Next Article in Journal
Specific Recognition of the 5′-Untranslated Region of West Nile Virus Genome by Human Innate Immune System
Next Article in Special Issue
Synthesized Magnolol Derivatives Improve Anti-Micropterus salmoides Rhabdovirus (MSRV) Activity In Vivo
Previous Article in Journal
Design of a US28 ORF Deletion Virus in a Temperature-Sensitive Cytomegalovirus Strain Fails to Promote Lytic Replication in Hematopoietic Cells
Previous Article in Special Issue
Aptamer-Based High-Throughput Screening Model for Efficient Selection and Evaluation of Natural Ingredients against SGIV Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Review of Medicinal Plants and Active Pharmaceutical Ingredients against Aquatic Pathogenic Viruses

1
Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Sciences, Nanning 530007, China
2
Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, College of Marine Sciences, Beibu Gulf University, Qinzhou 535000, China
3
Guangxi Fisheries Technology Extension Station, Nanning 530007, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2022, 14(6), 1281; https://doi.org/10.3390/v14061281
Submission received: 1 June 2022 / Accepted: 8 June 2022 / Published: 13 June 2022

Abstract

:
Aquaculture offers a promising source of economic and healthy protein for human consumption, which can improve wellbeing. Viral diseases are the most serious type of diseases affecting aquatic animals and a major obstacle to the development of the aquaculture industry. In the background of antibiotic-free farming, the development and application of antibiotic alternatives has become one of the most important issues in aquaculture. In recent years, many medicinal plants and their active pharmaceutical ingredients have been found to be effective in the treatment and prevention of viral diseases in aquatic animals. Compared with chemical drugs and antibiotics, medicinal plants have fewer side-effects, produce little drug resistance, and exhibit low toxicity to the water environment. Most medicinal plants can effectively improve the growth performance of aquatic animals; thus, they are becoming increasingly valued and widely used in aquaculture. The present review summarizes the promising antiviral activities of medicinal plants and their active pharmaceutical ingredients against aquatic viruses. Furthermore, it also explains their possible mechanisms of action and possible implications in the prevention or treatment of viral diseases in aquaculture. This article could lay the foundation for the future development of harmless drugs for the prevention and control of viral disease outbreaks in aquaculture.

1. Introduction

With the increasing demand for high-quality food in modern society, aquatic products account for an increasing proportion of meat consumption [1], because modern nutrition believes that excessive intake of red meat increases the risk of rectal cancer, advocating its replacement with other foods [2], while fish and shrimp foods are also able to provide the body with essential amino acids, lipids, and minerals, representing an excellent substitute [3]. With the increasing tension in global aquatic wild fishing resources and the increasing demand for aquaculture production, the aquaculture industry has been developing rapidly while facing increased challenges, especially the steep increase in the outbreak of aquatic animal diseases due to intensive farming, resulting in the death of a large number of farmed species and serious economic losses [4,5]. More importantly, antibiotics cannot control viral diseases; when aquatic animals are infected with viral diseases, the use of antibiotics by unprofessional farmers not only fails to alleviate the condition, but also exacerbates water pollution, resulting in an increase in mortality instead of a decrease [6].
Studies have shown that only 20–30% of antibiotics can be absorbed by farmed fish, with most of them entering the water environment [7]. The addition of antibiotics to fish and shellfish production sites via feed is a direct route to aquatic environmental pollution [8]. Intensive aquaculture leads to the overfeeding of aquatic animals and the flow of large amounts of antibiotics from ponds to lakes and then to the oceans, contributing to the deterioration of global biodiversity [9]. Abuse of antibiotics can easily cause irreversible consequences in the ecosystem. More seriously, the harmful components of antibiotic and chemical agents will remain in aquatic products, and then enter the human body through ingestion, which can directly endanger human health, causing allergic reactions, disrupting the balance of the human microbiota, affecting the growth and development of children, and even causing cancer [10]. For example, tetracycline antibiotics can inhibit bone marrow hematopoietic function, causing human aplastic anemia [11]; furan antibiotics can cause human hemolytic anemia and acute liver necrosis [12].
In order to ensure the green and sustainable development of aquaculture, it is urgent to find alternatives to antibiotics and chemical drugs. In fact, medicinal plants are very suitable candidates for antibiotic alternatives. Medicinal plants contain a variety of active ingredients, including polysaccharides, alkaloids, organic acids, flavonoids, and phenols, which are antibacterial and antiviral, in addition to promoting the body’s immune function and improving the body’s ability to resist diseases [13]. In recent years, because medicinal plants have the advantages of low toxicity, few side-effects, no drug resistance, few drug residues, and low prices, they have attracted much attention in the prevention and treatment of aquatic animal diseases, and substantial progress has been made in research. For example, methanolic extracts of Urtica dioica and Pleurotus ostreatus were able to exert antibacterial effects against Aeromonas hydrophila in rainbow trout (Oncorhynchus mykiss) [14]; Zingiber officinale Roscoe enhanced disease resistance of Lates calcarifer (Bloch) against Vibrio harveyi and enhanced the nonspecific immunity of Lates calcarifer (Bloch) [15]; the compound pentagalloylglucose extracted from Galla chinensis was highly resistant to Ichthyophthirius multifiliis (Ich) and significantly increased the survival of infected channel catfish (Ictalurus punctatus) [16]; Ophiopogon japonicus extract inhibited the proliferation of white spot syndrome virus (WSSV) in Chinese mitten crab (Eriocheir sinensis) and enhanced the immune response of Chinese mitten crab [17].
Viral disease is one of the biggest obstacles facing the aquaculture industry; viruses are highly contagious and fast-spreading, have a wide host range and high mortality rate, and are the most serious type of disease affecting aquaculture species [18]. At present, most viral diseases in aquaculture are not treatable by drugs and are usually preventative in nature. Most farmers can only prevent and control viral diseases via an improvement the aquaculture environment and disinfection, but they do not achieve good results [19]. It is a very urgent task to find more effective methods of virus diseases prevention and control, and herbal medicines have been confirmed by many studies in this regard. Therefore, this article summarizes several common aquatic viruses that are very destructive, collates the components of medicinal plants or natural compounds of plants that have antiviral effects against these viruses, and analyzes the current obstacles and problems that need to be solved in applying medicinal plants to aquaculture, aiming to provide new ideas for the prevention and treatment of viral diseases in aquatic animals.

2. Antiviral Effect of Medicinal Plants in Aquaculture

2.1. Medicinal Plants against RNA Viruses

2.1.1. Medicinal Plants against IPNV

Infectious pancreatic necrosis virus (IPNV) belongs to the family Dictyostelium, a genus of aquatic Dictyostelium RNA viruses with icosahedral symmetry, a diameter size of about 60 nm, no capsule membrane, high pathogenicity, and high infectivity [20]. The virus is extremely resistant to the external environment, stable to heat and acid, and insensitive to liposolvents [21]. IPNV mainly infects fry and juveniles up to 20 weeks of age, with a very high mortality rate [22]. Fish diseases caused by IPNV have been observed in Asia, America, and Europe, involving dozens of economic fish and shellfish species [23]. The most obvious pathological feature is pancreatic necrosis with abnormalities in the pancreatic vesicles, islets, and almost all cells, most of which are necrotic [24]. IPNV can be spread both horizontally and vertically; fry infected with IPNV can carry the virus for several years, be excreted in semen, eggs, and feces, and subsequently infect other fish [25]. However, neither the infection mechanism of IPNV nor the immune response of fish is well understood [26].
Species of the genus Heliotropiu were capable of inhibiting the replication of viruses such as Herpes simplex virus types 1 and 2, Junin, and Respiratory syncytial virus [27]. Accordingly, Modak et al. [28] evaluated the anti-IPNV effects of a group of natural compounds (geranyl aromatic derivatives) isolated from the resin exudates of Heliotropium filifolium (Heliotropiaceae) in vitro, including filifolinol, filifolinone, filifolinyl senecionate, and filifolinoic acid. Geranyl aromatic derivatives were added to the IPNV-infected CHSE-214 cell line at a series of different concentrations, and the viral plaque inhibition was tested. The results showed that the compound ester filifolinyl senecionate presented the best antiviral effect with an EC50 of 160 mg/mL and a cytotoxic concentration of up to 400 mg/mL that reduced cell viability by 50%. The experiment indicated that the ester filifolinyl senecionate is a good candidate for the prevention and treatment of IPNV infection in aquaculture.
Mycophenolic acid (MPA) is a non-nucleoside inhibitor of the cellular inosine monophosphate dehydrogenase (IMPDH), an enzyme that catalyzes an essential step in the biosynthesis of guanosine triphosphate (GTP) [29]. The MPA against IPNV and viral hemorrhagic septicemia virus (VHSV) was studied in cell culture [30]. Marroquí et al. evaluated the inhibitory effect of MPA using measurement methods such as RNA and protein synthesis determination, and they found that MPA inhibited the replication of both IPNV and VHSV in a dose-dependent manner. In particular, MPA was particularly effective for IPNV, with the inhibition of infectious virus production more than 105-fold. Additionally, MPA was effective in preventing viral protein synthesis. RT-qPCR results showed that MPA showed good efficacy in reducing the accumulation of viral RNA at low concentrations. The time-of-addition and washout experiments suggested that MPA might have a dual mechanism against virus infection that targets both cellular and viral functions. This study demonstrated that MPA could be used as a broad-spectrum antiviral agent for the treatment of aquatic animal diseases.
Infectious pancreatic necrosis (IPN) is a highly contagious acute viral disease that poses a great challenge to salmonid fish farming [31]. IPNV is strongly resistant and can survive for a long time in both fresh and marine water [32]. In the study of this virus, the international vaccine research is progressing well and has reached the molecular level; however, drug research is progressing slowly. There is no efficient drug, and the general research remains at the level of an anti-IPNV effect in cells outside the fish body, whereas the specific practical level in the fish body is inadequate. Moreover, broad-spectrum antiviral drugs have certain toxic effects on cells, and many of them have become prohibited drugs [33]. It is recommended to develop effective control drugs, such as traditional herbal medicines, from a pollution-free perspective.

2.1.2. Medicinal Plants against IHNV

Infectious hematopoietic necrosis virus (IHNV) belongs to the rhabdovirus genus Novirhabdovirus [34]. IHNV is a linear single-stranded RNA virus with a genome consisting of unsegmented negative-stranded RNA and a capsule membrane [35]. IHNV is a highly virulent fish bouncing virus that infects rainbow trout, causing acute, fatal systemic disease in fish fry, and renal hematopoietic tissue is the main target organ [36]. Additionally, IHNV is a pathogen that causes severe economic losses in salmonid trout farming in the United States, Canada, and many countries in Asia and Europe [37]. The virus is sensitive to temperature and an acidic environment. However, it is more tolerant in freshwater environments [38]. As with many viral diseases of finfish, younger life stages are more susceptible, with mortality rates of up to 95% during acute outbreaks [39]. Pathological signs in infected fish include protruding eyes, ocular hemorrhage, darkened skin, swollen abdomen, ulceration of the mouth and nose, and yellowish fluid in the intestinal viscera, histopathological examination reveals marked necrosis of the anterior kidney, liver, and spleen [40].
Carotenoids are outstanding antioxidants and have a very important role in the health of both humans and animals. It has been reported that carotenoids can improve the immune function of some aquatic animals [41]. Amar et al. [42] tested changes in resistance to IHNV in rainbow trout after carotenoid administration and found that the survival of rainbow trout did not change significantly when fish were immersed in high concentrations (2 × 104 TCID50·mL−1) of IHNV; however, when the virus dose was lower (2 × 103 TCID50·mL−1), the mortality was significantly reduced and the relative survival (RPS) was increased. This study suggested that carotenoids also have a role in enhancing disease resistance in fish.
A novel lentinan (LNT-I), consisting of glucose, mannose, and galactose, was extracted and purified from Lentinus edodes mycelia by Ren et al. [43]. LNT-I showed prominent antiviral activity against IHNV at a multiplicity of infectivity (MOI) of 0.05 and 0.10. At an MOI of 0.05, the direct inactivation of LNT-I against IHNV was 62.34% at 100 μg/mL. The antiviral mechanism of LNT-I is mainly involved in the direct inactivation of viral particles and inhibition of viral replication. In addition, LNT-I significantly downregulated the expression levels of tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2), and IL-11, whereas it upregulated the transcription of interferon-1 (IFN-1) and IFN-γ in epithelioma papulosum cyprini (EPC) cells infected with IHNV, suggesting that the inhibitory effect of LNT-I on IHNV infection might be attributed to its regulation of innate and specific immune responses.
Prunella vulgaris L. (PVL) has a broad range of biological activities including immune modulatory, antioxidant, antiviral, anti-inflammatory, antiallergic, and anticancer activities [44]. Li et al. [45] investigated the anti-IHNV activity of 32 medicinal plants in vitro and found that PVL exhibited the strongest antiviral effect with 99.3% inhibition of IHNV at a concentration of 100 mg/L. Further studies showed that ursolic acid (UA), a major component of PVL, also showed efficient anti-IHNV activity. The IC50 at 72 h of UA on IHNV was 8.0 μM. In addition, UA significantly reduced the cytopathic effect (CPE) and viral titer in EPC cells infected with IHNV. Moreover, the in vivo experiments also confirmed the strong anti-IHNV capability of UA. When infected with IHNV, the survival rate of the rainbow trout population injected intraperitoneally with UA increased, and viral gene expression was significantly suppressed.
Rhus verniciflua Stokes, a deciduous tree of the Anacardiaceae family, has long been used as a food supplement and a traditional herbal remedy for various ailments [46]. The methanolic extract of Rhus verniciflua Stokes bark exhibited significant anti-IHNV and VHSV ability and attenuated virus-induced CPE in vitro [47]. Further study of the active substances contained in the extract revealed that fisetin exhibited high antiviral activity against both IHNV and VHSV, as did fustin and sulfuretin. This test showed that Rhus verniciflua Stokes bark, as well as the isolated flavonoids, had significant antiviral effects [47]. It is also an excellent antiviral drug candidate.
Infectious hematopoietic necrosis (IHN) is also a viral disease that has dealt a fatal blow to the development of salmon and trout fisheries. To date, the IHNV DNA vaccine has been approved as a commercially available DNA vaccine; however, the lack of a convenient and effective immunization route for the vaccine, the paucity of research on the mechanisms that produce the protective effect of the vaccine, and its impact on the host immune system have limited the widespread use of the IHNV vaccine. In addition, there is no effective treatment for IHNV. According to current research, in addition to the above medicinal plants and natural active ingredients, some active ingredient derivatives are gradually being revealed scientists, such as arctigenin derivatives [48] and coumarin derivatives [49], which are also potential antiviral drugs with very good application prospects that deserve to be studied in depth.

2.1.3. Medicinal Plants against VHSV

VHSV is a member of the genus Novirhabdovirus, a family of elasmobranch viruses with a diameter of 65–80 nm, a capsule membrane, and a negative-sense single-stranded RNA genome [50]. The virus was mainly endemic in Europe and North America, but later spread to East Asia [51]. VHSV can be excreted by sick fish and viral feces, eggs, semen, etc. and transmitted through water [52]; it is very stable at low temperatures and can survive longer in freshwater than in seawater [53]. The host range of VHSV is very large, including salmonids, dogfish, cod, and plaice [54]. Fish infected with VHSV have rapid morbidity and mortality rates of 70% to 90% [55]. Infected fish show characteristics of darkening, protruding eyes, swollen abdomen, anemia, hemorrhage on the side of the body and at the base of the fins, hemorrhage, swelling, and discoloration of the liver, kidneys, and other organs, and punctate hemorrhage in the skeletal muscle. When a pathological histological examination was performed, necrosis of the urinary system and hematopoietic tissues such as the kidneys, some necrosis of the liver and spleen, and marked hemorrhage in the skeletal muscles were seen [56].
Celosia cristata and Raphanus sativus are herbs frequently used in traditional medicine. Park et al. [57] tested the antiviral activity of a mixed extract of Celosia cristata and Raphanus sativus against VHSV, and they found that, if the extract was added before the EPC cells were inoculated with the virus, it had a good effect in reducing the virus titer. However, the addition of this extract after the cells were inoculated with the virus had no effect. In an in vivo assay, the expression of several immune-related factors was examined in fish orally administered with the extract. The results showed that the expression of Mx and TLR7 peaked at 48 h after extract administration, while the expression of ISG15 and TLR2 peaked at 72 h, indicating that the antiviral activity of Celosia cristata and Raphanus sativus is achieved by inducing the expression of genes involved in the innate immune response.
Licorice (Glycyrrhiza uralensis, GUF) is a very common medicinal plant, and many important research projects have identified the many beneficial effects of this medicinal herb, including antimicrobial, antiviral, anti-inflammatory, antioxidant, antiprotozoal, hepatoprotective, and neuroprotective activities. Licorice contains important bioactive components, such as glycyrrhizin (GL), glycyrrhizinic acid (GLA), licorice acid, formononetin, and liquiritin [58]. Licorice extracts showed significant antiviral activity against VHSV, significantly reducing virus-induced CPE [59]. The active pharmaceutical ingredients GL and GLA also showed antiviral activity against VHSV. Moreover, a time-course study using a plaque reduction assay showed that GLA exhibited direct virucidal effects against VHSV or might inhibit viral invasion into cells.
The antiviral effects of curcumin have been extensively studied, and the mechanism of action mainly involves direct interference with the viral replication machinery, as well as inhibition of cellular signaling pathways essential for viral replication, such as phosphatidylinositol-3-kinase (PI3K)/Akt and nuclear factor kappa B (NF-κB) [60]. Curcumin also has an inhibitory effect on VHSV [61]. Pretreatment with 120 mM curcumin resulted in a significant increase in the viability of VHSV-infected cells, a decrease in viral copy number, and a decrease in apoptosis. A further study found that curcumin inhibited VHSV entry into cells by downregulating fibronectin (FN) 1 or upregulating F-actin. HSC71 is the primary protein interacting with FN1, gelsolin (GSN), and actins, which is a key target for protection from VHSV infection at the viral entry stage. Indeed, curcumin downregulated HSC71 expression while increasing the viability of cells infected with VHSV and inhibiting VHSV replication. The present experiments suggest that curcumin antiviral might be achieved by downregulating the expression of HSC71.
Olive (Olea europaea) (LExt) is the most famous member of the genus Olea, has a high nutritional value, and is commonly used by people; extracts from its leaves and fruits are widely used to counteract and prevent various pathologies [62]. It contains a significant number of bioactive compounds such as flavonoids, secoiridoids, carotenoids, and phenolic compounds. Olive tree leaf and its active component oleuropein (Ole) were able to inhibit VHSV infection in vitro, and incubation of the virus with LExt or Ole prior to infection reduced viral infectivity to 10% and 30%, respectively [63]. Furthermore, when LExt was added for 36 h after viral infection of cells, LExt sharply reduced VHSV titer and viral protein accumulation in a dose-dependent manner. On the other hand, both LExt and Ole were able to inhibit VHSV transmission between cells. This demonstrates that O. Europaea is a promising source of natural antiviral drugs.
α-Lipoic acid (LA) is a disulfide-containing compound that acts as a cofactor for several enzymes participating dehydrogenation and decarboxylation. Due to its excellent antioxidant capacity, LA has been used as an aquafeed additive to enhance detoxification and antioxidant capacity [64]. Furthermore, LA significantly increased the antiviral ability of fathead minnow cells against VHSV in a dose-dependent manner [65]. The dosing time assay showed that the antiviral activity of LA occurred during the viral replication phase. Injecting LA and VHSV into Micropterus salmoides also significantly enhanced survival compared to VHSV-only controls. In addition, LA injection significantly upregulated the expression of several immune-related genes, including IRF7, Viperin, and ISG15, as well as reduced VHSV-induced oxidative stress. (Figure 1).
VHSV has been found in most countries in the Northern Hemisphere, and VHSV is frequently endemic in aquaculture, causing morbidity and mortality in a wide range of fish species. Both VHSV and IHNV are members of the Novirhabdovirus family and are biologically similar in that they are both highly resistant to the external environment. After an epidemic, survivors are usually in a carrier state, and these carriers of the virus spread the pathogen throughout their lives via urine, feces, or gills. There is no commercially available vaccine for VHSV or effective prevention and control measures. In the study of this virus, more research has been conducted on diagnosis and detection, but less research has been invested in the study of prevention and control drugs; thus, we should pay more attention to the development of new efficient and nonpolluting drugs against VHSV.

2.1.4. Medicinal Plants against SVCV

Spring viremia of carp virus (SVCV) belongs to the family Rhabdoviridae, genus Vesiculovirus, and it is a negative-sense single-stranded RNA virus with a capsule membrane [66]. SVCV can infect koi (C. carpio koi), rainbow trout, sockeye salmon (O. nerka), blackhead minnow (Pimephales promelas), yellow perch (Perca flavescens) [67], and common carp (Cyprinus carpio carp), among them, common carp is the main host. SVCV is a serious pathogen of several economically important carp species, with a lethality rate of up to 100% [68]. SVCV was first identified in Europe and has since been found in Asia and South America [69]; it has spread widely throughout carp culture mainly in Europe and has caused significant economic losses [70]. The clinical symptoms of the diseased fish are mainly slow movement, neurological disorders, internal bleeding, abdominal inflammation, and fluid accumulation in the abdomen [71]. SVCV can be transmitted through the water by mucus-like feces excreted by diseased fish and also by blood-sucking insects, and it invades into fish via gills or other routes [72]; it can also replicate in fish epithelial cells [73]. SVCV is mainly transmitted horizontally, while it can also be transmitted vertically [74].
Beta-glucans (β-glucans) are glucose polymers found in yeast cell walls. There have been many literature studies on the expression of immune response genes in fish, where a β-glucan-enriched diet increases the transcript levels of some key genes of the innate and adaptive immune response [75]. β-Glucans have a therapeutic effect on zebrafish infected with SVCV via a mechanism that mainly regulates the release process of inflammatory factors [76]. In vitro tests revealed that β-glucose was able to regulate the location of IL-1 in cells; in vivo tests revealed that the expression of a group of genes involved in the innate immune response (IL-1β, IL-6, IL-8, IL-10, and TNF-α) were upregulated to inhibit the process of SVCV infection.
Tian et al. [77] purified the chemical element selenium from the fermentation of Herbaspirillum camelliae WT00C. Selenium is an essential trace element with antiviral properties. Selenium has various functions, such as antioxidant, anti-inflammatory, and immune boosting activities, in addition to reducing the incidence of cancer [78]. Tian and his subject group found that selenium significantly induced the expression of IFN, ISG15, and Mx, key factors in the immune response process. In in vivo experiments, IFN expression was increased 13- and 39-fold in Carassius carassius fed food containing 5 and 10 μg/g elemental selenium, respectively. On the 16th day post injection, the expression of IFN was selenium concentration-dependent within a certain range. The antiviral effects of selenium are mainly derived from its immunomodulatory effect through its incorporation into selenoproteins. The right amount of selenium helps to improve the immunity of fish, while excess selenium causes excessive immune and inflammatory responses.
Astragalus membranaceus is a traditional herb with a long history of clinical application. Astragalus polysaccharide (APS) is an important bioactive component of A. membranaceus and has a variety of pharmacological properties [79]. The antiviral effect of APS in zebrafish was investigated by Li et al. [80]. They fed zebrafish for 3 weeks on the control diet and experimental diets containing 0.01% or 0.02% APS. Zebrafish were challenged by SVCV at the end of a feeding. The antiviral immune responses were assessed 4 days after SVCV infection, and the survival rate was calculated 14 days after the challenge. The results showed that zebrafish fed 0.01% APS exhibited a higher survival rate post SVCV infection. Correspondingly, the expression of antiviral genes in the spleen was increased 4 days after the challenge. However, the addition of excessive amounts of APS was detrimental to liver health.
Librán-Pérez et al. [81] studied the effect of palmitic acid (PA) treatment on different immune parameters and mortality caused by SVCV in zebrafish. The results showed that PA had a positive effect on the immunomodulation of zebrafish via reducing mortality and viral titers. This FA-induced antiviral protective effect appears to be associated with the inhibition of autophagy and be independent of other immune processes such as neutrophil proliferation or IFN activity. PA is the most common saturated fatty acid and has long been negatively portrayed for its adverse health effects, but it actually has a variety of important physiological activities [82]. The usage of PA as an immunostimulant at low concentrations has shown great potential in the prevention of SVCV infection.
Psoralea corylifolia Linn. is a proverbial traditional medicinal plant that has been used since ancient times for the treatment of various diseases. It is widely distributed and is an important part of therapeutics in Ayurveda and Chinese medicine [83]. Psoralea corylifolia Linn. and one of its main components, bavachin (BVN), inhibited SVCV replication strongly [84]. BVN significantly inhibited both SVCV glycoprotein and nucleoprotein expression, but BVN did not affect the infectivity of SVCV and could not be used to prevent SVCV infection. Mechanistically speaking, BVN mainly inhibited the early events of SVCV replication but did not interfere with SVCV adsorption, and the inhibition of SVCV replication by BVN was achieved to some extent by blocking SVCV-induced apoptosis. This trial demonstrated that BVN could inhibit SVCV replication and block viral release to protect host cells, leading to therapeutic effects.
Saikosaponin D (SSD), a major bioactive triterpenoid saponin, an important component of Bupleurum yinchowense, significantly reduced SVCV-induced apoptosis and recovered SVCV-activated caspase-3/8/9 activity [85]. Moreover, SVCV-induced cell morphological damage and reactive oxygen species (ROS) generation were inhibited by SSD treatment. In vivo tests showed that SSD was also effective in the treatment of SVCV-infected zebrafish and carp. Intraperitoneal injection of SSD (6 mg/kg) increased the survival rate of zebrafish up to 36% and inhibited the expression of more than 90% of SVCV nucleoprotein and glycoprotein genes in the kidney and spleen, while the survival rate of carp treated with SSD (6 mg/kg) was also found to increase by 32%. In conclusion, SSD has been proven to be a natural product with very high anti-SVCV activity.
Spring viremia of carp (SVC) is one of the most damaging infectious diseases in the carp family, with diverse modes of transmission, long in vitro survival time, and high infective activity of virus particles expelled from infected fish. Although substantial research has been conducted worldwide on the control of SVC, the results have not been fruitful. There are no recognized drugs, vaccines, or other methods for effective control of the disease, and the internationally accepted prevention and control measures are to closely monitor SVC, detect diseased fish early, and isolate and cull them, so as to interrupt the trend of the disease outbreak and epidemic. Considering the sustainability of the entire carp culture industry, the prevention of the disease at its root also requires the use of resourceful and inexpensive herbal medicine containing a large number of effective natural components and various secondary metabolites.

2.1.5. Medicinal Plants against NNV

Nervous necrosis virus (NNV) is a widespread and serious infectious agent with a spherical shape, non-capsule membrane, diameter of approximately 30–40 nm, and a single positive-stranded, 2-segmented RNA molecule genome [86]. NNV belongs to the family of Nodaviridae, β genus Noda virus [87], which is divided into four genotypes (SJNNV, RGNNV, TPNNV, and BFNNV) [88]. The optimum temperature of NNV is 20–30 °C; NNV infections are usually observed in aquaculture farms when seawater temperatures are above 24 °C [89]. NNV usually infects fish fry and causes viral neuronecrosis, encephalomyelitis, and vacuolar retinopathy in fish after infection, resulting in massive fish mortality, which poses a devastating threat to aquaculture worldwide [90]. However, the pathogenic mechanism is still poorly understood [91]. Moreover, there is no effective vaccine to prevent the disease [92].
Gymnema sylvestre is mainly found in tropical and subtropical regions and has unique therapeutic properties for diabetes [93]. The gymnemagenol (C30H50O4) extracted from the leaves of Gymnema Sylvestre had antiviral potential [94]. In in vitro conditions, they used Sahul Indian Grouper Eye (SIGE) cell lines infected with grouper nervous necrosis virus (GNNV) to study the antiviral activity of gymnemagenol. The inhibition of GNNV by gymnemagenol was confirmed by measuring the viral titer (TCID50·mL−1) in the infected SIGE cells every 24 h. It was found that gymnemagenol exhibited a concentration-dependent inhibition on GNNV proliferation in virus-infected SIGE cells. At the end of the sixth day, 20 μg·mL−1 of gymnemagenol inhibited the proliferation of GNNV to 53%, while at the end of day 7, 20 μg·mL−1 of gymnemagenol reduced the survival rate of virus-infected SIGE cells to 47%. We conclude that gymnemagenol can be used as an antiviral agent against GNNV infection.
In addition, Asim et al. [95] studied the effect of glutamine on RGNNV replication. They found that a lack of glutamine did not affect cell viability but greatly inhibited RGNNV replication, revealing that glutamine is required for RGNNV replication. Glutamine can be converted to α-ketoglutarate (α-KG) by glutaminase (GLS) and then incorporated into the tricarboxylic acid (TCA) cycle. Inhibition of GLS activity with a GLS inhibitor significantly inhibits RGNNV replication while increasing the intermediates of the TCA cycle. The addition of α-KG, oxaloacetic acid (OAA), or pyruvate to glutamine-free medium significantly restored RGNNV replication, indicating that glutamine is required for RGNNV proliferation to complement the TCA cycle. These data suggest that glutamine could regulate RGNNV replication through the TCA cycle, which would pave a new pathway for the prevention of RGNNV infection in the future [95].
As mentioned earlier, β-glucan was able to increase the expression of some genes critical for immune response. Krishnan et al. [96] investigated the anti-NNV effect of β-glucan and found that β-glucan enhanced the viability of macrophages against NNV, which was associated with the activation of gene expression of persistent inflammatory cytokines. In the experiment, the expression of both NLR family CARD domain-containing 3 (NLRC3) and caspase-1 in the test group of seven-banded grouper was higher in the early stage and lower in the late stage than in the control group, suggesting that β-glucan may induce an antiviral state against NNV infection in grouper macrophages. In the present experiment, macrophages attenuated viral infectivity without cell death, as the lactate dehydrogenase (LDH) center remained stable in both the test and the control groups. In conclusion, it is known from the present experiment that β-glucan induces a protective response of the host against NNV infection.
Viral encephalopathy and retinopathy (VER) and viral nervous necrosis (VNN), caused by NNV, have no effective control measures, whether drugs or vaccines. Only a few herbal medicines have entered the anti-NNV research field in recent years, but there is still a lack of direct antiviral evidence, and the mechanism of action is still unclear; the lack of research in natural drug treatment has resulted in a serious shortage of natural drugs in the antiviral field. NNV has a very wide range of infection, including numerous marine invertebrates. The mortality rate of fish fry in farms infected with NNV is extremely high. Therefore, it is also extremely important to search for medicinal plants with high antiviral activity against NNV and to conduct systematic studies.

2.1.6. Medicinal Plants against GCRV

The grass carp reovirus (GCRV), a member of the Reoviridae family, Aquareovirus, is a double-stranded RNA virus with spherical particles, approximately 75 nm in diameter, which possesses a double non-enveloped capsid [97]. GCRV particles are insensitive to lipid solvents and are stable between pH 3 and 10. Moreover, these viruses are heat-stable and remain infectious after 30 min of passivation at 56 °C [98]. GCRV is the most pathogenic strain of the genus Echovirus [99] and can infect not only grass carp, but also cyprinid fish, whelk, and buccaneer [100]. The typical symptoms of GCRV infection are muscle and visceral hemorrhage, black body color, and unresponsiveness [101]. GCRV enters the fish mainly through the gills. Fish infected with GCRV produce a symptom reaction 4 days after infection. After 5–7 days, the fish show obvious hemorrhagic symptoms, and the fish population enters the peak of mortality [102]. Because of its high risk, wide prevalence, and long onset season, this virus has been the focus of research on fish viruses, but there is not yet an effective treatment [103].
Chen et al. [104] conducted in vitro experiments using Ctenopharyngodon idella kidney (CIK) cells to investigate the effects of 30 plant extracts on GCRV viral mRNA expression, and they found that Magnolia bark extract exhibited very strong inhibition of GCRV replication in vitro. Further studies showed that the main components of Magnolia bark, namely, magnolo and honokiol, were both effective in inhibiting GCRV replication in CIK cells at safe concentrations, but the mechanisms were not the same. In GCRV-infected cells, magnolol induced IFN-I activation by enhancing IRF7 expression, while honokiol enhanced the innate antiviral response of the host to GCRV infection via the NF-κB pathway [105]. They conducted a further study on magnolol and found that magnolol increased the resistance of grass carp to GCRV infection and inhibited GCRV-induced apoptosis; the inhibition of this apoptotic effect was probably due to the direct interaction of magnolol with caspase-3 [104].
Epigallocatechin-3-gallate (EGCG) is the most abundant and active compound in green tea. EGCG has significant antimicrobial, anti-inflammatory, antioxidant, anticancer, immunomodulatory, and neuroprotective effects, and has therapeutic potential for many diseases [106]. The adhesion of GCRV particles to the surface of CIK cells was inhibited by EGCG, as well as crude extracts of green tea, in a dose-dependent manner [107]. Through the virus overlay protein binding assay (VOPBA), they deduced that the blocking effect of EGCG on GCRV adhesion was due to EGCG serving as a ligand for the 37/67 kDa laminin receptor (LamR), which was related to the binding potential of GCRV particles to LamR [107]. Further studies showed that the addition of a certain dose of EGCG to fish feed could improve the survival rate of grass carp. This experiment also showed that even high doses of EGCG did not cause toxicity, and there was no significant change in cell survival at high concentrations. Grass carp injected with EGCG showed a decrease in malondialdehyde (MDA) concentration and an increase in glucuronide (GSH) and lysozyme (LZM) concentrations compared to the control group [108].
Zhang et al. [109] evaluated the effect of epicatechin-3-gallate (ECG) on GCRV in vitro, which differs from EGCG only in the absence of hydroxyl groups. ECG and EGCG have similar pharmacokinetic behavior, and EGCG is partially converted to ECG in vivo. GCRV was incubated with different concentrations of ECG in CIK cells and it was found that the typical CPE was mild in the range of 20–40 μg/mL ECG, while too high or too low concentrations caused harm to CIK cells. Western blot results showed that ECG efficiently inhibited the replication of GCRV in CIK cells. These articles suggested that both ECG and EGCG exhibit potential as antiviral agents in aquaculture.
Ginsenosides are the compounds responsible for the main pharmacological effects of Panax ginseng. Dai et al. [110] investigated the inhibitory effect of ginsenoside Rg3 on GCRV-infected grass carp ovarian (CO) epithelial cells. This experiment treated CO epithelial cells with 1, 10, and 100 μg/mL ginsenoside Rg3 and found that the highest inhibitory effect on GCRV was observed with 100 μg/mL ginsenoside Rg3 treatment. The antioxidant assay and RT-qPCR assay showed that ginsenoside Rg3 could effectively inhibit the replication of GCRV in CO epithelial cells. Expression analysis of immune-related genes showed that treatment with ginsenoside Rg3 promoted the expression of IRF-3 and IRF-7, induced the expression of IFN-1, and inhibited the expression of TNF-α. In conclusion, this experiment demonstrated that ginsenoside Rg3 enhanced the resistance of CO cells to GCRV infection by inhibiting GCRV activity and promoting the immune activity of CO cells.
Previous studies have shown that heat-shock factor 1 (HSF-1) is responsible for the transcriptional activation of heat-shock proteins in mammals in response to stress stimulation, and quercetin effectively inhibits HSF-1 transcription [111]. Shan et al. [112] found that heat-shock protein 70 (Hsp70), the major heat-shock protein during the heat-shock response, played an important role in facilitating the entry of GCRV into CIK cells and acted as a pro-viral factor during GCRV infection. These results suggested that quercetin (Qct) is likely to have anti-GCRV effects. Thus, Fu et al. [113] designed a relevant experiment and confirmed that quercetin could effectively improve the survival rate of the rare minnow Gobiocypris rarus after GCRV infection. Qct was injected at a dose of 30 µL/tail at concentrations of 1, 2, 4, 6, 8, and 16 mg/mL, and the mortality of rare minnow at different concentrations of Qct was observed and recorded over a period of 15 days; the results showed that quercetin dose-dependently protected rare minnow from GCRV challenge by reducing the production of progeny virus and improving fish survival. Hence, quercetin should be used as a promising environmentally friendly therapeutic agent for aquaculture.
Grass carp hemorrhagic disease (GCHD) is characterized by a wide range of prevalence, a long incidence season, a high incidence rate, and easy mutation of the pathogen. At present, although the research on vaccine control of GCHD has made great progress, the frequent mutation of GCRV and the bottleneck of vaccine commercialization for large-scale application have restricted the process of research and application of disease control, whereas the use of drugs for control is still the most direct and effective method for large-scale control of grass carp hemorrhagic disease. As a result, the development of antiviral drugs and related mechanisms of research are particularly important in the field of grass carp hemorrhagic disease control. The current research on the drug control of GCRV is still extremely backward, and the pharmacology of herbal drugs is not clear; thus, aquarists need to accelerate the pace of research.

2.2. Medicinal Plants against DNA Viruses

2.2.1. Medicinal Plants against SGIV

The grouper iridovirus (SGIV) is a novel marine fish DNA virus that belongs to the family Iridoviridae, genus Ranavirus [114]. SGIV is a class of plasma type linear double-stranded DNA viruses with a capsule membrane, virions in the form of regular icosahedrons, and a diameter of 154–176 nm. It is highly contagious; grouper fry are susceptible to this disease, with a large number of deaths in a short period of time after the disease [115]. SGIV is highly pathogenic, and high morbidity and mortality rates in aquatic animals have been caused by iridovirus infections worldwide in recent years [116]. The disease caused by SGIV results in significant economic losses to the aquaculture industry and poses a major threat to global biodiversity. Accordingly, SGIV has attracted the attention of virologists [117]. The main manifestation symptoms of iridovirus disease are fish swimming alone, white or bleeding spots on the gill filaments, swollen and white or earthy yellow liver, and enlarged liver and spleen [118]. An inactivated SGIV vaccine is currently available on the market, and the protective effect produced by the vaccine can be observed within 30 days of intraperitoneal injection to grouper [119]. However, inactivated whole virus vaccines have certain drawbacks, such as the risk of incomplete inactivation and the higher dose and cost of immunization in aquatic animals compared to terrestrial animals [120]. More importantly, vaccines cannot treat viral diseases; thus, it is imperative to develop new efficient and safe antiviral drugs to control the SGIV epidemic in aquaculture.
Illicium verum Hook. f. is rich in chemical constituents, and has antiviral, antibacterial, and antiphlogistic properties [121]. Liu et al. [122] investigated the application of I. verum extracts including trans-anethole (TAT), 3,4-dihydroxybenzoic acid (DDBA), and Qct in the treatment of grouper iridovirus infection, and the highest safe concentration of the extracts was selected for subsequent tests. The inhibitory activity of each I. verum extract against grouper iridovirus infection was analyzed using an aptamer (Q2)-based fluorescent molecular probe (Q2-AFMP) and the RT-qPCR method. All I. verum extracts showed dose-dependent antiviral activity against grouper iridovirus. IVAE, IVEE, DDBA, and Qct all showed maximum antiviral activity (all >90%) according to the percentage inhibition achieved.
Later, they further analyzed the possible antiviral mechanism of Qct [123,124]. Qct resulted in significant damage to SGIV particles. Furthermore, Qct could interfere with the binding of SGIV to host cell targets (76.14%), prevent SGIV invasion into host cells (56.03%), and affect SGIV replication in host cells (52.73%). The qPCR assay revealed that Qct induced the expression of genes for immune-related factors (IFN, STAT1, PKR, MxI, and ISG15) and TLR9. This suggested that quercetin exerted indirect antiviral activity against SGIV infection by promoting SGIV recognition and activating the IFN pathway to establish the antiviral state of host cells. Overall, Qct has direct and host-mediated antiviral effects against SGIV and has great potential for both prevention and treatment of SGIV infection.
In addition to the I. verum extracts, some active pharmaceutical ingredients of Curcuma kwangsiensis also exhibited strong anti-SGIV effects. C. kwangsiensis belongs to the family Zingiberaceae, which is used in medicine and food for the improvement and treatment of various diseases [125]. Liu et al. [126] tested C. kwangsiensis ethanol ingredient (CKEE), curcumin, curdione, curcumenol, and curcumol in vitro and in vivo. The results of the aptamer Q2-AFMP and RT-qPCR showed that all of them exhibited antiviral activity in a dose-dependent manner. CKEE and curdione showed the best results with inhibition rates higher than 93%, and they are excellent candidates for the development of effective drugs for the prevention and control of SGIV outbreaks in mariculture.
Liu et al. [127] found that Lonicera japonica Thunb. extracts also showed anti-SGIV activity, providing new ideas for the development of effective drugs for the prevention and control of SGIV infection in mariculture. They examined L. japonica aqueous extracts (LAE) and the active pharmaceutical ingredients chlorogenic acid (CGA), cryptochlorogenic acid (CCGA), isochlorogenic acid A (IAA), isochlorogenic acid B (IAB), isochlorogenic acid C (IAC), caffeic acid (CA), luteolin (LT), and inositol (IS) by light microscopy, Q3-AFMP, and RT-qPCR, revealing their anti-SGIV effects, with an inhibition rate above 90% except for IS.
Viola philippica is a perennial herb distributed throughout East Asia, containing anthocyanins, flavonoids, coumarins, alkaloids, phytosterol, sulfonated carbohydrate polymer, cyclotides, etc. [128]. The aqueous extracts of V. philippica also showed strong inhibition of SGIV infection both in vitro and in vivo [129]. RT-qPCR results showed that V. philippica did not damage SGIV particles, but it could interfere with SGIV binding, entry, and replication in host cells. V. philippica was found to have the best inhibitory effect on SGIV; it was very effective at the stage of virus binding and replication. In conclusion, the results indicate that the aqueous extract of V. philippica at appropriate concentrations has a strong anti-SGIV effect.
As previously mentioned, Glycyrrhiza uralensis (GUF) has a variety of beneficial effects, including but not limited to antiviral, antibacterial, and anti-inflammatory activities. Li et al. [130] investigated the antiviral effects of licorice against SGIV and found several major components of GUF, glycyrrhizin, glycyrrhetinic acid, liquiritin, isoliquiritigenin, and liquiritigenin. Although none of them exhibited significant anti-SGIV effects, the aqueous extract of GUF had significant anti-SGIV infection activity in a concentration-dependent manner. Moreover, the aqueous extract of licorice could disrupt the particle structure of SGIV. These results suggest that its antiviral effect may occur during the binding of viral particles to cellular receptors and during the replication phase of the virus in host cells, but the exact components of its action are not known and need to be further investigated.

2.2.2. Medicinal Plants against WSSV

WSSV is a rod-shaped double-stranded DNA virus containing a capsid membrane, approximately 250–300 nm × 75–100 nm in size, belonging to the family Nimaviridae, genus Whispovirus [131]. WSSV is highly infectious and lethal. It is reported that WSSV mainly infects crustacean decapods such as Penaeus monodon, P. vannamei, P. japonics, P. chinensis, P. merguiensis, P. penicillatius, and Scylla paramosain [132]. Under natural conditions, WSSV can be transmitted both horizontally through excreta, diseased shrimp remains, etc., by feeding and contacting with body parts, and vertically by infecting offspring individuals [133]. WSSV is very destructive; the cumulative mortality of infected shrimp can reach 100% within 3 to 10 days [134]. Shrimp infected with WSSV show symptoms such as lethargy, empty stomach, floating head, loss of balance, and white spots on the carapace, especially on the cephalothorax. In the final stage of the disease occur cell disintegration and organ necrosis, and eventual disease shrimp death [135]. To date, WSSV remains the number one killer of shrimp culture worldwide. At present, the occurrence of white spot syndrome is a multifactorial effect that is closely related to environmental conditions, culture density, etc. [136]. In contrast to the natural environment, shrimp culture in farms with high densities and unstable water conditions can easily form outbreaks once WSSV infection occurs, making it difficult to control [137].
EGCG has anti-WSSV activity in both Scylla paramosain [138] and Marsupeneaus japonicus [139], and the resistance of S. paramosain and M. japonicus to WSSV was significantly increased after EGCG treatment. Further studies revealed that moderate amounts of EGCG could effectively enhance innate immunity in shrimp and crab, and that EGCG had positive effects on several innate immune-related genes, including the immune deficiency pathway (IMD), prophenoloxidase (proPO), QM, myosin, Rho, Rab7, p53, TNF-α, mitogen-activated protein kinase (MAPK), and nitric oxide synthase (NOS). EGCG may induce certain immune pathways in vivo, such as the Janus kinase (JAK)–signal transducers and activators of transcription (STAT) pathway. EGCG also enhances phenol oxidase and superoxide dismutase activities and is an excellent anti-WSSV drug candidate [138,139].
Sun et al. [140] compared the inhibitory activity of 13 herbs against WSSV and found that the extract of Typha angustifolia showed a very strong inhibition of WSSV replication (84.62%, 100 mg/kg). Further studies showed that its main active ingredient, naringenin (NAR), a citrus flavonoid that possesses various biological activities, exhibited higher inhibition of WSSV (92.85%, 50 mg/kg). NAR inhibited the transcription of key genes during viral replication, especially the immediate-early gene ie1. NAR blocked the transcription of ie1 by reducing the expression of STAT gene and regulated several anti-inflammatory-, antioxidant-, and proapoptotic-related factors, such as cyclooxygenase (COX)-1/2, catalase (CAT), and Bax, to inhibit WSSV replication. These results suggest that NAR could be a candidate for the prevention and treatment of WSSV [140].
Hesperidin, an important constituent of citrus fruits, has a wide range of pharmacological effects, including antiviral, anti-inflammatory, antioxidant, antitumor, and radioprotective activities [141]. The addition of 50–150 mg·kg−1 hesperidin (the major compound of Pericarpium Citri Reticulatae) to diets fed to Procambarus clarkii not only reduced the percentage of mortality following WSSV infection in P. clarkii, but also improved their nonspecific immunity, antioxidant capacity, and growth performance [142]. Compared with the control group, the crayfish group supplemented with 50–150 mg·kg−1 hesperidin showed increased final body weight (FBW), specific growth rate (SGR), and weight gain (WG), as well as significantly increased total antioxidant capacity (T-AOC), glutathione peroxidase (GPx) activity, and superoxide dismutase (SOD) activity. Moreover, the activities of acid phosphatase (ACP), alkaline phosphatase (AKP), LZM, and phenoloxidase (PO) were increased in crayfish, and the mRNA expression of extracellular copper-zinc superoxide dismutase (ecCuZnSOD), Hsp70, cyclophilin A (CypA), GPxs, astacidin, crustin, and Toll 3 was upregulated.
Huang et al. [143] evaluated the anti-WSSV activity of 23 medicinal plant extracts and found that Gardenia jasminoides extracts showed the highest inhibition of WSSV replication (92.31%). After further study, they found that G. jasminoides reduced the transcription of the immediate-early gene ie1, DNA pol, and the late gene VP28 of WSSV, while inhibiting the expression of Hsp70. In addition, after injecting G. jasminoides into P. clarkii, the expression of antioxidant- and apoptosis-related factors, such as cMnSOD, mMnSOD, and Bax, increased significantly, indicating that G. jasminoides not only has antiviral but also antioxidant activity, in addition to regulating apoptosis-related factors. Next, they extracted the active compound genipin (GN) from the fruits of G. jasminoides and investigated the anti-WSSV effect of GN; they found that GN decreased the expression of STAT gene, thus reducing the transcription of the ie1 gene. In addition, GN upregulated the expression of antioxidant-related genes and downregulated the expression of inflammation-related genes, thereby reducing oxidative stress and inflammation caused by viral infection.
Huang et al. [144] also found that the extract of Eucommia ulmoides showed very high inhibition of WSSV replication (84.12%); its main active ingredient, geniposidic acid (GPA), exerted a very strong antiviral effect (over 97%). Moreover, when WSSV was reinfected with P. clarkii after treatment with GPA, the mortality rate of P. clarkii was reduced by 43.33%. GPA is an iridoid glucoside, a major component of several herbs that has long been used to treat inflammation, jaundice, and hepatic disorders [145]. Mechanistically, GPA reduces the transcription of ie1 gene by decreasing the expression of STAT, thus inhibiting the replication of WSSV. In addition, GPA also inhibited the expression of the apoptosis-related factor Bax inhibitor-1, which also indirectly inhibited WSSV replication. Pursuantly, GPA can also be considered as a potential drug for the prevention and treatment of WSSV infection.
There are many more candidates like this, such as Pongamia pinnata [146], Ulva intestinalis [147], and Gracilaria tenuistipitata [148]. WSSV is one of the most thoroughly studied aquatic viruses to date, as the major disaster it has brought to shrimp and crabs is one of the problems that fisheries and biologists still have to face.
WSSV is one of the most serious pathogens in shrimp aquaculture, causing catastrophic economic losses to the global shrimp industry and seriously hindering the sustainable development of the marine economy. Many scholars have conducted substantial research on the epidemiological characteristics, etiology, pathology, pathogenesis, and rapid detection techniques of WSSV, and considerable progress has been made. However, due to the habits of shrimp themselves and the complexity of WSSV transmission, the mechanism of WSSV transmission is not yet fully understood, resulting in the slow development of its prevention and control; furthermore, the existing technology has not been able to fully and effectively control the WSSV epidemic. Shrimp lack a vertebrate-like adaptive immune system, but an effective innate immune system may render protection against invading pathogens [149]. An effective way to improve the resistance of shrimp to pathogenic invasion is by enhancing nonspecific immunity. Medicinal plants and their natural chemical components are very effective in this regard, activating the body’s immune system and enhancing the resistance of shrimp to disease. Thus, the use of medicinal plants against WSSV is an excellent strategy.
Table 1 summarizes the medicinal plants and active pharmaceutical ingredients that can inhibit the abovementioned viruses, as well as their effects or possible antiviral mechanisms. It should be noted that the antiviral mechanisms of some of these medicinal plants and active pharmaceutical ingredients have not been confirmed, but are merely inferences made by researchers on the basis of past results.

3. Challenges and Perspectives

The rise of aquaculture is considered to be one of the most profound changes in global food production in a century [178] and one of the most environmentally friendly and sustainable food industries to meet the needs of humans today [179]. However, almost all kinds of farmed aquatic animals are threatened by infections from viruses, bacteria, parasites, or other nascent and regenerating pathogenic microorganisms [180]. Epidemics have become a constraint to the sustainable development of the aquaculture industry, especially viral diseases, which are highly contagious, spread rapidly, and have a wide range of hosts with high mortality rates [181]. The occurrence of aquatic animal viral diseases not only leads to a decline in the quality of aquatic products and food safety hazards, but also seriously hinders the sustainable development of fisheries.
Medicinal plants and active pharmaceutical ingredients have unique advantages in terms of antiviral activity. On the premise of being able to effectively inhibit viral infection, medicinal plants and active pharmaceutical ingredients possess the advantages of low drug resistance, fewer toxic side-effects, fewer drug residues, and less pollution of the farmed water environment [182]. Consequently, the search for new antiviral drugs with high efficiency and low toxicity from natural plants is an important way to develop antiviral drugs. At present, the development and the research of safe and efficient new herbal medicine products are getting more and more attention. An investigation found that healthy aquaculture has certain advantages and great potential for the future [183]. The antivirus mechanisms of medicinal plants are diverse, such as direct inactivation of viral particles [184], interdiction of viral attachment and penetration phases [185], inhibition of virus replication [186], involvement in transcriptional regulation [187], disruption of virus protein synthesis or expression [188], inhibition of viral cell-to-cell transmission [189], and immunomodulatory roles [190]. The same medicinal plants may exert different mechanisms of action against different viruses, and different medicinal plants exert even more different effects, which makes the utilization of medicinal plants promising.
Medicinal plants contain numerous active ingredients such as phenolic substances, flavonoids, alkaloids, terpenoids, pigments, starch, steroids, and essential oils [191]. In aquaculture, medicinal plants and active ingredients can be used as growth promoters [192], immunostimulants [193], antibacterial agents [194], antifungal agents [195], antistress agents [196], appetite stimulants [197], and even aphrodisiacs [198], in addition to being antiviral agents. In many cases, medicinal plants exert antiviral effects by acting as immunostimulants, with antiviral activity being only an added effect. At this point, although researchers have used particular viruses to confirm whether a medicinal plant exerts an antiviral effect in fish or shellfish, it is likely that the resistance effect is not limited to that virus. When host nonspecific immunity is increased, it is usually elevated against a wide range of pathogens. Preventing diseases by improving the immunity of fish and shellfish represents a new direction in the development of pollution-free aquaculture, which is important for disease control, health safety, and environmental protection in aquaculture.
Although the advantages and efficacy of herbal medicine in aquaculture have been proven in production practice and have great potential for development in the future, herbal medicine in aquaculture is still in its infancy and has many shortcomings at this stage, mainly manifested in the following aspects: (1) there is no unified standard for medication, because most herbal medicines are not measured by ingredient content, but rely on practical effectiveness accumulated over the years, which is difficult to quantify; (2) most products are crude products. The ingredients are limited by multiple influences such as geography, climate, and time, and the effect is always unstable; (3) the therapeutic effect is not rapid, and the effect on the treatment of acute infectious diseases is not obvious; (4) combined application is not effective. The ratio tests are not enough, and the interactions between medicinal plants of different genera are not clear. Thus, it is difficult to achieve a scientific ratio; (5) commercial production is faced with difficulties, and the technical process of isolation and mass extraction of active pharmaceutical ingredients in medicinal plants is not perfect. Therefore, it is difficult to control low-cost production.
Therefore, future research on the application of medicinal plants in aquaculture should mainly focus on combining medical theory with modern technology, establishing sound techniques for cultivation, extraction, and refinement of medicinal plants, and achieving standardized and commercialized production of new medicinal plant products through improved processing techniques. Moreover, it is necessary to strengthen the research on the specific mechanisms of the role of medicinal plants in aquaculture. It is not enough to focus only on the effect of action, because there are many uncertainties in practical application, and if the specific mechanism of action is not clear, then only the rigid use of medicinal plants is likely to be counterproductive in the end. In conclusion, the effects of medicinal plants in aquatic animals is an exciting topic in aquaculture.

4. Conclusions

Medicinal plants have great potential in terms of antivirals; whereas most of the current studies on the antiviral effects of medicinal plants focused on the inhibitory effects of plants on viruses, relatively few established a clear understanding of the antiviral mechanisms of the active pharmaceutical ingredients. In addition, few studies on the synergistic and antagonistic effects of different medicinal plants have been reported. Future research work should focus on the basic research of medicinal plants to clarify their active pharmaceutical ingredients, as well as their pharmacological and toxicological effects; accordingly, they can then be better applied in the prevention and treatment of aquatic animal diseases

Author Contributions

W.L. and L.H. conceived and wrote this review together, and W.L. made the final revisions to the article. S.H. (Shuyu Han); D.H.; Y.X.; M.L.; Q.Y.; S.H. (Shuaishuai Huang) all provided valuable suggestions to this review and participated in the proofreading. D.W.; P.L. guided the authors in the writing of this paper and provided funding support for this paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (Pengfei Li, 41966004), the Natural Science Foundation of Guangxi (Mingzhu Liu, 2020GXNSFBA297161, 2022GXNSFBA035521), Guangxi Innovation Team Project of National Modern Agricultural Industrial Technology System (Pengfei Li, nycytxgxcxtd-2021-08-02), and the Basic Research Fund of Guangxi Academy of Sciences (Mingzhu Liu, 2019YJJ1005).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Acknowledgments

This work was supported by grants from the National Natural Science Foundation of China (41966004; U20A20102), the Natural Science Foundation of Guangxi (2020GXNSFBA297161; 2022GXNSFBA035521), the Guangxi Innovation Team Project of the National Modern Agricultural Industrial Technology System (nycytxgxcxtd-2021-08-02), and the Basic Research Fund of Guangxi Academy of Sciences (2019YJJ1005).

Conflicts of Interest

The authors of this study declare no conflict of interest.

Abbreviations

IchIchthyophthirius multifiliis
WSSVWhite spot syndrome virus
IPNVInfectious pancreatic necrosis virus
MPAMycophenolic acid
IMPDHInosine monophosphate dehydrogenase
GTPGuanosine triphosphate
VHSVViral hemorrhagic septicemia virus
IPNInfectious hematopoietic necrosis virus
IHNVInvasive pneumococcal disease
RPSRelative survival
LNTLentinan
TNFTumor necrosis factor
ILInterleukin
IFNsInterferon
MOIMultiplicity of infectivity
EPCEpithelioma papulosum cyprini
PVLPrunella vulgaris L.
UAUrsolic acid
CPECytopathic effect
IHNInfectious hematopoietic necrosis
GUFGlycyrrhiza uralensis
GLGlycyrrhizin
GLAGlycyrrhizinic acid
PI3KPhosphatidylinositol-3-kinase
NF-κBNuclear factor kappa B
FNFibronectin
GSNGelsolin
LAα-Lipoic acid
SVCVSpring viremia of carp virus
APSAstragalus polysaccharide
PAPalmitic acid
IFNType I interferon
BVNBavachin
SSDSaikosaponin D
ROSReactive oxygen species
SVCSpring viremia of carp
NNVNervous necrosis virus
α-KGα-Ketoglutarate
GLSGlutaminase
TCATricarboxylic acid
OAAOxaloacetic acid
NLRC3NLR family CARD domain-containing 3
LDHLactate dehydrogenase
VERViral encephalopathy and retinopathy
VNNViral nervous necrosis
GCRVGrass carp reovirus
CIKCtenopharyngodon idella kidney
EGCGEpigallocatechin-3-gallate
VOPBAVirus overlay protein binding assay
LamRLaminin receptor
MDAMalondialdehyde
GSHGlucuronide
LZMLysozyme
ECGEpicatechin-3-gallate
COCarp ovarian
HSFHeat-shock factor
HspHeat-shock protein
QctQuercetin
GCHDGrass carp haemorrhagic disease
SGIVGrouper iridovirus
TATtrans-Anethole
DDBA3,4-Dihydroxybenzoic acid
Q2-AFMP(Q2)-based fluorescent molecular probe
CKEECurcuma kwangsiensis ethanol ingredient
LAELonicera japonica aqueous extract
CGAChlorogenic acid
CCGACryptochlorogenic acid
IAAIsochlorogenic acid A
IABIsochlorogenic acid B
IACIsochlorogenic acid C
CACaffeic acid
LTLuteolin
ISInositol
GUFGlycyrrhiza uralensis
IMDImmune deficiency pathway
proPOProphenoloxidase
MAPKMitogen-activated protein kinase
NOSNitric oxide synthase
JAKJanus kinase
STATSignal transducers and activators of transcription
NARNaringenin
COXCyclooxygenase
CATCatalase
FBWFinal body weight
SGRSpecific growth rate
WGWeight gain
T-AOCTotal antioxidant capacity
GPxGlutathione peroxidase
SODSuperoxide dismutase
ACPActivities of acid phosphatase
AKPAlkaline phosphatase
POPhenoloxidase
CypACyclophilin A
GNGenipin
GPAGeniposidic acid
LUTLuteolin
CPChicory polysaccharides
GMLGlycerol monolaurate
THCTotal hemocyte count
ARPsAnoectochilus roxburghii polysaccharides
AMPsAntimicrobial peptides

References

  1. Sudhakaran, G.; Guru, A.; Haridevamuthu, B.; Murugan, R.; Arshad, A.; Arockiaraj, J. Molecular Properties of Postbiotics and Their Role in Controlling Aquaculture Diseases. Aquac. Res. 2022, 53, 3257–3273. [Google Scholar] [CrossRef]
  2. Ekmekcioglu, C.; Wallner, P.; Kundi, M.; Weisz, U.; Haas, W.; Hutter, H.-P. Red Meat, Diseases, and Healthy Alternatives: A Critical Review. Crit. Rev. Food Sci. Nutr. 2018, 58, 247–261. [Google Scholar] [CrossRef] [PubMed]
  3. Fiorella, K.J.; Okronipa, H.; Baker, K.; Heilpern, S. Contemporary Aquaculture: Implications for Human Nutrition. Curr. Opin. Biotechnol. 2021, 70, 83–90. [Google Scholar] [CrossRef]
  4. Brugere, C.; Onuigbo, D.M.; Morgan, K.L. People Matter in Animal Disease Surveillance: Challenges and Opportunities for the Aquaculture Sector. Aquaculture 2017, 467, 158–169. [Google Scholar] [CrossRef] [Green Version]
  5. Sargenti, M.; Bartolacci, S.; Luciani, A.; Di Biagio, K.; Baldini, M.; Galarini, R.; Giusepponi, D.; Capuccella, M. Investigation of the Correlation between the Use of Antibiotics in Aquaculture Systems and Their Detection in Aquatic Environments: A Case Study of the Nera River Aquafarms in Italy. Sustainability 2020, 12, 5176. [Google Scholar] [CrossRef]
  6. Watts, J.; Schreier, H.; Lanska, L.; Hale, M. The Rising Tide of Antimicrobial Resistance in Aquaculture: Sources, Sinks and Solutions. Mar. Drugs 2017, 15, 158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Samuelsen, O.B.; Lunestad, B.T.; Jelmert, A. Pharmacokinetic and Efficacy Studies on Bath-Administering Potentiated Sulphonamides in Atlantic Halibut, Hippoglossus hippoglossus L. J. Fish Dis. 1997, 20, 287–296. [Google Scholar] [CrossRef]
  8. Topp, E.; Larsson, D.G.J.; Miller, D.N.; Van den Eede, C.; Virta, M.P.J. Antimicrobial Resistance and the Environment: Assessment of Advances, Gaps and Recommendations for Agriculture, Aquaculture and Pharmaceutical Manufacturing. FEMS Microbiol. Ecol. 2018, 94, 3. [Google Scholar] [CrossRef]
  9. Done, H.Y.; Halden, R.U. Reconnaissance of 47 Antibiotics and Associated Microbial Risks in Seafood Sold in the United States. J. Hazard. Mater. 2015, 282, 10–17. [Google Scholar] [CrossRef] [Green Version]
  10. Limbu, S.M.; Chen, L.-Q.; Zhang, M.-L.; Du, Z.-Y. A Global Analysis on the Systemic Effects of Antibiotics in Cultured Fish and Their Potential Human Health Risk: A Review. Rev. Aquac. 2021, 13, 1015–1059. [Google Scholar] [CrossRef]
  11. Liu, X.; Lv, Y.; Xu, K.; Xiao, X.; Xi, B.; Lu, S. Response of Ginger Growth to a Tetracycline-Contaminated Environment and Residues of Antibiotic and Antibiotic Resistance Genes. Chemosphere 2018, 201, 137–143. [Google Scholar] [CrossRef] [PubMed]
  12. Griboff, J.; Carrizo, J.C.; Bonansea, R.I.; Valdés, M.E.; Wunderlin, D.A.; Amé, M.V. Multiantibiotic Residues in Commercial Fish from Argentina. The Presence of Mixtures of Antibiotics in Edible Fish, a Challenge to Health Risk Assessment. Food Chem. 2020, 332, 127380. [Google Scholar] [CrossRef] [PubMed]
  13. Nik Mohamad Nek Rahimi, N.; Natrah, I.; Loh, J.-Y.; Ervin Ranzil, F.K.; Gina, M.; Lim, S.-H.E.; Lai, K.-S.; Chong, C.-M. Phytocompounds as an Alternative Antimicrobial Approach in Aquaculture. Antibiotics 2022, 11, 469. [Google Scholar] [CrossRef] [PubMed]
  14. Bilen, S.; Ünal, S.; Güvensoy, H. Effects of Oyster Mushroom (Pleurotus Ostreatus) and Nettle (Urtica Dioica) Methanolic Extracts on Immune Responses and Resistance to Aeromonas hydrophila in Rainbow Trout (Oncorhynchus Mykiss). Aquaculture 2016, 454, 90–94. [Google Scholar] [CrossRef]
  15. Talpur, A.D.; Ikhwanuddin, M.; Ambok Bolong, A.-M. Nutritional Effects of Ginger (Zingiber Officinale Roscoe) on Immune Response of Asian Sea Bass, Lates calcarifer (Bloch) and Disease Resistance against Vibrio Harveyi. Aquaculture 2013, 400–401, 46–52. [Google Scholar] [CrossRef]
  16. Zhang, Q.; Xu, D.-H.; Klesius, P.H. Evaluation of an Antiparasitic Compound Extracted from Galla Chinensis against Fish Parasite Ichthyophthirius Multifiliis. Vet. Parasitol. 2013, 198, 45–53. [Google Scholar] [CrossRef]
  17. Chen, C.; Shen, J.-L.; Wang, T.; Yang, B.; Liang, C.-S.; Jiang, H.-F.; Wang, G.-X. Ophiopogon japonicus Inhibits White Spot Syndrome Virus Proliferation in Vivo and Enhances Immune Response in Chinese Mitten Crab Eriocheir Sinensis. Fish Shellfish Immunol. 2021, 119, 432–441. [Google Scholar] [CrossRef]
  18. Oidtmann, B.; Dixon, P.; Way, K.; Joiner, C.; Bayley, A.E. Risk of Waterborne Virus Spread—Review of Survival of Relevant Fish and Crustacean Viruses in the Aquatic Environment and Implications for Control Measures. Rev. Aquac. 2018, 10, 641–669. [Google Scholar] [CrossRef] [Green Version]
  19. Kibenge, F.S. Emerging Viruses in Aquaculture. Curr. Opin. Virol. 2019, 34, 97–103. [Google Scholar] [CrossRef]
  20. Dobos, P.; Hill, B.J.; Hallett, R.; Kells, D.T.; Becht, H.; Teninges, D. Biophysical and Biochemical Characterization of Five Animal Viruses with Bisegmented Double-Stranded RNA Genomes. J. Virol. 1979, 32, 593–605. [Google Scholar] [CrossRef] [Green Version]
  21. Dobos, P. The Molecular Biology of Infectious Pancreatic Necrosis Virus (IPNV). Annu. Rev. Fish Dis. 1995, 5, 25–54. [Google Scholar] [CrossRef]
  22. Ahne, W. Isolation and Characterization of Infectious Pancreatic Necrosis Virus from Pike (Esox Lucius). Arch. Virol. 1978, 58, 65–69. [Google Scholar] [CrossRef] [PubMed]
  23. Lyoo, Y.S.; Chang, C.H.; Jeon, Y.H.; Lee, J.O.; Rhee, J.C. Isolation of Infectious Pancreatic Necrosis Virus from Rainbow Trout in Korea. Korean J. Vet. Res. 1991, 31, 195–199. [Google Scholar]
  24. Ahmadivand, S.; Weidmann, M.; El-Matbouli, M.; Rahmati-Holasoo, H. Low Pathogenic Strain of Infectious Pancreatic Necrosis Virus (IPNV) Associated with Recent Outbreaks in Iranian Trout Farms. Pathogens 2020, 9, 782. [Google Scholar] [CrossRef] [PubMed]
  25. Rønneseth, A.; Haugland, G.T.; Wergeland, H.I. Flow Cytometry Detection of Infectious Pancreatic Necrosis Virus (IPNV) within Subpopulations of Atlantic Salmon (Salmo salar L.) Leucocytes after Vaccination and during the Time Course of Experimental Infection. Fish Shellfish Immunol. 2013, 34, 1294–1305. [Google Scholar] [CrossRef]
  26. Kamil, A.; Raae, A.; Fjelldal, P.G.; Koppang, E.O.; Fladmark, K.E.; Hordvik, I. Comparative Analysis of IgM Sub-Variants in Salmonid Fish and Identification of a Residue in Μ3 Which Is Essential for MAb4C10 Reactivity. Fish Shellfish Immunol. 2013, 34, 667–672. [Google Scholar] [CrossRef]
  27. Modak, B.; Galeno, H.; Torres, R. Antiviral Activity on Hantavirus and Apoptosis of Vero Cells of Natural and Semi-Synthetic Compounds from Heliotropium Filifolium Resin. J. Chil. Chem. Soc. 2004, 49, 143–145. [Google Scholar] [CrossRef]
  28. Modak, B.; Sandino, A.M.; Arata, L.; Cárdenas-Jirón, G.; Torres, R. Inhibitory Effect of Aromatic Geranyl Derivatives Isolated from Heliotropium Filifolium on Infectious Pancreatic Necrosis Virus Replication. Vet. Microbiol. 2010, 141, 53–58. [Google Scholar] [CrossRef]
  29. Sintchak, M.D.; Fleming, M.A.; Futer, O.; Raybuck, S.A.; Chambers, S.P.; Caron, P.R.; Murcko, M.A.; Wilson, K.P. Structure and Mechanism of Inosine Monophosphate Dehydrogenase in Complex with the Immunosuppressant Mycophenolic Acid. Cell 1996, 85, 921–930. [Google Scholar] [CrossRef] [Green Version]
  30. Marroquí, L.; Estepa, A.; Perez, L. Inhibitory Effect of Mycophenolic Acid on the Replication of Infectious Pancreatic Necrosis Virus and Viral Hemorrhagic Septicemia Virus. Antivir. Res. 2008, 80, 332–338. [Google Scholar] [CrossRef]
  31. Min, L.; Li-Li, Z.; Jun-Wei, G.; Xin-Yuan, Q.; Yi-Jing, L.; Di-Qiu, L. Immunogenicity of Lactobacillus-Expressing VP2 and VP3 of the Infectious Pancreatic Necrosis Virus (IPNV) in Rainbow Trout. Fish Shellfish Immunol. 2012, 32, 196–203. [Google Scholar] [CrossRef] [PubMed]
  32. Dopazo, C.P. The Infectious Pancreatic Necrosis Virus (IPNV) and Its Virulence Determinants: What Is Known and What Should Be Known. Pathogens 2020, 9, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Zhang, M.; Zhong, J.; Xiong, Y.; Song, X.; Li, C.; He, Z. Development of Broad-Spectrum Antiviral Agents—Inspiration from Immunomodulatory Natural Products. Viruses 2021, 13, 1257. [Google Scholar] [CrossRef] [PubMed]
  34. Morzunov, S.P.; Winton, J.R.; Nichol, S.T. The Complete Genome Structure and Phylogenetic Relationship of Infectious Hematopoietic Necrosis Virus. Virus Res. 1995, 38, 175–192. [Google Scholar] [CrossRef]
  35. Schutze, H.; Enzmann, P.-J.; Kuchling, R.; Mundt, E.; Niemann, H.; Mettenleiter, T.C. Complete Genomic Sequence of the Fish Rhabdovirus Infectious Haematopoietic Necrosis Virus. J. Gen. Virol. 1995, 76, 2519–2527. [Google Scholar] [CrossRef]
  36. Amend, D.F.; Yasutake, W.T.; Mead, R.W. A Hematopoietic Virus Disease of Rainbow Trout and Sockeye Salmon. Trans. Am. Fish. Soc. 1969, 98, 796–804. [Google Scholar] [CrossRef]
  37. Crane, M.; Hyatt, A. Viruses of Fish: An Overview of Significant Pathogens. Viruses 2011, 3, 2025–2046. [Google Scholar] [CrossRef]
  38. Whipple, M.J.; Rohovec, J.S. The Effect of Heat and Low PH on Selected Viral and Bacterial Fish Pathogens. Aquaculture 1994, 123, 179–189. [Google Scholar] [CrossRef]
  39. Lapatra, S.E. Factors Affecting Pathogenicity of Infectious Hematopoietic Necrosis Virus (IHNV) for Salmonid Fish. J. Aquat. Anim. Health 1998, 10, 121–131. [Google Scholar] [CrossRef]
  40. Ahmadivand, S.; Soltani, M.; Mardani, K.; Shokrpoor, S.; Hassanzadeh, R.; Ahmadpoor, M.; Rahmati-Holasoo, H.; Meshkini, S. Infectious Hematopoietic Necrosis Virus (IHNV) Outbreak in Farmed Rainbow Trout in Iran: Viral Isolation, Pathological Findings, Molecular Confirmation, and Genetic Analysis. Virus Res. 2017, 229, 17–23. [Google Scholar] [CrossRef]
  41. Amar, E.C.; Kiron, V.; Satoh, S.; Watanabe, T. Enhancement of Innate Immunity in Rainbow Trout (Oncorhynchus mykiss Walbaum) Associated with Dietary Intake of Carotenoids from Natural Products. Fish Shellfish Immunol. 2004, 16, 527–537. [Google Scholar] [CrossRef] [PubMed]
  42. Amar, E.C.; Kiron, V.; Akutsu, T.; Satoh, S.; Watanabe, T. Resistance of Rainbow Trout Oncorhynchus mykiss to Infectious Hematopoietic Necrosis Virus (IHNV) Experimental Infection Following Ingestion of Natural and Synthetic Carotenoids. Aquaculture 2012, 330–333, 148–155. [Google Scholar] [CrossRef]
  43. Ren, G.; Xu, L.; Lu, T.; Yin, J. Structural Characterization and Antiviral Activity of Lentinan from Lentinus Edodes Mycelia against Infectious Hematopoietic Necrosis Virus. Int. J. Biol. Macromol. 2018, 115, 1202–1210. [Google Scholar] [CrossRef] [PubMed]
  44. Roh, K.-B.; Park, D.; Jung, E. Inhibitory Effects of Prunella vulgaris L. Extract on 11 β -HSD1 in Human Skin Cells. Evid.-Based Complementary Altern. Med. 2018, 2018, 1–8. [Google Scholar] [CrossRef] [Green Version]
  45. Li, B.-Y.; Hu, Y.; Li, J.; Shi, K.; Shen, Y.-F.; Zhu, B.; Wang, G.-X. Ursolic Acid from Prunella vulgaris L. Efficiently Inhibits IHNV Infection in Vitro and in Vivo. Virus Res. 2019, 273, 197741. [Google Scholar] [CrossRef]
  46. Kim, M.S.; Lee, C.W.; Kim, J.-H.; Lee, J.-C.; An, W.G. Extract of Rhus Verniciflua Stokes Induces P53-Mediated Apoptosis in MCF-7 Breast Cancer Cells. Evid.-Based Complementary Altern. Med. 2019, 2019, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Kang, S.Y.; Kang, J.-Y.; Oh, M.-J. Antiviral Activities of Flavonoids Isolated from the Bark of Rhus verniciflua Stokes against Fish Pathogenic Viruses In Vitro. J. Microbiol. 2012, 50, 293–300. [Google Scholar] [CrossRef]
  48. Shan, L.; Wang, H.; Hu, Y. A Preliminary Investigation on the Mechanism of Action of 4-(8-(2-Ethylimidazole)Octyloxy)-Arctigenin against IHNV. Virus Res. 2021, 294, 198287. [Google Scholar] [CrossRef]
  49. Wang, H.; Hu, Y.; Song, D.; Shan, L.; Liu, L. Synthesis and Application of a Potential Therapeutic Coumarin Derivative against IHNV in Aquaculture. Aquaculture 2021, 543, 736999. [Google Scholar] [CrossRef]
  50. Ammayappan, A.; Vakharia, V.N. Molecular Characterization of the Great Lakes Viral Hemorrhagic Septicemia Virus (VHSV) Isolate from USA. Virol. J. 2009, 6, 171. [Google Scholar] [CrossRef] [Green Version]
  51. Isshiki, T.; Nishizawa, T.; Kobayashi, T.; Nagano, T.; Miyazaki, T. An Outbreak of VHSV (Viral Hemorrhagic Septicemia Virus) Infection in Farmed Japanese Flounder Paralichthys Olivaceus in Japan. Dis. Aquat. Org. 2001, 47, 87–99. [Google Scholar] [CrossRef] [PubMed]
  52. López-Vázquez, C.; Dopazo, C.P.; Barja, J.L.; Bandín, I. Experimental Infection of Turbot, Psetta Maxima (L.), with Strains of Viral Haemorrhagic Septicaemia Virus Isolated from Wild and Farmed Marine Fish. J. Fish Dis. 2007, 30, 303–312. [Google Scholar] [CrossRef] [PubMed]
  53. Hawley, L.; Garver, K. Stability of Viral Hemorrhagic Septicemia Virus (VHSV) in Freshwater and Seawater at Various Temperatures. Dis. Aquat. Org. 2008, 82, 171–178. [Google Scholar] [CrossRef]
  54. Cabon, J.; Almeras, F.; Baud, M.; Pallandre, L.; Morin, T.; Louboutin, L. Susceptibility of Pike Esox Lucius to VHSV and IHNV and Potential Transmission to Rainbow Trout Oncorhynchus Mykiss. Dis. Aquat. Org. 2020, 139, 175–187. [Google Scholar] [CrossRef]
  55. Brudeseth, B.; Evensen, Ø. Occurrence of Viral Haemorrhagic Septicaemia Virus (VHSV) in Wild Marine Fish Species in the Coastal Regions of Norway. Dis. Aquat. Org. 2002, 52, 21–28. [Google Scholar] [CrossRef] [Green Version]
  56. Ahmadivand, S.; Soltani, M.; Mardani, K.; Shokrpoor, S.; Rahmati-Holasoo, H.; Mokhtari, A.; Hasanzadeh, R. Isolation and Identification of Viral Hemorrhagic Septicemia Virus (VHSV) from Farmed Rainbow Trout (Oncorhynchus Mykiss) in Iran. Acta Trop. 2016, 156, 30–36. [Google Scholar] [CrossRef]
  57. Park, Y.J.; Moon, C.; Kang, J.-H.; Choi, T.-J. Antiviral Effects of Extracts from Celosia cristata and Raphanus sativus Roots against Viral Hemorrhagic Septicemia Virus. Arch Virol 2017, 162, 1711–1716. [Google Scholar] [CrossRef] [PubMed]
  58. Pastorino, G.; Cornara, L.; Soares, S.; Rodrigues, F.; Oliveira, M.B.P.P. Liquorice (Glycyrrhiza Glabra): A Phytochemical and Pharmacological Review: Liquorice (Glycyrrhiza Glabra): A Review. Phytother. Res. 2018, 32, 2323–2339. [Google Scholar] [CrossRef]
  59. Lim, J.; Seo, J.; Jung, S.; Kang, S.Y. Efficacy of an Optimized Extract from Licorice Roots (Glycyrrhiza Uralensis Fischer) against Viral Hemorrhagic Septicemia Virus in Olive Flounder (Paralichthys Olivaceus). Aquac. Res. 2021, 52, 2609–2621. [Google Scholar] [CrossRef]
  60. Mathew, D.; Hsu, W.-L. Antiviral Potential of Curcumin. J. Funct. Foods 2018, 40, 692–699. [Google Scholar] [CrossRef]
  61. Jeong, E.-H.; Vaidya, B.; Cho, S.-Y.; Park, M.-A.; Kaewintajuk, K.; Kim, S.R.; Oh, M.-J.; Choi, J.-S.; Kwon, J.; Kim, D. Identification of Regulators of the Early Stage of Viral Hemorrhagic Septicemia Virus Infection during Curcumin Treatment. Fish Shellfish Immunol. 2015, 45, 184–193. [Google Scholar] [CrossRef] [PubMed]
  62. Alesci, A.; Miller, A.; Tardugno, R.; Pergolizzi, S. Chemical Analysis, Biological and Therapeutic Activities of Olea Europaea L. Extracts. Nat. Prod. Res. 2021, 36, 2932–2945. [Google Scholar] [CrossRef] [PubMed]
  63. Micol, V.; Caturla, N.; Pérez-Fons, L.; Más, V.; Pérez, L.; Estepa, A. The Olive Leaf Extract Exhibits Antiviral Activity against Viral Haemorrhagic Septicaemia Rhabdovirus (VHSV). Antivir. Res. 2005, 66, 129–136. [Google Scholar] [CrossRef]
  64. Monserrat, J.M.; Lima, J.V.; Ferreira, J.L.R.; Acosta, D.; Garcia, M.L.; Ramos, P.B.; Moraes, T.B.; dos Santos, L.C.; Amado, L.L. Modulation of Antioxidant and Detoxification Responses Mediated by Lipoic Acid in the Fish Corydoras Paleatus (Callychthyidae). Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2008, 148, 287–292. [Google Scholar] [CrossRef]
  65. Zhang, W.; Chen, X.; Yu, F.; Li, F.; Li, W.; Yi, M.; Jia, K. α-Lipoic Acid Exerts Its Antiviral Effect against Viral Hemorrhagic Septicemia Virus (VHSV) by Promoting Upregulation of Antiviral Genes and Suppressing VHSV-Induced Oxidative Stress. Virol. Sin. 2021, 36, 1520–1531. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, Z.-X.; Liu, S.-B.; Guan, H.; Lu, L.-F.; Tu, J.-G.; Ouyang, S.; Zhang, Y.-A. Structural and Functional Characterization of the Phosphoprotein Central Domain of Spring Viremia of Carp Virus. J. Virol. 2020, 94, e0085-20. [Google Scholar] [CrossRef] [PubMed]
  67. Emmenegger, E.J.; Sanders, G.E.; Conway, C.M.; Binkowski, F.P.; Winton, J.R.; Kurath, G. Experimental Infection of Six North American Fish Species with the North Carolina Strain of Spring Viremia of Carp Virus. Aquaculture 2016, 450, 273–282. [Google Scholar] [CrossRef] [Green Version]
  68. Shao, L.; Zhao, J. Isolation of a Highly Pathogenic Spring Viraemia of Carp Virus Strain from Grass Carp (Ctenopharyngodon Idella) in Late Summer, China, 2016. Virus Res. 2017, 238, 183–192. [Google Scholar] [CrossRef]
  69. Padhi, A.; Verghese, B. Detecting Positively Selected Codons in the Glycoprotein of Spring Viraemia of Carp Virus (SVCV) Isolates from the USA and China. J. Fish Dis. 2008, 31, 785–791. [Google Scholar] [CrossRef]
  70. Ahne, W.; Bjorklund, H.V.; Essbauer, S.; Fijan, N.; Kurath, G.; Winton, J.R. Spring Viremia of Carp (SVC). Dis. Aquat. Org. 2002, 52, 261–272. [Google Scholar] [CrossRef]
  71. Ghasemi, M.; Zamani, H.; Hosseini, S.M.; Haghighi Karsidani, S.; Bergmann, S.M. Caspian White Fish (Rutilus Frisii Kutum) as a Host for Spring Viraemia of Carp Virus. Vet. Microbiol. 2014, 170, 408–413. [Google Scholar] [CrossRef] [PubMed]
  72. Ahne, W. Uptake and Multiplication of Spring Viraemia of Carp Virus in Carp, Cyprinus carpio L. J. Fish Dis. 1978, 1, 265–268. [Google Scholar] [CrossRef]
  73. Ahne, W. Argulus foliaceus L. and Piscicola geometra L. as Mechanical Vectors of Spring Viraemia of Carp Virus (SVCV). J. Fish Dis. 2010, 8, 241–242. [Google Scholar] [CrossRef]
  74. Fijan, N. Spring Viraemia of Carp and Other Viral Diseases and Agents of Warm-Water Fish; CAB International: Wallingford, UK, 1999; Volume 3, pp. 177–244. [Google Scholar]
  75. Douxfils, J.; Fierro-Castro, C.; Mandiki, S.N.M.; Emile, W.; Tort, L.; Kestemont, P. Dietary β-Glucans Differentially Modulate Immune and Stress-Related Gene Expression in Lymphoid Organs from Healthy and Aeromonas hydrophila-infected Rainbow Trout (Oncorhynchus Mykiss). Fish Shellfish Immunol. 2017, 63, 285–296. [Google Scholar] [CrossRef] [PubMed]
  76. Medina-Gali, R.M.; del Mar Ortega-Villaizan, M.; Mercado, L.; Novoa, B.; Coll, J.; Perez, L. Beta-Glucan Enhances the Response to SVCV Infection in Zebrafish. Dev. Comp. Immunol. 2018, 84, 307–314. [Google Scholar] [CrossRef] [Green Version]
  77. Tian, J.; Zhang, Y.; Zhu, R.; Wu, Y.; Liu, X.; Wang, X. Red Elemental Selenium (Se0) Improves the Immunoactivities of EPC Cells, Crucian Carp and Zebrafish against Spring Viremia of Carp Virus (SVCV). J. Fish Biol. 2021, 98, 208–218. [Google Scholar] [CrossRef] [PubMed]
  78. Jablonska, E.; Li, Q.; Reszka, E.; Wieczorek, E.; Tarhonska, K.; Wang, T. Therapeutic Potential of Selenium and Selenium Compounds in Cervical Cancer. Cancer Control 2021, 28, 107327482110018. [Google Scholar] [CrossRef]
  79. Wang, J.; Jia, J.; Song, L.; Gong, X.; Xu, J.; Yang, M.; Li, M. Extraction, Structure, and Pharmacological Activities of Astragalus Polysaccharides. Appl. Sci. 2018, 9, 122. [Google Scholar] [CrossRef] [Green Version]
  80. Li, Y.; Ran, C.; Wei, K.; Xie, Y.; Xie, M.; Zhou, W.; Yang, Y.; Zhang, Z.; Lv, H.; Ma, X.; et al. The Effect of Astragalus Polysaccharide on Growth, Gut and Liver Health, and Anti-Viral Immunity of Zebrafish. Aquaculture 2021, 540, 736677. [Google Scholar] [CrossRef]
  81. Librán-Pérez, M.; Pereiro, P.; Figueras, A.; Novoa, B. Antiviral Activity of Palmitic Acid via Autophagic Flux Inhibition in Zebrafish (Danio Rerio). Fish Shellfish Immunol. 2019, 95, 595–605. [Google Scholar] [CrossRef]
  82. Carta, G.; Murru, E.; Banni, S.; Manca, C. Palmitic Acid: Physiological Role, Metabolism and Nutritional Implications. Front. Physiol. 2017, 8, 902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Alam, F.; Khan, G.N.; Asad, M.H.H.B. Psoralea corylifolia L: Ethnobotanical, Biological, and Chemical Aspects: A Review. Phytother. Res. 2018, 32, 597–615. [Google Scholar] [CrossRef] [PubMed]
  84. Cheng, C.; Yu-Feng, S.; Yang, H.; Lei, L.; Wei-Chao, C.; Gao-Xue, W.; Bin, Z. Highly Efficient Inhibition of Spring Viraemia of Carp Virus Replication in Vitro Mediated by Bavachin, a Major Constituent of Psoralea Corlifonia Lynn. Virus Res. 2018, 255, 24–35. [Google Scholar] [CrossRef] [PubMed]
  85. Shen, Y.-F.; Hu, Y.; Zhang, Z.; Liu, L.; Chen, C.; Tu, X.; Wang, G.-X.; Zhu, B. Saikosaponin D Efficiently Inhibits SVCV Infection in Vitro and in Vivo. Aquaculture 2019, 504, 281–290. [Google Scholar] [CrossRef]
  86. Mori, K.-I.; Nakai, T.; Muroga, K.; Arimoto, M.; Mushiake, K.; Furusawa, I. Properties of a New Virus Belonging to Nodaviridae Found in Larval Striped Jack (Pseudocaranx Dentex) with Nervous Necrosis. Virology 1992, 187, 368–371. [Google Scholar] [CrossRef]
  87. Barsøe, S.; Allal, F.; Vergnet, A.; Vandeputte, M.; Olesen, N.J.; Schmidt, J.G.; Larsen, C.A.; Cuenca, A.; Vendramin, N. Different Survival of Three Populations of European Sea Bass (Dicentrarchus Labrax) Following Challenge with Two Variants of Nervous Necrosis Virus (NNV). Aquac. Rep. 2021, 19, 100621. [Google Scholar] [CrossRef]
  88. Tanaka, S.; Kuriyama, I.; Nakai, T.; Miyazaki, T. Susceptibility of Cultured Juveniles of Several Marine Fish to the Sevenband Grouper Nervous Necrosis Virus. J. Fish Dis. 2003, 26, 109–115. [Google Scholar] [CrossRef]
  89. Krishnan, R.; Jang, Y.-S.; Kim, J.-O.; Oh, M.-J. Altered Expression of Immune Factors in Sevenband Grouper, Hyporthodus Septemfasciatus Following Nervous Necrosis Virus Challenge at Optimal and Suboptimal Temperatures. Fish Shellfish Immunol. 2021, 119, 442–451. [Google Scholar] [CrossRef]
  90. Zhu, S.; Miao, B.; Zhang, Y.-Z.; Zeng, W.-W.; Wang, D.-S.; Su, S.-Q. In Vitro Neutralization of Nervous Necrosis Virus by a Nanobody Binding to the Protrusion Domain of Capsid Protein. Aquaculture 2022, 548, 737654. [Google Scholar] [CrossRef]
  91. Zhang, W.; Jia, K.; Jia, P.; Xiang, Y.; Lu, X.; Liu, W.; Yi, M. Marine Medaka Heat Shock Protein 90ab1 Is a Receptor for Red-Spotted Grouper Nervous Necrosis Virus and Promotes Virus Internalization through Clathrin-Mediated Endocytosis. PLoS Pathog. 2020, 16, e1008668. [Google Scholar] [CrossRef]
  92. Yi, L.; Qin, Z.; Lin, H.; Zhou, Y.; Li, J.; Xu, Z.; Babu, V.S.; Lin, L. Features of Chicken Egg Yolk Immunoglobulin (IgY) against the Infection of Red-Spotted Grouper Nervous Necrosis Virus. Fish Shellfish Immunol. 2018, 80, 534–539. [Google Scholar] [CrossRef] [PubMed]
  93. Tiwari, P.; Ahmad, K.; Hassan Baig, M. Gymnema Sylvestre for Diabetes: From Traditional Herb to Future’s Therapeutic. Curr. Pharm. Des. 2017, 23, 1667–1676. [Google Scholar] [CrossRef] [PubMed]
  94. Gopiesh Khanna, V.; Kannabiran, K.; Sarath Babu, V.; Sahul Hameed, A.S. Inhibition of Fish Nodavirus by Gymnemagenol Extracted from Gymnema Sylvestre. J. Ocean Univ. China 2011, 10, 402–408. [Google Scholar] [CrossRef]
  95. Asim, M.; Jiang, S.; Yi, L.; Chen, W.; Sun, L.; Zhao, L.; Khan Khattak, M.N.; Tu, J.; Lin, L. Glutamine Is Required for Red-Spotted Grouper Nervous Necrosis Virus Replication via Replenishing the Tricarboxylic Acid Cycle. Virus Res. 2017, 227, 245–248. [Google Scholar] [CrossRef]
  96. Krishnan, R.; Jang, Y.-S.; Oh, M.-J. Beta Glucan Induced Immune Priming Protects against Nervous Necrosis Virus Infection in Sevenband Grouper. Fish Shellfish Immunol. 2022, 121, 163–171. [Google Scholar] [CrossRef]
  97. Qiu, T.; Lu, R.; Zhang, J.; Zhu, Z. Complete Nucleotide Sequence of the S10 Genome Segment of Grass Carp Reovirus (GCRV). Dis. Aquat. Org. 2001, 44, 69–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Winton, J.R.; Lannan, C.N.; Fryer, J.L.; Hedrick, R.P.; Meyers, T.R.; Plumb, J.A.; Yamamoto, T. Morphological and Biochemical Properties of Four Members of a Novel Group of Reoviruses Isolated from Aquatic Animals. J. Gen. Virol. 1987, 68, 353–364. [Google Scholar] [CrossRef] [PubMed]
  99. Rangel, A.A.C.; Rockemann, D.D.; Hetrick, F.M.; Samal, S.K. Identification of Grass Carp Haemorrhage Virus as a New Genogroup of Aquareovirus. J. Gen. Virol. 1999, 80, 2399–2402. [Google Scholar] [CrossRef] [Green Version]
  100. Wang, B.; Zhou, M.; Lin, Y.; Ma, Y.; Cao, H. TBK1 Regulates the Induction of Innate Immune Response against GCRV by Phosphorylating IRF3 in Rare Minnow (Gobiocypris Rarus). Dev. Comp. Immunol. 2021, 115, 103883. [Google Scholar] [CrossRef]
  101. Tang, Y.; Zeng, W.; Wang, Y.; Wang, Q.; Yin, J.; Li, Y.; Wang, C.; Bergmann, S.M.; Gao, C.; Hu, H. Comparison of the Blood Parameters and Histopathology between Grass Carp Infected with a Virulent and Avirulent Isolates of Genotype II Grass Carp Reovirus. Microb. Pathog. 2020, 139, 103859. [Google Scholar] [CrossRef]
  102. Chen, G.; Xiong, L.; Wang, Y.; He, L.; Huang, R.; Liao, L.; Zhu, Z.; Wang, Y. ITGB1b-Deficient Rare Minnows Delay Grass Carp Reovirus (GCRV) Entry and Attenuate GCRV-Triggered Apoptosis. Int. J. Mol. Sci. 2018, 19, 3175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chu, P.; Zhu, Y.; Zhuang, M.; He, L.; Zhang, X. Autophagy Signaling Pathway Is a Therapeutic Target to Inhibit GCRV Replication. Aquaculture 2022, 548, 737657. [Google Scholar] [CrossRef]
  104. Chen, X.; Hao, K.; Yu, X.; Huang, A.; Zhu, B.; Wang, G.; Ling, F. Magnolol Protects Ctenopharyngodon Idella Kidney Cells from Apoptosis Induced by Grass Carp Reovirus. Fish Shellfish Immunol. 2018, 74, 426–435. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, X.; Hu, Y.; Shan, L.; Yu, X.; Hao, K.; Wang, G. Magnolol and Honokiol from Magnolia Officinalis Enhanced Antiviral Immune Responses against Grass Carp Reovirus in Ctenopharyngodon Idella Kidney Cells. Fish Shellfish Immunol. 2017, 63, 245–254. [Google Scholar] [CrossRef]
  106. Chakrawarti, L.; Agrawal, R.; Dang, S.; Gupta, S.; Gabrani, R. Therapeutic Effects of EGCG: A Patent Review. Expert Opin. Ther. Pat. 2016, 26, 907–916. [Google Scholar] [CrossRef]
  107. Wang, H.; Liu, W.; Yu, F.; Lu, L. Identification of (-)-Epigallocatechin-3-Gallate as a Potential Agent for Blocking Infection by Grass Carp Reovirus. Arch Virol 2016, 161, 1053–1059. [Google Scholar] [CrossRef]
  108. Wang, H.; Chen, Y.; Ru, G.; Xu, Y.; Lu, L. EGCG: Potential Application as a Protective Agent against Grass Carp Reovirus in Aquaculture. J. Fish Dis. 2018, 41, 1259–1267. [Google Scholar] [CrossRef]
  109. Zhang, Y.; Wang, H.; Su, M.; Lu, L. (-)-Epicatechin Gallate, a Metabolite of (-)-epigallocatechin Gallate in Grass Carp, Exhibits Antiviral Activity in Vitro against Grass Carp Reovirus. Aquac. Res. 2020, 51, 1673–1680. [Google Scholar] [CrossRef]
  110. Dai, J.; Zhang, L.; Zhang, P.; Shu, H.; Mao, A.; Li, Y. Ginsenoside Rg3 Inhibits Grass Carp Reovirus Replication in Grass Carp Ovarian Epithelial Cells. Microb. Pathog. 2020, 144, 104174. [Google Scholar] [CrossRef]
  111. Sliutz, G.; Karlseder, J.; Tempfer, C.; Orel, L.; Holzer, G.; Simon, M. Drug Resistance against Gemcitabine and Topotecan Mediated by Constitutive Hsp70 Overexpression in Vitro: Implication of Quercetin as Sensitiser in Chemotherapy. Br. J. Cancer 1996, 74, 172–177. [Google Scholar] [CrossRef] [Green Version]
  112. Shan, L.-P.; Chen, X.-H.; Ling, F.; Zhu, B.; Wang, G.-X. Targeting Heat Shock Protein 70 as an Antiviral Strategy against Grass Carp Reovirus Infection. Virus Res. 2018, 247, 1–9. [Google Scholar] [CrossRef] [PubMed]
  113. Fu, J.; Xu, N.; Sun, H.; Xuan, J.; Lu, L. Quercetin Protects Rare Minnow Gobiocypris Rarus from Infection of Genotype II Grass Carp Reovirus. Aquac. Res. 2021, 52, 4867–4873. [Google Scholar] [CrossRef]
  114. Wang, S.; Huang, X.; Huang, Y.; Hao, X.; Xu, H.; Cai, M.; Wang, H.; Qin, Q. Entry of a Novel Marine DNA Virus, Singapore Grouper Iridovirus, into Host Cells Occurs via Clathrin-Mediated Endocytosis and Macropinocytosis in a PH-Dependent Manner. J. Virol. 2014, 88, 13047–13063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Qin, Q.; Chang, S.; Ngoh-Lim, G.; Gibson-Kueh, S.; Shi, C.; Lam, T. Characterization of a Novel Ranavirus Isolated from Grouper Epinephelus Tauvina. Dis. Aquat. Org. 2003, 53, 1–9. [Google Scholar] [CrossRef]
  116. Liu, M.; Xiao, H.; Wu, S.; Yu, Q.; Li, P. Aptamer-based High-throughput Screening Model for Medicinal Plant Drugs against SGIV. J. Fish Dis. 2020, 43, 1479–1482. [Google Scholar] [CrossRef]
  117. Chinchar, V.G. Ranaviruses (Family Iridoviridae): Emerging Cold-Blooded Killers. Arch. Virol. 2002, 147, 447–470. [Google Scholar] [CrossRef]
  118. Wei, S.; Wang, S.; Yang, M.; Huang, Y.; Wei, J.; Huang, X.; Qin, Q. Characterization of Cathepsin C from Orange-Spotted Grouper, Epinephelus Coioides Involved in SGIV Infection. Fish Shellfish Immunol. 2019, 84, 423–433. [Google Scholar] [CrossRef]
  119. Yu, Q.; Liu, M.; Wei, S.; Qin, X.; Qin, Q.; Li, P. Research Progress and Prospects for the Use of Aptamers in Aquaculture Biosecurity. Aquaculture 2021, 534, 736257. [Google Scholar] [CrossRef]
  120. Xiao, H.; Liu, M.; Li, S.; Shi, D.; Zhu, D.; Ke, K.; Xu, Y.; Dong, D.; Zhu, L.; Yu, Q.; et al. Isolation and Characterization of a Ranavirus Associated with Disease Outbreaks in Cultured Hybrid Grouper (♀ Tiger Grouper Epinephelus Fuscoguttatus × ♂ Giant Grouper E. Lanceolatus) in Guangxi, China. J. Aquat. Anim. Health 2019, 31, 364–370. [Google Scholar] [CrossRef]
  121. Zhou, B.-G.; Wang, S.; Dou, T.-T.; Liu, S.; Li, M.-Y.; Hua, R.-M.; Li, S.-G.; Lin, H.-F. Aphicidal Activity of Illicium Verum Fruit Extracts and Their Effects on the Acetylcholinesterase and Glutathione S-Transferases Activities in Myzus Persicae (Hemiptera: Aphididae). J. Insect Sci. 2016, 16, 11. [Google Scholar] [CrossRef] [Green Version]
  122. Liu, M.; Yu, Q.; Xiao, H.; Yi, Y.; Cheng, H.; Putra, D.F.; Huang, Y.; Zhang, Q.; Li, P. Antiviral Activity of Illicium Verum Hook. f. Extracts against Grouper Iridovirus Infection. J. Fish Dis. 2020, 43, 531–540. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, M.; Yu, Q.; Xiao, H.; Li, M.; Huang, Y.; Zhang, Q.; Li, P. The Inhibitory Activities and Antiviral Mechanism of Medicinal Plant Ingredient Quercetin Against Grouper Iridovirus Infection. Front. Microbiol. 2020, 11, 586331. [Google Scholar] [CrossRef]
  124. Huang, L.; Li, M.; Wei, H.; Yu, Q.; Huang, S.; Wang, T.; Liu, M.; Li, P. Research on the Indirect Antiviral Function of Medicinal Plant Ingredient Quercetin against Grouper Iridovirus Infection. Fish Shellfish Immunol. 2022, 124, 372–379. [Google Scholar] [CrossRef] [PubMed]
  125. Dong, C.-X.; Zhang, L.-J.; Xu, R.; Zhang, G.; Zhou, Y.-B.; Han, X.-Q.; Zhang, Y.; Sun, Y.-X. Structural Characterization and Immunostimulating Activity of a Levan-Type Fructan from Curcuma Kwangsiensis. Int. J. Biol. Macromol. 2015, 77, 99–104. [Google Scholar] [CrossRef] [PubMed]
  126. Liu, M.; Xiao, H.; Zhang, Q.; Wu, S.; Putra, D.F.; Xiong, X.; Xu, M.; Dong, L.; Li, S.; Yu, Q.; et al. Antiviral Abilities of Curcuma Kwangsiensis Ingredients against Grouper Iridoviral Infection in Vitro and in Vivo. Aquac. Res. 2020, 51, 351–361. [Google Scholar] [CrossRef]
  127. Liu, M.; Yu, Q.; Yi, Y.; Xiao, H.; Putra, D.F.; Ke, K.; Zhang, Q.; Li, P. Antiviral Activities of Lonicera Japonica Thunb. Components against Grouper Iridovirus in Vitro and in Vivo. Aquaculture 2020, 519, 734882. [Google Scholar] [CrossRef]
  128. Wang, Y.-L.; Zhang, L.; Li, M.-Y.; Wang, L.-W.; Ma, C.-M. Lignans, Flavonoids and Coumarins from Viola Philippica and Their α-Glucosidase and HCV Protease Inhibitory Activities. Nat. Prod. Res. 2019, 33, 1550–1555. [Google Scholar] [CrossRef]
  129. Yu, Q.; Liu, M.; Xiao, H.; Wu, S.; Qin, X.; Lu, Z.; Shi, D.; Li, S.; Mi, H.; Wang, Y.; et al. The Inhibitory Activities and Antiviral Mechanism of Viola Philippica Aqueous Extracts against Grouper Iridovirus Infection in Vitro and in Vivo. J. Fish Dis. 2019, 42, 859–868. [Google Scholar] [CrossRef]
  130. Li, M.; Liu, M.; Wei, H.; Huang, L.; Yu, Q.; Huang, S.; Li, J.; Li, P. Antiviral Activities of Glycyrrhiza Uralensis Components against Singapore Grouper Iridovirus. J. World Aquac. Soc. 2022, 1–16. [Google Scholar] [CrossRef]
  131. Rajendran, K.V.; Vijayan, K.K.; Santiago, T.C.; Krol, R.M. Experimental Host Range and Histopathology of White Spot Syndrome Virus (WSSV) Infection in Shrimp, Prawns, Crabs and Lobsters from India. J. Fish Dis. 2010, 22, 183–191. [Google Scholar] [CrossRef]
  132. Sahul Hameed, A.S.; Yoganandhan, K.; Sathish, S.; Rasheed, M.; Murugan, V.; Jayaraman, K. White Spot Syndrome Virus (WSSV) in Two Species of Freshwater Crabs (Paratelphusa hydrodomous and P. pulvinata). Aquaculture 2001, 201, 179–186. [Google Scholar] [CrossRef]
  133. Leu, J.H.; Yang, F.; Zhang, X.; Xu, X.; Lo, C.F. Whispovirus. Curr. Top. Microbiol. Immunol. 2009, 328, 197–227. [Google Scholar] [CrossRef] [PubMed]
  134. van Hulten, M.C.W.; Witteveldt, J.; Peters, S.; Kloosterboer, N.; Tarchini, R.; Fiers, M.; Sandbrink, H.; Lankhorst, R.K.; Vlak, J.M. The White Spot Syndrome Virus DNA Genome Sequence. Virology 2001, 286, 7–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Takahashi, Y.; Itami, T.; Kondo, M.; Maeda, M.; Fujii, R.; Tomonaga, S.; Supamattaya, K.; Boonyaratpalin, S. Electron Microscopic Evidence of Bacilliform Virus Infection in Kuruma Shrimp(Penaeus Japonicus). Fish Pathol. 1994, 29, 121–125. [Google Scholar] [CrossRef] [Green Version]
  136. Alavandi, S.V.; Muralidhar, M.; Syama Dayal, J.; Rajan, J.S.; Ezhil Praveena, P.; Bhuvaneswari, T.; Saraswathy, R.; Chitra, V.; Vijayan, K.K.; Otta, S.K. Investigation on the Infectious Nature of Running Mortality Syndrome (RMS) of Farmed Pacific White Leg Shrimp, Penaeus Vannamei in Shrimp Farms of India. Aquaculture 2019, 500, 278–289. [Google Scholar] [CrossRef]
  137. Cock, J.; Gitterle, T.; Salazar, M.; Rye, M. Breeding for Disease Resistance of Penaeid Shrimps. Aquaculture 2009, 286, 1–11. [Google Scholar] [CrossRef]
  138. Wang, Z.; Sun, B.; Zhu, F. Epigallocatechin-3-Gallate Inhibit Replication of White Spot Syndrome Virus in Scylla Paramamosain. Fish Shellfish Immunol. 2017, 67, 612–619. [Google Scholar] [CrossRef]
  139. Wang, Z.; Sun, B.; Zhu, F. Epigallocatechin-3-Gallate Protects Kuruma Shrimp Marsupeneaus Japonicus from White Spot Syndrome Virus and Vibrio Alginolyticus. Fish Shellfish Immunol. 2018, 78, 1–9. [Google Scholar] [CrossRef]
  140. Sun, Z.; Chen, C.; Xu, F.; Li, B.; Shen, J.; Wang, T.; Jiang, H.; Wang, G. Evaluation of the Antiviral Activity of Naringenin, a Major Constituent of Typha angustifolia, against White Spot Syndrome Virus in Crayfish Procambarus Clarkii. J. Fish Dis. 2021, 44, 1503–1513. [Google Scholar] [CrossRef]
  141. Peng, H.; Wei, Z.; Luo, H.; Yang, Y.; Wu, Z.; Gan, L.; Yang, X. Inhibition of Fat Accumulation by Hesperidin in Caenorhabditis elegans. J. Agric. Food Chem. 2016, 64, 5207–5214. [Google Scholar] [CrossRef]
  142. Liu, F.; Qu, Y.-K.; Geng, C.; Wang, A.-M.; Zhang, J.-H.; Chen, K.-J.; Liu, B.; Tian, H.-Y.; Yang, W.-P.; Yu, Y.-B. Effects of Hesperidin on the Growth Performance, Antioxidant Capacity, Immune Responses and Disease Resistance of Red Swamp Crayfish (Procambarus Clarkii). Fish Shellfish Immunol. 2020, 99, 154–166. [Google Scholar] [CrossRef] [PubMed]
  143. Huang, A.-G.; Tu, X.; Qi, X.-Z.; Ling, F.; Zhu, B.; Wang, G.-X. Gardenia Jasminoides Ellis Inhibit White Spot Syndrome Virus Replication in Red Swamp Crayfish Procambarus Clarkii. Aquaculture 2019, 504, 239–247. [Google Scholar] [CrossRef]
  144. Huang, A.-G.; Tan, X.-P.; Cui, H.-B.; Qi, X.-Z.; Zhu, B.; Wang, G.-X. Antiviral Activity of Geniposidic Acid against White Spot Syndrome Virus Replication in Red Swamp Crayfish Procambarus Clarkii. Aquaculture 2020, 528, 735533. [Google Scholar] [CrossRef]
  145. Nakamura, K.; Hosoo, S.; Yamaguchi, S.; Koyama, M.; Yamazaki, R.; Hirata, T.; Yamaguchi, Y.; Yamasaki, H.; Minamino, N.; Wada, K.; et al. Geniposidic Acid Upregulates Atrial Natriuretic Peptide Secretion and Lowers Blood Pressure in Spontaneously Hypertensive Rats. J. Funct. Foods 2018, 40, 634–638. [Google Scholar] [CrossRef]
  146. Rameshthangam, P.; Ramasamy, P. Antiviral Activity of Bis(2-Methylheptyl)Phthalate Isolated from Pongamia Pinnata Leaves against White Spot Syndrome Virus of Penaeus Monodon Fabricius. Virus Res. 2007, 126, 38–44. [Google Scholar] [CrossRef]
  147. Klongklaew, N.; Praiboon, J.; Tamtin, M.; Srisapoome, P. Chemical Composition of a Hot Water Crude Extract (HWCE) from Ulva Intestinalis and Its Potential Effects on Growth Performance, Immune Responses, and Resistance to White Spot Syndrome Virus and Yellowhead Virus in Pacific White Shrimp (Litopenaeus Vannamei). Fish Shellfish Immunol. 2021, 112, 8–22. [Google Scholar] [CrossRef]
  148. Sirirustananun, N.; Chen, J.-C.; Lin, Y.-C.; Yeh, S.-T.; Liou, C.-H.; Chen, L.-L.; Sim, S.S.; Chiew, S.L. Dietary Administration of a Gracilaria Tenuistipitata Extract Enhances the Immune Response and Resistance against Vibrio Alginolyticus and White Spot Syndrome Virus in the White Shrimp Litopenaeus Vannamei. Fish Shellfish Immunol. 2011, 31, 848–855. [Google Scholar] [CrossRef]
  149. Kulkarni, A.; Krishnan, S.; Anand, D.; Kokkattunivarthil Uthaman, S.; Otta, S.K.; Karunasagar, I.; Kooloth Valappil, R. Immune Responses and Immunoprotection in Crustaceans with Special Reference to Shrimp. Rev. Aquacult. 2021, 13, 431–459. [Google Scholar] [CrossRef]
  150. Liu, J.; Zhang, P.; Wang, B.; Lu, Y.; Li, L.; Li, Y.; Liu, S. Evaluation of the Effects of Astragalus Polysaccharides as Immunostimulants on the Immune Response of Crucian Carp and against SVCV in Vitro and in Vivo. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2022, 253, 109249. [Google Scholar] [CrossRef]
  151. Huang, A.-G.; Tan, X.-P.; Qu, S.-Y.; Wang, G.-X.; Zhu, B. Evaluation on the Antiviral Activity of Genipin against White Spot Syndrome Virus in Crayfish. Fish Shellfish Immunol. 2019, 93, 380–386. [Google Scholar] [CrossRef]
  152. Mariot, L.V.; Bolívar, N.; Coelho, J.D.R.; Goncalves, P.; Colombo, S.M.; do Nascimento, F.V.; Schleder, D.D.; Hayashi, L. Diets Supplemented with Carrageenan Increase the Resistance of the Pacific White Shrimp to WSSV without Changing Its Growth Performance Parameters. Aquaculture 2021, 545, 737172. [Google Scholar] [CrossRef]
  153. Medina-Beltrán, V.; Luna-González, A.; Fierro-Coronado, J.A.; Campa-Córdova, Á.I.; Peraza-Gómez, V.; Flores-Miranda, M.d.C.; Gutiérrez Rivera, J.N. Echinacea Purpurea and Uncaria Tomentosa Reduce the Prevalence of WSSV in Witheleg Shrimp (Litopenaeus Vannamei) Cultured under Laboratory Conditions. Aquaculture 2012, 358–359, 164–169. [Google Scholar] [CrossRef]
  154. Palanikumar, P.; Daffni Benitta, D.J.; Lelin, C.; Thirumalaikumar, E.; Michaelbabu, M.; Citarasu, T. Effect of Argemone Mexicana Active Principles on Inhibiting Viral Multiplication and Stimulating Immune System in Pacific White Leg Shrimp Litopenaeus Vannamei against White Spot Syndrome Virus. Fish Shellfish Immunol. 2018, 75, 243–252. [Google Scholar] [CrossRef]
  155. Fierro Coronado, J.; Luna Gonzalez, A.; Caceres Martinez, C.; Ruiz Verdugo, C.; Escamilla Montes, R.; Diarte Plata, G.; Flores Miranda, M.; Alvarez Ruiz, P.; Peraza Gomez, V. Effect of Medicinal Plants on the Survival of White Shrimp (Penaeus Vannamei) Challenged with WSSV and Vibrio Parahaemolyticus. Lat. Am. J. Aquat. Res. 2019, 47, 377–381. [Google Scholar] [CrossRef] [Green Version]
  156. Jiang, H.-F.; Chen, C.; Jiang, X.-Y.; Shen, J.-L.; Ling, F.; Li, P.-F.; Wang, G.-X. Luteolin in Lonicera Japonica Inhibits the Proliferation of White Spot Syndrome Virus in the Crayfish Procambarus Clarkii. Aquaculture 2022, 550, 737852. [Google Scholar] [CrossRef]
  157. Wang, Q.; Guo, L.Y.; Zhou, G.Q.; Jiang, G.C.; Ding, Z.F. Effects of Chicory Polysaccharides on the Growth, Antioxidant Activity, and Disease Resistance in the Chinese Mitten Crab Eriocheir Sinensis H. Milne Edwards, 1853 (Decapoda: Brachyura: Varunidae). J. Crustacean Biol. 2021, 41, ruab023. [Google Scholar] [CrossRef]
  158. Zhang, Y.; Xiao, C.; Zhu, F. Effects of Dietary Quercetin on the Innate Immune Response and Resistance to White Spot Syndrome Virus in Procambarus Clarkii. Fish Shellfish Immunol. 2021, 118, 205–212. [Google Scholar] [CrossRef]
  159. Dewi, N.R.; Huang, H.-T.; Wu, Y.-S.; Liao, Z.-H.; Lin, Y.-J.; Lee, P.-T.; Nan, F.-H. Guava (Psidium Guajava) Leaf Extract Enhances Immunity, Growth, and Resistance against Vibrio Parahaemolyticus in White Shrimp Penaeus Vannamei. Fish Shellfish Immunol. 2021, 118, 1–10. [Google Scholar] [CrossRef]
  160. Qian, X.; Zhu, F. Hesperetin Protects Crayfish Procambarus Clarkii against White Spot Syndrome Virus Infection. Fish Shellfish Immunol. 2019, 93, 116–123. [Google Scholar] [CrossRef]
  161. Bindhu, F.; Velmurugan, S.; Donio, M.B.S.; Michaelbabu, M.; Citarasu, T. Influence of Agathi Grandiflora Active Principles Inhibit Viral Multiplication and Stimulate Immune System in Indian White Shrimp Fenneropenaeus Indicus against White Spot Syndrome Virus Infection. Fish Shellfish Immunol. 2014, 41, 482–492. [Google Scholar] [CrossRef]
  162. Citarasu, T.; Sivaram, V.; Immanuel, G.; Rout, N.; Murugan, V. Influence of Selected Indian Immunostimulant Herbs against White Spot Syndrome Virus (WSSV) Infection in Black Tiger Shrimp, Penaeus Monodon with Reference to Haematological, Biochemical and Immunological Changes. Fish Shellfish Immunol. 2006, 21, 372–384. [Google Scholar] [CrossRef] [PubMed]
  163. Balasubramanian, G.; Sarathi, M.; Venkatesan, C.; Thomas, J.; Sahul Hameed, A.S. Oral Administration of Antiviral Plant Extract of Cynodon Dactylon on a Large Scale Production against White Spot Syndrome Virus (WSSV) in Penaeus Monodon. Aquaculture 2008, 279, 2–5. [Google Scholar] [CrossRef]
  164. Balasubramanian, G.; Sarathi, M.; Venkatesan, C.; Thomas, J.; Sahul Hameed, A.S. Studies on the Immunomodulatory Effect of Extract of Cyanodon Dactylon in Shrimp, Penaeus Monodon, and Its Efficacy to Protect the Shrimp from White Spot Syndrome Virus (WSSV). Fish Shellfish Immunol. 2008, 25, 820–828. [Google Scholar] [CrossRef] [PubMed]
  165. Tomazelli Junior, O.; Kuhn, F.; Mendonça Padilha, P.J.; Mota Vicente, L.R.; Winckler da Costa, S.; Corrêa da Silva, B.; Dias Schleder, D.; Boligon, A.A.; Scapinello, J.; Nunes Nesi, C.; et al. Effect of Cynodon Dactylon Extract on White Spot Virus-Infected Litopenaeus Vannamei. Aquac. Int. 2017, 25, 1107–1122. [Google Scholar] [CrossRef]
  166. Trejo-Flores, J.V.; Luna-González, A.; Álvarez-Ruíz, P.; Escamilla-Montes, R.; Peraza-Gómez, V.; Diarte-Plata, G.; Esparza-Leal, H.M.; Campa-Córdova, Á.I.; Gámez-Jiménez, C.; Rubio-Castro, A. Protective Effect of Aloe Vera in Litopenaeus Vannamei Challenged with Vibrio Parahaemolyticus and White Spot Syndrome Virus. Aquaculture 2016, 465, 60–64. [Google Scholar] [CrossRef]
  167. Balasubramanian, G.; Sarathi, M.; Kumar, S.R.; Hameed, A.S.S. Screening the Antiviral Activity of Indian Medicinal Plants against White Spot Syndrome Virus in Shrimp. Aquaculture 2007, 263, 15–19. [Google Scholar] [CrossRef]
  168. Júnior, O.T.; Kuhn, F.; Mendonça Padilha, P.J.; Mota Vicente, L.R.; Winckler da Costa, S.; Corrêa da Silva, B.; Schleder, D.D.; Nunes Nesi, C.; Dal Magro, J.; de Lamo-Castellví, S. Survival of White Spot Syndrome Virus–Infected Litopenaeus Vannamei Fed with Ethanol Extract of Uncaria Tomentosa. J. World Aquac. Soc. 2018, 49, 165–174. [Google Scholar] [CrossRef]
  169. Muliani; Nurbaya; Kadriah, I.A.K. The Effect of Several Types of Mangrove Exctracs on Tiger Shrimp Penaeus Monodon Survival Rate Challenged with White Spot Syndrome Virus (WSSV). IOP Conf. Ser. Earth Environ. Sci. 2020, 564, 012054. [Google Scholar] [CrossRef]
  170. Qian, X.; Zhu, F. Use of Glycerol Monolaurate as a Treatment against White Spot Syndrome Virus in Crayfish (Procambarus Clarkii). Aquaculture 2021, 541, 736853. [Google Scholar] [CrossRef]
  171. Gholamhosseini, A.; Kheirandish, M.R.; Shiry, N.; Akhlaghi, M.; Soltanian, S.; Roshanpour, H.; Banaee, M. Use of a Methanolic Olive Leaf Extract (Olea Europaea) against White Spot Virus Syndrome in Penaeus Vannamei: Comparing the Biochemical, Hematological and Immunological Changes. Aquaculture 2020, 528, 735556. [Google Scholar] [CrossRef]
  172. Jin, Q.-R.; Mao, J.-W.; Zhu, F. The Effects of Anoectochilus Roxburghii Polysaccharides on the Innate Immunity and Disease Resistance of Procambarus Clarkii. Aquaculture 2022, 555, 738210. [Google Scholar] [CrossRef]
  173. Zhang, Y.; Wen, J.; Xu, Y.; Wang, H.; Lu, L.; Song, R.; Zou, J. Epigallocatechin-3-Gallate Inhibits Replication of White Spot Syndrome Virus in the Freshwater Crayfish Procambarus Clarkii. J. Fish Dis. 2022, 45, 445–450. [Google Scholar] [CrossRef] [PubMed]
  174. Shan, L.-P.; Zhang, X.; Hu, Y.; Liu, L.; Chen, J. Antiviral Activity of Esculin against White Spot Syndrome Virus: A New Starting Point for Prevention and Control of White Spot Disease Outbreaks in Shrimp Seedling Culture. J. Fish Dis. 2022, 45, 59–68. [Google Scholar] [CrossRef] [PubMed]
  175. Hu, Y.; Liu, L.; Shan, L.-P.; Chen, J. Natural Ingredient Paeoniflorin Could Be a Lead Compound against White Spot Syndrome Virus Infection in Litopenaeus Vannamei. J. Fish Dis. 2022, 45, 349–359. [Google Scholar] [CrossRef]
  176. Huang, A.-G.; He, W.-H.; Zhang, F.-L.; Wei, C.-S.; Wang, Y.-H. Natural Component Geniposide Enhances Survival Rate of Crayfish Procambarus Clarkii Infected with White Spot Syndrome Virus. Fish Shellfish Immunol. 2022, 126, 96–103. [Google Scholar] [CrossRef]
  177. Xj, A.; Wj, A.; Fei, Z.B. Dietary Hizikia fusiforme Enhance Survival of White Spot Syndrome Virus Infected Crayfish Procambarus Clarkii—ScienceDirect. Fish Shellfish Immunol. 2020, 103, 88–94. [Google Scholar] [CrossRef]
  178. Walker, P.J.; Winton, J.R. Emerging Viral Diseases of Fish and Shrimp. Vet. Res. 2010, 41, 51. [Google Scholar] [CrossRef] [Green Version]
  179. Frankic, A.; Hershner, C. Sustainable Aquaculture: Developing the Promise of Aquaculture. Aquac. Int. 2003, 11, 517–530. [Google Scholar] [CrossRef]
  180. Gui, J.; Zhu, Z. Molecular Basis and Genetic Improvement of Economically Important Traits in Aquaculture Animals. Chin. Sci. Bull. 2012, 57, 1751–1760. [Google Scholar] [CrossRef] [Green Version]
  181. Bacharach, E.; Mishra, N.; Briese, T.; Zody, M.C.; Kembou Tsofack, J.E.; Zamostiano, R.; Berkowitz, A.; Ng, J.; Nitido, A.; Corvelo, A.; et al. Characterization of a Novel Orthomyxo-like Virus Causing Mass Die-Offs of Tilapia. mBio 2016, 7, e00431-16. [Google Scholar] [CrossRef] [Green Version]
  182. Lin, L.-T.; Hsu, W.-C.; Lin, C.-C. Antiviral Natural Products and Herbal Medicines. J. Tradit. Complementary Med. 2014, 4, 24–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Dawood, M.A.O.; Habotta, O.A.E.; Elsabagh, M.; Azra, M.N.; Van Doan, H.; Kari, Z.A.; Sewilam, H. Fruit Processing By-products in the Aquafeed Industry: A Feasible Strategy for Aquaculture Sustainability. Rev. Aquac. 2022, raq.12680. [Google Scholar] [CrossRef]
  184. Cheng, H.-Y.; Yang, C.-M.; Lin, T.-C.; Lin, L.-T.; Chiang, L.-C.; Lin, C.-C. Excoecarianin, Isolated from Phyllanthus Urinaria Linnea, Inhibits Herpes Simplex Virus Type 2 Infection through Inactivation of Viral Particles. Evid.-Based Complement. Altern. Med. 2011, 2011, 259103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Gescher, K.; Kühn, J.; Lorentzen, E.; Hafezi, W.; Derksen, A.; Deters, A.; Hensel, A. Proanthocyanidin-Enriched Extract from Myrothamnus Flabellifolia Welw. Exerts Antiviral Activity against Herpes Simplex Virus Type 1 by Inhibition of Viral Adsorption and Penetration. J. Ethnopharmacol. 2011, 134, 468–474. [Google Scholar] [CrossRef]
  186. Chiang, L.-C.; Ng, L.-T.; Cheng, P.-W.; Chiang, W.; Lin, C.-C. Antiviral Activities of Extracts and Selected Pure Constituents of Ocimum Basilicum. Clin. Exp. Pharmacol. Physiol. 2005, 32, 811–816. [Google Scholar] [CrossRef]
  187. Mouler Rechtman, M.; Har-Noy, O.; Bar-Yishay, I.; Fishman, S.; Adamovich, Y.; Shaul, Y.; Halpern, Z.; Shlomai, A. Curcumin Inhibits Hepatitis B Virus via Down-Regulation of the Metabolic Coactivator PGC-1α. FEBS Lett. 2010, 584, 2485–2490. [Google Scholar] [CrossRef] [Green Version]
  188. Kuo, Y.-C.; Lin, L.-C.; Tsai, W.-J.; Chou, C.-J.; Kung, S.-H.; Ho, Y.-H. Samarangenin B from Limonium Sinense Suppresses Herpes Simplex Virus Type 1 Replication in Vero Cells by Regulation of Viral Macromolecular Synthesis. Antimicrob. Agents Chemother. 2002, 46, 2854–2864. [Google Scholar] [CrossRef] [Green Version]
  189. Takebe, Y.; Saucedo, C.J.; Lund, G.; Uenishi, R.; Hase, S.; Tsuchiura, T.; Kneteman, N.; Ramessar, K.; Tyrrell, D.L.J.; Shirakura, M.; et al. Antiviral Lectins from Red and Blue-Green Algae Show Potent in Vitro and in Vivo Activity against Hepatitis C Virus. PLoS ONE 2013, 8, e64449. [Google Scholar] [CrossRef] [Green Version]
  190. Pei-wen, C.; Chiang, L.-C.; Yen, M.-H.; Lin, C.-C. Bupleurum Kaoi Inhibits Coxsackie B Virus Type 1 Infection of CCFS-1 Cells by Induction of Type I Interferons Expression. Food Chem. Toxicol. 2007, 45, 24–31. [Google Scholar] [CrossRef]
  191. Devi, A.B.; Sarala, R. Substantial Effect of Phytochemical Constituents against the Pandemic Disease Influenza—A Review. Future J. Pharm. Sci. 2021, 7, 120. [Google Scholar] [CrossRef]
  192. Safari, O.; Sarkheil, M.; Paolucci, M. Dietary Administration of Ferula (Ferula Asafoetida) Powder as a Feed Additive in Diet of Koi Carp, Cyprinus Carpio Koi: Effects on Hemato-Immunological Parameters, Mucosal Antibacterial Activity, Digestive Enzymes, and Growth Performance. Fish Physiol. Biochem. 2019, 45, 1277–1288. [Google Scholar] [CrossRef] [PubMed]
  193. Maldonado-Garcia, M.; Angulo, C.; Vazquez-Martinez, J.; Sanchez, V.; Lopez, M.G.; Reyes-Becerril, M. Antioxidant and Immunostimulant Potentials of Chenopodium ambrosioides L. in Pacific Red Snapper (Lutjanus Peru). Aquaculture 2019, 513, 734414. [Google Scholar] [CrossRef]
  194. Bilen, S.; Ispir, S.; Kenanoglu, O.N.; Taştan, Y.; Güney, K.; Terzi, E. Effects of Greek Juniper (Juniperus Excelsa) Extract on Immune Responses and Disease Resistance against Yersinia Ruckeri in Rainbow Trout (Oncorhynchus Mykiss). J. Fish Dis. 2021, 44, 729–738. [Google Scholar] [CrossRef] [PubMed]
  195. Chitmanat, C.; Tongdonmuan, K.; Khanom, P.; Pachontis, P.; Nunsong, W. Antiparasitic, Antibacterial, and Antifungal Activities Derived from a Terminalia Catappa Solution against Some Tilapia (Oreochromis Niloticus) Pathogens. Acta Hortic. 2005, 678, 179–182. [Google Scholar] [CrossRef]
  196. Wu, G.; Yuan, C.; Shen, M.; Tang, J.; Gong, Y.; Li, D.; Sun, F.; Huang, C.; Han, X. Immunological and Biochemical Parameters in Carp (Cyprinus Carpio) after Qompsell Feed Ingredients for Long-Term Administration. Aquac. Res. 2007, 38, 246–255. [Google Scholar] [CrossRef]
  197. VENKATRAMALINGAM, K.; Christopher, G.; Citarasu, T. Zingiber Officinalis an Herbal Appetizer in the Tiger Shrimp Penaeus Monodon (Fabricius) Larviculture. Aquac. Nutr. 2007, 13, 439–443. [Google Scholar] [CrossRef]
  198. Liñán-Cabello, M.; Medina-Zendejas, R.; Sánchez-Barajas, M.; Herrera, A. Effects of Carotenoids and Retinol in Oocyte Maturation of Crayfish Cherax Quadrucarinatus. Aquac. Res. 2004, 35, 905–911. [Google Scholar] [CrossRef]
Figure 1. Mechanism of VHSV replication inhibition by α-lipoic acid.
Figure 1. Mechanism of VHSV replication inhibition by α-lipoic acid.
Viruses 14 01281 g001
Table 1. Medicinal plants and active compounds exhibiting antivirus activity in fish or shellfish and their effects or possible mechanisms of action.
Table 1. Medicinal plants and active compounds exhibiting antivirus activity in fish or shellfish and their effects or possible mechanisms of action.
Pathogenic VirusesMedicinal PlantsActive CompoundsThe Possible MechanismsReferences
IPNVHeliotropium filifoliumFilifolinyl senecionateInhibiting the synthesis of viral genomic RNA[28]
/Mycophenolic acid (MPA)Restraining cellular GMP synthesis by inhibiting IMPDH; disturbing RNA polymerase[30]
IHNV/CarotenoidImproving nonspecific immunity[42]
Lentinus edodes myceliaLentinan (LNT-I)Regulating the innate immune response and specific immunity[43]
Prunella vulgaris L. (PVL)Ursolic acid (UA)Inhibiting virus replication[45]
Rhus verniciflua StokesFlavonoidsInducing apoptosis of cells[47]
VHSV/Mycophenolic acid (MPA)Restraining cellular GMP synthesis by inhibiting IMPDH; disturbing RNA polymerase[30]
Rhus verniciflua StokesFlavonoidsInducing apoptosis of cells[47]
Celosia cristata and Raphanus sativusExtractInducing gene expression involved in the innate immune response[57]
Licorice (Glycyrrhiza uralensis, GUF)Extract; glycyrrhizin (GL); glycyrrhetinic acid (GLA)Inhibiting the early fusion steps[59]
/CurcuminInhibiting virus entry in cells[61]
Olive tree leaf (Olea europaea) (LExt)Extract; oleuropein (Ole)Inactivating virus particles; inhibiting cell-to-cell spread[63]
/α-Lipoic acid (LA)Inducing antiviral gene expression; reducing VHSV-induced oxidative stress[65]
SVCV/β-GlucoseRegulating the innate immune response[76]
Herbaspirillum camelliaeSeleniumActivating IFN-related gene expression[77]
Astragalus membranaceusAstragalus polysaccharide (APS)Activating IFN-related gene expression[80]
Astragalus membranaceusAPSStimulating the immune response of host; reducing SVCV-induced apoptosis[150]
/Palmitic acid (PA)Inhibiting autophagy[81]
Psoralea corylifoliaBavachin (BVN)Blocking SVCV-induced apoptosis[84]
Bupleurum yinchowenseSaikosaponin D (SSD)Reducing SVCV-induced apoptosis[85]
NNVGymnema sylvestreGymnemagenolInhibiting virus replication[94]
/GLS inhibitorInvolved in regulation of the TCA cycle[95]
/β-GlucanStimulating the innate immune memory of macrophages[96]
GCRVMagnoliaExtract; magnolo; honokiolFacilitating the expression of innate immune-related genes; restraining GCRV-induced apoptosis[104,105]
Green teaExtract; EGCGInhibiting viral particle adhesion to cells[107]
Green teaECGInhibiting viral particle adhesion to cells[109]
Panax ginsengGinsenoside Rg3Activating IFN-related gene expression[110]
/Quercetin (Qct)Counteracting the pro-viral effect of heat-shock response[113]
SGIVIllicium verum Hook. f.IVAE; IVEE; DDBA; QctInactivating virus particles; inhibiting early viral entry phases; inhibiting virus replication[122]
Illicium verum Hook. f.QctPromoting the recognition of SGIV and activating the IFN pathway[123,124]
Curcuma kwangsiensisCKEE; urdioneInhibiting virus replication[126]
Lonicera japonica Thunb.IAA, IAB, IAC, CA, LT, ISInhibiting virus replication[127]
Viola philippicaExtractDisturbing virus binding, entry, and replication in host cells[129]
Glycyrrhiza uralensis (GUF)ExtractImpacting the binding of virus particles to cell receptors and the replication of viruses in host cells[130]
WSSVGreen teaEGCGInducing gene expression involved in the innate immune response[138,139]
Typha angustifoliaNaringenin (NAR)Restraining early viral gene replication[140]
Pericarpium Citri ReticulataeHesperidinImproving nonspecific immunity[142]
Gardenia jasminoidesExtractBlocking viral immediate-early stage gene transcript[143]
Gardenia jasminoidesGenipin (GN)Attenuating oxidative stress and inflammatory; decreasing signal transducer and activator of transcription gene expression[151]
Eucommia ulmoidesGeniposidic acid (GPA)Restraining early viral gene replication; promoting apoptosis[144]
Pongamia pinnataBis(2-methylheptyl)phthalateImproving nonspecific immunity[146]
Gracilaria tenuistipitataExtractEnhancing the innate immunity[148]
Kappaphycus alvareziiCarrageenanImproving nonspecific immunity[152]
Echinacea purpurea; Uncaria tomentosaExtractIncreasing the activity of phenoloxidase[153]
Argemone mexicanaExtractInhibiting viral multiplication; stimulating immune system[154]
Mixture of garlic, echinacea, ginger, and basilPowdered plantsImproving nonspecific immunity[155]
Lonicera japonicaLuteolin (LUT)Inhibiting the expression of important viral genes; enhancing antioxidant defenses; mitigating inflammation; inducing apoptosis[156]
/Chicory polysaccharides (CP)Enhancing antioxidant activity; enhancing anti-WSSV resistance[157]
/QuercetinRegulating the innate immune response[158]
Psidium guajavaExtractImproving nonspecific immunity[159]
Lemon; orangeHesperetinRegulating the innate immunity[160]
Agathi grandifloraExtractEnhancing antioxidative enzyme gene expression[161]
Mixture of Cyanodon dactylon, Aegle marmelos, Tinospora cordifolia, Picrorhiza kurooa, and Eclipta albaExtractImproving nonspecific immunity[162]
Cynodon dactylonExtractPreventing the entry of the virus into the host; preventing the multiplication of the virus in the host cell; enhancing the innate immunity[163,164,165]
Aloe veraPowdered whole leafImproving nonspecific immunity[166]
A. marmelos; C. dactylon; L. camara; M. charantia; P. amarusExtractImproving nonspecific immunity[167]
Uncaria tomentosaExtractScavenging free radicals; increasing the activity of phenoloxidase[168]
Sonneratia albaExtractReducing the destruction of blood cells by viruses[169]
/Glycerol monolaurate (GML)Increasing hemocyte apoptosis, total hemocyte count (THC), PO, and SOD activity; enhancing the expression of immune-related genes[170]
Olea europaeaExtractReducing WSSV-induced oxidative stress[171]
Anoectochilus roxburghiiAnoectochilus roxburghii polysaccharides (ARPs)Upregulating the expression level of multiple immune genes; promoting the apoptosis of hemocytes[172]
Ophiopogon japonicusExtractBlocking early gene transcription; inducing cellular autophagy; attenuating WSSV-induced oxidative stress[17]
Green teaEGCGInhibiting virus replication[173]
/EsculinAttenuating the infectivity of viral particles; increasing the expression of antimicrobial peptides (AMPs)[174]
Paeonia lactifloraPaeoniflorinImproving nonspecific immunity, especially by increasing the expression of AMPS[175]
Gardenia jasminoidesGeniposide (GP)Restraining early and late viral gene expression[176]
Hizikia fusiformeExtractregulates the innate immunity[177]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liao, W.; Huang, L.; Han, S.; Hu, D.; Xu, Y.; Liu, M.; Yu, Q.; Huang, S.; Wei, D.; Li, P. Review of Medicinal Plants and Active Pharmaceutical Ingredients against Aquatic Pathogenic Viruses. Viruses 2022, 14, 1281. https://doi.org/10.3390/v14061281

AMA Style

Liao W, Huang L, Han S, Hu D, Xu Y, Liu M, Yu Q, Huang S, Wei D, Li P. Review of Medicinal Plants and Active Pharmaceutical Ingredients against Aquatic Pathogenic Viruses. Viruses. 2022; 14(6):1281. https://doi.org/10.3390/v14061281

Chicago/Turabian Style

Liao, Wenyu, Lin Huang, Shuyu Han, Dasheng Hu, Youhou Xu, Mingzhu Liu, Qing Yu, Shuaishuai Huang, Dongdong Wei, and Pengfei Li. 2022. "Review of Medicinal Plants and Active Pharmaceutical Ingredients against Aquatic Pathogenic Viruses" Viruses 14, no. 6: 1281. https://doi.org/10.3390/v14061281

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

Article Metrics

Back to TopTop