<<

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

International Journal of Current Microbiology and Applied Sciences ISSN: 2319-7706 Volume 9 Number 9 (2020) Journal homepage: http://www.ijcmas.com

Review Article https://doi.org/10.20546/ijcmas.2020.909.096

Disease Diagnosis Prevention and Control of Diseases in L. vannamei

T. Suguna*

Fisheries Research Station, S.V. Veterinary University, West Godavari, Andhra Pradesh - 534 199, India

*Corresponding author

ABSTRACT

K e yw or ds Aquaculture plays a major role in augmenting food production, levelihood and

L. Vananmei, thereby the economy of a country. Andhra Pradesh is the leading state in cultured Diseases, Disease shrimp production, value and exports. Increasing intensification of shrimp causing agents, aquaculture, devoid of awareness on scientific farming practices among farmers,

Preliminary spurious seed and water pollution, exerted disease problems. Diseases in shrimp knowledge, Preventive farming are a recurring problem and the future is dwindling and farmers are facing measures heavy losses. Despite of these problems, farmers are forced to use of therapeutic chemicals indiscriminately causing abrupt change in water quality which lead to

Article Info stress. The increased stress conditions in the pond pave way for onset of serious

problems by many disease causing pathogens such as , bacteria, fungi and Accepted: protozoa in culture. In order to overcome the situation, shrimp farmer has to have 07 August 2020 scientific knowledge on various diseases, causative factors and remedial measures, Available Online: to sustain the crop. 10 September 2020

Introduction increased remarkably. According to MPEDA (2015), farming area has increased from 283 In Aquaculture, especially shrimp farming, ha in 2009-10 to 50,241 ha in 2014-15 and the has witnessed tremendous growth in the last production has also grown from 1731 to several years, and this has been achieved 353,413 metric tonnes. However, as happened through expansion, intensification and in the case of P. monodon, P. vannamei diversification. Like in other shrimp growing farming also has been experiencing large- countries, major transformation in shrimp scale production losses due to a variety of production has been witnessed in India, after diseases. These diseases have been either due switching over from native species, Penaeus to infectious aetiology, poor management monodon to the non-native species, whiteleg measures, spurious seed, water pollution. shrimp, Penaeus vannamei which was Diseases are the outcome of imbalance introduced during 2009-10. Ever since the between environment, host and pathogen. The introduction of P. vannamei, the area under impact of disease is a combination of several farming as well as the production have factors such as mass mortality of shrimp,

764

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

reduction in feed acceptance or morbidity. animals. Although many pathogens results in Morbidity and production losses due to heavy mortality in farmed shrimp, some of multiple disease conditions of obscure the emerging disease conditions which are aetiology have been a common phenomenon reported recently resulted in severe retarded and running mortality syndrome (RMS), slow growth or other chronic conditions. growth / retarded growth caused by multiple Nevertheless, these chronic conditions are causes, white gut and white faeces syndrome often resulted in heavy production losses and and vibriosis etc. have been of common significant economic impact in shrimp occurrence. Although shrimp farming in the farming. neighboring Asian countries and Latin America have been severely affected by Emerging diseases Acute Hepato Pancreatic Necrosis Disease (AHPND), popularly known as Early Globally, disease emergence is recognized as Mortality Syndrome (EMS), Indian shrimp an important constraint to the expansion of farming system has not been affected by this aquaculture. An “emerging disease” is serious problem. defined as a new disease, a new presentation of a known disease or an existing disease that Concomitant with the expansion and appears in a new geographical area (Brown, intensification of shrimp aquaculture, large 2000). According to Walker and Winton number of diseases, especially those caused (2010), emerging disease can be categorized by viral infection, have been recorded in into four: „i) new or previously unknown farmed shrimp. Disease such as white spot diseases; ii) known diseases appearing for the syndrome virus (WSSV) has devastated the first time in a new species (expanding host shrimp aquaculture world over due to its high range); iii) known diseases appearing for the virulence, wide host and geographical range. first time in a new location (expanding Some of the other serious viral diseases geographical range); and iv) known diseases include Taura Syndrome Virus (TSV), with a new presentation (different signs) or Yellow Head Virus (YHV), infectious higher virulence due to changes in the hypodermal and haematopoietic necrosis causative agent‟. Further, Walker and Winton virus (IHHNV), infectious myonecrosis virus reported that emerging new diseases are (IMNV) etc., In addition, a number of challenging problems because of the pathogens or disease conditions have emerged following reasons: „a) difficulty or delay in in farmed shrimp during the past several years developing confirmatory diagnostic tools; b) and caused significant production loss to the poor knowledge on the host susceptibility and industry. Further, some of the existing / host-range; c) lack of knowledge on the endemic diseases have shown increased epidemiological factors such as mode of virulence and a few of them also showed transmission, reservoirs and carrier hosts etc‟. distribution in new host species and Following are some of the diseases which can geographical areas. Some of the reasons be categorized under emerging diseases in attributed to these phenomena include high farmed shrimp. intensification of culture practices, introduction of non-native species for (WSD) farming, unregulated transboundary movement of aquatic animals and products White Spot Syndrome disease is the most together with changing climate and the serious threat faced by the shrimp farming increased stress imposed on the farmed industry worldwide. It was first reported in

765

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

farmed P. japanicus from Japan 1992 – 93 (Rajendran et al., 2016). EHP is reported to and later identified in all shrimp farming be associated with growth retardation in countries. The WSSV can infect a wide range farmed shrimp. It was first discovered from of aquatic crustaceans including marine, Penaeus monodon in Thailand in 2004 brackish and freshwater penaeids, crabs and (Chayaburakul, et al., 2004), and reported as cray fish. All decopod crustaceans including an undersigned microsporidian. Subsequently, crabs, cray fish, freshwater prawns, spriny the parasite was identified and characterized labsters and clawed labsters in marine from P. monodon from Thailand in 2009. It brackish and freshwater sources are infects only the epithelial cells of the susceptible but morbidity and mortality as a hepatopancreatic (HP) tubules of shrimp. The consequence of infection is highly variable. EHP infection affects the digestive and Penaeid shrimps are highly susceptible to absorptive functioning of the hepatopancreas infection, often resulting in high mortality. leading to poor growth and immunity (Fig. 2). Prevalence of WSSV is reported as highly Although there are reports indicating that valuable, from <1% in infected wild EHP would cause growth retardation in populations to up to 100% in captive farmed shrimp, a statistical correlation populations. It is caused by a double standard between the parasite and the clinical signs has DNA virus. It damages the stomach, gills, not been proved so far. However, the parasite antennal gland, heart and eyes. Affected will not cause any mortality or obvious shrimp are lethargic, show reddish body clinical signs and symptom. Nevertheless, the discoloration during advanced stage of micropsoridian had been identified as one the disease. WSD is diagnosed by presence of pathogenic agents associated with white faces characteristic white spots and rapid sysndrome (WFS) in cultured P. monodon in mortalities (Fig. 1). WSSV can be detected Vietnam. Similar indications have also been using polymerase chain reaction (PCR). It is reported from Thailand. transmitted vertically and horizontally by cannibalism, predation and by water borne EHP could not transmitted directly from routs. There is no treatment for WSSV except shrimp by the oral (horizontal) route to take preventive measures, not to enter in (Tangprasittipap et al., 2013). Clinical signs the culture system. So filter and sanitise the of EHP infection are not specific. The EHP pond water before stocking and follow good spores can be detected by light microscopy in management practices and strict biodiversity tissue sections or in squash preparation of measures to avoid the entry of disease in hepatopencreas using specific vital stains. culture. EHP spores could be reported from faecal matter of shrimp exhibiting white faeces Hepatopancreatic microsporidiosis (HPM) syndrome (WFS) also (Tang et al., 2015); Rajendran et al., 2016). Other reported Hepatopancreatic microsporidiosis, caused by diagnostic methods include histopathology, in the microsporidian parasite, Enterocytozoon situ hybridization, polymerase chain reaction hepatopenaei (EHP), is an emerging pathogen (PCR), loop – mediated isothermal of cultured penaeid shrimp. Incidence of EHP amplification (LAMP) and real-time PCR has been reported from cultured shrimps of (Suebsing et al., 2013a; Tangprasittipap et al., many Southeast Asian Countries including 2013; Tang et al., 2015; Liu et al., 2015). Vietnam, Thailand, Malaysia, Indonesia and China. Recently, the pathogen has been Though it does not cause mortality, but limits recorded in the farmed shrimp in India growth and production finally resulting in

766

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

economic loss. Complete drying of pond insecticidal related genes PirA and Pir B bottom can limit the spores, stock should be (Lightner 2014), and these genes have been PCR tested and follow strict biosecurity used for diagnosing AHPND by PCR measures as prevention. methods (Flegel and Lo, 2014; Flegel, 2014). Recently, a loop-meditated isothermal Early Mortality Syndrome (EMS) / Acute amplification (LAMP) method to detect hepatopancreatic necrosis Syndrome or AHPND has been developed (Koiwai et al., disease (AHPNS or AHPND) 2015).

Recently, a new emerging disease known as AHPND can be prevented by use of SPF Early Mortality Syndrome (EMS) or Acute broodstock and restrict adoption of BMPS in Hepatopancreatic Necrosis Disease hatcheries and grow out farms. Its horizontal (AHPND), has been a major issue of concern transfer can be reduced by disinfection during for economic loss in the shrimp farming pond preparation. Pond sludge management is industry in many Asian countries and Latin also an importance strategy along with good America. Incidence of AHPND has been management practices. reported from China in 2010 and subsequently in Vietnam, Thailand, Malaysia Infectious myonecrosis (IMN) and Mexico (Tran et al., 2013; Soto – Rodriguex et al., 2015). However, there are Outbreaks of infectious myonecrosis (IMN) no confirmed reports of EMS from India. It due to infectious myonecrosis virus (IMNV) affects both Penaeus monodon and P. were first reported from Brazil in whiteleg vannamei (P. chinensis was also reported to shrimp, Penaeus vannamei, in 2002. be affected in China). The EMS / AHPND Infectious myonecrosis disease is disease typically affect shrimp post larvae characterized by extensive necrosis of skeletal within 20-30 days after stocking and muscle, especially in the distal abdominal frequently causes up to 100% mortality. segments and tail fin. This disease resulted in Recently, the i8.paraharmolyticus that is cumulative mortality rate of up to 70% infected by a phage, which causes it to release throughout the production cycle. The species a potent toxin (Lightner, 2014). Gross clinical of shrimp P. vannamei, P. stylirostris and P. signs of the disease include lethargy, slow monodon are susceptible to IMNV and among growth, corkscrew swimming, loose shells these species P. vannamei is the most and pale colouration. Other signs include an susceptible. IMNV has been reported to be empty stomach and midgut, and a pale to transmitted from shrimp to shrimp by white atrophied hepatopancreas, and cannibalism. Transmission via water and ultimately dead animals can be seen at the vertical transmission from broodstock to bottom of the pond. The most distinctive progeny probably occurs. The causative agent histological feature of AHPND is extensive has been identified as an RNA virus known as rounding and sloughing of hepatopancreatic infectious myonecrosis virus (IMNV), tubule epithelial cells in the absence of any belonging to the family. Severely detected causative pathogen (Tran et al., infected shrimp become lethargic during or 2013). after stressful events such as capturing using cast-netting, feeding, sudden changes in It has been reported that the AHPND V. temperature and drop in salinity. However, parahaemolyticus strains possessed a unique shrimp will have full gut. In acute infection, plasmid called pVPA3-1, containing animals show extensive whitish necrosis in

767

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

skeletal muscles, especially in the distal have revealed geographic variations in abdominal segments and tail (Fig. 3). infectious hypodermal and harmatopoietic Lymphoid organ shows excessive necrosis virus. It is caused by a small single hypertrophy. Necrotic areas may appear standard DNA – containing parvovirus. The reddened similar to the color of cooked gross signs of disease are not specific to shrimp in some animals. Severe infection IHHNV. But included reduced feeding, results in morbidity and high mortality which elevated morbidity and mortality rates, continues for several days. Juveniles and sub fouling by epicommonsals and bluish body adults of L. vannamei farmed in marine or coloration. low saline, brackish water, appear to be the most severely affected by IMNV disease. The In L. vannamei IHHNV cause runt deformity principal target tissue for IMNV includes the syndrome (RDS) which is growth retarded striated muscle, connective tissue, disease. Along with this, infected shrimp haemocytes and lymphoid organs. There is no show change in color, deformed culture, reported treatment for IMNV. Stock should be rostrum, winkled antennal flagella, cuticular PCR tested and seed should be of SPF quality. roughness, deformities in cuticular, thoragic Good Management Practices proven to be the and abdominal areas of exoskeleton (Fig. 4). most successful method to control the disease. Reduced feed consumption change in behaviour and appearance are also noticed. Current diagnostic methods for the detection There is no treatment for this viral disease. As of IMNV range from clinical observation and a preventive measure, the ponds should be histological examination (Poulos et al., 2006; dried between two crops, stock should be da Silva et al; 2010) to molecular approaches. PCR tests, seed of SPF quality, strict Molecular diagnosis include in situ biosecurity and good management practices hybridization using IMNV – specific gene have to be followed. probes (Tang et al., 2005). Molecular diagnostic methods reported include RT-PCR Muscle necrosis of Penaeus vannamei and nested RT-PCR (Poulos and Lightner, caused by Penaeus vannamei nodavirus 2006; Senapin et al., 2007), quantitative real – (PvNV) time RT-PCR (Andrate et al., 2007), and reverse-transcriptase loop-mediated Viral muscle necrosis is an emerging disease isothermal amplication reaction (RT-LAMP; of Penaeus (Litopenaeus) vannamei caused by Puthawibool et al., 2009; Andrade and Penaeus vannamei nodavirus (PvNV). The lightner, 2009). Apart from these, monoclonal disease was first reported in Belize in 2004 antibiotics have been developed for detecting and caused 50% production losses in affected major capsid proteins of IMNV (Seibert et al., shrimp ponds (Tang et al., 2007). PvNV is a 2010). Moreover, an immunichromatographic member of the family and genus strip test has been developed for the rapid Alphanodavirus (Tang et al., 2011). Infected diagnosis of IMNV (Chaivisuthangkura et al., shrimp show clinical signs of white, opaque 2013). lesions in the tail muscle thus resembling infectious myonecrosis virus (IMNV) Infectious Hypodermal and infection. Histopathological changes include Haematopoietic Necrosis Virus (IHHNV) multifocal necrosis and haemocytic fibrosis in the skeletal muscle. Basophilic cytoplasmic It was first discovered in P. vannamei and P. inclusions in strained muscle, lymphboid stylirostris in America in 1981. Recent studies organ and connective tissues are also

768

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

observed. The disease also causes lymphoid disease has not been identified; however, organ spheroids. Although the virus does not reports indicate the involvement of multiple cause serious mortality, the infection impacts causes such as covert mortality disease caused survival in grow-out ponds. Sporadic by covert mortality nodavirus (CMNV), white mortality of infected shrimp has been muscle syndrome, white gut / faeces recorded when they are under environmental syndrome and white patch disease. stress such as crowding (stocking density (>50 m-2) and high temperature (>32ºC) and The dead and moribund shrimp are found survival decreased to 40% and increased food every day. The redding of appendage, tail and conversion ratio. The disease has not been white abdomen muscle are symptoms of reported from India. disease. Measures to be taken in selection of seed quality and follow strict biosecurity and Molecular techniques such as in situ good management practices. hybridication and nested RT-PCR method have been developed for the detection of White gut / faeces syndrome (WFS) PvNV (Tang et al., 2017). Recently, reverse transcription loop mediated isothermal This has been observed in farmed P. monodn amplification (RT-LAMP) combined with and P. vannamei. Gross signs of WFS in colorimetric gold nanoparticles(AuNP) probe shrimp include the appearance of faecal assay for visual detection of PvNV has been string-like bodies in the gut. It appears like developed (Suebsing et al., 2013b). PvNV vermiform bodies that resemble gregarines could be detected in mosquito fish, seabird within the hepatopancreatic tubules, at the feces, barnacles, and zooplankton by RT- hepatopancreas- stomach – midgut junction PCR, indicating their role as carriers of and in the midgut (Fig. 6). It generally occurs PvNV. This disease also has not been approximately from 2 months of culture and reported from India. The stock should be PCR initially the causative agent has been reported tested and seed should be of SPF quality. as gregarines and the condition is described as Good management practices are the remedied white faeces syndrome (WS). The disease has measures. been reported to cause 10-15% production loss due to decreased survival and smaller Running mortality syndrome (RMS) harvest size of shrimp. Although the causative organisms has not been identified, latest The term running mortality syndrome (RMS) report shows that WFS arises from has been used to describe prolonged chronic transformation, sloughing and aggregation of mortality during a crop. The mortality starts hepatopancreatic microvilli into vermiform 1-2 months after stocking and become severe bodies, which superficially resembles like during later part of summer crop. This with protozoan, gregarines (Sriurairatana et condition has been noticed in farmed P. al., 2014) and this will result in retarded vannamei in India, since 2001. The farmed growth and may predispose shrimp to shrimp in the affected ponds showed different opportunistic pathogens. Vibrio species have mortality patterns which are result of unusual been found in the faecal analysis from symptoms with no correlation to any other infected shrimps. Incidence of WFS in shrimp reported diseases. Reports indicate that some has been reported to be associated with farmers have lost up to four crops, with stressful conditions including high stocking mortality percentage reaching 50-70% in the densities, poor water quality, poor pond most the cases (Fig. 5). The exact cause of the bottom, high plankton blooms and bad feed

769

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

management and high pollution in pond EHP and WFS has not been established, water. White faeces syndrome has been spores of the microsporidian have been reported to be a serious problem in P. recorded in the faecal samples of WFS vannamei culture ponds of Andhra Pradesh infected / slow growing shrimp (Rahendran et and Tamilnadu (Mastan, 2015). Although al., 2016; Tang et al., 2016). direct correlation between the microsporidian

Fig.1 White spot disease

Fig.2 L. vannamei effected with growth retrodation and there by showing wide range of size variation due to Hepatopancreatic microsporidiosis

Fig.3

770

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

Fig.4

Fig.5

Fig.6 White gut / faeces syndrome

Fig.7

771

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

Fig.8

Fig.9

The disease can cause moderate to severe Necrotic areas appear white opaque patches economic loss by reducing the shrimp and in later stage the white patches changed survival by 20-30%. The cause of white faces into black spot or splinter. Other symptoms syndrome and treatment is uncertain. include whitish blue discoloration of infected However reduced stocking density, proper shrimp body, loss of appetite, roughness on water exchange together with better whole surface of infected shrimp with or management practices will be helpful in without red discoloration and pale white evading WFS. muscles. The mortality rate was observed up to 70%. The causative agent responsible for White patch disease this condition has been identified as Gram- positive bacterium, Bacillus cereus. The This condition has been witnessed in the isolates exhibited high degree of proteolytic, semi-intensive P. vannamei farming in haemolytic and lipase activity sufficient to Andhra Pradesh and Tamilnadu. Initially lyse the cuticle of shrimp. (before 2012), the outbreak was observed in summer season only, however, in 2014, the Vibriosis disease outbreak could be noticed both in the winter and summer seasons also (Velmurugan The prevalence of Vibrio parahaemolyticus et al., 2015). The major symptoms include and V. harveyi has caused the occurrence of focal to extensive necrotic areas in striated tail vibriosis and resulted in severe crop losses to muscle tissues and abdominal muscles tissues. the farmers. It easily affects the shrimp

772

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

cultured in saline waters and known as associated with black gill cause mortality in “bacterial septicaemia”. The shrimp are ponds. The gills become black in colour and affected at any stage. Environmental stress the shrimp swims on the surface of pond (Fig. aggrevate the disease and cause huge loss to 8). In low DO ponds mortality is notices. farmers. Regular monitoring of water quality parameters, bacterial load and following good The major symptoms include the exhibition of management practices are the preventive septicaemia conditions followed by loss of measures. reflex and cuticular fouling. The gills appear brown in color and the body becomes red. Loose Shell Syndrome (LSS) Antennal cut is also been observed. The affected shrimps do not eat and hence Loose shell syndrome is a chronic, non stomach appears empty and at times white infectious disease in shrimp which cause watery liquid oozes out sometimes serious threat to farmers. Due to loose shell luminescence also been observed in ponds. syndrome the feed conversion efficiency of shrimp substantially declines, leading to poor In serious conditions mortality is observed. meat quality and doesn‟t fetch price. Affected Blackening or whitening of the basal part of ponds suffer poor survival and chronic the antenna, the oviduct and edges of the mortalities. Stress factors like plankton crash, abdominal segments. The shrimps may water quality and weather changes, the aged exhibit either one of the symptoms or all the ponds with high organic load, high densities, symptoms based on disease severity. The toxic gases like ammonia and nitrite are prone preventive measures like drying between to loose shell disease. production cycles, strict biosecurity measures and good management practices are to be The affected shrimp has a paper like carapace adopted (Fig. 7). with a gap between muscle tissue and exoskeleton (Fig. 9). They become sluggish Black Gill Disease and the growth retards. Regular drying of ponds between two crops, monitoring of Affected shrimps have gills with black to water quality and following good brown discoloration, in acute cases necrosis management practices are preventive and atrophy of the gill lamualle may be measures. apparent. Blackening is due to the deposition of melanin at sites of massive haemocyte In conclusion it is extremely difficult to control accumulation, followed by dysfunction and disease once it strikes. Hence prevention is destruction of gill processes. A small always better than cure. Disease prevention percentage of shrimp population in ponds largely depends on the implementation of occasionally suffer with black gill disease. biosecurity measures and best management But in recent vannamei crops, incidence of practices in the farms, disinfection, drying, black gill disease is in rise. The accumulation removing the accumulated organic matter, tilling, of organic load, feed wastage leads to the liming, management of feed, water quality formation of toxic gases like ammonia, nitrite parameters and aquatic animal health and hydrogen sulphide are the main causative management and further responsible farming factors for this disease. Sometimes vibrio results in a successful growth, production and bacteria may also pose blackening of gills in enhanced economic status and simultaneously shrimp. Low DO levels in pond waters sustainability of culture.

773

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

References Flegel, T.W., Lo, C.F. 2014. Interim primers for specific detection of bacterial Andrade, T.P., Lightner, D.V. 2009. isolates that cause acute Development of a method for the hepatopancreatic necrosis disease direction of infectious myonecrosis (AHPND) virus by revere-transcription loop- (http://enaca.org/modu;es/library/publi mediated isothermal amplification and cation. php?publication_id+1128) nucleic acid lateral flow hybrid assay. Koiwai, K., Tinwongger, S., Nozaki, R., J.Fish. Dis. 32, 911-924. Kondo, H., Hirono, I. 2016. Detection Andrade, T.P.D., Srisuvan, T., Tang, K.F.J., of acute hepatopancreatic necrosis Lightner, D.V. 2007. Real-time revere disease strain of Vibrio transcription polymerase chain parahaemilyticus using loop-mediated reaction assay using TaqMan probe isothermal amplification. J. Fish Di, for detection and quantification of 39, 603-606. infectious myonecrosis virus (IMNV). Lightner, D.V. 2014. Documentation of a Aquaculture 264, 9-15. unique strain of Vibrio Brown, C. 2000. Emerging diseases of parahaemolyticus as the agent of early animals – an overview. In: Brown, C., mortality syndrome (EMS) or acute Bolin, C. (Eds.), Emerging Diseases of hepatopancreatic necrosis disease Animals. American Society for (AHPND) affecting penaeid shrimp Microbiology, p. 324. with notes on the putative toxins. 9th Chaivisuthangkura, P., Senapin, S., symposium on diseases in Asian Wangman, P., Longyant, S., Aquaculture, Ho Chi Minh City, Sithigorngul, P. 2013. Simple and Vietnam. rapid detection of infectious Liu, Z., Zhang, Q.-L., Wan, X.-Y., Huang, J., myonecrosis virus using an 2015. Development of real-time PCR immunochromatographic strip test assay for detection of microsporidian Arch. Virol. 158, 1925-1930. Enterocytozoon hepatopenaei and Chayaburakul, K., Nash, G., Pratanpipat, P., detection in shrimp samples under Sriurairatana, S., Withyachumnarnkul, different growth rates. Prog. Fish. Sci. B., 2004. Multiple pathogens found in (in press, in Chinese, English growth-retarded black tiger shrimp Abstract). Penaeus monodon cultivated in Mastan, S.A. 2-015. Incidences of white feces Thailand. Dis. Aquat. Org. 60, 89-96. syndrome (WFS) in farm-reared Da Silva, V.A., dos Santos, F.L., Bezerra, shrimp Litopenaeus vannamei, Andhra S.S., Pedrosa, V.F., Mends, P.D., Pradesh. Indo American Journal of Mendes, E.S. 2010. A multi-season Pharmaceutical Research. 5 (09). survey for infectious myonecrosis in MPEDA, 2015. The Marine Products Export farmed shrimp, Litopenaeus Development Authority. vannamei, in Pernambuco, Brazil. J. www.mpeda.gov.in. Invertebr. Pathol. 104, 161-165. Poulos, B.t., Lightner, D.V. 2006. Detection Flegel, T.W. 2014. A game changer for the of infectious myonecrosis virus future development of aquaculture. 9th (IMNV) of penaeid shrimp by reverse symposium on diseases in Asian –transcriptase polymerase chain Aquaculture, Ho Chi Minh City, reaction (RT-PCR). Dis. Aquat. Vietnam. Organ. 73, 69-72.

774

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

Puthawibool, T., Senapin, S., Kiatpathomchai, Transformation, Sloughing and W., Flegel, T.W. 2009. Detection of Aggregation of Hepatopancreatic shrimp infectious myonecrosis virus Microvilli into Vermiform Bodies by reverse transcription loop-mediated Superficially Resembling Gregarines. isothermal amplification combined PLoS One. 9(6), e99170. with a lateral flow dipstick. J. Virol. Suebsing, R., Prombun, P., Srisala, J., Tmethods 156, 27-31. Kiatpathomchai, W., 2013a. Loop Rajendran, K.V., Shivam, S., Praveeena, P.E., mediated isothermal amplification Rajan, J.J.S., Kumar, T.S., Avunje, S., combined with colorimetric nanogold Jagadeesan, V., Babu, P.S.V.A.N.V., for detection of the microsporidian Pande, A., Krishnan, N., Alavandi, Enterocytozoon hepatopenaei in S.V., Vijayan, K.K. 2016. Emergence penaeid shrimp. J. Appl. Microbiol. of Enterocytozoon hepatopenaei 114, 1254-1263. (EHP) in farmed Penaeus Suebsing, R., Prombun, P., Kiatpathomchai, (Litopenaeus) vannamei in India. W. 2013b. Reverse transcription loop- Aquaculture 454, 272-280. mediated isothermal amplification Seibert, C.H., Borsa, M., Rosa, R.D., (RT-LAMP) combined with Cargnin-Ferreira, E., Pereira, A.M.L., colorimetric gold nanoparticle (AuNP) Grisard, E.C., Zanetti, C.R., Pinto, probe assay for visual detection of A.R. 2010. Detection of major capsid Penaeus vannamei nodavirus (PvNV). protein of infectious myonecrosis Lett. Appl. Microbiol. 56, 428-435. virus in shrimps using monoclonal Tang, K.F.J., Hana, J.E., Aranguren, L.F., antibodies. J. Virol. Methods 169, White-Noble, B., Schmidt, M.M., 169-175. Piamsomboon, P., Risdiana, E., Senapin, S., Phewsaiya, K., Briggs, M., Hanggono. 2016. Dense populations Flegel, T.W. 2007. Outbreaks of of the microsporidian Enterocytosoon infectious myonecrosis virus (IMNV) hepatopenaei (EHP) in feces of in Indonesia confirmed by genome Penaeus vannamei exhibiting white sequencing and use of an alternative feces syndrome and pathways of their RT-PCR detection method. transmission to healthy shrimp. J. Aquaculture 266, 32-38. Invert. Pathol. 140, 1-7. Soto-Rodriguez, S.A., Gomez-Gil, B., Tang, K.F., Pantoja, C.r., Poulos, B.T., Lozano-Olvera, R., Betancourt- Redman, R.M., Lighner, D.V. 2005. In Lozano, M., Morales-Covarrubias, situ hybridization demonstrates that M.S. 2015. Filed and experimental Litopenaeus vannamei, L. stylirostris evidence of Vibrio parahaemolyticus and Penaeus monodon are susceptible as the causative agent of acute to experimental infection with hepatopancreatic necrosis disease infectious myonecrosis virus (IMNV). (AHPND) of cultured shrimp Dis. Aquat. Organ. 63, 261-265. Litopenaeus vannamei) in north- Tang, K.F.J., Pantoja, C.R., Redman, R.M., western Mexico. Appl. Environ. Han, J.E., Tran, L.H., Lightner, D.V. Microbiol. 81, 1689-1699. 2015. Development of in situ Sriurairatana, S., Boonyawiwat, V., hybridization and PCR assays for the Gangnonngiw, W., Laosutthipong, C., detection of Enterocytozoon Hiranchan, J. 2014. White Feces hepatopenaei (EHP), a microsporidian Syndrome of Shrimp Arise from parasite infecting penaeid shrimp. J.

775

Int.J.Curr.Microbiol.App.Sci (2020) 9(9): 764-776

Invert Pathol. 130. 37-41. Tran, L.H., Nuvan, l., Redman, R.M., Tang, K.F.J., Pantoja, C.R., Redman, R.M., Mohney, L.L., Pantoja, C.R., Lightner, D.V. 2007. Development of Fitzsimmons, K., Lightner, D.V. 2013. in situ hybridization and RT-PCR Determination of the infectious nature assay for the detection of a nodavirus of the agent of acute hepatopancreatic (PvNV) that causes muscle necrosis in necrosis syndrome affecting penaeid Penaeus vannamei. Dis. Aquat. Org. shrimp, Dis. Aquat. Organ. 105, 45- 75, 183-190. 55. Tang, K.F.J., Pantoja, C.R., Redman, R.M., Velmurgugan, S., Palanikumar, P., Navarro, S.A. and Lightner, D.V. Velayuthani, P., Donio, M.B.S., (2011) Ultrastructural and sequence Michael Babu, M., Lelin, C., Sudhkar, characterization of Penaeus vannamei S., Citarasu, T. 2015. Bacterial white nodavirus (PvNV) from Belize. Dis. patch disease caused by Bacillus Aquat. Organ. 94, 179-187. cereus, a new emerging disease in Tangprasittipap, A., Srisala, J., Chouwdee, S., semi intensive culture of Litopenaeus Somboon, M., Chuchird, N., vananmei. Aquaculture. Limsuwan, C., Srisuvan, T., Flegel, doi:10.1016/j.aquaculture. T.W., Sritunyalucksana, K., 2013. The 2015.03.017. microsporidian Enterocytozon Walker, P.J., Winton, J.R. 2010. Emerging hepatopenaei is not the cause of white viral diseases o fish and shrimp. Vet. feces syndrome in whiteleg shrimp Res. 41(6), 51. Penaeus (Litopenaeus) vannamei. BMC Vet. Res. 9, 139-148.

How to cite this article:

Suguna, T. 2020. Disease Diagnosis Prevention and Control of Diseases in L. vannamei. Int.J.Curr.Microbiol.App.Sci. 9(09): 764-776. doi: https://doi.org/10.20546/ijcmas.2020.909.096

776