterium maltaromaticum and C. divergens [72]. The efficacy of Carnobacterium sp. at reducing diseases caused by A. salmonicida, V. ordalii, and Y. ruckeri in salmonids has also been demonstrated [72]. However, there is no solid knowledge regarding the potential of probiotic against P. salmonis.

The Food and Agriculture Organization of the United Nations (FAO) has now highlighted the use of probiotics in aquaculture as a means of improving the quality of the aquatic environment [72]. However, concerns have been voiced about the possible acquisition of antibiotic resistance and virulence genes via horizontal gene transfer, which might lead to safety problems if using live probiotics in an open aquatic environment. Probiotics can also affect host tissue and result in severe cell damage. To avoid this, probiotic strains must be recognized as safe for the cellular integrity of the host [72].

In the aquaculture industry, vaccination strategies include traditional inactivated and attenuated vaccines, as well as next-generation vaccines comprising recombinant, subunit, vectored, genetically engineered, DNA and peptide vaccines, reverse vaccinology, plant-based edible vaccines, and nanovaccines [74]. Current vaccination protocols for P. salmonis include whole cell, inactivated and adjuvant vaccines for injection (primary immunization), followed by oral boost (where the timing of boost delivery is determined by measuring circulating antibody levels against the pathogen). Live vaccines and DNA vaccine studies have been unsuccessful under laboratory conditions. There are more than 25 different vaccines against SRS that are available in the Chilean market. These vaccines confer good short-term protection against disease and mortality but are inefficient at conferring long-term protection, or the duration of protection is insufficient to protect the fish throughout their economic life [75, 76, 77].

Advertisement

5. Conclusions

Veterinarians in charge of the salmon industry in Chile have used large quantities of antibiotics relative to its production volumes. In the years of highest production, an average of 600 g ton−1 produced was used. The antibiotics used by this industry are florfenicol and oxytetracycline for the control of P. salmonis at the seawater stage; studies have demonstrated the resistance of this pathogen to quinolones, oxytetracycline, and florfenicol, as well as their mechanisms of resistance.

There are 12 different types of generic and 25 branded antimicrobials authorized for use in salmonids in Chile, with no specifications related to pathogens or diseases. This is in contrast to the US situation, where the FDA has approved just four antibiotics for specific uses and against certain pathogens.

The studies of the effectiveness, adequate dosage, animal safety, and human food safety (toxicology) of the authorized pharmaceutical products, as established by the Chilean authority, are not available to the public or were not found. This was also the case for the administrative procedures, work guides, and specific requirements of the Quality Assurance Program (PAC), whereas the effectiveness and toxicology studies of the FDA-approved antibiotics are freely available online. This absence of Chilean regulation and antibiotic data is concerning. To avoid chemical hazards and ensure food safety, we propose that a mandatory legal framework based on international regulations is needed in Chilean aquaculture. Antimicrobial treatment is required for an efficient production of animal products; however, antibiotics should never be used as a substitute for proper nutrition and hygiene management.

The alternatives to the use of antibiotics in Chilean salmon farming, such as the use of nutritional immunity, phytopharmaceuticals, probiotics, antimicrobial peptides, and selective breeding for disease resistance, require advanced research with in vivo studies. Although several vaccines have been authorized, this remains an inefficient strategy for the control of pathogens in aquaculture.

References

  1. 1. Aminov RI. A brief history of the antibiotic era: Lessons learned and challenges for the future. Frontiers in Microbiology. 2010;1. DOI: 10.3389/fmicb.2010.00134
  2. 2. Millanao A, Barrientos M, Gómez C, Tomova A, Buschmann A, Dölz H, et al. Uso inadecuado y excesivo de antibióticos: Salud pública y salmonicultura en Chile. Rev Médica Chile. 2011;139:107-118
  3. 3. Méndez ZR, Munita OC, Cortés DR. Salmonicultura en Chile. 1989. [cited 07-05-2017]; Available from: http://agris.fao.org/agris-search/search.do?recordID=US201300717211
  4. 4. Claude M, Oporto J, Ibáñez C, Brieva L, Espinosa PC, Arqueros WM. La ineficiencia de la salmonicultura en Chile: aspectos sociales, económicos y ambientales. Santiago Terram Publicaciones; 2000 [cited 07-05-2017]; Available from: http://www.sidalc.net/cgi-bin/wxis.exe/?IsisScript=BIBACL.xis&method=post&formato=2&cantidad=1&expresion=mfn=025865
  5. 5. Cabello F. Antibioticos Y Acuicultura Un Analisis De Sus Potenciales Impactos Para El Medio Ambiente, La Salud Humana Y Animal En Chile; 2003; Available from: http://www.terram.cl/docs/App17_Antibioticos_y_Acuicultura.pdf
  6. 6. Ibieta P, Tapia V, Venegas C, Hausdorf M, Takle H. Chilean salmon farming on the horizon of sustainability: Review of the development of a highly intensive production, the ISA crisis and implemented actions to reconstruct a more sustainable aquaculture industry. In: Barbara Sladonja editor. Aquaculture and the Environment-A Shared Destiny. Institute of Agriculture and Tourism Porec. Croatia: InTech; 2011 [cited 2017 Jul 28]. Available from: https://www.intechopen.com/download/pdf/25465
  7. 7. Cabello FC. Antibiotics and aquaculture in Chile: Implications for human and animal health. Revista Médica de Chile. 2004;132:1001-1006
  8. 8. Servicio Nacional de Pesca y Acuicultura, Ministerio de Economía, Fomento y Turismo. Informe sobre el uso de antimicrobianos en la salmonicultura nacional 2005-2009. 2011
  9. 9. Servicio Nacional de Pesca y Acuicultura, Ministerio de Economía, Fomento y Turismo. Informe sanitario de salmonicultura en centros marinos. 2015
  10. 10. Servicio Nacional de Pesca y Acuicultura, Ministerio de Economía, Fomento y Turismo. Informe sobre uso de antimicrobianos en la salmonicultura nacional. 2016
  11. 11. Report on the Use of Antimicrobials in National Salmon Farming. 2005. Available from: http://www.sernapesca.cl/presentaciones/Comunicaciones/Informe_Sobre_Uso_de_Antimicrobianos_2015.pdf
  12. 12. Gómez F, Henríquez V, Marshall SH. Additional evidence of the facultative intracellular nature of the fish bacterial pathogen Piscirickettsia salmonis. Archivos de Medicina Veterinaria. 2009 [cited 2017 Aug 1];41. Available from: http://www.redalyc.org/html/1730/173013342011/
  13. 13. Mauel MJ, Ware C, Smith PA. Culture of Piscirickettsia salmonis on enriched blood agar. Journal of Veterinary Diagnostic Investigation. 2008;20:213-214
  14. 14. Marshall SH, Gómez FA, Ramírez R, Nilo L, Henríquez V. Biofilm generation by Piscirickettsia salmonis under growth stress conditions: A putative in vivo survival/persistence strategy in marine environments. Research in Microbiology. 2012;163:557-566
  15. 15. Flemming H-C, Wingender J, Szewzyk U, Steinberg P, Rice SA, Kjelleberg S. Biofilms: An emergent form of bacterial life. Nature Reviews. Microbiology. 2016;14:563-575
  16. 16. Albornoz R, Valenzuela K, Pontigo JP, Sánchez P, Ruiz P, Avendaño-Herrera R, et al. Identification of chemotaxis operon cheYZA and cheA gene expression under stressful conditions in Piscirickettsia salmonis. Microbial Pathogenesis. 2017;107:436-441
  17. 17. Tremaroli V, Fedi S, Tamburini S, Viti C, Tatti E, Ceri H, et al. A histidine-kinase cheA gene of Pseudomonas pseudoalcaligens KF707 not only has a key role in chemotaxis but also affects biofilm formation and cell metabolism. Biofouling. 2011;27:33-46
  18. 18. Ruiz P, Poblete M, Yáñez AJ, Irgang R, Toranzo AE, Avendaño-Herrera R. Cell-surface properties of Vibrio ordalii strains isolated from Atlantic salmon Salmo salar in Chilean farms. Diseases of Aquatic Organisms. 2015;113:9-23
  19. 19. Sundell K, Wiklund T. Effect of biofilm formation on antimicrobial tolerance of Flavobacterium psychrophilum. Journal of Fish Diseases. 2011;34:373-383
  20. 20. Hazen TC, Esch GW, Dimock RV, Mansfield A. Chemotaxis of Aeromonas hydrophila to the surface mucus of fish. Current Microbiology. 1982;7:371-375
  21. 21. Almendras FE, Fuentealba IC, Jones SRM, Markham F, Spangler E. Experimental infection and horizontal transmission of Piscirickettsia salmonis in freshwater-raised Atlantic salmon, Salmo salar L. Journal of Fish Diseases. 1997;20:409-418
  22. 22. Smith PA, Pizarro P, Ojeda P, Contreras J, Oyanedel S, Larenas J. Routes of entry of Piscirickettsia salmonis in rainbow trout Oncorhynchus mykiss. Diseases of Aquatic Organisms. 1999;37:165-172
  23. 23. Smith PA, Rojas ME, Guajardo A, Contreras J, Morales MA, Larenas J. Experimental infection of coho salmon Oncorhynchus kisutch by exposure of skin, gills and intestine with Piscirickettsia salmonis. Diseases of Aquatic Organisms. 2004;61:53-57
  24. 24. Sakai DK. Electrostatic mechanism of survival of virulent Aeromonas salmonicida strains in river water. Applied and Environmental Microbiology. 1986;51:1343-1349
  25. 25. Valenzuela-Miranda D, Gallardo-Escárate C. Novel insights into the response of Atlantic salmon (Salmo salar) to Piscirickettsia salmonis: Interplay of coding genes and lncRNAs during bacterial infection. Fish & Shellfish Immunology. 2016;59:427-438
  26. 26. Rozas M, Enríquez R. Piscirickettsiosis and Piscirickettsia salmonis in fish: A review. Journal of Fish Diseases. 2014;37:163-188
  27. 27. Saavedra J, Hernandez N, Osses A, Castillo A, Cancino A, Grothusen H, et al. Prevalence, geographic distribution and phenotypic differences of Piscirickettsia salmonis EM-90-like isolates. Journal of Fish Diseases. 2017;40:1055-1063
  28. 28. Henríquez P, Kaiser M, Bohle H, Bustos P, Mancilla M. Comprehensive antibiotic susceptibility profiling of Chilean Piscirickettsia salmonis field isolates. Journal of Fish Diseases. 2016;39:441-448
  29. 29. Cartes C, Isla A, Lagos F, Castro D, Muñoz M, Yañez A, et al. Search and analysis of genes involved in antibiotic resistance in Chilean strains of Piscirickettsia salmonis. Journal of Fish Diseases. 2016 [cited 2017 Aug 1]; Available from:. DOI: http://onlinelibrary.wiley.com/doi/10.1111/jfd.12579/full
  30. 30. Sandoval R, Oliver C, Valdivia S, Valenzuela K, Haro RE, Sánchez P, et al. Resistance-nodulation-division efflux pump acrAB is modulated by florfenicol and contributes to drug resistance in the fish pathogen Piscirickettsia salmonis. FEMS Microbiology Letters. 2016;363:fnw102
  31. 31. Buschmann AH, Tomova A, López A, Maldonado MA, Henríquez LA, Ivanova L, et al. Salmon aquaculture and antimicrobial resistance in the marine environment. PLoS One. 2012;7:e42724
  32. 32. Tomova A, Ivanova L, Buschmann AH, Godfrey HP, Cabello FC. Plasmid-mediated quinolone resistance (PMQR) genes and Class 1 integrons in quinolone-resistant marine bacteria and clinical isolates of Escherichia coli from an aquacultural area. Microbial Ecology. 2017:1-9
  33. 33. Servicio Nacional de Pesca y Acuicultura, Ministerio de Economía, Fomento y Turismo. Resolución exenta N°8228, programa sanitario general para uso de antimicrobianos en la salmonicultura y otros peces de cultivo. 2015
  34. 34. National Fisheries and Aquaculture Service, Ministry of Economy, Development and Tourism. Manual of Good Practices in the Use of Antimicrobials and Antiparasitics in Chilean Salmon Farming. 3rd ed2015 Available from: http://www.sernapesca.cl
  35. 35. National Fisheries and Aquaculture Service, Ministry of Economy, Development and Tourism. Manual of Food Safety and Certification. Available from: http://www.sernapesca.cl/index.php?option=com_content&view=article&id=133&Itemid=295
  36. 36. Servicio Nacional de Pesca y Acuicultura, Ministerio de Economía, Fomento y Turismo. Programa de Aseguramiento de la Calidad (PAC) para Plantas Pesueras y Buques Factorías. 2007. Available from: http://www.sernapesca.cl/index.php?option=com_content&view=article&id=273&Itemid=474
  37. 37. Servicio Nacional de Pesca y Acuicultura, Ministerio de Economía, Fomento y Turismo. D.S. N°319 de 2001, Reglamento de medidas de protección, control y erradicación de enfermedades de alto riesgo para las especies hidrobiológicas. (última modificación D.S. N°74-2016) (F.D.O, 23-08-2016). 2001
  38. 38. National Fisheries and Aquaculture Service, Ministry of Economy, Development and Tourism. Specific Sanitary Surveillance and Control Program for Piscirickettsiosis; 2013. Available from: http://www.sernapesca.cl/index.php?option=com_remository&Itemid=246&func=fileinfo&id=6726
  39. 39. National Fisheries and Aquaculture Service, Ministry of Economy, Development and Tourism. Procedure for certification of fish free of antimicrobial and/or antiparasitic treatments for fish farm centers, CER-FAR-MP1. 2017. Available from: http://www.sernapesca.cl/index.php?option=com_remository&Itemid=246&func=startdown&id=20475
  40. 40. CFR—Code of Federal Regulations Title 21 [cited 2017 Apr 26]. Available from: https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=558.450
  41. 41. FD&C Act Chapter V: Drugs and Devices [cited 2017 Oct 14]. Available from: https://www.fda.gov/regulatoryinformation/lawsenforcedbyfda/federalfooddrugandcosmeticactfdcact/fdcactchaptervdrugsanddevices/default.htm
  42. 42. CFR—Code of Federal Regulations Title 21 [cited 2017 Oct 14]. Available from: https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?CFRPart=511
  43. 43. CFR—Code of Federal Regulations Title 21 [cited 2017 Oct 14]. Available from: https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=514.1
  44. 44. Food and Drug Administration, Center for Food Safety and Applied Nutrition, Office of Food Safety. Fish and Fishery Products Hazards and Control Guidance, 4th ed. 2011. Available from: http://wwwfda.gov/FoodGuidances
  45. 45. Medicine C for V: Aquaculture—Approved Aquaculture Drugs [cited 2017 Jun 24]. Available from: https://www.fda.gov/animalveterinary/developmentapprovalprocess/aquaculture/ucm132954.htm
  46. 46. Japan External Trade Organization. Handbook for Agricultural and Fishery Products Imports Regulations; 2010
  47. 47. Baynes RE, Dedonder K, Kissell L, Mzyk D, Marmulak T, Smith G, et al. Health concerns and management of select veterinary drug residues. Food and Chemical Toxicology. 2016;88:112-122
  48. 48. Japanese Ministry of Health Welfare and Labor. Food Safety Information; 2008. Available from: http://www.mhlw.go.jp/english/topics/foodsafety/index.html
  49. 49. Cañada-Cañada F, de la Peña AM, Espinosa-Mansilla A. Analysis of antibiotics in fish samples. Analytical and Bioanalytical Chemistry 2009;395:987-1008
  50. 50. Dasenaki ME, Thomaidis NS. Multi-residue determination of seventeen sulfonamides and five tetracyclines in fish tissue using a multi-stage LC–ESI–MS/MS approach based on advanced mass spectrometric techniques. Analytica Chimica Acta. 2010;672:93-102
  51. 51. Kemper N. Veterinary antibiotics in the aquatic and terrestrial environment. Ecological Indicators. 2008;8:1-13
  52. 52. Kiron V. Fish immune system and its nutritional modulation for preventive health care. Animal Feed Science and Technology. 2012;173:111-133
  53. 53. Morales B, Macgayver M, Rondón-Barragán IS. Comparative biology of the complement system in fish. CES Medicina Veterinaria y Zootecnia. 2011;6:74-90
  54. 54. Ellis AE. Innate host defense mechanisms of fish against viruses and bacteria. Developmental and Comparative Immunology. 2001;25:827-839
  55. 55. Defoirdt T, Sorgeloos P, Bossier P. Alternatives to antibiotics for the control of bacterial disease in aquaculture. Current Opinion in Microbiology. 2011;14:251-258
  56. 56. Skaar EP. The battle for iron between bacterial pathogens and their vertebrate hosts. PLoS Pathogens. 2010;6. DOI: 10.1371/journal.ppat.1000949
  57. 57. Tarifeño-Saldivia E, Valenzuela-Miranda D, Gallardo-Escárate C. In the shadow: The emerging role of long non-coding RNAs in the immune response of Atlantic salmon. Developmental and Comparative Immunology. 2017;73:193-205
  58. 58. Valenzuela-Muñoz V, Gallardo-Escárate C. Iron metabolism modulation in Atlantic salmon infested with the sea lice Lepeophtheirus salmonis and Caligus rogercresseyi: A matter of nutritional immunity? Fish & Shellfish Immunology. 2017;60:97-102
  59. 59. Cecchini S, Caputo AR. Potential use of bovine lactoferrin as adjuvant for the modulation of the specific immune response in rainbow trout (Oncorhynchus mykiss W.). Aquaculture International. 2017;25:223-232
  60. 60. Ammons MC, Copié V. Mini-review: Lactoferrin: A bioinspired, anti-biofilm therapeutic. Biofouling. 2013;29:443-455
  61. 61. Dashper SG, Pan Y, Veith PD, Chen Y-Y, Toh EC, Liu SW, et al. Lactoferrin inhibits Porphyromonas gingivalis proteinases and has sustained biofilm inhibitory activity. Antimicrobial Agents and Chemotherapy. 2012;56:1548-1556
  62. 62. González-Chávez SA, Arévalo-Gallegos S, Rascón-Cruz Q. Lactoferrin: Structure, function and applications. International Journal of Antimicrobial Agents. 2009;33:301-3e1
  63. 63. Yen C-C, Shen C-J, Hsu W-H, Chang Y-H, Lin H-T, Chen H-L, et al. Lactoferrin: An iron-binding antimicrobial protein against Escherichia coli infection. Biometals. 2011;24:585-594
  64. 64. Yen M-H, Chiu C-H, Huang Y-C, Lin T-Y, et al. Effects of lactoferrin-containing formula in the prevention of enterovirus and rotavirus infection and impact on serum cytokine levels: a randomized trial. Chang Gung Medical Journal. 2011;34:395-402
  65. 65. Lozano I, Wacyk JM, Carrasco J, Cortez-San Martín MA. Red macroalgae Pyropia columbina and Gracilaria chilensis: Sustainable feed additive in the Salmo salar diet and the evaluation of potential antiviral activity against infectious salmon anemia virus. Journal of Applied Phycology. 2016;28:1343-1351
  66. 66. Hernández AJ, Romero A, Gonzalez-Stegmaier R, Dantagnan P. The effects of supplemented diets with a phytopharmaceutical preparation from herbal and macroalgal origin on disease resistance in rainbow trout against Piscirickettsia salmonis. Aquaculture. 2016;454:109-117
  67. 67. Choudhury TG, Nagaraju VT, Gita S, Paria A, Parhi J. Advances in bacteriophage research for bacterial disease control in aquaculture. Reviews in Fisheries Science & Aquaculture. 2017;25:113-125
  68. 68. Castillo D, Higuera G, Villa M, Middelboe M, Dalsgaard I, Madsen L, et al. Diversity of Flavobacterium psychrophilum and the potential use of its phages for protection against bacterial cold water disease in salmonids. Journal of Fish Diseases. 2012;35:193-201
  69. 69. Romero J, Feijoó CG, Navarrete P. Antibiotics in aquaculture–Use, abuse and alternatives. In: Health and Environment in Aquaculture. InTech; 2012 [cited 2017 Aug 7]. Available from: https://www.intechopen.com/download/pdf/35141
  70. 70. Howard-Varona C, Hargreaves KR, Abedon ST, Sullivan MB. Lysogeny in nature: mechanisms, impact and ecology of temperate phages. The ISME Journal. 2017;11:1511-1520
  71. 71. Merrifield DL, Dimitroglou A, Foey A, Davies SJ, Baker RT, Bøgwald J, et al. The current status and future focus of probiotic and prebiotic applications for salmonids. Aquaculture. 2010;302:1-18
  72. 72. Sharifuzzaman SM, Austin B. Probiotics for disease control in aquaculture. Diagnosis and Control of Diseases of Fish and Shellfish. 2017:189-222
  73. 73. Das S, Mondal K, Haque S. A review on application of probiotic, prebiotic and synbiotic for sustainable development of aquaculture. Growth. 2017;14:15
  74. 74. Dadar M, Dhama K, Vakharia VN, Hoseinifar SH, Karthik K, Tiwari R, et al. Advances in aquaculture vaccines against fish pathogens: Global status and current trends. Reviews in Fisheries Science & Aquaculture. 2017;25:184-217
  75. 75. Evensen Ø. Immunization strategies against Piscirickettsia salmonis infections: Review of vaccination approaches and modalities and their associated immune response profiles. Frontiers in Immunology. 2016;7. DOI: 10.3389/fimmu.2016.00482
  76. 76. Maisey K, Montero R, Christodoulides M. Vaccines for piscirickettsiosis (salmonid rickettsial septicaemia, SRS): The Chile perspective. Expert Review of Vaccines. 2017;16:215-228
  77. 77. Cabello FC, Godfrey HP, Buschmann AH, Dölz HJ. Aquaculture as yet another environmental gateway to the development and globalisation of antimicrobial resistance. The Lancet Infectious Diseases. 2016;16:e127-e133

Written By

Ivonne Lozano, Nelson F. Díaz, Susana Muñoz and Carlos Riquelme

Submitted: 11 April 2017 Reviewed: 19 October 2017 Published: 20 December 2017