1Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Barrackpore-700120, West Bengal, India

2Department of Zoology, The University of Burdwan, Golapbag, Burdwan-713104, West Bengal, India

3Regional Research Station-Rahara, ICAR-Central Institute of Freshwater Aquaculture, Kolkata-700118, West Bengal, India

Corresponding Author Email: beherabk18@yahoo.co.in

DOI : https://doi.org/10.58321/AATCCReview.2022.10.02.39

Keywords

and cellular organization, Aquaculture, diagnostic methods, histological, immunological study, molecular tools, parasitic diseases

Download this article as:

Abstract

Aquaculture is considered an integral part of achieving food security and poverty reduction by providing nutritional, and livelihood security to millions of people around the world. However, poor health management and a lack of proper diagnostic facilities create problems in the early identification of pathogens leading to mass mortality in fish. However, fish, like any animal, are exposed to and susceptible to a wide range of diseases and parasites. Parasitic diseases of fish are usually encountered more often than microbial diseases. From 30%-50% of the cases received at several fish disease, diagnostic laboratories involve parasites. Some of the parasites could not be differentiated by conventional diagnostic methods from other phenotypically similar pathogens of the same genera. Some attempts have been made using biochemical tests, DNA homology, and protease variability. Moreover, most molecular technologies are mostly lab-based and need time to provide information on parasitic infections. Additionally, such technology couldn’t be used in the field, providing direct information on potential parasitic pathogens to adopt immediate management measures and avoid disease outbreaks and production loss. However, in the last fifteen years or so, great advances have taken place in understanding the molecular biology of fish pathogens and their hosts. Among them, vast fields of molecular study like Nucleic acid-based approach, PCR, ITS, RT-PCR, LAMP, Luminex, RAPD, AFLP, RFLP, and microsatellites have been used for parasitic disease diagnosis. Detection of nucleic acid molecules has demonstrated its usefulness for highlighting hardly cultivable, non-cultivable, and even dead microorganisms, generating appropriate novel or replacement technologies. Thus, a better understanding of molecular tools developed for the detection of specific parasites would be helpful to increase diagnostic precision, aid carrier detection, and promote species discoveries in different parasitic groups. Further, studies are still necessary to increase our knowledge about molecular tools in the detection of a diverse group of parasites associated with the disease in fish and also from fish to humans.

  1. Introduction

Aquaculture is the highest-growing sector in the field of fishery sciences. Fish (both Chondrichthyes and Osteichthyes) that is high in protein is widely consumed for human consumption throughout the Indian subcontinent, which significantly boosts the region’s economy. But, nowadays several pathogens like parasites, bacteria, and fungi manifest fishes and adversely affect fish’s healthy growth [1-7]. The traditional method for disease diagnosis is carried out through macroscopic as well as microscopic examination for the primary screening procedures of the causative pathogens [8-9]. The conventional diagnostic protocol has been discovered for the same clades of pathogens, and genera [10]. With few diagnostic tools like DNA homology analysis and biochemical assays [11-13], a few consequences arise, and previously isolated pathogens are lesser sensitive to these pathogenicity tests. In the last two decades, molecular tools are the detection of fish parasitic pathogens, and their hosts are widely accepted. Molecular-level diagnostic tools are widely accepted methods for a resistor of fish illnesses and their epidemiology of parasitic, bacterial, fungus, and viral diseases [14]. Recognition of nucleic acid assays are also useful for microbes detection [3]. The parasites are differentiated into seven different taxa they are- Subkingdom Protozoa (Phylum Rhizopoda, Phylum Mastigophora, Phylum Ciliophora, Phylum Microspora, Phylum Apicomplexa), Phylum Cnidaria, Phylum Platyhelminthes (Turbellaria, Monogenea, Digenia, Cestoda), Phylum Nematoda, Phylum Acanthocephala, Phylum Annelida and Subkingdom Arthropoda (Branchiura, Copepoda, Malacostraca (order Isopoda) [15]. The fish specimens are highly susceptible to different parasitic diseases, they are economically significant in many tropical and sub-tropical countries. The impact of the different diseases in aquaculture is difficult to analyze by regular examination in case of mortality. The prime parasitic diseases documented in freshwater fishes (salmonids, cyprinids, and eels) are white spot diseases (ich), trichodiniasis, chilodonellosis, gyrodactylosis, dactylogyrosis, argulosis, microsporidiosis [16-17], and several marine fishes (seabass, sea bream, turbot) aretrichodiniasis, costiasis, ceratomyxosis, amyloodiniosis, mycrocotylosis, and sea lice disease [18]. A few parasites are caused severe damage to the pisciculture, like- Amyloodinium sp.(Dinoflagellates), Scuticociliatida (Ciliates), Enteromyxum sp. (Myxosporea), and Mycrocotylidae (Monogenea). However, their pathological signs should not be negligible and concern the enhancement of cultures and their deleterious effects, which are not the possible cause of high mortality [19].

In the parasitological research field, laboratory tools depend on traditional processes like- light microscopy for the morphological identification in the parasitological study [20]. Molecular biological research are based on the detection of parasites responsible for disease diagnosis [21]. Conventional studies have the merit of being cheaper because they do not require costly chemicals and instruments. Simply completed when a skilled microscopist is able. In another way, molecular techniques opined the incidence of parasitic worms involved in their antigenic determinants or DNA parts [22]. These assessments are not involved in ecological attributes and may often interfere with the possible outcome of a faecal samples test [23].

Recently, the determination and diagnosis of parasitic infestations depend on several laboratory purposes and scientific signs; including medical history, travel history, and geographic coordination of fish species [18]. In another way, new diagnostic kits have shown new ways to the improvement of parasite detection [18]. Firstly, serological analyses that are more precise and highly sensitive such as the luciferase immunoprecipitation system (LIPS), rapid antigen detection system (RDTS), and Dot-ELISA [24]. Secondly, molecular tools like loop-mediated isothermal amplification (LAMP) [25], Luminex, and real-time polymerase chain reaction (RT-PCR) [26] have shown the highest potentiality for use in parasitic determination which enhances specificity and sensitivity. The molecular assays that identify parasites include PCR, RAPD (Dominant marker), AFLP (Dominant marker), RFLP (Co-dominant marker), microsatellite marker method, Luminex xMAP-based technology, LAMP, and the currently added RT-PCR [25, 27-33]. In this review, we mainly focus on different molecular techniques and their uses in the detection of fish parasitic pathogens.

  • Fish parasitic disease
    • Importance of disease

Fish diseases indicate poor quality of water and poor management systems. The addition of dissolved oxygen (DO) leads to hypoxic conditions in the aquatic fauna. Fish diseases can be significant since they have the potential to cause mortality. Sublethal illnesses can also result in poor growth, poor flesh quality, and unfavourable alterations. Additionally, some fish infections can spread to humans.

  • Parasites’ major causes of concern

Parasites are living things that eat from and dwell inside of other living things, such as your body. A bug bite, contaminated food or water, or sexual contacts are all ways to contact them.

The size of parasites varies, from the microscopic protozoa (one-celled creatures) to the visible worms. The United State is home to some parasite illnesses. Infections with Giardia can result from contaminated water sources. Toxoplasmosis, which can be harmful to expectant mothers, can be spread by cats. Others, like malaria, are widespread around the world (NIH 2017). For diseases caused by parasites, there are no vaccinations. Certain medications can treat parasite infections.

  • Host-parasite relationship

Host-parasite co-evolution leads to host or parasitic adaptation in spatiotemporally changing situations, e.g. when aggressive species express their variety [34]. A major factor that explains the achievement of these species is the blown of the again-originated populations from the pressure worked out by their original parasites are left behind in the original environment [35-36]. This model states the knowledge that co-evolution primes to widespread resident adaptations, and as such the parasites in a specific area contaminate hosts from the same area more proficiently than they infect hosts from another geographically diverse population [37-38]. In another way, environments are not parasite friendly, host-parasite interactions lead to parasite mal-adaptation, where parasites can show their highest capability in the native (allopatric) hosts than in the sympatric hosts [39-40].

Host defence against the pathogenic which is a well-regulated inflammatory retort signed by leukocyte movements into the site of infection, resolution of inflammation response, and repairing of the tissue architecture [17, 41]. Generally, the association between the host and the parasite governs the consequence of the dirt. Some parasites penetrate the host’s immune systems and hide intracellularly, like nematode and cestode species, and others invade the cell immunity including extracellular parasites such as Argulus sp., Ichthyophthirius multifiliis, and monogeneans species [41].

Many molecules are triggering signalling pathways, where parasite and host cells are critical for parasitic entrance, leading to the parasites’ survival [17]. An important advance has been realized in several factors that are key features in parasite virulence and the pathogenesis of the diseases they cause. Among the most extensively studied of these are parasitic conjugate proteases [16]. Parasitic proteases can play a wide role that establishing, maintaining and exacerbating an infection [41]. Parasitic protozoans were attacked and migrated a variation of tissues and organs, regardless of being intracellular or extracellular parasites. The mechanisms for connective tissue distortion might be vital for parasite survival [41].

  • Fish immune response to parasitic infection

Teleosts show innate and adaptive immunity. Fishes provide an innate immune system, which is the first line of immune defence. Adaptive immunity that highly diverse T and B cell receptors encode recombinant activation genes (RAGs) which caused infections [42-43]. Innate immune response recognition is based on pathogen-associated molecular patterns (PAMPs) by pathogen-recognizing receptors (PRRs). PRRs which are Toll-like receptors (TLRs) (Fig. 1) in nature that helps in molecules like PAMPs are well in the field of fish parasitology.

C-type lectin receptors (CLRs) are useful against host-parasitic interactions. Most of the CLRs are involved in antigen presentation, and capture [44-46]. CLRs guided intracellular internalization motifs, in other ways CLRs consist of ITIM (immunoreceptor tyrosine-based inhibitory motif) or, ITAM (immunoreceptor tyrosine-based activation motif)-like motifs in their cytoplasmic domains, elicit the capability of immune-activation or immune suppressive functions of these receptors. In the general structure of carbohydrate recognition domains (CRDs) they are divided into diverse families including C, F, P, I-type lectins, galectin, and pentraxin etc [47]. However, the mannose receptor (MR), macrophage galactose-type C-type lectin (MGL), dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), DEC205, and Dectin-1 are the most significant molecules from the CLR family [45]. Antigen-presenting cells APCs, such as macrophages or dendritic cells, must first recognise an invading pathogen to determine the type of effector T cell that will then mediate an immune response [48]. Highly specialised receptors, such as a variety of pattern recognition receptors (PRRs), such as C-type receptors (CLRs) and Toll-like receptors, are present in APCs (TLRs). When these receptors bind to conserved pathogen structures known as pathogen-associated molecular patterns, APCs are activated and mature, which is a critical step in the process (PAMPs). CLRs recognise and internalise particular carbohydrate antigens articulated by pathogens and host tissues in a Ca2+-dependent manner, in contrast to TLRs [44, 46-47, 49]. Interactions between proteins and carbohydrates play significant roles in two separate facets of the immune response. Both pathogen recognition and the cellular interactions that result in pathogen neutralisation depend on these interactions [50]. By identifying and attaching particular carbohydrate moieties (typically a nonreducing terminal monosaccharide or oligosaccharide) on the surface of potential pathogens through CRDs, lectin receptors play a significant role in the innate immune response [49]. CRDs can recognise carbohydrate moieties and trigger agglutination, immobilisation, complement-mediated opsonization, and lysis when combined with other domains [46] (Fig. 2).

Fig. 1 In this schematic presentation TLR cells MyD88 and TAM are situated in the cytoplasmic surface. Then, TIRAP binds with MyD88 and TRIF binds to several immune cells like TRAF6, IRF3, NF-KB, and MAPK. Finally, NF-KB elicits the secretion of inflammatory cytokines, which fight against inflammation. In another way, IRF3 increases type 1 IFNs production, which enters the nucleus, and further inflammation process is carried forward.       

Fig. 2 Structureof membranes of the C-type lectin (DC-SIGN, MR, Dectin-1, and mMGL). These receptors contain one or more carbohydrate-recognition domains (CRDs), transmembrane domains, and a cytoplasmic domain that may contain a tyrosine-based motif, a triad of acidic amino acids, dileucine motif, and an immunoreceptor tyrosine-based inhibitory motif (ITIM) [45].

Inflammatory reactions of phagocytosis and their activity are involved in ciliates, flagellates and finally myxozoans [51-53]. Few humoral parameters like peroxidases, lysozyme and acute-phase proteins are involved in some parasites (Fig. 3).      

Ciliates like Chilodonella, Ichthyophthirius, Trichodina, Uronema, Tertrahymena, and Epistylus are obligate parasites and facultative in forms. Some ciliate parasites are highly pathogenic, they are highly prone to mortalities. Due to such harsh conditions, hosts are evolving an effective response to combat parasitic diseases [53]. The flagellate parasitic infection does not depend on the age and size of the fish; it is measured by a dominant gene. The α2-macroglobulin in the metalloprotease is blood neutralised and secreted by the pathogenic C. salmositica. Humoral (e.g. complement-fixing antibodies to lyse the parasite) and cell-mediated (e.g. T-cell cytotoxicity, phagocytosis) are part of the protective mechanism in acquired immunity [51].

Fig. 3 Physical barriers produce acquired immunity to prevent pathogen entering. The phagocytic process activates during cell-mediated defence mechanism, which turns further activates macrophage and dendritic cells. At last, immunomodulation comes to B and T lymphocytes, B lymphocytes enhance antibody production which creates a defence process to resist pathogen destruction. The humoral defence mechanism elicits antimicrobial enzymes and non-specific proteins which activate the complement system, and finally, antibody shows a defence mechanism against causative pathogens. In, the inflammation process activates the host immune system for pathogenic distribution.         

2.5 Development of a vaccine against parasites of fish

Vaccination is widely useful in aquaculture for a few fish species [54-56]. Molecular techniques against fish vaccines are widely available, but it is most useful for antiviral vaccines [57]. Immunological aspects against I. multifiliis is a mostly humoral immune response, it is abundant in parasitic membrane proteins called immobilization antigens (i-antigens) [58-59]. Purified i-antigen confers activity immune system as well [60].

DNA vaccines are lesser proven against Ichthyophthirius, due to their enormous potential against a different variety of microbial agents. A similar strategy is used against fish parasites, those fishes that have economic importance in the aquaculture sector. Cryptobia salmositica is a flagellate parasite, that has protective immunity and vaccination capability [61].    

Lepeophtheirus salmonis is sea lice, that are an economically important parasite of farmed salmonids. In addition, injection of extracts of the parasitic induction of exact Abs in the same fish [62]. Antibody stimuli are unable to protect against adult and preadult stages. Partial protection against fish louse is not a replication of treatment groups. As per the available study, the vaccination of salmon is useful against this crustacean [63]. Vaccination development is still in a dark position, Abs could target critical host-parasitic interactions that are not classified.

The knowledge gathered from omics data can be quite helpful for creating multivalent vaccines. A multivalent vaccine combines numerous antigens to stimulate the host’s immune system in a broad-spectrum protective manner [64]. The same parasite, various parasites, parasite strains, and phases of parasite development can be used to select distinct antigens [65]. A multivalent vaccination would generally be an efficient method of combating aquatic parasites given the myriad of serious issues connected to them, especially the variable antigenic profile of the developmental stages and parasite strains. Although it is hypothesised that fish may be protected from infection by cell-mediated immune responses for some parasites, such as T. bryosalmonae [66], the precise protective response is still unknown. In such cases, investigations on the parasite transcriptome, genome, and proteome as well as information on host-parasitic interactions might be used to identify and target many antigenic possibilities [65]. Genomic, transcriptomic, structural proteomic, and immunoproteomic approaches can be employed to find an appropriate antigen while creating a multivalent vaccine. Illustrates a strategy for the assortment of antigenic candidates and the creation of vaccines using omics techniques (Fig. 4). In this method, the targeted parasite or the infected host tissue can be started with data from whole-genome sequencing, RNA sequencing, and proteomics. The data can be further annotated using various bioinformatics tools, including Blast, Blast2GO, and UniProt/Swiss-Prot. After annotation, the function of the gene, RNA transcripts, and proteins, as well as the many biological pathways, may be examined. Fish can be given vaccine candidates to test the vaccine’s potency and effectiveness.

Fig. 4 The desired parasite or tissue from the infected host fish can be used to extract the desired material (DNA, RNA, or protein). The isolated material is used as a sample for Edman sequencing, spectrophotometry, and next-generation sequence analysis (for DNA, RNA and Protein). The immunogenic targets (capable of inducing host immune response) can be chosen and their protective epitopes predicted based on molecular function and biological pathway analysis. Consequently, the vaccine created in this manner may be employed. For the study and functional annotation of parasite compounds, various bioinformatic methods can be applied to the acquired sequences. Following their mass production, these compounds can be coupled with the appropriate adjuvants to create vaccines. Consequently, the vaccine created in this manner may be employed [65].

  • Economic losses due to parasitic diseases

Parasite infection indicates a population bottleneck in the development of the aquaculture sector [67]. Therefore, disease outbreaks are thought to affect farms’ production and financial health by affecting feed conversion, increasing host mortality, and decreasing animal welfare [68]. Clinical disease outbreaks result from a number of cues relating to the manufacturing process, host susceptibility, and the virulence of the specific pathogen [69]. Coincidentally, disease resistance and appropriate measures are crucial for the expansion of the sustainable aquaculture business, and these must be put into practice with the cooperation of farmers and authorities on animal health [67].        

  • Timely diagnosis is of utmost importance

The methods of medical treatment and prevention used in fish are quite specific and frequently dissimilar from those used in warm-blooded species. They necessitate in-depth familiarity with fish environments. A comprehensive understanding of the aetiology of the disease and the biology of the host (fish) forms the basis of the complex combination of therapies that make up preventive plans. It focuses on reducing or eliminating infection (invasion) sources, and the likelihood of further spread, as well as improving fish organism health so that it can tolerate infection (invasion). The primary factor in the elimination of diseases is prevention. For a lot of disorders, no specific treatments have yet been created, hence the use of efficient diagnostic methods is needed.

  • Methods used for identification of fish parasites
    • Traditional approaches

3.1.1 Microscopy

Microscopy is a primitive and well-accepted tool for detecting parasites and tissue specimens [70-71]. For the direct microscopy method, observation is very much time taking, laborious which needs a highly specialized laboratory technician. In endemic areas, where resources are concise these were observed difficulties and also misdiagnosed. Still, several helminthic infections are solely dependent on microscopy-based studies and also dependent on molecular studies (Fig. 5).

Table. 1 Microscopy-based detection of fish parasite

Sl no.Name of parasitesHostInfected organReference
1.Amyloodinium sp.Amphiprion percula (Lacepède, 1802)Head, and caudal fin[72].
2.Chilodonella sp.Carassius auratus (Linnaeus, 1758)Gill, and fin[73].
3.Ichthyophthirius multifiliisSchizothorax macropogon (Regan 1905)Skin, and fin[74].
4.Trichodina sp.Carassius auratus (Linnaeus, 1758)Gill[75].
5.Myxobolus sp.Salminus franciscanus (Agassiz 1829)Fin[76].
6.Lernaea sp.Rhinogobius similis (Gill, 1859)Skin[76].
7.Caligus sp.Lutjanus erythropterus (Bloch, 1790)Skin[77].
8.Argulus sp.Schizothorax richardsonii (Gray, 1832)Skin[78].
9.Trypanosoma sp.Platydoras armatulus (Valenciennes, 1840)Blood[79].
10.Eimeria sp.Chloroceryle americana (Gmelin, 1788)Intestine[80].
11.Henneguya sp.Prochilodus lineatus (Valenciennes, 1837)Gill tissue[81].
12.Dactylogyrus sp.Osteobrama cotio (Hamilton, 1822)Gill[82].
13.Gyrodactylus sp.Oreochromis niloticus (Bloch 1790)Fin[83].

Fig. 5 Microscopical observation of parasite (A) and molecular characterization of the isolated parasites (B).           

  • Histology

The histological study is co-related with microscopic examination like thin or thick tissue staining process and also by using staining for the histopathological study to determine the changes due to pathogens infections. Histological screening is a very common diagnostic tool for aquatic animals [84]. The direct smear method is the first step of the examination for parasite screening, if parasites were observed then performed next level analysis fixation and stained for confirmatory diagnosis. Giemsa staining is widely used for many parasite staining [85], and also for blood parasites like Plasmodium sp., Trypanosoma sp., etc. Tissue stains like haematoxylin and eosin and PAS help in parasite identification and also visualization of cellular architecture [86-87].

In these current conditions, biological specimens or tissue samples are not available. The serological tests can be separated into twice, antigen-detection and antibody-detection tests [18]. In another way, like the hemagglutination (HA) test, direct and indirect immunofluorescent antibody tests, complementation complex, and lastly immunoblotting and rapid diagnostic tests (RDTs) [18]. It becomes very important to identify the parasites from blood smears. Lastly, these serological tests are available for monitoring parasitic diseases for future therapy purposes [18].

  • Molecular-based approachesNucleic Acid-Based Approaches

There will be several limitations to microscopy and serology-based assays predisposed by parasitologists. Besides the PCR, the usage of such new tools as real-time PCR (RT-PCR) for the exposure of numerous parasitic detections. Such new processes as LAMP and Luminex-based assays have also appeared as a newer approach for the identification of parasites. The multiplexed PCR analysis is useful for multiple sequences in the same reaction tube that is needed for several parasitic infections [88]. In the detection of Cryptosporidium and Giardia, the nucleic acid-based diagnosis was applicable [89].

3.2.2 PCR

The PCR-based process is selective amplification from complex genomes. This happened by denaturing a double-stranded genomic DNA procedure that uses heat. In temperature lowering, primers can anneal to their complementary base sequences. Thus, the extended DNA template trails in both orders from the primer site using enzymatic catalysis with a thermostable DNA polymerase, producing double-stranded products [19, 31].

PCR-based processes are widely used for the diagnosis of diseases in the field of veterinary parasitology [88]. The PCR-based technique is also combined with RFLP or nested PCR for genotyping of organisms. The understanding process in PCR is higher than in light microscopy, and it is also needed for detecting a very low amount of parasites [24]. The PCR-based method is also useful for non-intestinal parasites. Here, we identify species-level analysis [90-98].

There are few consequences, this process is very time-consuming and provides lesser data [99]. In the advanced level study, we need quantitative real-time PCR analysis [27, 99]. In these methods, we easily detect causative pathogens, genetic regulation and expression, allelic function etc. and also get several useful data from these applications [99].

In 18S rRNA-based detection was performed for the diagnosis of Ichthyobodo spp., highly variable regions of the 18S rRNA gene were sequenced by a parasitic protozoan of Cryptocaryon irritans for their molecular identification. As per available literature [100], monogenean (Dactylogyrus spp.) and digenean (Centrocestus formosanus) parasites were detected with the help of the 18S rRNA gene.

  • ITS

The internal transcribed spacer (ITS) is a region of the polycistronic rRNA precursor transcript that is transcribed between the small- and large-subunit ribosomal RNA (rRNA) genes on a chromosome. An earlier study stated an ascaridoid nematode confirmatory test was performed using this method [101].  

  • RT-PCR

This quantitative process requires original template strands by using several fluorescent chemicals like- Sybergreen, Scorpion primers, and fluorescence resonance energy transfer (FRET). The measurement of concentration concluded with a standard curve. At the time of multiplexed RT-PCR, several gene sequences were in one locked tube reaction. Using RT-PCR is useful for quantifying parasitic nucleic acids from different samples, and also estimating the prevalence of parasitic viability [19, 31]. Based on RT-PCR, the identification of Giardia lamblia, Dientamoeba fragilis, Blastocystis hominis, and Entamoeba histolytica [102], and the molecular study was performed with the help of RT-PCR detection of canine leishmaniasis [103].

  • LAMP

Loop-mediated isothermal amplification (LAMP) is a newer concept for the extension method with highly exact, and sensitive to being able to differentiate between single nucleotide changes [25]. It is identified as six different primers that were famously designed in eight different portions on a specific target gene, and amplification occurs only in specific primers which bind with their products [104]. It is a method that can intensify smaller replicas of genetic elements like 109 in less than an hour [104]. LAMP is a DNA amplification-based method and it needs a highly specific and sensitive process. Amplification of DNA is achieved by using a hot water bath for these isothermal conditions [105]. Here, a large amount of whitish magnesium pyrophosphate precipitated from DNA has been observed [106]. The LAMP method can produce 20 μg of DNA for 25 μL of the reaction combination in a single hour [106]. LAMP had more specificity and sensitivity than nested PCR, similar output has been observed but at a faster rate. Their results are static with other demonstrations that rapidly improved specificity and sensitivity observed using the LAMP assay [19]. By using this process, detection of Toxoplasma gondii from blood samples of stray dogs, and cats [107]. Cestodan parasite names like Raillietina echinobothrida, Raillietina tetragona, and Raillietina cesticillus were identified by using the LAMP technique [108].    

  • Luminex

Luminex is a new method that used a bead-based xMAP (multi-analyte profiling), that conjugates fluorescent microspheres (beads) lasers, flow cytometry, and digital signal processing, and it instantaneously measures 100 different analytes in only a sample (Luminex Corporation 2014). The microspheres are covalently connected with antigens, antibodies or oligonucleotides which serve as probes in the assay [32]. Several DNA tests have been developed in the field of Luminex platform in the years for the identification and genotyping of bacteria, viruses and fungi such as Escherichia coli, Mycobacterium, Trichosporon, Salmonella, Listeria monocytogenes, Campylobacter jejuni, and Candida spp. [32, 106, 109-110]. After having been changed for parasitological surveys, recently showed the same reaction using a very minute volume. This technique is useful in the study of antigenic variety for the field of diagnosis of parasitic diseases [110]. 

An earlier study stated [111] that Luminex beads were useful against the detection of “Antibodies and Proteins” in diagnosing Plasmodium falciparum.

  • RAPD

It is known as AP-PCR (arbitrarily primed PCR), and RAPD has been widely used for the explanation of strains in epidemiological studies. The surveying of parasites of genomes is improved by the benefit of RAPD is an easy, fast, and reasonable procedure that does not entail prime knowledge of the DNA sequence or DNA hybridization [112]. In the case of different genotyping from RAPD makes of random amplification of the genome. The fragments are differentiated by gel electrophoresis and the observed banding pattern is useful in genomic sketching. In this method, primers generally amplify ten bases in a PCR protocol with minute selectivity, primer anneals numerous homologous sites of the genomic portions, and finally generated large portions of DNA fragments by sequential amplification [113].   

RAPD is the molecular technique that helps in delineating the strains of microorganisms. It is also useful for parasitic nematodes of plants and humans signifies its high efficacy in the amplification profiles, and is also applicable for the differentiation of polymorphisms between several microorganisms [114-115]. It is mainly used for gene mapping for the depiction of the species, enhances genetic changeability, determines the genetic structure and pinpoints polymorphs in the noncoding genomic parts among the populations of the different microorganisms [116]. By using this process, they identify the genetic structures of Toxocara sp. from canine, ruminant, and feline organisms [117]. 

  • AFLP

AFLP detects a DNA polymorphism from very low information on the genomic sequences. It is very effective for a large number of band detection and also provides large coverage of the genome [118]. It divides into four steps – DNA ingestion, ligation, amplification, and gel analysis. Polymorphisms are characterized based on the presence or absence of DNA portions in the polyacrylamide gel electrophoresis [29]. The merit of this process is to analyze the entire genome for polymorphisms against parasites which is no prior genetic information [29, 118-119].

  • RFLP

It is the most widely used molecular method, that helps in the diagnosis of genotypes of the parasites such as Toxoplasma gondii [30]. This process is useful to detect different variations at the genomic DNA level [120]. This method depends only on the PCR products by restriction enzymes or endonucleases. Due to these enzymatic reactions, DNA gets fragmented into several pieces, which will help at the time of agarose or polyacrylamide gel electrophoresis identification [121]. It is suitable for environmental samples because capable of detecting multiple genotyping in the same genera sample [28].

As per this process utilization, molecular-level identification of Theileria sp. diagnosis was performed [122]. In an earlier study [123], Hymenolepis nana and Hymenolepis diminuta diagnosis were carried out by using this technique.

  • Microsatellites

Microsatellites are short sequences of DNA (around 300 bps) consisting of tandem repetition of 1-6 nucleotides, and around 100 replicas [124]. Microsatellites are new tools in the field of parasitology. In human and animals parasitic studies it is also applicable [33]. It is predominant in eukaryotic genomes and mutates randomly which loss or gain of function [121]. It is also helpful for parasitic nematodes like Trichostrongyloid [33]. With the help of this process, detection and diagnosis were performed for leishmanial parasites from an asymptomatic canine host [125].

  • Diagnosis of parasitic diseases in fish

4.1 Ectoparasites

Serial no.Name of parasiteHostInfected organReferenceTreatmentMolecular study
1.Neoparamoeba sp.Atlantic salmon, seabass, ballan wrasse, and salmon salarGill, gill, gill, and gill.[126-129].Hydrogen peroxide (H2O2) bath [130].Table. 2
2.Amyloodinium sp.Sea bassBlood[130].Copper sulphate (CuSO4) treatment; hydrogen peroxide (H2O2) [128].Table. 2
3.Chilodonella sp.Carassius auratus (Linnaeus, 1758), and Lates calcarifer (Bloch, 1790)Gill, gill, and skin[73].Formalin baths, potassium permanganate (KMnO4), sodium chloride (NaCl), hydrogen peroxide (H2O2), and copper sulphate (CuSO4) [73, 131].Table. 2
4.Ichthyophthirius multifiliisPangasianodon hypophthalmus (Sauvage, 1878).Skin[1].Herbal medicine (Turmeric oil) [1].Table. 2
5.Trichodina sp. Epithelial cells[132].Peracetic acid [133].Table. 2
6.Myxobolus sp. Gill lamellae[134].Kutuklin, and diflubenzuron [135].Table. 2
7.Lernaea sp.Smallmouth bassGill[136].Emamectin benzoate [137].Table. 2
8.Caligus sp.Trachurus picturatus (Bowdich, 1825)Gill lamellae[136].Emamectin benzoate [137].Table. 2
9.Argulus sp.Carassius auratus (Linnaeus, 1758)Skin[138].Extract of Azadirachta indica [139].Table. 2
  • Endoparasites
Serial no.Name of parasiteHostInfected organReferenceTreatmentMolecular study
1.Trypanosoma sp.Cyprinids (Cruz and Eiras 1997; Poynton and Sterud 2001)Blood system[140].Pentamidine, suramin sulphate, melarsoprol, and nifurtimox [141].Table. 2
2.Eimeria sp.Common carp, and crucian carp.Intestine[142].Toltrazuril [142].Table. 2
3.Henneguya sp.Ictalurus punctatus (Rafinesque, 1818)Gill lamellae[143].Fumagillin and toltrazuril [144].Table. 2
4.Dactylogyrus sp.Carassius auratus (Linnaeus, 1758)Gill[145].Coumarin extract [145].Table. 2
5.Gyrodactylus sp.Atherina boyeri (Risso, 1810)Skin, and gill[146].Isoimperatorin [145].Table. 2

Table. 2 Molecular study for parasite identification

Name of parasitesMolecular toolsReference
Neoparamoeba sp.18S rRNA[147].
Amyloodinium sp.  18S rDNA and 28S rDNA[148].  
Chilodonella sp.ITS, LSU rDNA gene, and mtSSU[149-150].
Ichthyophthirius multifiliis18S rRNA[1].
Trichodina sp.18S rDNA, Euk A and Euk B of SSUrRNA[151-153].
Myxobolus sp.18S rRNA[154-156].
Trypanosoma sp.TBR-1/2, and 18S rRNA[157-158].
Eimeria sp.SSU rRNA[156, 159].
Bucephalus sp.ITS2, and cox1[162].
Aporocotylidae (Seriola dumerili)18S, 28S rDNA, and ITS[163-165].
Atractolytocestus sp.COX1, and ITS2[166].
Diphillobothrium sp.COX1[167].
Bothriocephalus sp.28S rDNA, and COX1[168].
Ligula sp.COI[169].
Anisakis sp.COX1, and ITS-RFLP[170-172].
Philometroides sp.18S and 28S rDNA[173].
Dracunculus sp.COX1[174].
Acanthocephala18S and 28S rRNA[175-176].
Lernaea sp.18S rRNA, 28S rRNA, and COX1[177].
Caligus sp.18S rRNA, and 18S rDNA[178].
Argulus sp.D1-D2 of the 28S rDNA, and 18S rRNA[179].

Conclusion

The aquatic parasite is comprised of subkingdom protozoa, phylum Cnidaria, phylum Platyhelminthes, phylum Nematoda, phylum Acanthocephala, phylum Annelida, and phylum Arthropoda. Routine parasitological detection is based on light microscopy of parasitic identification. The first step of parasitic identifications are based on wet mount methods that help in the identification of intracellular parasites. Histopathological changes can be diagnosed with the help of microscopy. Molecular biology methods are used to detect parasitic diseases, enhancing the diagnosis of parasitic clarities. The developmental stage is highly-sensitive to parasitic infections useful for monitoring and control for the pathogen in cultured and ornamental fish.

Acknowledgment

This work was funded by National Surveillance Program for Aquatic Animal Disease, Pradhan Mantri Matsya Sampada Yojana, Govt. of India with grant number (G/Nat. Surveillance phase-II). The authors are also thankful to Mr. Jayanta Pramanik and Mr. Sk Sarif for their continuous support.

Conflict of interest

The authors declare that there is no conflict of interest.

Reference

  1.  Kumar, V., Das, B.K., Swain, H.S., Chowdhury, H., Roy, S., Bera, A.K., Das, R., Parida, S.N., Dhar, S., Jana, A.K. and Behera, B.K., 2022a. Outbreak of Ichthyophthirius multifiliis associated with Aeromonas hydrophila in Pangasianodon hypophthalmus: The role of turmeric oil in enhancing immunity and inducing resistance against co-infection. Frontiers in immunology13, p.956478.
  2. Kumar, V., Roy, S., Behera, B.K. and Das, B.K., 2022b. Heat Shock Proteins (Hsps) in Cellular Homeostasis: A Promising Tool for Health Management in Crustacean Aquaculture. Life, 12(11), p.1777.
  3. Kumar, V., Roy, S., Barman, D. and Kumar A. 2014. Immunoserological and molecular techniques used in fish disease diagnosis- A mini-review. International Journal of Fisheries and Aquatic Studies. 1(3):111-117.
  4. Kumar, V., Roy, S., and Barman, D. (2015). Effect of Mikania cordata (Burm) B. L. Robins on Non-Specific Immune Response of Catla catla (Hamilton, 1822) Against Aphanomyces invadans. Fish. Technol. 52, 20–25.
  5. Kumar, V., Roy, S., and Barman, D. (2016). Effect of Mikania cordata on non-specific immune response and survival of Labeo rohita against Aphanomyces invadans. J. Anim. Plant Sci. 26, 1833–1842.
  6. Kumar, V., Roy, S., Sarkar, U. K., and Das, A. K. (2018). Occurrence of Ichthyophthiriasis in Pangasianodon hypophthalmus (Sauvage, 1878) Cultured in Net-Cages of Maithon Reservoir, Jharkhand, India. Natl. Acad. Sci. Lett. 41, 275–278. doi:10.1007/s40009-018-0647-9.
  7. Kumar, V., Roy, S., Behera, B. K., Swain, H. S., and Das, B. K. (2021). Biofloc Microbiome with Bioremediation and Health Benefits. Front. Microbiol. 12. doi:10.3389/fmicb.2021.741164.
  8. Pazos F, Santos Y, Macías AR, Núñez S and Toranzo AE. Evaluation of media for the successful culture of Flexibacter maritimus. J. Fish Dis., 1996; 19: 193-197.
  9. Bernardet JF, Campbell AC and Buswell JA.Flexibacter maritimus is the agent of ‘black patch necrosis’ in Dover sole in Scotland. Dis. Aquat. Org. 1990; 8: 233-237.
  10. Shewan JM and McMeekin TA. Taxonomy and ecology of the Flavobacterium and related genera. Ann. Rev. Microbiol., 1983; 37: 233-252.
  11. Pyle SW and Shotts EB. A new approach for differentiating Flexibacteria isolated from cold water and warm water fish. Can. J. Fish. Aquat. Sci., 1980; 37: 1040-1042.
  12. Chen MF, Henry-Ford D and Groff JM. Isolation and characterization of Flexibacter maritimus from marine fishes of California. J. Aquat. Anim. Health. 1995; 7: 318- 326.
  13. Bertolini JM and Rohovec JS. Electrophoretic detection of proteases from different Flavobacterium columnare strains and assessment of their variability. Dis. Aquat. Org. 1992; 12: 121-128.
  14. Plumb JA. Health maintenance and principle microbial diseases of cultured fishes.Iowa State University Press. Ames, Iowa. 1999: 344 pp.
  15. Purivirojkul W. A survey of fish species infected with trematode metacercariae from some areas in northeast Thailand. Journal of Fisheries Technology Research 2011; 5(2) 75-86.
  16. Roberts RJ. Fish pathology, 3rd edition. London: W.B. Saunders publishing; 2001.
  17. Noga EJ. Fish disease: diagnosis and treatment. Iowa: Iowa state university press; 2000.
  18. Ndao M, 2009. Diagnosis of parasitic diseases: old and new approaches. Interdisciplinary Perspectives on Infectious Diseases. 2009: 278246.
  19. Tavares RG, Staggemeier R, Borges ALP, Rodrigues MT, Castelan LA, Vasconcelos J, Anschau ME, Spalding SM. 2011. Molecular techniques for the study and diagnosis of parasite infection.The Journal of Venomous Animals and Toxins including Tropical Diseases. 17(3): 239-248.
  20. Kompalic-Cristo A, Britto C, Fernandes O. Diagnóstico molecular da toxoplasmose. J Bras Patol Med Lab. 2005;41(4):229-35.
  21. Jardim EAGV, Linhares GFC, Torres FAG, Araújo JLB, Barbosa SM. Diferenciação específica entre Taenia saginata e Taenia solium por ensaio de PCR e duplex-PCR. Ciênc Rural. 2006; 36: 166-72.
  22. Scott JA. The molecular genetics of resistance: resistance as a response to stress. Florida Entomologist. 1995;78(3):399-414.
  23. Portela-Lindoso AAB, Shikanai-Yasuda MA. Doença de Chagas crônica: do xenodiagnóstico e hemocultura à reação em cadeia da polimerase. Rev Saúde Pública. 2003;37(1):107-15.
  24. Shokoples SE, Ndao M, Kowalewska-Grochowska K, Yanow SK. Multiplexed real-time PCR assay for discrimination of Plasmodium species with improved sensitivity for mixed infections. J Clin Microbiol. 2009;47(4):975-80.
  25. Parida M, Sannarangaiah S, Dash PK, Rao PV, Morita K. Loop-mediated isothermal amplification (LAMP): a new generation of innovative gene amplification technique; perspectives in clinical diagnosis of infectious diseases. Rev Med Virol. 2008;18(6):407-21.
  26. Tait, B. D. Hudson, F. Cantwell, L. Brewin G, Holdsworth R, Bennett G, Jose M. 2009. Review article: luminex technology for HLA antibody detection in organ transplantation. Nephrology, 14(2): 247–254.
  27. Guy RA, Xiao C, Horgen PA. Real-time PCR assay for detection and genotype differentiation of Giardia lamblia in stool specimens. J Clin Microbiol. 2004; 42: 3317-20.
  28. Monis PT, Andrews RH. Molecular epidemiology: assumptions and limitations of commonly applied methods. Int J Parasitol. 1998;28(6):981-7.
  29. Blears MJ, Pokorny NJ, Carreno RA, Chen S, De Grandis SA, Lee H, et al. DNA fingerprinting of Cryptosporidium parvum isolates using amplified fragment length polymorphism (AFLP). J Parasitol. 2000; 86: 838-41.
  30. Quan JH, Kim TY, Choi IU, Lee YH. Genotyping of a Korean isolate of Toxoplasma gondii by multilocus PCR-RFLP and microsatellite analysis. Korean J Parasitol. 2008;46(2):105-8.
  31. Gasser RB. Molecular tools–advances, opportunities and prospects. Vet Parasitol. 2006; 136: 69-89.
  32. Dunbar SA. Applications of Luminex xMAP technology for rapid, high-throughput multiplexed nucleic acid detection. Clin Chim Acta. 2006; 363: 71-82.
  33. Temperley ND, Webster LM, Adam A, Keller LF, Johnson PC. Cross-species utility of microsatellite markers in Trichostrongyloid nematodes. J Parasitol. 2009;95(2):487-9.
  34. Lemoine M., Doligez B. and Richner H. 2012. On the Equivalence of Host Local Adaptation and Parasite Maladaptation: An Experimental Test. Am. Nat., 179, 270–281.
  35. Mastitsky S.E., Karatayev A.Y. and Burlakova L.E., 2014. Parasites of Aquatic Exotic Invertebrates: Identification of Potential Risks Posed to the Great Lakes. Human and Ecological Risk Assessment: An International Journal, 20, 743–763.
  36. Torchin M.E., Lafferty K.D., Dobson A.P., McKenzie V.J. and Kuris A.M., 2003. Introduced species and their missing parasites. Nature, 421, 628–630.
  37. Roth O., Keller I., Landis S.H., Salzburger W. and Reusch T.B.H., 2012. Hosts are ahead in a marine host–parasite coevolutionary arms race innate immune system adaptation in pipefish Syngnathus typhie against Vibrio phylotypes. Evolution, 66, 2528–2539.
  38. Morand S., Manning S.D. and Woolhouse M.E.J., 1996.Parasite-Host Coevolution and Geographic Patterns of Parasite Infectivity and Host Susceptibility. Proc. R Soc. London, Ser. B. Biol. Sci., 263, 119–128.
  39. Dybdahl M.F. and Storfer A., 2003. Parasite local adaptation: Red Queen versus Suicide King. Trends Ecol. Evol. 18: 523–530.
  40. Stavrescu-Bedivan, M.M., Popa, O.P. and Popa, L.O. 2014.Infestation of Lernaea cyprinacea (Copepoda: Lernaeidae) in two invasive fish species in Romania, Lepomis gibbosus and Pseudorasbora parva. Knowledge and Management of Aquatic Ecosystems.414, 12.
  41. Pina-Vazquez C, Reyes-Lopez M, Ortiz-Estrada G, de la Garza M, Serrano-Luna J (2012) Host-parasite interaction: parasite-derived and -induced proteases that degrade human extracellular matrix. J Parasitol Res 2012: 748206.
  42. Medzhitov, J.R., 2007. Recognition of microorganisms and activation of the immune response. Nature 449, 819–826.
  43. McGuinness, D.H., Dehal, P.K., Pleass, R.J., 2003. Pattern recognition molecules and innate immunity to parasites. Trends Parasitol. 15, 312–319.
  44. Dam, T.K. and Brewer, C.F., 2010. Lectins as pattern recognition molecules: the effects of epitope density in innate immunity. Glycobiology20(3), pp.270-279.
  45. Vázquez-Mendoza, A., Carrero, J.C. and Rodriguez-Sosa, M., 2013. Parasitic infections: a role for C-type lectins receptors. BioMed Research International2013.
  46. Sancho, D. and Reis e Sousa, C., 2012. Signaling by myeloid C-type lectin receptors in immunity and homeostasis. Annual review of immunology30, pp.491-529.
  47. Chen, J., Xiao, S. and Yu, Z., 2011. F-type lectin involved in defense against bacterial infection in the pearl oyster (Pinctada martensii). Fish & Shellfish Immunology30(2), pp.750-754.
  48. Duverger, E., Lamerant-Fayel, N., Frison, N. and Monsigny, M., 2010. Carbohydrate–lectin interactions assayed by SPR. In Surface Plasmon Resonance (pp. 157-178). Humana Press.
  49. Van Kooyk, Y., 2008. C-type lectins on dendritic cells: key modulators for the induction of immune responses. Biochemical Society Transactions36(6), pp.1478-1481.
  50. Weis, W.I., Taylor, M.E. and Drickamer, K., 1998. The C‐type lectin superfamily in the immune system. Immunological reviews163(1), pp.19-34.
  51. Woo, P. T. (1996). Protective immune response of fish to parasitic flagellates. Annual review of fish diseases, 6, 121-131.
  52. Sitjà-Bobadilla, A. (2008). Fish immune response to Myxozoan parasites. Parasite, 15(3), 420-425.
  53. Dickerson, H. W., & Clark, T. G. (1996). Immune response of fishes to ciliates. Annual Review of Fish Diseases6: 107-120.
  54. Midtlyng, P.J., 2000. Vaccination in salmonid aquaculture: a review. Rec. Adv. Mar. Biotechnol. 5, 227–242.
  55. Biering, E., Villoing, S., Sommerset, I., Christie, K.E., 2005. Update of viral vaccines for fish. In: Midtlyng, P.J. (Ed.), Progress in Fish Vaccinology, Developments in Biologicals, vol. 121. Karger, Basel, pp. 97–113.
  56. Tonheim, T.C., Bøgwald, J., Dalmo, R.A., 2008. What happens to the DNA vaccines in fish?—a review of the current knowledge. Fish Shellfish Immunol. 25, 1–18.
  57. Lorenzen, N., LaPatra, S.E., 2005. DNA vaccines for aquacultured fish. Rev. Sci. Tech. Off. Int. Epiz. 24, 201–213.
  58. Clark, T.G., Lin, T.-L.and  DickersonH.W. 1995. Surface immobilization antigens of Ichthyophthirius multifiliis: Their role in protective immunity. Annual Review of Fish Diseases. 5: 113–131.
  59. Clark TG, Dickerson HW. 1997. Antibody-mediated effects on parasite behavior: Evidence of a novel mechanism of immunity against a parasitic protist. Parasitol Today. 13: 477-480.
  60. Wang, X., Clark, T. G., Noe, J. and Dickerson, H. W. 2002. Immunisation of channel catfish, Ictalurus punctatus, with Ichthyophthirius multifiliis immobilisation antigens elicits serotype-specific protection. Fish and Shellfish Immunology, 3(5):337-350.
  61. Woo, P.T.K., Poynton, S.L., 1999. Diplomonadida, Kinetoplastida and Amoebida (Phylum Sarcomastigophora). In: Woo, P.T.K. (Ed.), Fish Diseases and Disorders, vol. 1. Protozoan and Metazoan Infections. CABI Publishing, New York, pp. 27–96.
  62. Reilly, P., Mulcahy, M.F., 1993. Humoral antibody response in Atlantic salmon (Salmo salar L.) immunized with extracts derived from the ectoparasite caligid copepods, Caligus elongatus (Nordmann,1832) and Lepeopththeirus salmonis (Kroyer, 1838). Fish Shellfish Immunol. 3, 59–70.
  63. Raynard, R.S., Bricknell, I.R., Billingsley, P.F., Nisbet, A.J., 2002. Development of vaccines against sea lice. Pest Manage. Sci. 58, 569–575.
  64. Ivory, C. and Chadee, K., 2004. DNA vaccines: designing strategies against parasitic infections. Genetic vaccines and therapy2(1), pp.1-8.
  65. Shivam, S., El-Matbouli, M. and Kumar, G., 2021. Development of fish parasite vaccines in the OMICs era: Progress and opportunities. Vaccines9(2), p.179.
  66. Palikova, M., Papezikova, I., Markova, Z., Navratil, S., Mares, J., Mares, L., Vojtek, L., Hyrsl, P., Jelinkova, E. and Schmidt-Posthaus, H., 2017. Proliferative kidney disease in rainbow trout (Oncorhynchus mykiss) under intensive breeding conditions: Pathogenesis and haematological and immune parameters. Veterinary parasitology238, pp.5-16.
  67. , J.L.F., Le Breton, A., Brun, E., Vendramin, N., Spiliopoulos, G., Furones, D. and Basurco, B., 2022. Assessing the economic impact of diseases in Mediterranean grow-out farms culturing European sea bass. Aquaculture547, p.737530.
  68. Lama, R., Pereiro, P., Valenzuela-Muñoz, V., Gallardo-Escárate, C., Tort, L., Figueras, A. and Novoa, B., 2020. RNA-Seq analysis of European sea bass (Dicentrarchus labrax L.) infected with nodavirus reveals powerful modulation of the stress response. Veterinary research51(1), pp.1-22.
  69. Firmino, J., Furones, M.D., Andree, K.B., Sarasquete, C., Ortiz-Delgado, J.B., Asencio-Alcudia, G. and Gisbert, E., 2019. Contrasting outcomes of Vibrio harveyi pathogenicity in gilthead seabream, Sparus aurata and European seabass, Dicentrachus labraxAquaculture511, p.734210.
  70. Ferguson H. Systemic pathology of fish: a text and atlas of comparative tissue responses in diseases of teleosts. Ames: Iowa state university Press; 1989.
  71. Cobo, F. Aliaga, L. Talavera, P. and Concha, A. 2007. The histological spectrum of non-granulomatous localized mucosal leishmaniasis caused by Leishmania infantum,” Annals of Tropical Medicine and Parasitology. 101: 689– 694.
  72. Dhayanithi, N.B., Sudhagar, A., Kumar, T.T.A. and Lal, K.K., 2022. Study on amyloodiniosis outbreak in captive-bred percula clownfish (Amphiprion percula) and improved control regimens. Journal of Parasitic Diseases, pp.1-7.
  73. Bu, X.L., Wang, R.Q., Gomes, G.B., Ban, S.N., Li, W.X., Wu, S.G., Zou, H., Li, M. and Wang, G.T., 2021. First record of facultative parasitism of Chilodonella uncinata based on goldfish (Carassius auratus) infection model. Aquaculture538, p.736535.
  74. Li, W.J., Lyu, X.M., Nie, P. and Liu, Y., 2022. Morphological, histopathological and molecular characterization of parasitic ciliate Ichthyophthirius multifiliis infecting an indigenous and endangered fish, Schizothorax macropogon (Cyprinidae: Schizothoracinae) in high plateau, Tibet, China. Aquaculture Reports25, p.101192.
  75. Saha, M. and Bandyopadhyay, P.K., 2019, March. Three new species of Trichodina (Protozoa: Ciliophora) isolated from ornamental fish, Carassius auratus from India. In Proceedings of the Zoological Society (Vol. 72, No. 1, pp. 79-89). Springer India.
  76. Naldoni, J., Carriero, M.M., Moreira, G.S., da Silva, M.R., Maia, A.A. and Adriano, E.A., 2020. Increasing the known biodiversity of cnidarian parasites of bryconid fishes from South America: two novel Myxobolus species with ultrastructure and ssrDNA-based phylogeny. Parasitology Research119(11), pp.3627-3637.
  77. Yahaya, Z.S., Azizah, M.N.S., Alkazmi, L., Ravi, R. and Awosolu, O.B., 2022. First Molecular Identification of Caligus clemensi on Cultured Crimson Snapper Lutjanus erythropterus on Jerejak Island, Penang, Peninsular Malaysia. Pathogens11(2), p.188.
  78. Tandel, R.S., Chanu, K.V., Hussain Bhat, R.A., Dash, P., Shah, T.K. and Thakuria, D., 2021. Morphometric and molecular identification of Argulus japonicus (Thiele 1900) in vulnerable Himalayan snow trout, Schizothorax richardsonii (Gray 1832). Aquaculture Research52(12), pp.6770-6778.
  79. Virgilio, L.R., Teixeira, G.O., Almeida, L.S., Melo, H.P.S., Júnior, S.L., Meneguetti, D.U.O., Camargo, L.M.A. and Takemoto, R.M., 2022. Infection with Trypanosoma spp. in Platydoras armatulus (Siluriformes, Doradidae), in southwestern Amazon, Brazil. Journal of Parasitic Diseases, pp.1-6.
  80. Andrade, L.D.A.S., Genovez-Oliveira, J.L., Oliveira, M.D.S., de Mello, E.R., Cardozo, S.V., de Oliveira, Á.A., de Lima, V.M., Ferreira, I. and Berto, B.P., 2022. Observations on an Eimeria sp. (Apicomplexa) from the green kingfisher Chloroceryle americana (Gmelin, 1788)(Coraciiformes) in Southeastern Brazil: an example of how the ecological aspects of the host can be essential for the identification of its coccidians. Parasitology Research121(3), pp.1059-1063.
  81. Vieira, D.H.M.D., Rangel, L.F., Tagliavini, V.P., Abdallah, V.D., Santos, M.J. and de Azevedo, R.K., 2021. Morphological and molecular analysis of Henneguya tietensis n. sp.(Cnidaria: Myxosporea), parasitizing the gill filaments of Prochilodus lineatus (Valenciennes, 1837) from Brazil. Parasitology Research120(1), pp.27-36.
  82. Trivedi, A.K., Prakash, S. and Tripathi, A., 2022. Dactylogyrus kolodynensis sp. n.(Platyhelminthes: Monogenea) infecting gills of Osteobrama cotio (Hamilton, 1822)(Cypriniformes: Cyprinidae) from India. Journal of Parasitic Diseases46(3), pp.854-859.
  83. García-Vásquez, A., Pinacho-Pinacho, C.D., Guzmán-Valdivieso, I., Calixto-Rojas, M. and Rubio-Godoy, M., 2021. Morpho-molecular characterization of Gyrodactylus parasites of farmed tilapia and their spillover to native fishes in Mexico. Scientific Reports11(1), pp.1-17.
  84. Klontz GW. Diagnostic methods in fish diseases: Present status and needs. In: Ellis AE.  (ed.) Fish and shellfish pathology. London: Academic press; 1985. p1-10.
  85. Barcia JJ. The Giemsa stain: its history and applications. Int J Surg Pathol. 2007; 15: 292-6.
  86. Powers CN. Diagnosis of infectious diseases: A cytopathologist’s perspective. Clin Microbiol Rev 1998;11:341-65.
  87. Woods GL, Walker DH. Detection of infection or infectious agents by the use of cytologic and histologic stains. Clin Microbiol Rev 1996;9:382-404.
  88. Zarlenga DS, Higgins J (2001) PCR as a diagnostic and quantitative technique in veterinary parasitology. Vet Parasitol 101:215–230.
  89. Adeyemo, F.E., Singh, G., Reddy, P. and Stenström, T.A., 2018. Methods for the detection of Cryptosporidium and Giardia: from microscopy to nucleic acid-based tools in clinical and environmental regimes. Acta Tropica184, pp.15-28.
  90. Roper C, Elhassan IM, Hviid L, Giha H, Richardson W, Babiker H, et al. Detection of very low-level Plasmodium falciparum infections using the nested polymerase chain reaction and a reassessment of the epidemiology of unstable malaria in Sudan. Am J Trop Med Hyg. 1996;54(4):325-31.
  91. Piarroux R, Gambarelli F, Dumon H, Fontes M, Dunan S, Mary C, et al. Comparison of PCR with direct examination of bone marrow aspiration, myeloculture, and serology for diagnosis of visceral Leishmaniasis in immunocompromised patients. J Clin Microbiol. 1994;32(3):746-9.
  92. Antinori S, Calattini S, Longhi E, Bestetti G, Piolini R, Magni C, et al. Clinical use of polymerase chain reaction performed on peripheral blood and bone marrow samples for the diagnosis and monitoring of visceral Leishmaniasis in HIV-infected and HIV-uninfected patients: a single-center, 8-year experience in Italy and review of the literature. Clin Infect Dis. 2007; 44: 1602-10.
  93. Andrade BB, Boaventura V, Barral-Netto M, Barral A. Métodos diagnósticos da leishmaniose tegumentar: fatos, falácias e perspectivas. Gaz Méd Bahia. 2005;75: 75-82.
  94. Contamin H, Fandeur T, Bonnefoy S, Skouri F, Ntoumi F, Mercereau-Puijalon O. PCR typing of field isolates of Plasmodium falciparum. J Clin Microbiol. 1995; 33: 944-51.
  95. Duraisingh MT, Curtis J, Warhurst DC. Plasmodium falciparum: detection of polymorphisms in the dihydrofolate reductase and dihydropteroate synthetase genes by PCR and restriction digestion. Exp Parasitol. 1998; 8: 1-8.
  96. Harris P.D. 1988. Changes in the site specificity of Gyrodactylus turnbulli Harris, 1986 (Monogenea) during infections of individual guppies (Poecilia reticulata Peters, 1859). Canadian Journal of Zoology. 66: 2854-2857.
  97. Lachaud L, Dereure J, Chabbert E, Reynes J, Mauboussin JM, Oziol E, et al. Optimized PCR using patient blood samples for diagnosis and follow-up of visceral Leishmaniasis, with special reference to AIDS patients. J Clin Microbiol. 2000;38(1):236-40.
  98. Wooden J, Kyes S, Sibley CH. PCR and strain identification in Plasmodium falciparum. Parasitol Today. 1993;9(8):303-5.
  99. Lin MH, Chen TC, Kuo TT, Tseng CC, Tseng CP. Real-time PCR for quantitative detection of Toxoplasma gondii. J Clin Microbiol. 2000;38(11):4121-5.
  100. Jaruboonyakorn, P., Tejangkura, T. and Chontananarth, T., 2022. Multiplex PCR development for the simultaneous and rapid detection of two pathogenic flukes, Dactylogyrus spp. and Centrocestus formosanus, in ornamental fishes. Aquaculture548, p.737660.
  101. Chen, H.X., Zhang, L.P., Gibson, D.I., Lü, L., Xu, Z., Li, H.T., Ju, H.D. and Li, L., 2018. Detection of ascaridoid nematode parasites in the important marine food-fish Conger myriaster (Brevoort)(Anguilliformes: Congridae) from the Zhoushan Fishery, China. Parasites & vectors11(1), pp.1-12.
  102. Venturini, E., Scarso, S., Prelazzi, G.A., Niccolai, C., Bianchi, L., Montagnani, C., Lapini, M., Chiappini, E., Antonelli, A., Rossolini, G.M. and Galli, L., 2021. Epidemiology and clinical features of intestinal protozoan infections detected by Real-time PCR in non-native children within an Italian tertiary care children’s hospital: A cross-sectional study. Travel Medicine and Infectious Disease43, p.102107.
  103. Djibougou, A.D., Nikièma, A.S., Hien, A.S., Sangaré, I., Yameogo, B.K., Koala, L., Ouari, A., Diagbouga, S.P., Diabaté, A., Price, H. and Fournet, F., 2022. Serological and molecular detection of Leishmania species in dog peripheral blood from Bobo-Dioulasso city, a confirmation of canine leishmaniasis enzootic area for Burkina Faso. Infection, Genetics and Evolution, p.105327.
  104. Paris DH, Imwong M, Faiz AM, Hasan M, Yunus EB, Silamut K, et al. Loop-mediated isothermal PCR (LAMP) for the diagnosis of falciparum malaria. Am J Trop Med Hyg. 2007;77(5):972-6.
  105. Notomi T, Okayama H, Masubuchi H, Yonekawa T, Watanabe K, Amino N, et al. Loop-mediated isothermal amplification of DNA. Nucleic Acids Res. 2000;28(12):E63.
  106. Mori Y, Nagamine K, Tomita N, Notomi T. Detection of loop-mediated isothermal amplification reaction by turbidity derived from magnesium pyrophosphate formation. Biochem Biophys Res Commun. 2001;289(1):150-4.
  107. Xue, Y., Kong, Q., Ding, H., Xie, C., Zheng, B., Zhuo, X., Ding, J., Tong, Q., Lou, D., Lu, S. and Lv, H., 2021. A novel loop-mediated isothermal amplification-lateral-flow-dipstick (LAMP-LFD) device for rapid detection of Toxoplasma gondii in the blood of stray cats and dogs. Parasite28.
  108. Panich, W., Tejangkura, T. and Chontananarth, T., 2021. Novel high-performance detection of Raillietina echinobothrida, Raillietina tetragona, and Raillietina cesticillus using loop-mediated isothermal amplification coupled with a lateral flow dipstick (LAMP-LFD). Veterinary Parasitology292, p.109396.
  109. Cowan LS, Diem L, Brake MC, Crawford JT. Transfer of a Mycobacterium tuberculosis genotyping method, spoligotyping, from a reverse line-blot hybridization, membrane-based assay to the Luminex multianalyte profiling system. J Clin Microbiol. 2004; 42: 474-7.
  110. Diaz MR, Fell JW. High-throughput detection of pathogenic yeasts of the genus Trichosporon. J Clin Microbiol. 2004; 42: 3696-706.
  111. Wakeman, B.S., Shakamuri, P., McDonald, M.A., Weinberg, J., Svoboda, P., Murphy, M.K., Kariuki, S., Mace, K., Elder, E., Rivera, H. and Qvarnstrom, Y., 2021. Development of a new peptide-bead coupling method for an all peptide–based Luminex multiplexing assay for detection of Plasmodium falciparum antibody responses. Journal of Immunological Methods499, p.113148.
  112. Nuchprayoon S, Junpee A, Poovorawan Y. Random Amplified Polymorphic DNA (RAPD) for differentiation between Thai and Myanmar strains of Wuchereria bancrofti. Filaria J. 2007;6(1):6.
  113. Lupchinski JR E, Vargas L, Ribeiro RP, Moreira HLM, Valentim M, Povh JA. A importância da utilização da técnica RAPD para a identificação de dactilogirídeos em tilápias do Nilo. (Oreochromis niloticus).Arq Cienc Vet Zool Unipar. 2006;9(1):49-57.
  114. Jobet E, Bougnoux ME, Morand S, Rivault C, Cloarec A, Hugot JP. Use of random amplified polymorphic DNA (RAPD) for generating specific DNA probes for oxyuroid species (Nematoda). Parasite. 1998; 5: 47-50.
  115. Martinez EM, Correia JA, Villela EV, Duarte AN, Ferreira LF, Bello AR. Random amplified polymorphic DNA analysis of DNA extracted from Trichuris trichiura (Linnaeus, 1771) eggs and its prospective application to paleoparasitological studies. Mem Inst Oswaldo Cruz. 2003;98 Suppl 1:59-62.
  116. Jain SK, Neekhra B, Pandey D, Jain K. RAPD marker system in insect study: a review. Indian J Biotechnol. 2010; 9: 7-12.
  117. Jeeva, K., Chatterjee, N., Bera, A.K., Maiti, S., Ram, H., Banerjee, P.S., Goswami, T.K., Das, P.J., Chamua, J.K., Deb, S.M. and Bhattacharya, D., 2021. Genetic structure of Toxocara of canine, feline and ruminant origin: An evaluation with RAPD marker.
  118. Buttow MV, Castro CM, Schwartz E, Tonietto A, Barbieri RL. Caracterização molecular de populações de Butia capitata (Arecaceae) do Sul do Brasil através de marcadores AFLP. Rev Bras Frutic. 2010; 32: 230-9.
  119. Bonin A, Ehrich D, Manel S. Statistical analysis of amplified fragment length polymorphism data: a toolbox for molecular ecologists and evolutionists. Mol Ecol. 2007; 16: 3737-58.
  120. Carpentieri-Pípolo V, Gallo-Meagher M, Dickson DW, Gorbet DW, Mendes ML, de Souza SGH. Comparação entre métodos de marcação da sonda de RFLP R2430E utilizada na seleção de cultivares de amendoim resistente à Meloidogyne arenaria. Ciênc Agrár. 2008; 29: 783-8.
  121. Huber F. Caracterização genotípica e estudo filogenético de Cryptosporidium spp. obtidos de diferentes hospedeiros [dissertation]. Rio de Janeiro: Universidade Federal Rural do Rio de Janeiro; 2007. 72p.
  122. Nangru, A., Maharana, B.R., Vohra, S. and Kumar, B., 2022. Molecular identification of Theileria species in naturally infected sheep using nested PCR–RFLP. Parasitology Research121(5), pp.1487-1497.
  123. Oliveira EJ, Pádua JG, Zucchi MI, Vencovsky R, Vieira MLC. Origin evolution and genome distribution of microsatellites. Genet Mol Biol. 2006;29(2):294-307.
  124. Najafzadeh, N., Taslimian, R., Fotouhi-Ardakani, R., Spotin, A. and Parvizi, P., 2022. Detection and differentiation of Leishmania parasites in asymptomatic canine by High-Resolution Melting analysis of microsatellite fragment in ITS gene. Microbial Pathogenesis162, p.105300.
  125. Karlsbakk, E., Olsen, A.B., Einen, A.C.B., Mo, T.A., Fiksdal, I.U., Aase, H., Kalgraff, C., Skår, S.Å. and Hansen, H., 2013. Amoebic gill disease due to Paramoeba perurans in ballan wrasse (Labrus bergylta). Aquaculture412, pp.41-44.
  126. Young ND, Crosbie PBB, Adams MB, Nowak BF, Morrison RN. Neoparamoeba perurans n. sp., an agent of amoebic gill disease of Atlantic salmon (Salmo salar L.). International Journal for Parasitology 2007; 37 1469–1481.
  127. Kim, W.S., Kong, K.H., Kim, J.O., Jung, S.J., Kim, J.H. and Oh, M.J., 2017. Amoebic gill disease outbreak in marine fish cultured in Korea. Journal of Veterinary Diagnostic Investigation29(3), pp.357-361.
  128. Crosbie, P.B., Bridle, A.R., Leef, M.J. and Nowak, B.F., 2010. Effects of different batches of Neoparamoeba perurans and fish stocking densities on the severity of amoebic gill disease in experimental infection of Atlantic salmon, Salmo salar L. Aquaculture Research41(10), pp.e505-e516.
  129. Bridle, A.R., Hill, T., Smith, A., Crosbie, P. and Nowak, B.F., 2021. Experimental exposure to low concentrations of Neoparamoeba perurans induces amoebic gill disease in Atlantic salmon. Journal of Fish Diseases44(7), pp.1025-1031.
  130. Valladão, G.M.R., Pereira, M.M., Gallani, S.U. and Pilarski, F., 2019. A massive Chilodonella hexasticha infestation associated with yellowtail tetra Astyanax lacustris mortality in aquaculture: Identification and pathology. Aquaculture Research50(7), pp.2019-2022.
  131. Wang, Z., Deng, Q., Zhou, T., Yang, H. and Gu, Z., 2018. First record of two ectoparasitic ciliates of the genus Trichodina (Ciliophora: Trichodinidae) parasitizing gills of an invasive freshwater fish, Micropercops swinhonis, in Tibet. Parasitology research117(7), pp.2233-2242.
  132. Abu-Elala, N.M., Attia, M.M., Abd-Elsalam, R.M., Gamal, A. and Younis, N.A., 2021. Peracetic acid treatment of Ichthyophthirius multifiliis (Ciliophora: Ichthyophthiriidae) and Trichodina spp. reduces the infection by Aeromonas hydrophila and improves survival in Nile tilapia (Oreochromis niloticus). Aquaculture538, p.736591.
  133. Wang, M.M., Zhang, J.Y. and Zhao, Y.J., 2022. Morphological description and molecular identification of Myxobolus dajiangensis n. sp. (Myxozoa: Myxobolidae) from the gill of Cyprinus carpio in southwest China. PeerJ10, p.e13023.
  134. Yanuhar, U., Caesar, N.R., Junirahma, N.S. and Soelistyoadi, R.N., 2022. Immunomolecular response of CD4+, CD8+, TNF-α and IFN-γ in Myxobolus-infected koi (Cyprinus carpio) treated with probiotics. Aquaculture and Fisheries.
  135. Muzzall, P.M. and Whelan, G., 2011. Parasites of fish from the Great Lakes: a synopsis and.
  136. Raja, R.A., Patil, P.K., Avunje, S., Kumaran, M., Solanki, H.G., Jithendran, K.P., Alavandi, S.V. and Vijayan, K.K., 2022. Efficacy of emamectin benzoate in controlling natural infestations of ectoparasites in economically important fish species of India. Aquaculture551, p.737940.
  137. Shukla, S., Ahmad, S., Tiwari, K.J. and Shukla, R., 2022. Fish lice Argulus foliaceus infestation in gold fish Carassius auratus from Lucknow, UP India: A report.
  138. Kumari, P., Kumar, S., Ramesh, M., Shameena, S., Deo, A.D., Rajendran, K.V. and Raman, R.P., 2019. Antiparasitic effect of aqueous and organic solvent extracts of Azadirachta indica leaf against Argulus japonicus in Carassius auratusAquaculture511, p.634175.
  139. de Jesus, R.B., Gallani, S.U., Valladão, G.M.R., Pala, G., da Silva, T.F.A., da Costa, J.C., Kotzent, S. and Pilarski, F., 2018. Trypanosomiasis causing mortality outbreak in Nile tilapia intensive farming: identification and pathological evaluation. Aquaculture491, pp.169-176.
  140. Venturelli, A., Tagliazucchi, L., Lima, C., Venuti, F., Malpezzi, G., Mago-ulas, G.E., Santarem, N., Calogeropoulou, T., Cordeiro-da-Silva, A. and Costi, M.P., 2022. Current Treatments to Control African Trypanosomiasis and One Health Perspective. Microorganisms10(7), p.1298.
  141. Yevtushenko, A.V., FEATURES OF THE PARASITIC SYSTEM FORMATION IN COMMON CARP IN THE AQUACULTURE OF THE NORTH-EASTERN AND EASTERN REGIONS OF UKRAINE. GUIDELINES FOR THE PREPARATION OF THE PAPERS SUBMITTED FOR PUBLICATION AT THE ‘JOURNAL FOR VETERINARY MEDICINE, BIOTECHNOLOGY AND BIOSAFETY’, p.9.
  142. Woodyard, E.T., Rosser, T.G., Stilwell, J.M., Camus, A.C., Khoo, L.H., Waldbieser, G., Lorenz, W.W. and Griffin, M., 2022. New data on Henneguya postexilis Minchew, 1977, a parasite of channel catfish Ictalurus punctatus, with notes on resolution of moleculasr markers for myxozoan phylogeny. Systematic Parasitology99(1), pp.41-62.
  143. Staton, L., Erdahl, D. and El-Matbouli, M. (2002). Efficacy of Fumagillin and TNP-470 to prevent experimentally induced whirling disease in rainbow trout Oncorhynchus mykiss. In: Whirling Disease: Reviews and Current Topics, Batholomew, J.L. and Wilson, J.C. (eds). American Fisheries Society, Symposium 29, Bethesda, Maryland, 18-22 August 2002, pp. 239-247.
  144. Liu, Y., Tan, X., Zhang, Y., Ling, F., Liu, T. and Wang, G., 2022. Isoimperatorin: A promising anti Gyrodactylus kobayashii natural compound from Angelica dahuricaAquaculture560, p.738552.
  145. Kvach, Y., Ondračková, M., Seifertová, M. and Hulak, B., 2019. Gyrodactylus ginestrae n. sp.(Monogenea: Gyrodactylidae), a parasite of the big-scale sand smelt, Atherina boyeri Risso, 1810 (Actinopterygii: Atherinidae) from the Black Sea. Parasitology research118(12), pp.3315-3325.
  146. Volkova, E. and Kudryavtsev, A., 2021. A morphological and molecular reinvestigation of Janickina pigmentifera (Grassi, 1881) Chatton 1953–an amoebozoan parasite of arrow-worms (Chaetognatha). International Journal of Systematic and Evolutionary Microbiology71(11), p.005094.
  147. Eissa, A.E., Attia, M.M., Abdelsalam, M., Elgendy, M.Y., Abou-Okada, M., Ismail, G.A. and Younis, N.A., 2022. Investigating the etiologies behind emergent mass mortalities of farmed Liza carinata juveniles from coastal farms at Damietta, Egypt. Scientific Reports12(1), pp.1-10.
  148. Gomes, G.B., Miller, T.L., Vaughan, D.B., Jerry, D.R., McCowan, C., Bradley, T.L. and Hutson, K.S., 2017. Evidence of multiple species of Chilodonella (Protozoa, Ciliophora) infecting Australian farmed freshwater fishes. Veterinary parasitology237, pp.8-16.
  149. Li, M., Wang, R., Gomes, G.B., Zou, H., Li, W.X., Wu, S.G., Wang, G.T. and Ponce-Gordo, F., 2018. Epidemiology and identification of two species of Chilodonella affecting farmed fishes in China. Veterinary parasitology264, pp.8-17.
  150. Wang, Z., Bourland, W.A., Zhou, T., Yang, H., Zhang, C. and Gu, Z., 2020. Morphological and molecular characterization of two Trichodina (Ciliophora, Peritrichia) species from freshwater fishes in China. European Journal of Protistology72, p.125647.
  151. Wang, S., Zhao, Y., Du, Y. and Tang, F., 2019. Morphological Redescription and Molecular Identification of Trichodina reticulata Hirschmann & Partsch, 1955 (Ciliophora, Mobilida, Trichodinidae) with the Supplemental New Data of SSU rDNA and ITS‐5.8 S rDNA. Journal of Eukaryotic Microbiology66(3), pp.447-459.
  152. Abdelkhalek, N.K., El-Adl, M.A., Salama, M.F., Elmishmishy, B., Ali, M.O., El-Ashram, A., Hamed, M.F. and Al-Araby, M.A., 2018. Molecular identification of Trichodina compacta Van As and Basson, 1989 (Ciliophora: Peritrichia) from cultured Oreochromis niloticus in Egypt and its impact on immune responses and tissue pathology. Parasitology Research117(6), pp.1907-1914.
  153. , U.B., Guo, Q., Zhao, D., Liu, Y. and Gu, Z., 2019. Description of Myxobolus xiantaoensis n. sp. from the fins of yellow catfish in China: a species previously attributed to Myxobolus physophilus Reuss, 1906 in Chinese records. Parasitology research118(4), pp.1137-1146.
  154. Xiang, Y., Zhang, J. and Zhao, Y., 2021. Molecular data and phylogenetic analysis of Myxobolus pseudonobilis n. sp. infecting the gill filaments of silver carp, Hypophthalmichthys molitrix Valenciennes, 1844. Parasitology International83, p.102314.
  155. , S., Pardavila, X., Ares-Mazás, E. and Gómez-Couso, H., 2022. Molecular identification of Eimeria species in Spanish bats. Parasitology International91, p.102621.
  156. Bastos, T.S.A., Faria, A.M., Couto, L.F.M., Nicaretta, J.E., de Assis Cavalcante, A.S., Zapa, D.M.B., Ferreira, L.L., Heller, L.M., de Carvalho Madrid, D.M., Cruvinel, L.B. and Rossi, G.A.M., 2020. Epidemiological and molecular identification of Trypanosoma vivax diagnosed in cattle during outbreaks in central Brazil. Parasitology147(12), pp.1313-1319.
  157. Margarido, Y.M., Adriano, E.A., Valladão, G.M., Naldoni, J. and Pilarski, F., 2021. Morphological, molecular, and histopathological characterization of a new species of Henneguya infecting farmed Astyanax lacustris in Brazil. Microbial Pathogenesis158, p.104991.
  158. Jin, X., Li, W., Cheng, Y., Li, M., Wu, S., Zou, H. and Wang, G., 2022. Description of Gyrodactylus banmae n. sp.(Monogenea, Gyrodactylidae) parasitic on zebrafish, Danio rerioParasitology International87, p.102531.
  159. Řehulková, E., Benovics, M. and Šimková, A., 2020. Uncovering the diversity of monogeneans (Platyhelminthes) on endemic cypriniform fishes of the Balkan Peninsula: new species of Dactylogyrus and comments on their phylogeny and host-parasite associations in a biogeographic context. Parasite27.
  160. Achatz, T.J., Martens, J.R., Kostadinova, A., Pulis, E.E., Orlofske, S.A., Bell, J.A., Fecchio, A., Oyarzún-Ruiz, P., Syrota, Y.Y. and Tkach, V.V., 2022. Molecular phylogeny of Diplostomum, Tylodelphys, Austrodiplostomum, and Paralaria (Digenea: Diplostomidae) necessitates systematic changes and reveals a history of evolutionary host switching events. International journal for parasitology52(1), pp.47-63.
  161. Corner, R.D., Cribb, T.H. and Cutmore, S.C., 2020. A new genus of Bucephalidae Poche, 1907 (Trematoda: Digenea) for three new species infecting the yellowtail pike, Sphyraena obtusata Cuvier (Sphyraenidae), from Moreton Bay, Queensland, Australia. Systematic Parasitology97(5), pp.455-476.
  162. Lockyer, A.E., Olson, P.D. and Littlewood, D.T.J., 2003. Utility of complete large and small subunit rRNA genes in resolving the phylogeny of the Neodermata (Platyhelminthes): implications and a review of the cercomer theory. Biological Journal of the Linnean Society78(2), pp.155-171.
  163. Cribb, T.H., Adlard, R.D. and Bray, R.A., 1998. A DNA-based demonstration of a three-host life-cycle for the Bivesiculidae (Platyhelminthes: Digenea). International Journal for Parasitology28(11), pp.1791-1795.
  164. Bazsalovicsová, E., Králová-Hromadová, I., Xi, B.W. and Štefka, J., 2018. Tour around the globe: the case of invasive tapeworm Atractolytocestus huronensis (Cestoda: Caryophyllidea), a parasite of common carp. Parasitology international67(4), pp.366-374.
  165. Ikuno, H., Akao, S. and Yamasaki, H., 2018. Epidemiology of Diphyllobothrium nihonkaiense diphyllobothriasis, Japan, 2001–2016. Emerging Infectious Diseases24(8), p.1428.
  166. Kvach, Y., Tkachenko, M.Y., Bartáková, V., Zięba, G. and Ondračková, M., 2021. The role of the non-indigenous pumpkinseed Lepomis gibbosus (Actinopterygii: Centrarchidae) in the life cycle of Bothriocephalus claviceps (Cestoda: Bothriocephalidae) in Europe. Parasitology Research120(9), pp.3163-3171.
  167. Ozcan, M. and Kartal, B., 2020. Molecular identification of the parasite Ligula intestinalis identified in Alburnus adanensisAsian Journal of Science and Technology11(1), pp.10700-10707.
  168. Muñoz-Caro, T., Machuca, A., Morales, P., Verdugo, J., Reyes, R., García, M., Rutaihwa, L., Schindler, T., Poppert, S., Taubert, A. and Hermosilla, C., 2022. Prevalence and molecular identification of zoonotic Anisakis and Pseudoterranova species in fish destined to human consumption in Chile. Parasitology Research121(5), pp.1295-1304.
  169. Troccoli, A., De Luca, F., Handoo, Z. A., & Di Vito, M. (2008). Morphological and molecular characterization of Pratylenchus lentis n. sp. (Nematoda: Pratylenchidae) from Sicily. Journal of nematology, 40(3), 190.
  170. Ratnasingham, S. and Hebert, P.D., 2007. BOLD: The Barcode of Life Data System (http://www. barcodinglife. org). Molecular ecology notes7(3), pp.355-364.
  171. Montes, M.M., Acosta Albarracin, M., Barneche, J., Croci, Y., Balcazar, D., Reig Cardarella, G.F. and Martorelli, S.R., 2022. Molecular phylogenetic relationship between Philometroides tahieli (Nematoda, Philometridae) and other philometrids from South America. Parasitology Research, pp.1-13.
  172. Cleveland, C.A., Eberhard, M.L., Thompson, A.T., Garrett, K.B., Swanepoel, L., Zirimwabagabo, H., Moundai, T., Ouakou, P.T., Ruiz-Tiben, E. and Yabsley, M.J., 2019. A search for tiny dragons (Dracunculus medinensis third-stage larvae) in aquatic animals in Chad, Africa. Scientific reports9(1), pp.1-7.
  173. Cheng, L.W., Rao, S., Wang, P.C. and Chen, S.C., 2022. First report of acanthocephalan parasite, Longicollum pagrosomi Yamaguti, 1935 in cultured red snapper (Lutjanus erythropterus) in Taiwan. Journal of Fish Diseases45(4), pp.579-593.
  174. García-Varela, M., Pérez-Ponce de León, G., De la Torre, P., Cummings, M.P., Sarma, S.S.S., Laclette, J.P., 2000. Phylogenetic relationships of acanthocephalan based on analysis of 18S ribosomal RNA gene sequences. J. Mol. Evol. 50: 532-540.
  175. Hua, C.J., Zhang, D., Zou, H., Li, M., Jakovlić, I., Wu, S.G., Wang, G.T. and Li, W.X., 2019. Morphology is not a reliable taxonomic tool for the genus Lernaea: molecular data and experimental infection reveal that L. cyprinacea and L. cruciata are conspecific. Parasites & vectors12(1), pp.1-13.
  176. Soares, G.B., Domingues, M.V. and Adriano, E.A., 2021. Morphological and molecular characterization of Udonella brasiliensis n. sp.(Monogenoidea), an epibiont on Caligus sp. parasite of Ariidae from the southeastern coast of Brazil. Parasitology International83, p.102371.
  177. Song Y., Wang G. T., Yao W. J., Gao Q. and Nie P. 2008. Phylogeny of freshwater parasitic copepods in the Ergasilidae (Copepoda: Poecilostomatoida) based on 18S and 28S rDNA sequences. Parasitol Res. 102:299–306.

Similar Posts