Next Article in Journal
Evaluation of the Effects of an Undenatured Collagen Type-2-Based Nutraceutical (ARTHROSHINE® HA²) on Recovery Time after TPLO in Dogs: A Prospective, Randomized Study with Objective Gait Analysis as the Primary Outcome Measure
Next Article in Special Issue
Characterization and Spike Gene Analysis of a Candidate Attenuated Live Bovine Coronavirus Vaccine
Previous Article in Journal
Neurobiology of Pathogen Avoidance and Mate Choice: Current and Future Directions
Previous Article in Special Issue
Leveraging Accelerometer Data for Lameness Detection in Dairy Cows: A Longitudinal Study of Six Farms in Germany
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Global Epidemiology of Bovine Leukemia Virus: Current Trends and Future Implications

by
Guanxin Lv
1,2,3,
Jianfa Wang
1,2,3,
Shuai Lian
1,2,3,
Hai Wang
1,2,3,* and
Rui Wu
1,2,3,4,*
1
College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
2
Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
3
China Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
4
College of Biology and Agriculture, Jiamusi University, Jiamusi 154007, China
*
Authors to whom correspondence should be addressed.
Animals 2024, 14(2), 297; https://doi.org/10.3390/ani14020297
Submission received: 8 December 2023 / Revised: 2 January 2024 / Accepted: 11 January 2024 / Published: 18 January 2024
(This article belongs to the Collection Cattle Diseases)

Abstract

:

Simple Summary

Bovine leukemia virus (BLV) is the causative agent of enzootic bovine leukosis (EBL), which is the most significant neoplastic disease in cattle. EBL is often overlooked in daily breeding processes due to the absence of obvious clinical symptoms. However, studies have revealed that EBL can severely impact the production performance of dairy cows, leading to a substantial economic burden on the cattle industry. In recent years, the global prevalence of EBL has been on the rise, and fragments of BLV nucleic acid have been detected in human breast cancer patients, raising public concerns. Due to the absence of an effective vaccine, controlling the disease is challenging. This review aims to provide a comprehensive overview of BLV, including its role in causing EBL, the genome of BLV, its current prevalence, transmission routes, clinical symptoms, detection methods, hazards, control strategies, and the current state of BLV research. The primary objective of this review is to offer breeders and researchers reliable veterinary knowledge on BLV and identify future research directions in this field.

Abstract

Bovine leukemia virus (BLV) is a retrovirus that causes enzootic bovine leucosis (EBL), which is the most significant neoplastic disease in cattle. Although EBL has been successfully eradicated in most European countries, infections continue to rise in Argentina, Brazil, Canada, Japan, and the United States. BLV imposes a substantial economic burden on the cattle industry, particularly in dairy farming, as it leads to a decline in animal production performance and increases the risk of disease. Moreover, trade restrictions on diseased animals and products between countries and regions further exacerbate the problem. Recent studies have also identified fragments of BLV nucleic acid in human breast cancer tissues, raising concerns for public health. Due to the absence of an effective vaccine, controlling the disease is challenging. Therefore, it is crucial to accurately detect and diagnose BLV at an early stage to control its spread and minimize economic losses. This review provides a comprehensive examination of BLV, encompassing its genomic structure, epidemiology, modes of transmission, clinical symptoms, detection methods, hazards, and control strategies. The aim is to provide strategic information for future BLV research.

1. Introduction

Bovine leukemia virus (BLV) is a retrovirus known to cause enzootic bovine leucosis (EBL) [1,2,3,4]. EBL has been successfully eradicated in most European countries [5]. However, recent investigations have shown an increase in infection rates in several countries, including Argentina, Brazil, Canada, Japan, and the United States [6,7,8,9]. The majority of BLV-infected cattle remain asymptomatic, harboring the virus in a latent state of infection, and are thus classified as carriers. As the disease progresses, around one-third of cattle infected with BLV exhibit a non-malignant proliferation of B lymphocytes, a condition referred to as persistent lymphocytosis (PL). After a long period of latent infection lasting 3 to 8 years, less than 5% of these cattle will eventually develop malignant B lymphocyte lymphoma [10,11].
Previous research suggested that lymphosarcoma caused by BLV primarily affected the economics of cattle farming. However, recent research has shown that BLV infection affects the immune system of cattle, even during the latency period, leading to varying degrees of immune suppression and direct economic losses. These losses include reduced milk production, increased susceptibility to infectious diseases (such as mastitis, skin infections, and hoof diseases), and decreased reproductive efficiency [12,13,14,15]. In the United States, where the BLV infection rate exceeds 40%, the annual losses due to reduced milk production have surpassed USD 500 million. Additionally, certain countries and regions have implemented strict import and export regulations for animals infected with BLV, resulting in various indirect economic losses [5,13,15,16,17,18]. As a result, the World Organisation for Animal Health (WOAH) has classified Enzootic Bovine Leukosis (EBL) as a disease that significantly impacts international trade [19].
In addition to its impact on cattle herds, studies have shown that unsterilized or uncooked dairy and beef products contain proviral nucleic acid fragments of BLV, which raises concerns about public health and safety [20]. While it has not been confirmed that BLV can cause human infections, the positive detection rate of BLV in samples from breast cancer patients is significantly higher compared to healthy breast control groups [21,22,23,24,25,26]. Moreover, there is a correlation between the consumption of beef, milk, and dairy products and the incidence of breast cancer in different countries and regions. For example, countries with lower consumption rates like India, Japan, South Korea, and China also exhibit lower incidences of breast cancer. Conversely, countries with higher consumption rates such as the United States, the United Kingdom, Australia, and Germany have a higher incidence of breast cancer [27,28]. However, some studies have not detected proviral DNA of BLV in breast cancer tissue samples [29,30,31]. Therefore, further research is necessary to determine the impact of BLV exposure on human health, and government or industry associations should develop preventive measures to reduce the risk of BLV infection. Consequently, the objective of this review is to provide a comprehensive overview of current global BLV epidemiology and summarize recent advancements in BLV detection, diagnosis, and prevention methods in order to offer strategic information for future BLV research.

2. Genomic Composition of BLV

BLV is a single-stranded RNA-RT virus that exhibits a spherical or rod-shaped morphology, measuring approximately 60 to 125 nanometers in diameter. It is enveloped by a double-layered membrane structure and its nucleocapsid demonstrates icosahedral symmetry [32,33]. It contains a diploid single-stranded RNA genome. The viral genome of BLV consists of a diploid single-stranded RNA molecule spanning a total of 8714 nucleotides. This genome encompasses structural protein genes, enzyme coding genes, the pX region, and two identical, long, terminal repeat sequences positioned at both ends of the genome (Figure 1) [34,35].
Structural proteins and enzyme coding genes, such as gag, pro, pol, and env, are crucial in the virus lifecycle, infectivity, and the production of infectious viral particles [36,37,38]. The gag gene is highly conserved and responsible for encoding three major non-glycosylated proteins: p12 nucleocapsid protein (which binds with viral genomic RNA), p15 matrix protein (which binds with viral genomic RNA and interacts with the lipid bilayer of the viral membrane), and p24 nucleocapsid protein (which serves as the main target of the host immune response) [37]. The pro gene encodes the viral proteinase p14, which is involved in the post-translation maturation of the virus [36]. The pol gene encodes reverse transcriptase and integrase, which play a role in the reverse transcription process and integration of proviral BLV DNA into the host genome [39]. The env gene encodes two glycosylated proteins: envelope protein gp51 and transmembrane protein gp30, which facilitate virus–cell fusion through interaction with cell membrane receptors [40,41]. The pX region, located between the env gene and 3’LTR, encodes other auxiliary regulatory or non-structural genes, including regulatory proteins Tax and Rex, as well as auxiliary proteins R3 and G4 [42,43,44,45,46,47,48]. Regulatory proteins are important in virus transcription regulation, inducing the malignant transformation of tumors and virus infection output, while R3 and G4 auxiliary proteins help maintain a high viral load [49]. The env gene also shows genetic polymorphism, which is beneficial for phylogenetic studies and classifying BLV isolates [40]. The nucleotide sequence and amino acid composition of the env gene are valuable genomic markers for studying the distribution of BLV and identifying different genotypes based on geographical origin [50]. Researchers have identified 10 BLV genotypes through sequencing and phylogenetic analysis of partial and full-length gp51 env genes. Genotype 1 is the most prevalent and commonly found in the United States, Japan, and South Korea [51,52,53]. South America has genotypes 1, 2, 3, 4, 5, 6, and 9 (with genotype 9 being particularly prevalent in Bolivia). Russia and Eastern Europe commonly have genotypes 4, 7, and 8 [54]. Genotype 10 is prevalent in China, Vietnam, Thailand, and Myanmar [51,55]. Genotype 4 is found in various countries in North and South America, Africa, and Asia [56,57]. Genotype 6 has been found in South American countries such as Argentina, Brazil, Peru, Paraguay, and Bolivia, as well as in Asian countries such as the Philippines, Thailand, and India [56,58,59,60]. Additionally, genotype 1 has been reported in Egypt [61]. Monitoring the emergence of new viral mutations in veterinary medicine and animal husbandry is important since each new genotype may exhibit unique characteristics in its interaction with the host organism and leukemogenesis is influenced by specific genotypes (Figure 2).
Research indicates that 10 different microRNA (miRNA) can be transcribed under the control of RNA polymerase III in the genomic region spanning from the env gene to the R3 coding area [63]. RNA sequencing analysis has revealed that these miRNAs are highly expressed in primary sheep and cattle cells containing pre-leukemic and malignant B cells, accounting for 40% of the total cellular miRNA [64]. Additionally, elevated levels of BLV miRNA were found in the serum of infected animals, suggesting a potential paracrine role. These findings indicate the important role of BLV miRNAs in the retroviral cycle and/or disease progression, challenging the traditional belief that RNA viruses do not encode miRNAs. They also demonstrate that BLV miRNAs regulate various biological processes, including apoptosis, immunity, signal transduction, and tumorigenesis [65]. Notably, Blv-miR-B4-3p, which shares a seed sequence with the host miRNA miR-29a, is known for downregulating the tumor suppressor genes HMG-box transcription factor 1 (HBP1) and peroxidasin homologs (PXDN) in B-cell tumors [63,65,66]. In vitro studies have confirmed that blv-miR-B4-3p can directly downregulate these genes, although this has not been experimentally validated in cattle infected with BLV. Recently, Petersen et al. used RT-qPCR to analyze hbp1 and pxdn expression in cattle naturally infected with BLV and noted a significant downregulation of pxdn compared to uninfected animals [67]. However, a comprehensive analysis of targets for each mature BLV miRNA is still needed to fully understand the role of BLV in tumorigenesis.

3. Current Epidemiological Distribution of BLV

In the 19th century, research reports had already described clinical symptoms associated with EBL in cattle [68,69]. In 1871, Leisering published a study in Germany where he observed slightly yellow modules in the splenomegaly of cattle, along with an increase in white blood cells [68]. Bollinger further described and defined the clinical symptoms of EBL in 1874 [70]. In 1876, Siedamgrotzky and Hofmeister documented the first case of a bovine lymphocytic malignant tumor [71]. Subsequently, EBL started spreading globally, mainly due to the trade of dairy and beef cattle and their related products. In 1969, researchers successfully isolated the EBL pathogen from the lymphocytes of infected cattle and named it BLV [72].
Countries and regions with advanced economies and high consumption of beef, milk, and dairy products have shown greater concern for BLV. Since the 1960s, several countries and regions, including the United Kingdom, Denmark, the Netherlands, New Zealand, and Australia, have implemented successful BLV control programs, claiming to have eradicated the virus [73,74]. Their primary strategy has been to eliminate or isolate all cattle that test positive in the ELISA test [75,76]. On the other hand, countries like the United States, Canada, Argentina, and Japan have not adopted BLV control measures, resulting in a BLV prevalence exceeding 40% (Figure 3) [6,8,9]. Table 1 provides the BLV infection rates in different countries and regions [62].
The natural hosts for BLV infection are cattle (Bos taurus), zebu (Bos indicus), and buffalo (Bubalus bubalis). However, it has also been experimentally transmitted to sheep, goats, chickens, rabbits, and rats [109,110,111]. Studies have shown that genetic susceptibility plays a significant role in the prevalence of BLV in cattle populations [112]. For instance, cattle carrying the BoLA-DRB3 gene have a significantly lower risk of BLV infection [113,114,115,116]. In dairy cattle populations, the frequency of BLV transmission is considerably higher compared to beef cattle populations [117]. A nationwide survey conducted in Japan from 2009 to 2011 reported seropositivity rates of 40.9% in dairy cattle and 28.7% in beef cattle [7,118]. Similarly, a study in China found infection rates of 49.1% in dairy cattle and 1.6% in beef cattle [96]. Furthermore, the seropositivity rate of BLV increases with the age of the cattle, as older age raises the risk of contact and infection [119,120].

4. Susceptibility and Transmission Pathways of BLV

BLV primarily resides in the peripheral blood lymphocytes of infected animals and can also be detected in various bodily fluids of BLV-positive cattle, including blood, milk, saliva, semen, and nasal secretions [121,122,123]. The main transmission routes of BLV are horizontal, which include (1) iatrogenic transmission, such as the reuse of medical devices contaminated with BLV (e.g., syringes, gloves, dehorning tools, castration tools, blood collection tools, and rectal examination instruments) during drug or vaccine injections [112,124]; (2) transmission through direct contact with infected cattle and their secretions, such as mucous membranes or the accidental ingestion of saliva, semen, urine, feces, and milk [124,125]; and (3) transmission through mosquito bites, including Stegomyia mosquitoes, Aedes vexans, and Aedes albopictus [126,127,128]. BLV can also be vertically transmitted from BLV-positive cows to their fetuses [129]. Additionally, BLV can be indirectly transmitted to fetuses through the ingestion of colostrum and milk containing free viral particles. This type of transmission typically occurs in dairy cows with a high proviral load (PVL) [130]. Studies have shown that cells in the milk of high-PVL dairy cows can carry BLV and transmit it indirectly through cell-to-cell contact [121]. The use of BLV-positive breeding bulls on farms can potentially result in the vertical transmission of BLV. While the risk of transmission through semen or embryos is considered to be negligible, natural mating with infected bulls can lead to transmission due to intense direct contact during mating [131,132]. Therefore, it is crucial to screen breeding bulls used for natural mating or artificial insemination to minimize the risk of BLV transmission [133].

5. Clinical Symptoms of EBL

EBL is divided into two types: fatal lymphoma-type EBL and sporadic bovine leukosis (SBL). The latter is non-contagious and primarily affects calves [11]. EBL progresses through three stages. In the first stage, approximately 70% of BLV-infected cattle test positive on serological tests but show no clinical symptoms or lymphocytosis. In the second stage, about 30% of BLV-infected cattle experience PL, which is characterized by non-malignant polyclonal expansion of CD5 + B cells. Most of these cattle carry high loads of BLV provirus. In the third stage, 5–10% of BLV-infected cattle eventually develop lymphosarcoma (LS) after a latent period of 3–8 years. This typically occurs in adult cattle and represents malignant proliferation [10,11,134,135,136].
The clinical symptoms of EBL include anorexia, digestive disorders, decreased milk production, bloating, abomasal displacement, diarrhea, constipation, superficial lymph node enlargement, lameness, paralysis, weight loss, and general weakness. In some cases, there may also be neurological dysfunctions [101,137]. Malignant lymphoma, caused by EBL, can invade various organs such as the uterus, mesentery, abomasum, spleen, lungs, kidneys, urinary tract, spine, scapula, and iliac lymph nodes, leading to disorders in the urinary, respiratory, and digestive systems [62,138]. Recent studies have revealed that infection with BLV reduces the diversity of microbial communities in the rumen and intestines of dairy cows, resulting in decreased energy conversion efficiency [139]. Furthermore, BLV infection impairs the function of macrophages and neutrophils, thereby suppressing the immune function of dairy cows. These findings provide insights into why BLV-positive dairy cows are more susceptible to other infectious diseases, experience decreased milk production, and have reduced reproductive efficiency [140,141].

6. Detection Methods of BLV

6.1. Serological Techniques Used for the Diagnosis of BLV

Researchers have developed various methods for detecting BLV, which can be broadly categorized into two types: serological tests based on BLV antibodies and polymerase chain reaction (PCR) tests based on the proviral DNA of BLV. Serological tests typically identify antibodies against the p24 capsid protein encoded by the gag gene and the gp51 membrane protein encoded by the env gene [142,143,144,145]. Common serological testing methods include agar gel immunodiffusion (AGID), passive hemagglutination assay (PHA), enzyme-linked immunosorbent assay (ELISA), and radioimmunoassay (RIA) [146,147,148,149,150]. These methods are suitable for detecting antibodies in bovine serum, milk, and the supernatants of BLV-infected cell cultures, as shown in Table 2 [142]. According to the WOAH, AGID and ELISA are the recommended tests for serological diagnosis of bovine leukemia virus (BLV) infection [151]. These tests have been validated using an international reference serum standard called E05, which establishes the minimum level of sensitivity required for the routine testing of serum and milk samples using AGID or ELISA.
However, each method has its own advantages and disadvantages. AGID is cost-effective and suitable for screening multiple samples but it has low sensitivity and is not suitable for milk samples. The efficiency of PHA is affected by pH values, temperature, and trypsin, which limits its reliability. RIA is suitable for immediate diagnosis after animal exposure but is not suitable for large-scale screening. On the other hand, ELISA is highly sensitive and easy to operate, making it suitable for serum and milk samples. However, it may produce false-negative results for early-stage serum samples and false-positive results for calves with maternal antibodies [99,142,144]. In summary, it is important to note that these antibody-based detection methods are not suitable for calves under six months old due to the potential for false-positive results caused by maternal antibodies.

6.2. Molecular Techniques Used for the Diagnosis of BLV

After BLV infection, genetic information is integrated into the host genome and remains transcriptionally silent for a long time to evade immune system recognition. In addition to conventional serological testing, various PCR techniques are widely used to directly detect the presence of proviral DNA in BLV-infected cattle with low, transient, or absent antibody titer. Various types of PCR are available for different purposes. Conventional single, semi-nested, and nested PCR tests are all valuable and sensitive tools for the early detection of BLV proviral DNA in blood, organs, or tumor samples. However, the semi-nested and nested tests offer significantly higher levels of sensitivity compared to single PCRs, as summarized in Table 3 [155,156,157,158,159,160]. Recently, a novel, blood-based PCR system was developed that can directly amplify DNA without the need for DNA separation and purification [161]. This method is highly specific and cost-effective, enabling the timely identification of BLV-infected cows. Takeshima developed a new qPCR-based method called BLVCoCoMo-qPCR for the quantitative detection of BLV. It is highly specific and sensitive, capable of detecting BLV strains in samples that test negative in nested PCR assays. This method has successfully detected various integrated BLV strains within the host genome from clinical cases of different geographic origins. With continuous technological improvements, BLVCoCoMo-qPCR can now measure the proviral load of both known and novel BLV variants, allowing for investigation into the correlation between BLV proviral load and disease progression [123,153,162,163,164]. For calves under six months of age that have not yet produced BLV antibodies, the PCR method is also applicable, but it requires specific laboratory equipment and instruments.
Other diagnostic methods for BLV include the detection of BLV viral proteins through Western blotting (WB), syncytium formation assays in a co-culture, and indirect immunofluorescence (IF) for detecting BLV antigens [165,166,167,168,169,170].
Table 3. Molecular techniques used for the diagnosis of BLV.
Table 3. Molecular techniques used for the diagnosis of BLV.
Test AssaySample TypeAdvantagesDisadvantagesReferences
Conventional PCR
(single, semi-nested, and nested PCR)
PBMC from blood, tumor sample, buffy coat, milk, somatic cells, semen, saliva, and nasal secretions.
Direct, sensitive, rapid, and can detect recent infection of BLV even before the development of antibodies.
Can be used for BLV detection during the early phase of infection or in the presence of colostrum antibodies.
False negatives with low proviral load
Cross-contamination
Requires specific primers
Requires equipment (PCR machine)
Needs sequencing for confirmation
[152,158,171]
Quantitative (real-time)
Direct, sensitive, rapid
Differentiates EBL from SBL
Detects BLV during the early phase of infection or in the presence of colostrum antibodies
Quantitative measurement of proviral load
Requires complicated sample preparation
Requires positive controls
Requires specific primers and probes
Expensive and requires equipment (real-time PCR machine)
[123,153,162,163,164,169,172]
Direct, blood-based PCRBlood
Low cost
Applied on the blood directly without DNA extraction or purification
Low risk of contamination
False negatives with low proviral load
Low sensitivity
[161]
PCR testing can be used to quantify the PVL of animals infected with BLV using molecular techniques. PVL refers to the number of copies of the provirus per nanogram of host genomic DNA or in a certain number of infected cells. It is an important indicator for measuring the progression of retroviral diseases and can be used to assess the risk of transmission of human T-cell leukemia virus type 1 (HTLV-1) or the progression of human immunodeficiency virus (HIV) [173,174,175,176,177]. Similarly, the PVL of BLV can also serve as an indicator for assessing the risk of transmission and progression of EBL [156,178,179,180]. Previous research has classified a BLV proviral copy number exceeding 1000 copies/10 ng of DNA as a high proviral load (H-PVL) and values below this as a low proviral load (L-PVL) [163]. Studies have shown that cows with a H-PVL are more likely to transmit BLV to other cows, while cows with a L-PVL have a lower probability of transmission [181]. Other research has found a positive correlation between BLV PVL and increased lymphocyte counts in cows with a H-PVL compared to those with normal lymphocyte counts [182,183]. Additionally, BLV PVL is closely associated with factors such as the duration of cow herd use, milk production, milk quality, and the impairment of immune function [178,181,184].
PCR detection of BLV PVL has certain limitations. Studies have shown that there is significant variation in BLV PVL detection results when lymphocyte samples from cows that have tested positive for BLV by ELISA are tested in different laboratories [185]. This variation can be attributed to three main factors. Firstly, different laboratories use different PCR detection materials, such as various brands of Taq DNA polymerase and PCR equipment. Secondly, differences in the operating habits of personnel, even within the same laboratory, can result in up to a 10% variance in BLV PVL detection results for the same positive sample when different personnel use the same equipment and materials. Lastly, the use of different gene sequence primers by different laboratories can lead to a tenfold difference in BLV PVL detection results. Currently, there are no standardized procedures or sequence primers for BLV PVL detection. Therefore, to accurately measure BLV PVL, it is recommended that researchers compare and analyze BLV PVL detection values, the OD values of ELISA antibody titration, and lymphocyte counts. Existing studies indicate a positive correlation between BLV PVL and BLV antibody titers, as well as between lymphocyte counts and BLV PVL [113,186,187,188,189]. By establishing a relationship among these three factors, BLV PVL can be more accurately defined, facilitating scientific research and daily production practices.

7. Hazards of BLV

7.1. Effects on the Function of the Immune System of Dairy Cows

BLV significantly affects immune system function in dairy cows. Under antigen stimulation, B cells can transform into plasma cells, which are responsible for synthesizing and secreting antibodies, thus contributing to humoral immunity. BLV primarily targets IgM CD5+ B cells, leading to interference in antibody production and resulting in functional disorders [190]. Research indicates that BLV infection causes abnormalities in the synthesis and secretion levels of various antibodies in the body. For instance, PBMC freshly isolated from PL cattle shows increased expression of Igγ mRNA while exhibiting decreased levels of Igμ mRNA, indicating transcriptional disruption in antibody production [191]. Calves infected with BLV in experimental settings exhibit similar total serum IgG levels as uninfected calves, but they experience a temporary elevation followed by a decline in total serum IgM [192]. Moreover, BLV-infected cattle demonstrate compromised antibody production in response to specific antigens. Notably, PL cattle require twice the time compared to uninfected cattle to generate antigen-specific antibodies when exposed to synthetic antigens. Additionally, antibody production in PL cattle is inconsistent, lacking a stable antibody ratio, whereas uninfected cattle consistently maintain an IgM:IgG ratio of 1:10 [193].
BLV not only infects CD5+ B lymphocytes but also infects various subgroups of immune cells, including CD2+, CD3+, CD4+, CD8+, γ/δ T cells, monocytes, and neutrophils [15]. As a result, immune cell dysfunction occurs, manifesting as changes in cytokine production, surface receptor expression, cell proliferation, and apoptotic capabilities [15]. The production of cytokines by the immune system plays a crucial role in regulating the growth, differentiation, and immune responses of immune cells. In the case of BLV infection, Th1 cell dysfunction can be observed, leading to abnormalities in the secretion of various Th1 cell cytokines. Research has demonstrated that compared to BLV-infected cattle, the PBMC of AL cattle exhibits a significant decrease in the transcription levels of IL-4 and IFN-γ mRNA. Similarly, in PL cattle, the transcription levels of IL-2, IL-4, and IFN-γ mRNA in the PBMC are also significantly reduced [194]. Following ConA stimulation, the transcription level of IL-2 mRNA in the PBMC of PL cattle is significantly higher than that in uninfected cattle and asymptomatic (AL) cattle [195]. BLV infection also impacts IFN-γ secretion, with a significant reduction in the INF-γ mRNA transcription level observed in the PBMC of BLV-uninfected cattle compared to AL and PL cattle [194]. While there are no significant changes in the INF-γ mRNA transcription levels in AL and PL cattle after ConA stimulation, they still remain significantly higher than in uninfected cattle [195].
During the immune response to BLV, plasmacytoid dendritic cells (pDCs) are activated and secrete increased levels of IFN-γ, while conventional dendritic cells (cDCs) show decreased secretion of IFN-γ [196,197]. In BLV-infected cattle, there is a significant increase in IL-12 mRNA transcription levels compared to those in uninfected cattle, although this change is not prominent among BLV-infected cattle [197]. IL-12 has two subtypes: IL-12 (p40) and IL-12 (p70). The transcription level of IL-12 (p40) mRNA is significantly increased in the PBMC of AL cattle, while it is significantly decreased in PL cattle [198]. Furthermore, BLV infection activates the MoDc group, leading to a reduction in the expression levels of both IL-12 (p40) and IL-12 (p70) genes, which promotes the process of BLV infection [199,200]. BLV infection may also upregulate the expression of immunosuppressive molecules such as PD-L1, LAG-3, Tim-3, and CTLA-4 on the surface of PBMC through an unknown mechanism [201,202,203,204]. This results in a reduced response of Th1 cells and cytokine secretion, impairing the activity of T lymphocytes and diminishing antiviral functions. Helper T cells 2 (Th2) secrete cytokines such as IL-4, IL-5, IL-6, IL-10, and IL-13, which play a crucial role in assisting B cell activation. These cytokines promote the proliferation, differentiation, and generation of antibodies. BLV infection affects the transcription expression of certain Th2 cell factors, including IL-4, IL-6, and IL-10. Research has demonstrated that the level of IL-4 mRNA in the PBMC of BLV-infected cattle decreases, while no significant changes are observed in AL or PL cattle [195]. This suggests that BLV indirectly impacts the expression of IL-4 rather than directly regulating it. Additionally, studies have revealed that the expression level of IL-6 mRNA in the PBMC of PL cattle is higher than in other groups under the stimulation of ConA, lipopolysaccharide (LPS), and BLV-gp51 protein, indicating that BLV infection may lead to inflammation [193]. IL-10 possesses immunosuppressive properties and can inhibit the transcription of Tax and pol genes during BLV replication in PBMC. Furthermore, it has been observed that the secretion of IL-10 increases with the progression of BLV infection [199].

7.2. Effects on the Milk Production and Milk Quality of Dairy Cows

Milk production is a crucial measure of dairy cattle productivity and plays a significant role in the economic benefits of the dairy farming industry. However, there is still ongoing debate regarding the impact of BLV infection on milk yield in dairy cows. The National Animal Health Monitoring System Dairy Research Center of the United States Department of Agriculture conducted a study revealing that a higher rate of BLV-positive infection in cattle herds could potentially lead to a decrease in overall milk production, affecting high-yielding dairy herds to a greater extent [13,131]. However, when examining individual cows, no correlation was found between BLV infection and milk yield [13,131]. Other research demonstrated that milk production in BLV-positive cows is comparable to that of healthy cows, and, in some cases, it may even be higher [13,205]. Additionally, a four-year study on the milking performance of cows experimentally infected with BLV reported that artificial infection with BLV can actually enhance the milk yield of dairy cows [206]. Hence, further research is necessary to thoroughly understand the impact of BLV infection on milk production in dairy cows and its underlying mechanisms.
The demand for milk and dairy products has shifted from quantity to quality as society has progressed. To judge the quality of fresh milk, the components of milk, such as somatic cell count and bacterial count, are considered important criteria. Several studies have shown that cows infected with BLV have a significantly higher somatic cell count compared to that of uninfected cows [96,207]. The infection of BLV also affects the quality of dairy products. For instance, the milk of BLV-positive cows has significantly reduced levels of antimicrobial peptides and lactoferrin, which are related to viral load [178]. Furthermore, untreated fresh milk and raw beef can contain BLV nucleic acid sequences. Although there are currently insufficient data to establish a direct link between BLV and human breast cancer, BLV gene fragments have been detected in the tumor tissues of breast cancer patients [20,21,24,25,26]. Therefore, it is crucial to strictly test and control dairy products from BLV-positive cows to ensure food safety. Additionally, conducting further research on the impact of BLV on human health will contribute to understanding its potential risks and ensuring public health and safety.

8. Prevention and Control Strategies for BLV

Various strategies have been identified in previous studies for controlling and preventing BLV infection. These strategies include implementing strict management procedures, testing and culling or separating infected animals, genetic trait screening, and vaccination, summarized in Table 4 [16,182,183,208].
Numerous attempts have been made worldwide over the last few decades to reduce the impact of EBL on dairy herds by decreasing the prevalence of BLV-infected animals within the herd. Some positive effects have been observed in dairy herds when implementing strict management procedures. These practices aim to minimize the spread of BLV within the herd, and a thorough understanding of transmission modes is crucial for optimizing such practices. According to animal health law (regulation (EU) no. 2016/429) [76], the most important practices that should be followed are as follows. (1) Use only milk from cows that have tested BLV-negative or, alternatively, use a milk replacer to feed calves. If using milk from BLV-infected cows, it should be treated through freezing or heat treatment before feeding to the calves. (2) Consider using chemical dehorning or cautery methods to reduce the risk of infection. (3) When administering injections, always use disposable needles or sterilize reusable needles by boiling them between animals. (4) It is important to clean and disinfect ear tattoo implements thoroughly between animals to prevent the spread of infection. (5) Properly wash and disinfect stomach tubes and drenching guns after each use on an animal to avoid cross-contamination. (6) Use separate gloves for rectal exploration to prevent the transmission of diseases. (7) Ensure that all equipment used to assist with calving is thoroughly washed and disinfected to maintain a hygienic environment. (8) Separate calving paddocks should be designated for BLV-infected and uninfected cattle to minimize the risk of transmission. (9) It is advisable to remove calves from cows within 24 h of birth, but only after they have received an adequate intake of colostrum. (10) Implementing a fly control program can help reduce the presence of flies, which can transmit disease [209,210].
The eradication of a disease with a long incubation period can only be achieved through programs that target all infected animals, rather than just focusing on the visible outbreaks of the disease. Implementing stringent management tools in dairy herds can result in a reduction of within-herd prevalence, but it is important to note that it cannot completely eradicate the infection. Therefore, the most effective and sustainable approach to achieving freedom from BLV is by eliminating infected animals. Testing and culling is considered a primary approach, involving regular screening of cattle herds using ELISA or PCR techniques, followed by the culling of positive dairy cows. Some European countries have successfully eradicated BLV using this strategy [5,16]. Achieving freedom can only be possible when the rate of removing positive animals exceeds the annual infection rate. However, this task becomes increasingly challenging as the within-herd prevalence increases. Additionally, in countries where there is a high prevalence of BLV or weaker economies, it may not be feasible to implement testing and culling due to the high economic costs associated with it [12,183]. As a result, the success of this strategy depends on the support of national governments through economic compensation policies. Countries that do not have such policy support, such as the United States, Canada, Argentina, and Japan, have not implemented this strategy. In order to reduce the prevalence of the disease in a herd, a test and separate scheme can be implemented. This scheme aims to gradually decrease the prevalence to a level where it would be feasible to switch to a test and cull strategy. The success of this control strategy depends on the initial level of disease prevalence within the herd and the rate at which animals are culled [161,211]. Recent research has demonstrated that cows infected with BLV can be categorized based on BLV PVL, a crucial indicator for assessing transmission risk [172,212]. This categorization can serve as a standard for culling or separating infected cows. Cows with a high PVL generally exhibit higher peripheral blood lymphocyte counts, and routine blood tests can be employed to identify and cull these cows, thereby further reducing costs. In conclusion, both ‘test and cull’ and ‘test and separate’ strategies are viable for eradicating the disease, depending on the prevalence within a specific herd.
In the field of genetic breeding, selecting traits that are beneficial for production and farming, such as yield, growth speed, and reproductive efficiency, can significantly enhance efficiency. Similarly, achieving comparable results can be possible by choosing dairy cow breeds that are resistant to BLV infection. Immune responses and genetic resistance are influenced by the host’s major histocompatibility complex (MHC), specifically the bovine leukocyte antigen (BoLA) in cattle. Research studies have consistently shown that cattle carrying the BoLA class II DRB3*0902 allele exhibit resistance to BLV or demonstrate significantly lower BLV proviral loads [213,214,215,216]. However, genetic breeding encounters certain challenges. It necessitates extensive research on a large scale to assess the effectiveness of marker genes in different breeds [217]. The genetic regulation of BLV is complex, making it challenging to consider specific alleles as absolute genetic markers [218,219]. Additionally, selecting based solely on genetic traits may have adverse effects on beneficial production traits [220]. Therefore, it is important to acknowledge that the strategies for BLV resistance selection may not always yield the expected benefits.
In recent decades, scientists have dedicated considerable efforts to the development of vaccines against BLV. Initially, inactivated BLV vaccines were created using cell lines continuously infected with BLV, such as FLK, LK15, Bat2Cl1, and others [221,222,223,224,225,226]. These vaccines were successful in generating specific neutralizing antibodies in sheep and cattle, effectively protecting them against low-dose BLV infections. However, they were found to be ineffective against high-dose infections. As a result, researchers shifted their focus towards the design and development of BLV subunit vaccines. This approach was driven by the high conservation of the gp51 gene sequence, which is found in various BLV isolates and contains at least three neutralizing epitopes [224,225,226,227,228,229,230]. On the other hand, attempts to target the p24 protein in similar research were unsuccessful [227]. Consequently, synthetic peptide vaccines were developed to mimic the gp51, B-cell, and T-cell epitopes [231,232]. In sheep models, vaccines have been shown to induce significant humoral and Th1 responses, leading to a reduction in BLV replication in the short term [233,234]. However, subsequent experiments revealed that the synthetic peptide vaccine failed to sustain antibody induction or provide effective protection against BLV infection in most vaccinated sheep [233]. This poor performance could be attributed to the inadequate presentation of certain epitopes and stereochemical structures. As a result, researchers developed recombinant vaccinia virus (RVV) vaccines carrying BLV. Studies on RVV vaccines encoding gp51 alone revealed that RVV-gp51 could not induce a humoral response or protect sheep from BLV infection [235]. However, RVV-env vaccines encoding the complete env gene, including gp51 and gp30, successfully induced humoral immune responses in sheep models [236]. Despite this, these RVV-env vaccines were unable to induce specific neutralizing antibodies and showed limited efficacy in experiments with cattle [237,238]. On the other hand, DNA vaccines have the potential to induce long-lasting immunity. DNA vaccines containing the env and tax genes, controlled by cytomegalovirus or Srα promoters, demonstrated strong immune responses but were unable to prevent subsequent BLV infections [239,240]. Excitingly, an attenuated virus vaccine developed through gene deletion or mutation has been reported to effectively protect cattle from BLV infection [241]. However, the efficacy and biosafety of this vaccine require further validation. In summary, there are currently no safe and effective commercial vaccines available to control BLV infection in cattle.

9. Reducing the BLV PVL Contributes to BLV Prevention and Control

The life cycle of retroviruses consists of early and late stages. In the early stage, the virus invades cells and integrates with the host genome. In the late stage, viral RNA is expressed and viral particles are produced. Currently, most antiretroviral drugs are developed to target HIV. Highly active antiretroviral therapy (HAART) has significantly improved the prognosis of HIV patients [242]. Antiviral drugs used in HAART inhibit important steps such as viral entry, reverse transcription, integration, and processing [243]. These drugs specifically target receptors, reverse transcriptase, integrase, and protease. Therefore, developing compounds that target these steps could be a promising treatment for BLV. However, previous studies have shown that reverse transcriptase inhibitors used for HIV have limited effects on BLV [35]. Recently, a study discovered a natural compound called Violacein E that can inhibit BLV replication in experiments [244]. However, these studies are still in the early stages and further extensive research is needed before practical application of these compounds can be considered. Additionally, some natural compounds are expensive and have complex extraction processes, and their effectiveness has not met expectations. Overall, due to the high incidence of BLV, the search for anti-BLV compounds remains a primary focus in treatment. However, there is still a need to develop commercially available drugs that can effectively reduce the BLV PVL.
During BLV infection, the body undergoes varying degrees of immune suppression, which can be attributed to the Th1 cell immune response. Investigating the mechanisms behind immune function suppression during BLV infection is crucial for identifying novel treatment strategies. Studies have revealed that the expression of immune suppressive molecules such as PD-L1, LAG-3, TIM-3, and CTLA-4 increases during BLV infection [201,202,203,204]. These molecules have been shown to hinder BLV-specific Th1 responses and contribute to disease progression. Research conducted by Okagawa T and Nishimori A suggests that blocking the PD-1/PD-L1 pathway with antibodies significantly reduces the BLV PVL, indicating that antibody blockade is an effective approach for treating BLV [245]. However, the production of such antibodies is expensive and not suitable for large-scale application in BLV control and treatment. In addition to this, BLV infection disrupts the functions of Th1 and Th2 cells, and cytokines demonstrate complex characteristics such as pleiotropy, antagonism, and synergy. Abnormal cytokine secretion during BLV infection is another important factor contributing to immune function suppression. Therefore, drugs that modulate the functions of Th1 and Th2 cells, restore cytokine secretion patterns, and enhance antiviral functions could potentially help reduce BLV PVL [35].

10. Conclusions

The prevalence of EBL poses a significant threat to the livestock industry and has resulted in substantial economic losses. Currently, the most effective strategy for controlling EBL involves identifying and eliminating infected animals, as well as implementing trade protection measures to prevent the entry of these animals. Most European countries have successfully employed this method to control EBL, despite its high cost. Furthermore, EBL control can also be achieved through genetic selection, management interventions, and the development of effective vaccines. While EBL cannot be completely cured, medication can be used to reduce the BLV PVL, thereby reducing the risk of transmission.

11. Future Prospective Studies

Vaccines are widely recognized as the most effective means of reducing disease transmission. However, developing a vaccine for retroviruses is particularly challenging. Despite decades of efforts, effective vaccines for retroviruses like HIV and HTLV-1 have not yet been developed. One of the reasons for this is the lack of suitable animal models and the long incubation period from infection to onset, which makes it difficult to accurately understand the disease progression. Additionally, BLV, a retrovirus that causes significant economic losses, has received less research attention compared to human or other bovine diseases. Instead, BLV has been primarily studied as a research model for HTLV-1 and other retroviruses. Drawing from the lessons learned from previous vaccine failures, it is recommended to develop attenuated vaccines for BLV by deleting single or multiple genes associated with viral immunosuppression or virulence. Another approach could be to learn from the development methods of COVID-19 mRNA vaccines. In addition to vaccine development, it is crucial to focus on effectively reducing BLV PVL as numerous studies have confirmed their correlation with the risk of BLV transmission. High-throughput in vitro cell assays can be used to screen natural compounds that effectively reduce BLV PVL. By reducing PVL and implementing detection and elimination strategies, the global spread of BLV can be controlled. Furthermore, the integration of the latest deep sequencing technology can facilitate in-depth research on the BLV transcriptome, metabolome, and proteome. A comprehensive understanding of the interaction between BLV and the host will provide valuable knowledge and insights for future research on BLV.

Author Contributions

Writing—original draft preparation, H.W. and G.L.; Writing—review and editing, H.W.; Supervision, J.W. and S.L.; Project administration, H.W. and R.W.; Funding acquisition, R.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the Natural Science Foundation of China (31972747, 32172937, and 32172814) and the Natural Science Foundation of the Heilongjiang Province of China, grant number TD2022C005.T.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Burny, A.; Bex, F.; Chantrenne, H.; Cleuter, Y.; Dekegel, D.; Ghysdael, J.; Kettmann, R.; Leclercq, M.; Leunen, J.; Mammerickx, M.; et al. Bovine leukemia virus involvement in enzootic bovine leukosis. Adv. Cancer Res. 1978, 28, 251–311. [Google Scholar] [CrossRef] [PubMed]
  2. Burny, A.; Bruck, C.; Cleuter, Y.; Couez, D.; Deschamps, J.; Gregoire, D.; Ghysdael, J.; Kettmann, R.; Mammerickx, M.; Marbaix, G.; et al. Bovine leukaemia virus and enzootic bovine leukosis. Onderstepoort J. Vet. Res. 1985, 52, 133–144. [Google Scholar] [PubMed]
  3. Kettmann, R.; Portetelle, D.; Mammerickx, M.; Cleuter, Y.; Dekegel, D.; Galoux, M.; Ghysdael, J.; Burny, A.; Chantrenne, H. Bovine leukemia virus: An exogenous RNA oncogenic virus. Proc. Natl. Acad. Sci. USA 1976, 73, 1014–1018. [Google Scholar] [CrossRef]
  4. Reed, V.I. Enzootic bovine leukosis. Can. Vet. J.-Rev. Vet. Can. 1981, 22, 95–102. [Google Scholar]
  5. Panel, E.A. Scientific opinion on enzootic bovine leukosis. EFSA J. 2015, 13, 4188. [Google Scholar]
  6. Nekouei, O.; Vanleeuwen, J.; Sanchez, J.; Kelton, D.; Tiwari, A.; Keefe, G. Herd-level risk factors for infection with bovine leukemia virus in Canadian dairy herds. Prev. Vet. Med. 2015, 119, 105–113. [Google Scholar] [CrossRef]
  7. Murakami, K.; Kobayashi, S.; Konishi, M.; Kameyama, K.; Tsutsui, T. Nationwide survey of bovine leukemia virus infection among dairy and beef breeding cattle in Japan from 2009–2011. J. Vet. Med. Sci. 2013, 75, 1123–1126. [Google Scholar] [CrossRef]
  8. Ma, B.; Gong, Q.; Sheng, C.; Liu, Y.; Ge, G.; Li, D.; Diao, N.; Shi, K.; Li, J.; Sun, Z.; et al. Prevalence of bovine leukemia in 1983–2019 in China: A systematic review and meta-analysis. Microb. Pathog. 2021, 150, 104681. [Google Scholar] [CrossRef]
  9. Ladronka, R.M.; Ainsworth, S.; Wilkins, M.J.; Norby, B.; Byrem, T.M.; Bartlett, P.C. Prevalence of bovine leukemia virus antibodies in US dairy cattle. Vet. Med. Int. 2018, 2018, 5831278. [Google Scholar] [CrossRef]
  10. Tsutsui, T.; Kobayashi, S.; Hayama, Y.; Yamamoto, T. Fraction of bovine leukemia virus-infected dairy cattle developing enzootic bovine leukosis. Prev. Vet. Med. 2016, 124, 96–101. [Google Scholar] [CrossRef]
  11. Gillet, N.; Florins, A.; Boxus, M.; Burteau, C.; Nigro, A.; Vandermeers, F.; Balon, H.; Bouzar, A.B.; Defoiche, J.; Burny, A.; et al. Mechanisms of leukemogenesis induced by bovine leukemia virus: Prospects for novel anti-retroviral therapies in human. Retrovirology 2007, 4, 18. [Google Scholar] [CrossRef]
  12. Norby, B.; Bartlett, P.C.; Byrem, T.M.; Erskine, R.J. Effect of infection with bovine leukemia virus on milk production in Michigan dairy cows. J. Dairy Sci. 2016, 99, 2043–2052. [Google Scholar] [CrossRef] [PubMed]
  13. Erskine, R.J.; Bartlett, P.C.; Byrem, T.M.; Render, C.L.; Febvay, C.; Houseman, J.T. Association between bovine leukemia virus, production, and population age in Michigan dairy herds. J. Dairy Sci. 2012, 95, 727–734. [Google Scholar] [CrossRef] [PubMed]
  14. Bartlett, P.C.; Norby, B.; Byrem, T.M.; Parmelee, A.; Ledergerber, J.T.; Erskine, R.J. Bovine leukemia virus and cow longevity in Michigan dairy herds. J. Dairy Sci. 2013, 96, 1591–1597. [Google Scholar] [CrossRef]
  15. Frie, M.C.; Coussens, P.M. Bovine leukemia virus: A major silent threat to proper immune responses in cattle. Vet. Immunol. Immunopathol. 2015, 163, 103–114. [Google Scholar] [CrossRef]
  16. Ruggiero, V.J.; Norby, B.; Benitez, O.J.; Hutchinson, H.; Sporer, K.; Droscha, C.; Swenson, C.L.; Bartlett, P.C. Controlling bovine leukemia virus in dairy herds by identifying and removing cows with the highest proviral load and lymphocyte counts. J. Dairy Sci. 2019, 102, 9165–9175. [Google Scholar] [CrossRef] [PubMed]
  17. Nuotio, L.; Rusanen, H.; Sihvonen, L.; Neuvonen, E. Eradication of enzootic bovine leukosis from Finland. Prev. Vet. Med. 2003, 59, 43–49. [Google Scholar] [CrossRef]
  18. Acaite, J.; Tamosiunas, V.; Lukauskas, K.; Milius, J.; Pieskus, J. The eradication experience of enzootic bovine leukosis from Lithuania. Prev. Vet. Med. 2007, 82, 83–89. [Google Scholar] [CrossRef]
  19. Enzootic Bovine Leukosis, Chapter 3.4.9. Available online: https://www.woah.org/fileadmin/Home/eng/Health_standards/tahm/3.04.09_EBL.pdf (accessed on 28 December 2023).
  20. Olaya-Galan, N.N.; Corredor-Figueroa, A.P.; Guzman-Garzon, T.C.; Rios-Hernandez, K.S.; Salas-Cardenas, S.P.; Patarroyo, M.A.; Gutierrez, M.F. Bovine leukaemia virus DNA in fresh milk and raw beef for human consumption. Epidemiol. Infect. 2017, 145, 3125–3130. [Google Scholar] [CrossRef]
  21. Khatami, A.; Pormohammad, A.; Farzi, R.; Saadati, H.; Mehrabi, M.; Kiani, S.J.; Ghorbani, S. Bovine Leukemia virus (BLV) and risk of breast cancer: A systematic review and meta-analysis of case-control studies. Infect. Agents Cancer 2020, 15, 48. [Google Scholar] [CrossRef]
  22. Khan, Z.; Abubakar, M.; Arshed, M.J.; Aslam, R.; Sattar, S.; Shah, N.A.; Javed, S.; Tariq, A.; Bostan, N.; Manzoor, S. Molecular investigation of possible relationships concerning bovine leukemia virus and breast cancer. Sci. Rep. 2022, 12, 4161. [Google Scholar] [CrossRef]
  23. Adekanmbi, F.; Mcneely, I.; Omeler, S.; Kalalah, A.; Poudel, A.; Merner, N.; Wang, C. Absence of bovine leukemia virus in the buffy coats of breast cancer cases from Alabama, USA. Microb. Pathog. 2021, 161, 105238. [Google Scholar] [CrossRef]
  24. Canova, R.; Weber, M.N.; Budaszewski, R.F.; Da, S.M.; Schwingel, D.; Canal, C.W.; Kreutz, L.C. Bovine leukemia viral DNA found on human breast tissue is genetically related to the cattle virus. One Health 2021, 13, 100252. [Google Scholar] [CrossRef] [PubMed]
  25. Buehring, G.C.; Shen, H.M.; Jensen, H.M.; Choi, K.Y.; Sun, D.; Nuovo, G. Bovine leukemia virus DNA in human breast tissue. Emerg. Infect. Dis. 2014, 20, 772–782. [Google Scholar] [CrossRef] [PubMed]
  26. Olaya-Galan, N.N.; Salas-Cardenas, S.P.; Rodriguez-Sarmiento, J.L.; Ibanez-Pinilla, M.; Monroy, R.; Corredor-Figueroa, A.P.; Rubiano, W.; de la Pena, J.; Shen, H.; Buehring, G.C.; et al. Risk factor for breast cancer development under exposure to bovine leukemia virus in Colombian women: A case-control study. PLoS ONE 2021, 16, e257492. [Google Scholar] [CrossRef]
  27. Afzal, S.; Fiaz, K.; Noor, A.; Sindhu, A.S.; Hanif, A.; Bibi, A.; Asad, M.; Nawaz, S.; Zafar, S.; Ayub, S.; et al. Interrelated Oncogenic Viruses and Breast Cancer. Front. Mol. Biosci. 2022, 9, 781111. [Google Scholar] [CrossRef]
  28. Lawson, J.S.; Salmons, B.; Glenn, W.K. Oncogenic Viruses and Breast Cancer: Mouse Mammary Tumor Virus (MMTV), Bovine Leukemia Virus (BLV), Human Papilloma Virus (HPV), and Epstein-Barr Virus (EBV). Front. Oncol. 2018, 8, 1. [Google Scholar] [CrossRef] [PubMed]
  29. Zhang, R.; Jiang, J.; Sun, W.; Zhang, J.; Huang, K.; Gu, X.; Yang, Y.; Xu, X.; Shi, Y.; Wang, C. Lack of association between bovine leukemia virus and breast cancer in Chinese patients. Breast Cancer Res. 2016, 18, 101. [Google Scholar] [CrossRef]
  30. Gillet, N.A.; Willems, L. Whole genome sequencing of 51 breast cancers reveals that tumors are devoid of bovine leukemia virus DNA. Retrovirology 2016, 13, 75. [Google Scholar] [CrossRef]
  31. Bender, A.P.; Robison, L.L.; Kashmiri, S.V.; Mcclain, K.L.; Woods, W.G.; Smithson, W.A.; Heyn, R.; Finlay, J.; Schuman, L.M.; Renier, C.; et al. No involvement of bovine leukemia virus in childhood acute lymphoblastic leukemia and non-Hodgkin’s lymphoma. Cancer Res. 1988, 48, 2919–2922. [Google Scholar] [PubMed]
  32. Zhao, X.; Buehring, G.C. Natural genetic variations in bovine leukemia virus envelope gene: Possible effects of selection and escape. Virology 2007, 366, 150–165. [Google Scholar] [CrossRef]
  33. Boris-Lawrie, K.; Altanerova, V.; Altaner, C.; Kucerova, L.; Temin, H.M. In vivo study of genetically simplified bovine leukemia virus derivatives that lack tax and rex. J. Virol. 1997, 71, 1514–1520. [Google Scholar] [CrossRef] [PubMed]
  34. Aida, Y.; Murakami, H.; Takahashi, M.; Takeshima, S.N. Mechanisms of pathogenesis induced by bovine leukemia virus as a model for human T-cell leukemia virus. Front. Microbiol. 2013, 4, 328. [Google Scholar] [CrossRef]
  35. Plant, E.; Bellefroid, M.; Van Lint, C. A complex network of transcription factors and epigenetic regulators involved in bovine leukemia virus transcriptional regulation. Retrovirology 2023, 20, 11. [Google Scholar] [CrossRef]
  36. Llames, L.; Goyache, J.; Domenech, A.; Montana, A.V.; Suarez, G.; Gomez-Lucia, E. Cellular distribution of bovine leukemia virus proteins gp51SU, Pr72(env), and Pr66(gag-pro) in persistently infected cells. Virus Res. 2001, 79, 47–57. [Google Scholar] [CrossRef] [PubMed]
  37. Katoh, I.; Kyushiki, H.; Sakamoto, Y.; Ikawa, Y.; Yoshinaka, Y. Bovine leukemia virus matrix-associated protein MA(p15): Further processing and formation of a specific complex with the dimer of the 5′-terminal genomic RNA fragment. J. Virol. 1991, 65, 6845–6855. [Google Scholar] [CrossRef]
  38. Mamoun, R.Z.; Morisson, M.; Rebeyrotte, N.; Busetta, B.; Couez, D.; Kettmann, R.; Hospital, M.; Guillemain, B. Sequence variability of bovine leukemia virus env gene and its relevance to the structure and antigenicity of the glycoproteins. J. Virol. 1990, 64, 4180–4188. [Google Scholar] [CrossRef]
  39. Lairmore, M.D. Animal models of bovine leukemia virus and human T-lymphotrophic virus type-1: Insights in transmission and pathogenesis. Annu. Rev. Anim. Biosci. 2014, 2, 189–208. [Google Scholar] [CrossRef] [PubMed]
  40. Rola-Luszczak, M.; Sakhawat, A.; Pluta, A.; Rylo, A.; Bomba, A.; Bibi, N.; Kuzmak, J. Molecular Characterization of the env Gene of Bovine Leukemia Virus in Cattle from Pakistan with NGS-Based Evidence of Virus Heterogeneity. Pathogens 2021, 10, 910. [Google Scholar] [CrossRef] [PubMed]
  41. de Brogniez, A.; Bouzar, A.B.; Jacques, J.R.; Cosse, J.P.; Gillet, N.; Callebaut, I.; Reichert, M.; Willems, L. Mutation of a Single Envelope N-Linked Glycosylation Site Enhances the Pathogenicity of Bovine Leukemia Virus. J. Virol. 2015, 89, 8945–8956. [Google Scholar] [CrossRef]
  42. Gillet, N.A.; Gutierrez, G.; Rodriguez, S.M.; de Brogniez, A.; Renotte, N.; Alvarez, I.; Trono, K.; Willems, L. Massive depletion of bovine leukemia virus proviral clones located in genomic transcriptionally active sites during primary infection. PLoS Pathog. 2013, 9, e1003687. [Google Scholar] [CrossRef]
  43. Lefebvre, L.; Ciminale, V.; Vanderplasschen, A.; D’Agostino, D.; Burny, A.; Willems, L.; Kettmann, R. Subcellular localization of the bovine leukemia virus R3 and G4 accessory proteins. J. Virol. 2002, 76, 7843–7854. [Google Scholar] [CrossRef]
  44. Willems, L.; Kerkhofs, P.; Dequiedt, F.; Portetelle, D.; Mammerickx, M.; Burny, A.; Kettmann, R. Attenuation of bovine leukemia virus by deletion of R3 and G4 open reading frames. Proc. Natl. Acad. Sci. USA 1994, 91, 11532–11536. [Google Scholar] [CrossRef]
  45. Reichert, M.; Cantor, G.H.; Willems, L.; Kettmann, R. Protective effects of a live attenuated bovine leukaemia virus vaccine with deletion in the R3 and G4 genes. J. Gen. Virol. 2000, 81, 965–969. [Google Scholar] [CrossRef]
  46. Zyrianova, I.M.; Kovalchuk, S.N. Bovine leukemia virus tax gene/Tax protein polymorphism and its relation to Enzootic Bovine Leukosis. Virulence 2020, 11, 80–87. [Google Scholar] [CrossRef]
  47. Willems, L.; Grimonpont, C.; Kerkhofs, P.; Capiau, C.; Gheysen, D.; Conrath, K.; Roussef, R.; Mamoun, R.; Portetelle, D.; Burny, A.; et al. Phosphorylation of bovine leukemia virus Tax protein is required for in vitro transformation but not for transactivation. Oncogene 1998, 16, 2165–2176. [Google Scholar] [CrossRef]
  48. Pyeon, D.; Splitter, G.A. Regulation of bovine leukemia virus tax and pol mRNA levels by interleukin-2 and -10. J. Virol. 1999, 73, 8427–8434. [Google Scholar] [CrossRef]
  49. Kerkhofs, P.; Heremans, H.; Burny, A.; Kettmann, R.; Willems, L. In vitro and in vivo oncogenic potential of bovine leukemia virus G4 protein. J. Virol. 1998, 72, 2554–2559. [Google Scholar] [CrossRef] [PubMed]
  50. Licursi, M.; Inoshima, Y.; Wu, D.; Yokoyama, T.; Gonzalez, E.T.; Sentsui, H. Provirus variants of bovine leukemia virus in naturally infected cattle from Argentina and Japan. Vet. Microbiol. 2003, 96, 17–23. [Google Scholar] [CrossRef]
  51. Lee, E.; Kim, E.J.; Joung, H.K.; Kim, B.H.; Song, J.Y.; Cho, I.S.; Lee, K.K.; Shin, Y.K. Sequencing and phylogenetic analysis of the gp51 gene from Korean bovine leukemia virus isolates. Virol. J. 2015, 12, 64. [Google Scholar] [CrossRef] [PubMed]
  52. Corredor-Figueroa, A.P.; Salas, S.; Olaya-Galan, N.N.; Quintero, J.S.; Fajardo, A.; Sonora, M.; Moreno, P.; Cristina, J.; Sanchez, A.; Tobon, J.; et al. Prevalence and molecular epidemiology of bovine leukemia virus in Colombian cattle. Infect. Genet. Evol. 2020, 80, 104171. [Google Scholar] [CrossRef]
  53. Marawan, M.A.; Mekata, H.; Hayashi, T.; Sekiguchi, S.; Kirino, Y.; Horii, Y.; Moustafa, A.M.; Arnaout, F.K.; Galila, E.; Norimine, J. Phylogenetic analysis of env gene of bovine leukemia virus strains spread in Miyazaki prefecture, Japan. J. Vet. Med. Sci. 2017, 79, 912–916. [Google Scholar] [CrossRef]
  54. Pluta, A.; Rola-Luszczak, M.; Kubis, P.; Balov, S.; Moskalik, R.; Choudhury, B.; Kuzmak, J. Molecular characterization of bovine leukemia virus from Moldovan dairy cattle. Arch. Virol. 2017, 162, 1563–1576. [Google Scholar] [CrossRef]
  55. Le, D.T.; Yamashita-Kawanishi, N.; Okamoto, M.; Nguyen, S.V.; Nguyen, N.H.; Sugiura, K.; Miura, T.; Haga, T. Detection and genotyping of bovine leukemia virus (BLV) in Vietnamese cattle. J. Vet. Med. Sci. 2020, 82, 1042–1050. [Google Scholar] [CrossRef]
  56. Polat, M.; Takeshima, S.N.; Hosomichi, K.; Kim, J.; Miyasaka, T.; Yamada, K.; Arainga, M.; Murakami, T.; Matsumoto, Y.; de la Barra, D.V.; et al. A new genotype of bovine leukemia virus in South America identified by NGS-based whole genome sequencing and molecular evolutionary genetic analysis. Retrovirology 2016, 13, 4. [Google Scholar] [CrossRef]
  57. Hamada, R.; Metwally, S.; Polat, M.; Borjigin, L.; Ali, A.O.; Abdel-Hady, A.; Mohamed, A.; Wada, S.; Aida, Y. Detection and Molecular Characterization of Bovine Leukemia Virus in Egyptian Dairy Cattle. Front. Vet. Sci. 2020, 7, 608. [Google Scholar] [CrossRef]
  58. Rodriguez, S.M.; Golemba, M.D.; Campos, R.H.; Trono, K.; Jones, L.R. Bovine leukemia virus can be classified into seven genotypes: Evidence for the existence of two novel clades. J. Gen. Virol. 2009, 90, 2788–2797. [Google Scholar] [CrossRef]
  59. Gautam, S.; Mishra, N.; Kalaiyarasu, S.; Jhade, S.K.; Sood, R. Molecular Characterization of Bovine Leukaemia Virus (BLV) Strains Reveals Existence of Genotype 6 in Cattle in India with evidence of a new subgenotype. Transbound. Emerg. Dis. 2018, 65, 1968–1978. [Google Scholar] [CrossRef]
  60. Lee, E.; Kim, E.J.; Ratthanophart, J.; Vitoonpong, R.; Kim, B.H.; Cho, I.S.; Song, J.Y.; Lee, K.K.; Shin, Y.K. Molecular epidemiological and serological studies of bovine leukemia virus (BLV) infection in Thailand cattle. Infect. Genet. Evol. 2016, 41, 245–254. [Google Scholar] [CrossRef]
  61. Selim, A.; Manaa, E.A.; Alanazi, A.D.; Alyousif, M.S. Seroprevalence, Risk Factors and Molecular Identification of Bovine Leukemia Virus in Egyptian Cattle. Animals 2021, 11, 319. [Google Scholar] [CrossRef]
  62. Polat, M.; Takeshima, S.N.; Aida, Y. Epidemiology and genetic diversity of bovine leukemia virus. Virol. J. 2017, 14, 209. [Google Scholar] [CrossRef]
  63. Kincaid, R.P.; Burke, J.M.; Sullivan, C.S. RNA virus microRNA that mimics a B-cell oncomiR. Proc. Natl. Acad. Sci. USA 2012, 109, 3077–3082. [Google Scholar] [CrossRef]
  64. Rosewick, N.; Momont, M.; Durkin, K.; Takeda, H.; Caiment, F.; Cleuter, Y.; Vernin, C.; Mortreux, F.; Wattel, E.; Burny, A.; et al. Deep sequencing reveals abundant noncanonical retroviral microRNAs in B-cell leukemia/lymphoma. Proc. Natl. Acad. Sci. USA 2013, 110, 2306–2311. [Google Scholar] [CrossRef]
  65. Gillet, N.A.; Hamaidia, M.; de Brogniez, A.; Gutierrez, G.; Renotte, N.; Reichert, M.; Trono, K.; Willems, L. Bovine Leukemia Virus Small Noncoding RNAs Are Functional Elements That Regulate Replication and Contribute to Oncogenesis In Vivo. PLoS Pathog. 2016, 12, e1005588. [Google Scholar] [CrossRef]
  66. Santanam, U.; Zanesi, N.; Efanov, A.; Costinean, S.; Palamarchuk, A.; Hagan, J.P.; Volinia, S.; Alder, H.; Rassenti, L.; Kipps, T.; et al. Chronic lymphocytic leukemia modeled in mouse by targeted miR-29 expression. Proc. Natl. Acad. Sci. USA 2010, 107, 12210–12215. [Google Scholar] [CrossRef]
  67. Petersen, M.I.; Carignano, H.A.; Mongini, C.; Gonzalez, D.D.; Jaworski, J.P. Bovine leukemia virus encoded blv-miR-b4-3p microRNA is associated with reduced expression of anti-oncogenic gene in vivo. PLoS ONE 2023, 18, e281317. [Google Scholar] [CrossRef]
  68. Leisering, A. Hypertrophy der Malpigischen Korperchen der Milz. Ber. Uber Das Vet. Im Konigreich Sachs. 1871, 16, 15–16. [Google Scholar]
  69. Olson, C.; Miller, J. Enzootic Bovine Leukosis and Bovine Leukemia Virus; Martinus Nijhoff Publishing: Boston, MA, USA, 1987. [Google Scholar]
  70. Bollinger, O. ber Leukmie bei den Haustieren. Virchows Arch. 1874, 59, 4. [Google Scholar]
  71. Siedamgrotzky, O.; Hofmeister, V. Anleitung zur Mikroskopischen und Chemischen Diagnostik der Krankheiten der Hausthiere: Für Thierärzte und Landwirte; Schönfeld VIII: Dresden, Germany, 1876; p. 192. [Google Scholar]
  72. Miller, J.M.; Miller, L.D.; Olson, C.; Gillette, K.G. Virus-like particles in phytohemagglutinin-stimulated lymphocyte cultures with reference to bovine lymphosarcoma. JNCI-J. Natl. Cancer Inst. 1969, 43, 1297–1305. [Google Scholar]
  73. Bartlett, P.C.; Sordillo, L.M.; Byrem, T.M.; Norby, B.; Grooms, D.L.; Swenson, C.L.; Zalucha, J.; Erskine, R.J. Options for the control of bovine leukemia virus in dairy cattle. JAVMA-J. Am. Vet. Med. Assoc. 2014, 244, 914–922. [Google Scholar] [CrossRef]
  74. Whittington, R.; Donat, K.; Weber, M.F.; Kelton, D.; Nielsen, S.S.; Eisenberg, S.; Arrigoni, N.; Juste, R.; Saez, J.L.; Dhand, N.; et al. Control of paratuberculosis: Who, why and how. A review of 48 countries. BMC Vet. Res. 2019, 15, 198. [Google Scholar] [CrossRef]
  75. Ruggiero, V.J.; Bartlett, P.C. Control of Bovine Leukemia Virus in Three US Dairy Herds by Culling ELISA-Positive Cows. Vet. Med. Int. 2019, 2019, 3202184. [Google Scholar] [CrossRef]
  76. More, S.; Botner, A.; Butterworth, A.; Calistri, P.; Depner, K.; Edwards, S.; Garin-Bastuji, B.; Good, M.; Gortazar, S.C.; Michel, V.; et al. Assessment of listing and categorisation of animal diseases within the framework of the Animal Health Law (Regulation (EU) No 2016/429): Enzootic bovine leukosis (EBL). EFSA J. 2017, 15, e4956. [Google Scholar] [CrossRef]
  77. WOAH. WOAH-WAHIS. Available online: https://wahis.woah.org/#/dashboards/country-or-disease-dashboard (accessed on 28 December 2023).
  78. OIE. World Animal Health Infromation Database-Version: 1.4. In World Animal Health Information Database; OIE: Paris, France, 2009. [Google Scholar]
  79. Commission Implementing Regulation (EU) 2021/620 of 15 April 2021 Laying Down Rules for the Application of Regulation (EU) 2016/429 of the European Parliament and of the Council as Regards the Approval of the Disease-Free and Non-Vaccination Status of Certain Member States or Zones or Compartments Thereof as Regards Certain Listed Diseases and the Approval of Eradication Programmes for Those Listed Diseases. Available online: http://data.europa.eu/eli/reg_impl/2021/620/oj (accessed on 28 December 2023).
  80. Nekouei, O.; Vanleeuwen, J.; Stryhn, H.; Kelton, D.; Keefe, G. Lifetime effects of infection with bovine leukemia virus on longevity and milk production of dairy cows. Prev. Vet. Med. 2016, 133, 1–9. [Google Scholar] [CrossRef]
  81. John, E.E.; Keefe, G.; Cameron, M.; Stryhn, H.; Mcclure, J.T. Development and implementation of a risk assessment and management program for enzootic bovine leukosis in Atlantic Canada. J. Dairy Sci. 2020, 103, 8398–8406. [Google Scholar] [CrossRef]
  82. Suzan, V.M.; Onuma, M.; Aguilar, R.E.; Murakami, Y. Prevalence of bovine herpesvirus-1, parainfluenza-3, bovine rotavirus, bovine viral diarrhea, bovine adenovirus-7, bovine leukemia virus and bluetongue virus antibodies in cattle in Mexico. JPN J. Vet. Res. 1983, 31, 125–132. [Google Scholar]
  83. Gonzalez, M.A.; Ceron-Tellez, F.; Sarmiento, S.R.; Tortora, P.J.; Rojas-Anaya, E.; Alvarez, H.R. Presence of co-infection between bovine leukemia virus and bovine herpesvirus 1 in herds vaccinated against bovine respiratory complex. Can. J. Vet. Res.-Rev. Can. Rech. Vet. 2023, 87, 105–109. [Google Scholar]
  84. Moratorio, G.; Obal, G.; Dubra, A.; Correa, A.; Bianchi, S.; Buschiazzo, A.; Cristina, J.; Pritsch, O. Phylogenetic analysis of bovine leukemia viruses isolated in South America reveals diversification in seven distinct genotypes. Arch. Virol. 2010, 155, 481–489. [Google Scholar] [CrossRef]
  85. Zinovieva, N.A.; Vinogradova, I.V.; Mikhailova, M.E.; Molofeeva, L.A.; Ernst, L.K. Prevalence of bovine leukemia virus in black and white cows with the different level of milk productive traits. Сельскoхoзяйственная биoлoгия 2012, 6, 49–55. [Google Scholar] [CrossRef]
  86. Trono, K.G.; Perez-Filgueira, D.M.; Duffy, S.; Borca, M.V.; Carrillo, C. Seroprevalence of bovine leukemia virus in dairy cattle in Argentina: Comparison of sensitivity and specificity of different detection methods. Vet. Microbiol. 2001, 83, 235–248. [Google Scholar] [CrossRef] [PubMed]
  87. Gutierrez, G.; Alvarez, I.; Politzki, R.; Lomonaco, M.; Dus, S.M.; Rondelli, F.; Fondevila, N.; Trono, K. Natural progression of Bovine Leukemia Virus infection in Argentinean dairy cattle. Vet. Microbiol. 2011, 151, 255–263. [Google Scholar] [CrossRef]
  88. Samara, S.I.; Lima, E.G.; Do Nascimento, A.A. Monitoring of enzootic bovine leukosis in dairy cattle from the Pitangueiras region in São Paulo, Brazil. Braz. J. Vet. Res. Anim. Sci. 1997, 34, 349–351. [Google Scholar] [CrossRef]
  89. D’Angelino, J.L.; Garcia, M.; Birgel, E.H. Epidemiological study of enzootic bovine leukosis in Brazil. Trop. Anim. Health Prod. 1998, 30, 13–15. [Google Scholar] [CrossRef]
  90. Ramalho, G.C.; Silva, M.L.C.R.; Falcão, B.M.R.; Limeira, C.H.; Nogueira, D.B.; Dos Santos, A.M.; Martins, C.M.; Alves, C.J.; Clementino, I.J.; Santos, C.D.S.A. High herd-level seroprevalence and associated factors for bovine leukemia virus in the semi-arid Paraíba state, Northeast Region of Brazil. Prev. Vet. Med. 2021, 190, 105324. [Google Scholar] [CrossRef] [PubMed]
  91. Bonifaz, N.; Ulcuango, F. Prevalencia de leucosis bovina en la comunidad Santo Domingo n° 1, Cayambe-Ecuador 2012. La Granja Rev. De Cienc. De La Vida 2015, 22, 33–39. [Google Scholar]
  92. Ch, A.H. Bovine leukaemia virus infection in Peru. Trop. Anim. Health Prod. 1983, 15, 61. [Google Scholar] [CrossRef]
  93. Marin, C.; de Lopez, N.M.; Alvarez, L.; Lozano, O.; Espana, W.; Castanos, H.; Leon, A. Epidemiology of bovine leukemia in Venezuela. Ann. Rech. Vet. 1978, 9, 743–746. [Google Scholar]
  94. Nava, Z.; Obando, C.; Molina, M.; Bracamonte, M.; Tkachuk, O. Seroprevalence of enzootic bovine leukosis and its association with clinical signs and risk factors in dairy herds from Barinas State, Venezuela. Rev. De La Fac. De Cienc. Vet. 2011, 52, 13–23. [Google Scholar]
  95. Furtado, A.; Rosadilla, D.; Franco, G.; Piaggio, J.; Puentes, R. Leucosis Bovina Enzoótica en cuencas lecheras de productores familiares del Uruguay. Veterinaria 2013, 49, 29–37. [Google Scholar]
  96. Yang, Y.; Fan, W.; Mao, Y.; Yang, Z.; Lu, G.; Zhang, R.; Zhang, H.; Szeto, C.; Wang, C. Bovine leukemia virus infection in cattle of China: Association with reduced milk production and increased somatic cell score. J. Dairy Sci. 2016, 99, 3688–3697. [Google Scholar] [CrossRef]
  97. Meas, S.; Ohashi, K.; Tum, S.; Chhin, M.; Te, K.; Miura, K.; Sugimoto, C.; Onuma, M. Seroprevalence of bovine immunodeficiency virus and bovine leukemia virus in draught animals in Cambodia. J. Vet. Med. Sci. 2000, 62, 779–781. [Google Scholar] [CrossRef] [PubMed]
  98. Nekouei, O.; Stryhn, H.; Vanleeuwen, J.; Kelton, D.; Hanna, P.; Keefe, G. Predicting within-herd prevalence of infection with bovine leukemia virus using bulk-tank milk antibody levels. Prev. Vet. Med. 2015, 122, 53–60. [Google Scholar] [CrossRef] [PubMed]
  99. Kobayashi, S.; Hidano, A.; Tsutsui, T.; Yamamoto, T.; Hayama, Y.; Nishida, T.; Muroga, N.; Konishi, M.; Kameyama, K.; Murakami, K. Analysis of risk factors associated with bovine leukemia virus seropositivity within dairy and beef breeding farms in Japan: A nationwide survey. Res. Vet. Sci. 2014, 96, 47–53. [Google Scholar] [CrossRef] [PubMed]
  100. Ochirkhuu, N.; Konnai, S.; Odbileg, R.; Nishimori, A.; Okagawa, T.; Murata, S.; Ohashi, K. Detection of bovine leukemia virus and identification of its genotype in Mongolian cattle. Arch. Virol. 2016, 161, 985–991. [Google Scholar] [CrossRef]
  101. Polat, M.; Ohno, A.; Takeshima, S.N.; Kim, J.; Kikuya, M.; Matsumoto, Y.; Mingala, C.N.; Onuma, M.; Aida, Y. Detection and molecular characterization of bovine leukemia virus in Philippine cattle. Arch. Virol. 2015, 160, 285–296. [Google Scholar] [CrossRef] [PubMed]
  102. Khan, M.F.; Siddique, U.; Shah, A.A.; Khan, I.; Anwar, F.; Ahmad, I.; Zeb, M.T.; Hassan, M.F.; Ali, T. Seroprevalence of bovine leukemia virus (BLV) in cattle from the North West of Pakistan. Pak. Vet. J 2019, 40, 127–129. [Google Scholar]
  103. Hsieh, J.C.; Li, C.Y.; Hsu, W.L.; Chuang, S.T. Molecular Epidemiological and Serological Studies of Bovine Leukemia Virus in Taiwan Dairy Cattle. Front. Vet. Sci. 2019, 6, 427. [Google Scholar] [CrossRef]
  104. Trainin, Z.; Brenner, J. The direct and indirect economic impacts of bovine leukemia virus infection on dairy cattle. Isr. J. Vet. Med. 2005, 60, 94. [Google Scholar]
  105. Hafez, S.M.; Sharif, M.; Al-Sukayran, A.; Dela-Cruz, D. Preliminary studies on enzootic bovine leukosis in Saudi dairy farms. DTW Dtsch. Tierarztl. Wochenschr. 1990, 97, 61–63. [Google Scholar]
  106. Burgu, I.; Alkan, F.; Karaoglu, T.; Bilge-Dagalp, S.; Can-Sahna, K.; Gungor, B.; Demir, B. Control and eradication programme of enzootic bovine leucosis (EBL) from selected dairy herds in Turkey. Dtsch. Tierarztl. Wochenschr. 2005, 112, 271–274. [Google Scholar]
  107. Khudhair, Y.I.; Hasso, S.A.; Yaseen, N.Y.; Al-Shammari, A.M. Serological and molecular detection of bovine leukemia virus in cattle in Iraq. Emerg. Microbes Infect. 2016, 5, e56. [Google Scholar] [CrossRef] [PubMed]
  108. Selim, A.; Megahed, A.A.; Kandeel, S.; Abdelhady, A. Risk factor analysis of bovine leukemia virus infection in dairy cattle in Egypt. Comp. Immunol. Microbiol. Infect. Dis. 2020, 72, 101517. [Google Scholar] [CrossRef] [PubMed]
  109. Lewin, H.A.; Bernoco, D. Evidence for BoLA-linked resistance and susceptibility to subclinical progression of bovine leukaemia virus infection. Anim. Genet. 1986, 17, 197–207. [Google Scholar] [CrossRef] [PubMed]
  110. Eguchi, M.; Kawamura, H.; Wada, F.; Shimauchi, T.; Shiota, E.; Shibata, K.; Sugioka, Y. Promotion of calcification by imidazole and its suppression by diltiazem in the growth cartilage of rats with HEBP induced rickets. Int. Orthop. 1989, 13, 217–220. [Google Scholar] [CrossRef] [PubMed]
  111. Porta, N.G.; Alvarez, I.; Suarez, A.G.; Ruiz, V.; Abdala, A.; Trono, K. Experimental infection of sheep with Bovine leukemia virus (BLV): Minimum dose of BLV-FLK cells and cell-free BLV and neutralization activity of natural antibodies. Rev. Argent. Microbiol. 2019, 51, 316–323. [Google Scholar] [CrossRef]
  112. Abdalla, E.A.; Weigel, K.A.; Byrem, T.M.; Rosa, G. Genetic correlation of bovine leukosis incidence with somatic cell score and milk yield in a US Holstein population. J. Dairy Sci. 2016, 99, 2005–2009. [Google Scholar] [CrossRef] [PubMed]
  113. Lo, C.W.; Borjigin, L.; Saito, S.; Fukunaga, K.; Saitou, E.; Okazaki, K.; Mizutani, T.; Wada, S.; Takeshima, S.N.; Aida, Y. BoLA-DRB3 Polymorphism is Associated with Differential Susceptibility to Bovine Leukemia Virus-Induced Lymphoma and Proviral Load. Viruses 2020, 12, 352. [Google Scholar] [CrossRef] [PubMed]
  114. Le, T.D.; Nguyen, V.S.; Lo, C.W.; Dao, D.T.; Bui, N.V.; Ogawa, H.; Imai, K.; Sugiura, K.; Aida, Y.; Haga, T. Association between BoLA-DRB3 polymorphism and bovine leukemia virus proviral load in Vietnamese Holstein Friesian cattle. HLA 2022, 99, 105–112. [Google Scholar] [CrossRef]
  115. Maezawa, M.; Fujii, Y.; Akagami, M.; Kawakami, J.; Inokuma, H. BoLA-DRB3*15:01 allele is associated with susceptibility to early enzootic bovine leukosis onset in Holstein-Friesian and Japanese Black cattle. Vet. Microbiol. 2023, 284, 109829. [Google Scholar] [CrossRef]
  116. Daous, H.E.; Mitoma, S.; Elhanafy, E.; Thi, N.H.; Thi, M.N.; Notsu, K.; Kaneko, C.; Norimine, J.; Sekiguchi, S. Relationship between Allelic Heterozygosity in BoLA-DRB3 and Proviral Loads in Bovine Leukemia Virus-Infected Cattle. Animals 2021, 11, 647. [Google Scholar] [CrossRef]
  117. Bauermann, F.V.; Ridpath, J.F.; Dargatz, D.A. Bovine leukemia virus seroprevalence among cattle presented for slaughter in the United States. J. Vet. Diagn. Investig. 2017, 29, 704–706. [Google Scholar] [CrossRef] [PubMed]
  118. Murakami, K.; Kobayashi, S.; Konishi, M.; Kameyama, K.; Yamamoto, T.; Tsutsui, T. The recent prevalence of bovine leukemia virus (BLV) infection among Japanese cattle. Vet. Microbiol. 2011, 148, 84–88. [Google Scholar] [CrossRef]
  119. Oguma, K.; Suzuki, M.; Sentsui, H. Enzootic bovine leukosis in a two-month-old calf. Virus Res. 2017, 233, 120–124. [Google Scholar] [CrossRef]
  120. Shaghayegh, A. Detection and Identification of Enzootic Bovine Leukosis (EBL) in Calves in Iran. Arch. Razi Inst. 2019, 74, 321–325. [Google Scholar] [CrossRef] [PubMed]
  121. Watanuki, S.; Takeshima, S.N.; Borjigin, L.; Sato, H.; Bai, L.; Murakami, H.; Sato, R.; Ishizaki, H.; Matsumoto, Y.; Aida, Y. Visualizing bovine leukemia virus (BLV)-infected cells and measuring BLV proviral loads in the milk of BLV seropositive dams. Vet. Res. 2019, 50, 102. [Google Scholar] [CrossRef] [PubMed]
  122. Kobayashi, S.; Tsutsui, T.; Yamamoto, T.; Hayama, Y.; Muroga, N.; Konishi, M.; Kameyama, K.; Murakami, K. The role of neighboring infected cattle in bovine leukemia virus transmission risk. J. Vet. Med. Sci. 2015, 77, 861–863. [Google Scholar] [CrossRef]
  123. Yuan, Y.; Kitamura-Muramatsu, Y.; Saito, S.; Ishizaki, H.; Nakano, M.; Haga, S.; Matoba, K.; Ohno, A.; Murakami, H.; Takeshima, S.N.; et al. Detection of the BLV provirus from nasal secretion and saliva samples using BLV-CoCoMo-qPCR-2: Comparison with blood samples from the same cattle. Virus Res. 2015, 210, 248–254. [Google Scholar] [CrossRef]
  124. Haga, T. Enzootic Bovine Leukosis: How to prevent the disease and control the spread of BLV infection. In Proceedings of the 1st International Conference Postgraduate School Universitas Airlangga: “Implementation of Climate Change Agreement to Meet Sustainable Development Goals” (ICPSUAS 2017), Surabaya, Indonesia, 1–2 August 2017; pp. 13–14. [Google Scholar]
  125. Mekata, H.; Sekiguchi, S.; Konnai, S.; Kirino, Y.; Honkawa, K.; Nonaka, N.; Horii, Y.; Norimine, J. Evaluation of the natural perinatal transmission of bovine leukaemia virus. Vet. Rec. 2015, 176, 254. [Google Scholar] [CrossRef]
  126. Kohara, J.; Takeuchi, M.; Hirano, Y.; Sakurai, Y.; Takahashi, T. Vector control efficacy of fly nets on preventing bovine leukemia virus transmission. J. Vet. Med. Sci. 2018, 80, 1524–1527. [Google Scholar] [CrossRef]
  127. Selim, A.; Ali, A.F. Seroprevalence and risk factors for C. burentii infection in camels in Egypt. Comp. Immunol. Microbiol. Infect. Dis. 2020, 68, 101402. [Google Scholar] [CrossRef]
  128. Selim, A.; Radwan, A.; Arnaout, F. Seroprevalence and molecular characterization of West Nile Virus in Egypt. Comp. Immunol. Microbiol. Infect. Dis. 2020, 71, 101473. [Google Scholar] [CrossRef]
  129. Mekata, H.; Yamamoto, M.; Hayashi, T.; Kirino, Y.; Sekiguchi, S.; Konnai, S.; Horii, Y.; Norimine, J. Cattle with a low bovine leukemia virus proviral load are rarely an infectious source. JPN J. Vet. Res. 2018, 66, 157–163. [Google Scholar]
  130. Konishi, M.; Ishizaki, H.; Kameyama, K.I.; Murakami, K.; Yamamoto, T. The effectiveness of colostral antibodies for preventing bovine leukemia virus (BLV) infection in vitro. BMC Vet. Res. 2018, 14, 419. [Google Scholar] [CrossRef] [PubMed]
  131. Ott, S.L.; Johnson, R.; Wells, S.J. Association between bovine-leukosis virus seroprevalence and herd-level productivity on US dairy farms. Prev. Vet. Med. 2003, 61, 249–262. [Google Scholar] [CrossRef] [PubMed]
  132. Erskine, R.J.; Bartlett, P.C.; Byrem, T.M.; Render, C.L.; Febvay, C.; Houseman, J.T. Herd-level determinants of bovine leukaemia virus prevalence in dairy farms. J. Dairy Res. 2012, 79, 445–450. [Google Scholar] [CrossRef]
  133. Benitez, O.J.; Roberts, J.N.; Norby, B.; Bartlett, P.C.; Takeshima, S.N.; Watanuki, S.; Aida, Y.; Grooms, D.L. Breeding bulls as a potential source of bovine leukemia virus transmission in beef herds. JAVMA-J. Am. Vet. Med. Assoc. 2019, 254, 1335–1340. [Google Scholar] [CrossRef]
  134. Alvarez, I.; Gutierrez, G.; Gammella, M.; Martinez, C.; Politzki, R.; Gonzalez, C.; Caviglia, L.; Carignano, H.; Fondevila, N.; Poli, M.; et al. Evaluation of total white blood cell count as a marker for proviral load of bovine leukemia virus in dairy cattle from herds with a high seroprevalence of antibodies against bovine leukemia virus. Am. J. Vet. Res. 2013, 74, 744–749. [Google Scholar] [CrossRef] [PubMed]
  135. Schwartz, I.; Levy, D. Pathobiology of bovine leukemia virus. Vet. Res. 1994, 25, 521–536. [Google Scholar]
  136. Willems, L.; Burny, A.; Collete, D.; Dangoisse, O.; Dequiedt, F.; Gatot, J.S.; Kerkhofs, P.; Lefebvre, L.; Merezak, C.; Peremans, T.; et al. Genetic determinants of bovine leukemia virus pathogenesis. Aids Res. Hum. Retrovir. 2000, 16, 1787–1795. [Google Scholar] [CrossRef]
  137. Zaghawa, A.; Beier, D.; Abd El Rahim, I.; El Ballal, S.; Karim, I.; Conraths, F.J.; Marquardt, O. An outbreak of enzootic bovine leukosis in upper egypt: Clinical, laboratory and molecular–epidemiological studies. J. Vet. Med. Ser. B 2002, 49, 123–129. [Google Scholar] [CrossRef]
  138. Zaher, K.S.; Ahmed, W.M. Bovine leukemia virus infection in dairy cows in Egypt. Acad. J. Cancer Res. 2014, 7, 126–130. [Google Scholar]
  139. Uchiyama, J.; Murakami, H.; Sato, R.; Mizukami, K.; Suzuki, T.; Shima, A.; Ishihara, G.; Sogawa, K.; Sakaguchi, M. Examination of the fecal microbiota in dairy cows infected with bovine leukemia virus. Vet. Microbiol. 2020, 240, 108547. [Google Scholar] [CrossRef] [PubMed]
  140. Blagitz, M.G.; Souza, F.N.; Batista, C.F.; Azevedo, L.; Sanchez, E.; Diniz, S.A.; Silva, M.X.; Haddad, J.P.; Della, L.A. Immunological implications of bovine leukemia virus infection. Res. Vet. Sci. 2017, 114, 109–116. [Google Scholar] [CrossRef]
  141. Patel, J.R.; Heldens, J.G.; Bakonyi, T.; Rusvai, M. Important mammalian veterinary viral immunodiseases and their control. Vaccine 2012, 30, 1767–1781. [Google Scholar] [CrossRef]
  142. Bai, L.; Yokoyama, K.; Watanuki, S.; Ishizaki, H.; Takeshima, S.N.; Aida, Y. Development of a new recombinant p24 ELISA system for diagnosis of bovine leukemia virus in serum and milk. Arch. Virol. 2019, 164, 201–211. [Google Scholar] [CrossRef] [PubMed]
  143. Gutierrez, G.; Alvarez, I.; Fondevila, N.; Politzki, R.; Lomonaco, M.; Rodriguez, S.; Dus, S.M.; Trono, K. Detection of bovine leukemia virus specific antibodies using recombinant p24-ELISA. Vet. Microbiol. 2009, 137, 224–234. [Google Scholar] [CrossRef]
  144. Kuczewski, A.; Orsel, K.; Barkema, H.W.; Kelton, D.F.; Hutchins, W.A.; van der Meer, F. Short communication: Evaluation of 5 different ELISA for the detection of bovine leukemia virus antibodies. J. Dairy Sci. 2018, 101, 2433–2437. [Google Scholar] [CrossRef]
  145. Endoh, D.; Mizutani, T.; Kirisawa, R.; Maki, Y.; Saito, H.; Kon, Y.; Morikawa, S.; Hayashi, M. Species-independent detection of RNA virus by representational difference analysis using non-ribosomal hexanucleotides for reverse transcription. Nucleic Acids Res. 2005, 33, e65. [Google Scholar] [CrossRef]
  146. Heinecke, N.; Tortora, J.; Martinez, H.A.; Gonzalez-Fernandez, V.D.; Ramirez, H. Detection and genotyping of bovine leukemia virus in Mexican cattle. Arch. Virol. 2017, 162, 3191–3196. [Google Scholar] [CrossRef]
  147. Bannenberg, T. Experiments using a modified disposition arrangement of the AGID test in the diagnosis of enzootic bovine leukosis. Zentralbl Vet. B 1982, 29, 676–680. [Google Scholar] [CrossRef]
  148. Martin, D.; Arjona, A.; Soto, I.; Barquero, N.; Viana, M.; Gomez-Lucia, E. Comparative study of PCR as a direct assay and ELISA and AGID as indirect assays for the detection of bovine leukaemia virus. J. Vet. Med. Ser. B 2001, 48, 97–106. [Google Scholar] [CrossRef]
  149. Buzala, E.; Deren, W. Comparison of PLA with AGID and ELISA results in serology diagnosis of bovine leukosis. Pol. J. Vet. Sci. 2003, 6, 9–11. [Google Scholar]
  150. Swart, K.; Hagemeijer, A.; Lowenberg, B. Density profiles and purification of chronic myeloid leukemia cells forming colonies in the PHA-leukocyte feeder assay. Exp. Hematol. 1981, 9, 588–594. [Google Scholar]
  151. World Organisation for Animal Health. Chapter 3.4.9 Enzootic bovine leukosis. In Manual of Diagnostic Tests and Vaccines for Terrestrial Animals; World Organisation for Animal Health: Paris, France, 2018; ISBN 978-92-95108-18-9. [Google Scholar]
  152. Monti, G.E.; Frankena, K.; Engel, B.; Buist, W.; Tarabla, H.D.; de Jong, M.C. Evaluation of a new antibody-based enzyme-linked immunosorbent assay for the detection of bovine leukemia virus infection in dairy cattle. J. Vet. Diagn. Investig. 2005, 17, 451–457. [Google Scholar] [CrossRef] [PubMed]
  153. Jimba, M.; Takeshima, S.; Murakami, H.; Kohara, J.; Kobayashi, N.; Matsuhashi, T.; Ohmori, T.; Nunoya, T.; Aida, Y. BLV-CoCoMo-qPCR: A useful tool for evaluating bovine leukemia virus infection status. BMC Vet. Res. 2012, 8, 167. [Google Scholar] [CrossRef] [PubMed]
  154. Levy, D.; Deshayes, L.; Parodi, A.L.; Levy, J.P.; Stephenson, J.R.; Devare, S.G.; Gilden, R.V. Bovine leukemia virus specific antibodies among French cattle. II. Radioimmunoassay with the major structural protein (BLV p24). Int. J. Cancer 1977, 20, 543–550. [Google Scholar] [CrossRef]
  155. Lew, A.E.; Bock, R.E.; Miles, J.; Cuttell, L.B.; Steer, P.; Nadin-Davis, S.A. Sensitive and specific detection of bovine immunodeficiency virus and bovine syncytial virus by 5′ Taq nuclease assays with fluorescent 3′ minor groove binder-DNA probes. J. Virol. Methods 2004, 116, 1–9. [Google Scholar] [CrossRef] [PubMed]
  156. Juliarena, M.A.; Gutierrez, S.E.; Ceriani, C. Determination of proviral load in bovine leukemia virus-infected cattle with and without lymphocytosis. Am. J. Vet. Res. 2007, 68, 1220–1225. [Google Scholar] [CrossRef]
  157. Lew, A.E.; Bock, R.E.; Molloy, J.B.; Minchin, C.M.; Robinson, S.J.; Steer, P. Sensitive and specific detection of proviral bovine leukemia virus by 5′ Taq nuclease PCR using a 3′ minor groove binder fluorogenic probe. J. Virol. Methods 2004, 115, 167–175. [Google Scholar] [CrossRef]
  158. Evermann, J.F.; Jackson, M.K. Laboratory diagnostic tests for retroviral infections in dairy and beef cattle. Vet. Clin. N. Am. Food Anim. Pract. 1997, 13, 87–106. [Google Scholar] [CrossRef]
  159. Wu, X.; Notsu, K.; Matsuura, Y.; Mitoma, S.; El, D.H.; Norimine, J.; Sekiguchi, S. Development of droplet digital PCR for quantification of bovine leukemia virus proviral load using unpurified genomic DNA. J. Virol. Methods 2023, 315, 114706, Erratum in J. Virol. Methods 2023, 315, 114708. [Google Scholar] [CrossRef]
  160. De Brun, M.L.; Cosme, B.; Petersen, M.; Alvarez, I.; Folgueras-Flatschart, A.; Flatschart, R.; Panei, C.J.; Puentes, R. Development of a droplet digital PCR assay for quantification of the proviral load of bovine leukemia virus. J. Vet. Diagn. Investig. 2022, 34, 439–447. [Google Scholar] [CrossRef] [PubMed]
  161. Nishimori, A.; Konnai, S.; Ikebuchi, R.; Okagawa, T.; Nakahara, A.; Murata, S.; Ohashi, K. Direct polymerase chain reaction from blood and tissue samples for rapid diagnosis of bovine leukemia virus infection. J. Vet. Med. Sci. 2016, 78, 791–796. [Google Scholar] [CrossRef] [PubMed]
  162. Takeshima, S.; Kitamura-Muramatsu, Y.; Yuan, Y.; Polat, M.; Saito, S.; Aida, Y. BLV-CoCoMo-qPCR-2: Improvements to the BLV-CoCoMo-qPCR assay for bovine leukemia virus by reducing primer degeneracy and constructing an optimal standard curve. Arch. Virol. 2015, 160, 1325–1332. [Google Scholar] [CrossRef]
  163. Jimba, M.; Takeshima, S.N.; Matoba, K.; Endoh, D.; Aida, Y. BLV-CoCoMo-qPCR: Quantitation of bovine leukemia virus proviral load using the CoCoMo algorithm. Retrovirology 2010, 7, 91. [Google Scholar] [CrossRef] [PubMed]
  164. Panei, C.J.; Takeshima, S.; Omori, T.; Nunoya, T.; Davis, W.C.; Ishizaki, H.; Matoba, K.; Aida, Y. Estimation of bovine leukemia virus (BLV) proviral load harbored by lymphocyte subpopulations in BLV-infected cattle at the subclinical stage of enzootic bovine leucosis using BLV-CoCoMo-qPCR. BMC Vet. Res. 2013, 9, 95. [Google Scholar] [CrossRef] [PubMed]
  165. Johnson, M.; Rommel, F.; Mone, J. Development of a syncytia inhibition assay for the detection of antibodies to bovine leukemia virus in naturally infected cattle; comparison with Western blot and agar gel immunodiffusion. J. Virol. Methods 1998, 70, 177–182. [Google Scholar] [CrossRef]
  166. Trainin, Z.; Meirom, R.; Gluckmann, A. Comparison between the immunodiffusion and the immunofluorescence tests in the diagnosis of bovine leukemia virus (BLV). Ann. Rech. Vet. 1978, 9, 659–662. [Google Scholar]
  167. Sato, H.; Bai, L.; Borjigin, L.; Aida, Y. Overexpression of bovine leukemia virus receptor SLC7A1/CAT1 enhances cellular susceptibility to BLV infection on luminescence syncytium induction assay (LuSIA). Virol. J. 2020, 17, 57. [Google Scholar] [CrossRef]
  168. Sato, H.; Watanuki, S.; Murakami, H.; Sato, R.; Ishizaki, H.; Aida, Y. Development of a luminescence syncytium induction assay (LuSIA) for easily detecting and quantitatively measuring bovine leukemia virus infection. Arch. Virol. 2018, 163, 1519–1530. [Google Scholar] [CrossRef] [PubMed]
  169. Sato, H.; Watanuki, S.; Bai, L.; Borjigin, L.; Ishizaki, H.; Matsumoto, Y.; Hachiya, Y.; Sentsui, H.; Aida, Y. A sensitive luminescence syncytium induction assay (LuSIA) based on a reporter plasmid containing a mutation in the glucocorticoid response element in the long terminal repeat U3 region of bovine leukemia virus. Virol. J. 2019, 16, 66. [Google Scholar] [CrossRef]
  170. Sato, H.; Fukui, J.N.; Hirano, H.; Osada, H.; Arimura, Y.; Masuda, M.; Aida, Y. Application of the Luminescence Syncytium Induction Assay to Identify Chemical Compounds That Inhibit Bovine Leukemia Virus Replication. Viruses 2022, 15, 4. [Google Scholar] [CrossRef] [PubMed]
  171. Tajima, S.; Ikawa, Y.; Aida, Y. Complete bovine leukemia virus (BLV) provirus is conserved in BLV-infected cattle throughout the course of B-cell lymphosarcoma development. J. Virol. 1998, 72, 7569–7576. [Google Scholar] [CrossRef]
  172. Takeshima, S.; Watanuki, S.; Ishizaki, H.; Matoba, K.; Aida, Y. Development of a direct blood-based PCR system to detect BLV provirus using CoCoMo primers. Arch. Virol. 2016, 161, 1539–1546. [Google Scholar] [CrossRef]
  173. Casoli, C.; Pilotti, E.; Bertazzoni, U. Proviral load determination of HTLV-1 and HTLV-2 in patients’ peripheral blood mononuclear cells by real-time PCR. Methods Mol. Biol. 2014, 1087, 315–323. [Google Scholar] [CrossRef]
  174. Pineda, M.V.; Bouzas, M.B.; Remesar, M.; Fridman, A.; Remondegui, C.; Mammana, L.; Altamirano, N.; Paradiso, P.; Costantini, P.; Tadey, L.; et al. Relevance of HTLV-1 proviral load in asymptomatic and symptomatic patients living in endemic and non-endemic areas of Argentina. PLoS ONE 2019, 14, e225596. [Google Scholar] [CrossRef]
  175. Rodrigues, E.S.; Salustiano, S.; Santos, E.V.; Slavov, S.N.; Picanco-Castro, V.; Maconetto, J.M.; de Haes, T.M.; Takayanagui, O.M.; Covas, D.T.; Kashima, S. Monitoring of HTLV-1-associated diseases by proviral load quantification using multiplex real-time PCR. J. Neurovirol. 2022, 28, 27–34. [Google Scholar] [CrossRef]
  176. Torti, C.; Quiros-Roldan, M.E.; Cologni, G.; Nichelatti, M.; Ceresoli, F.; Pinti, M.; Nasi, M.; Cossarizza, A.; Lapadula, G.; Costarelli, S.; et al. Plasma HIV load and proviral DNA decreases after two standard antiretroviral regimens in HIV-positive patients naive to antiretrovirals. Curr. HIV Res. 2008, 6, 43–48. [Google Scholar] [CrossRef]
  177. Bruisten, S.M.; Reiss, P.; Loeliger, A.E.; van Swieten, P.; Schuurman, R.; Boucher, C.A.; Weverling, G.J.; Huisman, J.G. Cellular proviral HIV type 1 DNA load persists after long-term RT-inhibitor therapy in HIV type 1 infected persons. AIDS Res. Hum. Retrovir. 1998, 14, 1053–1058. [Google Scholar] [CrossRef]
  178. Watanabe, A.; Murakami, H.; Kakinuma, S.; Murao, K.; Ohmae, K.; Isobe, N.; Akamatsu, H.; Seto, T.; Hashimura, S.; Konda, K.; et al. Association between bovine leukemia virus proviral load and severity of clinical mastitis. J. Vet. Med. Sci. 2019, 81, 1431–1437. [Google Scholar] [CrossRef] [PubMed]
  179. Marin-Flamand, E.; Araiza-Hernandez, D.M.; Vargas-Ruiz, A.; Rangel-Rodriguez, I.C.; Gonzalez-Tapia, L.A.; Ramirez-Alvarez, H.; Hernandez-Balderas, R.J.; Garcia-Camacho, L.A. Relationship of persistent lymphocytosis, antibody titers, and proviral load with expression of interleukin-12, interferon-gamma, interleukin-2, interleukin-4, interleukin-10, and transforming growth factor-beta in cows infected with bovine leukemia virus from a high-prevalence dairy complex. Can. J. Vet. Res.-Rev. Can. Rech. Vet. 2022, 86, 269–285. [Google Scholar]
  180. Nieto, F.M.; Souza, F.N.; Lendez, P.A.; Martinez-Cuesta, L.; Santos, K.R.; Della, L.A.; Ceriani, M.C.; Dolcini, G.L. Lymphocyte proliferation and apoptosis of lymphocyte subpopulations in bovine leukemia virus-infected dairy cows with high and low proviral load. Vet. Immunol. Immunopathol. 2018, 206, 41–48. [Google Scholar] [CrossRef]
  181. Nakada, S.; Fujimoto, Y.; Kohara, J.; Makita, K. Economic losses associated with mastitis due to bovine leukemia virus infection. J. Dairy Sci. 2023, 106, 576–588. [Google Scholar] [CrossRef]
  182. Nishiike, M.; Haoka, M.; Doi, T.; Kohda, T.; Mukamoto, M. Development of a preliminary diagnostic measure for bovine leukosis in dairy cows using peripheral white blood cell and lymphocyte counts. J. Vet. Med. Sci. 2016, 78, 1145–1151. [Google Scholar] [CrossRef] [PubMed]
  183. Hutchinson, H.C.; Norby, B.; Droscha, C.J.; Sordillo, L.M.; Coussens, P.M.; Bartlett, P.C. Bovine leukemia virus detection and dynamics following experimental inoculation. Res. Vet. Sci. 2020, 133, 269–275. [Google Scholar] [CrossRef]
  184. Somura, Y.; Sugiyama, E.; Fujikawa, H.; Murakami, K. Comparison of the copy numbers of bovine leukemia virus in the lymph nodes of cattle with enzootic bovine leukosis and cattle with latent infection. Arch. Virol. 2014, 159, 2693–2697. [Google Scholar] [CrossRef]
  185. Jaworski, J.P.; Pluta, A.; Rola-Luszczak, M.; Mcgowan, S.L.; Finnegan, C.; Heenemann, K.; Carignano, H.A.; Alvarez, I.; Murakami, K.; Willems, L.; et al. Interlaboratory Comparison of Six Real-Time PCR Assays for Detection of Bovine Leukemia Virus Proviral DNA. J. Clin. Microbiol. 2018, 56, 10–1128. [Google Scholar] [CrossRef]
  186. Nakada, S.; Kohara, J.; Makita, K. Estimation of circulating bovine leukemia virus levels using conventional blood cell counts. J. Dairy Sci. 2018, 101, 11229–11236. [Google Scholar] [CrossRef]
  187. Kobayashi, T.; Inagaki, Y.; Ohnuki, N.; Sato, R.; Murakami, S.; Imakawa, K. Increasing Bovine leukemia virus (BLV) proviral load is a risk factor for progression of Enzootic bovine leucosis: A prospective study in Japan. Prev. Vet. Med. 2019, 178, 104680. [Google Scholar] [CrossRef]
  188. Yoneyama, S.; Kobayashi, S.; Matsunaga, T.; Tonosaki, K.; Leng, D.; Sakai, Y.; Yamada, S.; Kimura, A.; Ichijo, T.; Hikono, H.; et al. Comparative Evaluation of Three Commercial Quantitative Real-Time PCRs Used in Japan for Bovine Leukemia Virus. Viruses 2022, 14, 1182. [Google Scholar] [CrossRef] [PubMed]
  189. Gutierrez, G.; Carignano, H.; Alvarez, I.; Martinez, C.; Porta, N.; Politzki, R.; Gammella, M.; Lomonaco, M.; Fondevila, N.; Poli, M.; et al. Bovine leukemia virus p24 antibodies reflect blood proviral load. BMC Vet. Res. 2012, 8, 187. [Google Scholar] [CrossRef] [PubMed]
  190. Meirom, R.; Moss, S.; Brenner, J. Bovine leukemia virus-gp51 antigen expression is associated with CD5 and IgM markers on infected lymphocytes. Vet. Immunol. Immunopathol. 1997, 59, 113–119. [Google Scholar] [CrossRef] [PubMed]
  191. Teutsch, M.R.; Lewin, H.A. Aberrant expression of immunoglobulin mRNA in bovine leukemia virus-infected cattle. Vet. Immunol. Immunopathol. 1996, 53, 87–94. [Google Scholar] [CrossRef]
  192. Meiron, R.; Brenner, J.; Gluckman, A.; Avraham, R.; Trainin, Z. Humoral and cellular responses in calves experimentally infected with bovine leukemia virus (BLV). Vet. Immunol. Immunopathol. 1985, 9, 105–114. [Google Scholar] [CrossRef] [PubMed]
  193. Trainin, Z.; Brenner, J.; Meirom, R.; Ungar-Waron, H. Detrimental effect of bovine leukemia virus (BLV) on the immunological state of cattle. Vet. Immunol. Immunopathol. 1996, 54, 293–302. [Google Scholar] [CrossRef] [PubMed]
  194. Amills, M.; Ramiya, V.; Norimine, J.; Olmstead, C.A.; Lewin, H.A. Reduced IL-2 and IL-4 mRNA expression in CD4+ T cells from bovine leukemia virus-infected cows with persistent lymphocytosis. Virology 2002, 304, 1–9. [Google Scholar] [CrossRef]
  195. Trueblood, E.S.; Brown, W.C.; Palmer, G.H.; Davis, W.C.; Stone, D.M.; Mcelwain, T.F. B-lymphocyte proliferation during bovine leukemia virus-induced persistent lymphocytosis is enhanced by T-lymphocyte-derived interleukin-2. J. Virol. 1998, 72, 3169–3177. [Google Scholar] [CrossRef]
  196. Usuga-Monroy, C.; Gonzalez, H.L.; Echeverri, Z.J.; Diaz, F.J.; Lopez-Herrera, A. IFN-gamma mRNA expression is lower in Holstein cows infected with bovine leukemia virus with high proviral load and persistent lymphocytosis. Acta Virol. 2020, 64, 451–456. [Google Scholar] [CrossRef]
  197. Farias, M.; Lendez, P.A.; Marin, M.; Quintana, S.; Martinez-Cuesta, L.; Ceriani, M.C.; Dolcini, G.L. Toll-like receptors, IFN-gamma and IL-12 expression in bovine leukemia virus-infected animals with low or high proviral load. Res. Vet. Sci. 2016, 107, 190–195. [Google Scholar] [CrossRef]
  198. Pyeon, D.; Splitter, G.A. Interleukin-12 p40 mRNA expression in bovine leukemia virus-infected animals: Increase in alymphocytosis but decrease in persistent lymphocytosis. J. Virol. 1998, 72, 6917–6921. [Google Scholar] [CrossRef] [PubMed]
  199. Iwan, E.; Szczotka, M.; Kocki, J.; Pluta, A. Determination of cytokine profiles in populations of dendritic cells from cattle infected with bovine leukaemia virus. Pol. J. Vet. Sci. 2018, 21, 681–690. [Google Scholar] [CrossRef] [PubMed]
  200. Iwan, E.; Szczotka, M.; Kocki, J. Cytokine profiles of dendritic cells (DCs) during infection with bovine leukaemia virus (BLV). Pol. J. Vet. Sci. 2017, 20, 221–231. [Google Scholar] [CrossRef]
  201. Konnai, S.; Suzuki, S.; Shirai, T.; Ikebuchi, R.; Okagawa, T.; Sunden, Y.; Mingala, C.N.; Onuma, M.; Murata, S.; Ohashi, K. Enhanced expression of LAG-3 on lymphocyte subpopulations from persistently lymphocytotic cattle infected with bovine leukemia virus. Comp. Immunol. Microbiol. Infect. Dis. 2013, 36, 63–69. [Google Scholar] [CrossRef]
  202. Shirai, T.; Konnai, S.; Ikebuchi, R.; Okagawa, T.; Suzuki, S.; Sunden, Y.; Onuma, M.; Murata, S.; Ohashi, K. Molecular cloning of bovine lymphocyte activation gene-3 and its expression characteristics in bovine leukemia virus-infected cattle. Vet. Immunol. Immunopathol. 2011, 144, 462–467. [Google Scholar] [CrossRef]
  203. Okagawa, T.; Konnai, S.; Ikebuchi, R.; Suzuki, S.; Shirai, T.; Sunden, Y.; Onuma, M.; Murata, S.; Ohashi, K. Increased bovine Tim-3 and its ligand expressions during bovine leukemia virus infection. Vet. Res. 2012, 43, 45. [Google Scholar] [CrossRef]
  204. Suzuki, S.; Konnai, S.; Okagawa, T.; Ikebuchi, R.; Nishimori, A.; Kohara, J.; Mingala, C.N.; Murata, S.; Ohashi, K. Increased expression of the regulatory T cell-associated marker CTLA-4 in bovine leukemia virus infection. Vet. Immunol. Immunopathol. 2015, 163, 115–124. [Google Scholar] [CrossRef] [PubMed]
  205. Pollari, F.L.; Wangsuphachart, V.L.; Digiacomo, R.F.; Evermann, J.F. Effects of bovine leukemia virus infection on production and reproduction in dairy cattle. Can. J. Vet. Res.-Rev. Can. Rech. Vet. 1992, 56, 289–295. [Google Scholar]
  206. Yang, Y.; Gong, Z.; Lu, Y.; Lu, X.; Zhang, J.; Meng, Y.; Peng, Y.; Chu, S.; Cao, W.; Hao, X.; et al. Dairy Cows Experimentally Infected With Bovine Leukemia Virus Showed an Increased Milk Production in Lactation Numbers 3–4: A 4-Year Longitudinal Study. Front. Microbiol. 2022, 13, 946463. [Google Scholar] [CrossRef]
  207. Jacobs, R.M.; Heeney, J.L.; Godkin, M.A.; Leslie, K.E.; Taylor, J.A.; Davies, C.; Valli, V.E. Production and related variables in bovine leukaemia virus-infected cows. Vet. Res. Commun. 1991, 15, 463–474. [Google Scholar] [CrossRef]
  208. Ruiz, V.; Porta, N.G.; Lomonaco, M.; Trono, K.; Alvarez, I. Bovine Leukemia Virus Infection in Neonatal Calves. Risk Factors and Control Measures. Front. Vet. Sci. 2018, 5, 267. [Google Scholar] [CrossRef]
  209. Esteban, E.N.; Poli, M.; Poiesz, B.; Ceriani, C.; Dube, S.; Gutierrez, S.; Dolcini, G.; Gagliardi, R.; Perez, S.; Lützelschwab, C. Bovine leukemia virus (BLV), proposed control and eradication programs by marker assisted breeding of genetically resistant cattle. Anim. Genet. 2009, 2009, 107–130. [Google Scholar]
  210. Rodriguez, S.M.; Florins, A.; Gillet, N.; de Brogniez, A.; Sanchez-Alcaraz, M.T.; Boxus, M.; Boulanger, F.; Gutierrez, G.; Trono, K.; Alvarez, I.; et al. Preventive and therapeutic strategies for bovine leukemia virus: Lessons for HTLV. Viruses 2011, 3, 1210–1248. [Google Scholar] [CrossRef] [PubMed]
  211. Balic, D.; Lojkic, I.; Periskic, M.; Bedekovic, T.; Jungic, A.; Lemo, N.; Roic, B.; Cac, Z.; Barbic, L.; Madic, J. Identification of a new genotype of bovine leukemia virus. Arch. Virol. 2012, 157, 1281–1290. [Google Scholar] [CrossRef]
  212. Bartlett, P.C.; Ruggiero, V.J.; Hutchinson, H.C.; Droscha, C.J.; Norby, B.; Sporer, K.; Taxis, T.M. Current Developments in the Epidemiology and Control of Enzootic Bovine Leukosis as Caused by Bovine Leukemia Virus. Pathogens 2020, 9, 58. [Google Scholar] [CrossRef]
  213. Hayashi, T.; Mekata, H.; Sekiguchi, S.; Kirino, Y.; Mitoma, S.; Honkawa, K.; Horii, Y.; Norimine, J. Cattle with the BoLA class II DRB3*0902 allele have significantly lower bovine leukemia proviral loads. J. Vet. Med. Sci. 2017, 79, 1552–1555. [Google Scholar] [CrossRef]
  214. Nikbakht, B.G.; Ghorbanpour, R.; Esmailnejad, A. Association of BoLA-DRB3.2 Alleles with BLV Infection Profiles (Persistent Lymphocytosis/Lymphosarcoma) and Lymphocyte Subsets in Iranian Holstein Cattle. Biochem. Genet. 2016, 54, 194–207. [Google Scholar] [CrossRef] [PubMed]
  215. Nakatsuchi, A.; Watanuki, S.; Borjigin, L.; Sato, H.; Bai, L.; Matsuura, R.; Kuroda, M.; Murakami, H.; Sato, R.; Asaji, S.; et al. BoLA-DRB3 Polymorphism Controls Proviral Load and Infectivity of Bovine Leukemia Virus (BLV) in Milk. Pathogens 2022, 11, 210. [Google Scholar] [CrossRef]
  216. Borjigin, L.; Lo, C.W.; Bai, L.; Hamada, R.; Sato, H.; Yoneyama, S.; Yasui, A.; Yasuda, S.; Yamanaka, R.; Mimura, M.; et al. Risk Assessment of Bovine Major Histocompatibility Complex Class II DRB3 Alleles for Perinatal Transmission of Bovine Leukemia Virus. Pathogens 2021, 10, 502. [Google Scholar] [CrossRef]
  217. Glass, E.J.; Baxter, R.; Leach, R.; Taylor, G. Breeding for Disease Resistance in Farm Animals; CAB International: Wallingford, UK, 2010. [Google Scholar]
  218. Basrur, P.K.; King, W.A. Genetics then and now: Breeding the best and biotechnology. Rev. Sci. Tech. Off. Int. Epizoot. 2005, 24, 31–49. [Google Scholar] [CrossRef]
  219. Williams, J.L. The use of marker-assisted selection in animal breeding and biotechnology. Rev. Sci. Tech. Off. Int. Epizoot. 2005, 24, 379–391. [Google Scholar] [CrossRef]
  220. Maradei, E.; Perez, B.C.; Malirat, V.; Salgado, G.; Seki, C.; Pedemonte, A.; Bonastre, P.; D’Aloia, R.; La Torre, J.L.; Mattion, N.; et al. Characterization of foot-and-mouth disease virus from outbreaks in Ecuador during 2009–2010 and cross-protection studies with the vaccine strain in use in the region. Vaccine 2011, 29, 8230–8240. [Google Scholar] [CrossRef]
  221. Miller, J.M.; Van Der Maaten, M.J. Evaluation of an inactivated bovine leukemia virus preparation as an immunogen in cattle. Ann. Rech. Vet. 1978, 9, 871–877. [Google Scholar] [PubMed]
  222. Patrascu, I.V.; Coman, S.; Sandu, I.; Stiube, P.; Munteanu, I.; Coman, T.; Ionescu, M.; Popescu, D.; Mihailescu, D. Specific protection against bovine leukemia virus infection conferred on cattle by the Romanian inactivated vaccine BL-VACC-RO. Virologie 1980, 31, 95–102. [Google Scholar]
  223. Parfanovich, M.I.; Zhdanov, V.M.; Lazarenko, A.A.; Nomm, E.M.; Simovart, Y.; Parakin, V.K.; Lemesh, V.M. The possibility of specific protection against bovine leukaemia virus infection and bovine leukaemia with inactivated BLV. Br. Vet. J. 1983, 139, 137–146. [Google Scholar] [CrossRef]
  224. Kabeya, H.; Ohashi, K.; Ohishi, K.; Sugimoto, C.; Amanuma, H.; Onuma, M. An effective peptide vaccine to eliminate bovine leukaemia virus (BLV) infected cells in carrier sheep. Vaccine 1996, 14, 1118–1122. [Google Scholar] [CrossRef]
  225. Miller, J.M.; Van der Maaten, M.J.; Schmerr, M.J. Vaccination of cattle with binary ethylenimine-treated bovine leukemia virus. Am. J. Vet. Res. 1983, 44, 64–67. [Google Scholar] [PubMed]
  226. Fukuyama, S.; Kodama, K.; Hirahara, T.; Nakajima, N.; Takamura, K.; Sasaki, O.; Imanishi, J. Protection against bovine leukemia virus infection by use of inactivated vaccines in cattle. J. Vet. Med. Sci. 1993, 55, 99–106. [Google Scholar] [CrossRef] [PubMed]
  227. Onuma, M.; Hodatsu, T.; Yamamoto, S.; Higashihara, M.; Masu, S.; Mikami, T.; Izawa, H. Protection by vaccination against bovine leukemia virus infection in sheep. Am. J. Vet. Res. 1984, 45, 1212–1215. [Google Scholar]
  228. Burkhardt, H.; Rosenthal, S.; Wittmann, W.; Starick, E.; Scholz, D.; Rosenthal, H.A.; Kluge, K.H. Immunization of young cattle with gp51 of the bovine leukosis virus and the subsequent experimental infection. Arch. Exp. Veterinarmed. 1989, 43, 933–942. [Google Scholar] [PubMed]
  229. Merza, M.; Sober, J.; Sundquist, B.; Toots, I.; Morein, B. Characterization of purified gp 51 from bovine leukemia virus integrated into iscom. Physicochemical properties and serum antibody response to the integrated gp51. Arch. Virol. 1991, 120, 219–231. [Google Scholar] [CrossRef]
  230. Bruck, C.; Mathot, S.; Portetelle, D.; Berte, C.; Franssen, J.D.; Herion, P.; Burny, A. Monoclonal antibodies define eight independent antigenic regions on the bovine leukemia virus (BLV) envelope glycoprotein gp51. Virology 1982, 122, 342–352. [Google Scholar] [CrossRef]
  231. Gatei, M.H.; Naif, H.M.; Kumar, S.; Boyle, D.B.; Daniel, R.C.; Good, M.F.; Lavin, M.F. Protection of sheep against bovine leukemia virus (BLV) infection by vaccination with recombinant vaccinia viruses expressing BLV envelope glycoproteins: Correlation of protection with CD4 T-cell response to gp51 peptide 51–70. J. Virol. 1993, 67, 1803–1810. [Google Scholar] [CrossRef]
  232. Callebaut, I.; Mornon, J.P.; Burny, A.; Portetelle, D. The bovine leukemia virus (BLV) envelope glycoprotein gp51 as a general model for the design of a subunit vaccine against retroviral infection: Mapping of functional sites through immunological and structural data. Leukemia 1994, 8 (Suppl. S1), S218–S221. [Google Scholar]
  233. Okada, K.; Sonoda, K.; Koyama, M.; Yin, S.; Ikeda, M.; Goryo, M.; Chen, S.L.; Kabeya, H.; Ohishi, K.; Onuma, M. Delayed-type hypersensitivity in sheep induced by synthetic peptides of bovine leukemia virus encapsulated in mannan-coated liposome. J. Vet. Med. Sci. 2003, 65, 515–518. [Google Scholar] [CrossRef]
  234. Ohishi, K.; Kabeya, H.; Amanuma, H.; Onuma, M. Peptide-based bovine leukemia virus (BLV) vaccine that induces BLV-Env specific Th-1 type immunity. Leukemia 1997, 11 (Suppl. S3), 223–226. [Google Scholar]
  235. Portetelle, D.; Limbach, K.; Burny, A.; Mammerickx, M.; Desmettre, P.; Riviere, M.; Zavada, J.; Paoletti, E. Recombinant vaccinia virus expression of the bovine leukaemia virus envelope gene and protection of immunized sheep against infection. Vaccine 1991, 9, 194–200. [Google Scholar] [CrossRef]
  236. Kumar, S.; Andrew, M.E.; Boyle, D.B.; Brandon, R.B.; Lavin, M.F.; Daniel, R.C. Expression of bovine leukaemia virus envelope gene by recombinant vaccinia viruses. Virus Res. 1990, 17, 131–142. [Google Scholar] [CrossRef]
  237. Ohishi, K.; Ikawa, Y. T cell-mediated destruction of bovine leukemia virus-infected peripheral lymphocytes by bovine leukemia virus env-vaccinia recombinant vaccine. Aids Res. Hum. Retrovir. 1996, 12, 393–398. [Google Scholar] [CrossRef]
  238. Cherney, T.M.; Schultz, R.D. Viral status and antibody response in cattle inoculated with recombinant bovine leukemia virus-vaccinia virus vaccines after challenge exposure with bovine leukemia virus-infected lymphocytes. Am. J. Vet. Res. 1996, 57, 812–818. [Google Scholar]
  239. Brillowska, A.; Dabrowski, S.; Rulka, J.; Kubis, P.; Buzala, E.; Kur, J. Protection of cattle against bovine leukemia virus (BLV) infection could be attained by DNA vaccination. Acta Biochim. Pol. 1999, 46, 971–976. [Google Scholar] [CrossRef] [PubMed]
  240. Van den Broeke, A.; Oumouna, M.; Beskorwayne, T.; Szynal, M.; Cleuter, Y.; Babiuk, S.; Bagnis, C.; Martiat, P.; Burny, A.; Griebel, P. Cytotoxic responses to BLV tax oncoprotein do not prevent leukemogenesis in sheep. Leuk. Res. 2010, 34, 1663–1669. [Google Scholar] [CrossRef] [PubMed]
  241. Suarez, A.G.; Gutierrez, G.; Camussone, C.; Calvinho, L.; Abdala, A.; Alvarez, I.; Petersen, M.; Franco, L.; Destefano, G.; Monti, G.; et al. A safe and effective vaccine against bovine leukemia virus. Front. Immunol. 2022, 13, 980514. [Google Scholar] [CrossRef] [PubMed]
  242. Kechine, T.; Ali, T.; Worku, T.; Abdisa, L.; Assebe, Y.T. Anxiety and Associated Factors Among Clients on Highly Active Antiretroviral Therapy (HAART) in Public Hospitals of Southern Ethiopia: A Multi-Center Cross-Sectional Study. Psychol. Res. Behav. Manag. 2022, 15, 3889–3900. [Google Scholar] [CrossRef]
  243. Ntolou, P.; Pani, P.; Panis, V.; Madianos, P.; Vassilopoulos, S. The effect of antiretroviral therapyon the periodontal conditions of patients with HIV infection: A systematic review and meta-analysis. J. Clin. Periodontol. 2023, 50, 170–182. [Google Scholar] [CrossRef]
  244. Murakami, H.; Murakami-Kawai, M.; Kamisuki, S.; Hisanobu, S.; Tsurukawa, Y.; Uchiyama, J.; Sakaguchi, M.; Tsukamoto, K. Specific antiviral effect of violaceoid E on bovine leukemia virus. Virology 2021, 562, 1–8. [Google Scholar] [CrossRef]
  245. Nishimori, A.; Konnai, S.; Okagawa, T.; Maekawa, N.; Ikebuchi, R.; Goto, S.; Sajiki, Y.; Suzuki, Y.; Kohara, J.; Ogasawara, S.; et al. In vitro and in vivo antivirus activity of an anti-programmed death-ligand 1 (PD-L1) rat-bovine chimeric antibody against bovine leukemia virus infection. PLoS ONE 2017, 12, e174916. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the BLV genome [35].
Figure 1. Schematic representation of the BLV genome [35].
Animals 14 00297 g001
Figure 2. A maximum likelihood phylogenetic tree was constructed using partial BLV env sequences from various geographical locations worldwide, from [62].
Figure 2. A maximum likelihood phylogenetic tree was constructed using partial BLV env sequences from various geographical locations worldwide, from [62].
Animals 14 00297 g002
Figure 3. World distribution map of EBL based on the last 5 years (2019–2023), from the WOAH database WAHIS [77].
Figure 3. World distribution map of EBL based on the last 5 years (2019–2023), from the WOAH database WAHIS [77].
Animals 14 00297 g003
Table 1. Prevalence of EBL worldwide, updated and modified from [62].
Table 1. Prevalence of EBL worldwide, updated and modified from [62].
StatusContinentCountriesYearReferences
BLV-freeEuropeBelgium2016
2021
[5,78,79]
Czech Republic
Denmark
Germany
Estonia
Ireland
Spain
France
Italy
Cyprus
Latvia
Lithuania
Luxembourg
The Netherlands
Austria
Poland
Portugal
Romania
Slovenia
Finland
Sweden
United Kingdom (Northern Ireland)
OceaniaAustralia2013
New Zealand2008
Tunisia2005
AsiaKyrgyzstan2008
Kazakhstan2007
BLV existing in countries with unknown prevalenceEuropeBelarusPresent
Bulgaria
Croatia
Greece
Ukraine
BLV existing in countries with variable prevalenceNorth AmericaCanada
78% at herd level
88.39% at herd level
89.30% at herd level
1998–2003
2016
2018
[80,81]
Mexico
Dairy 36.1%, beef 4%
Dairy 50.6%
1983
2023
[82,83]
USA
Dairy 83.9%, beef 39%
2007[84]
EuropeRussia
22.1% to 25.4%
2012[85]
South AmericaArgentina (Buenos Aires)
90.9% at herd level
Argentina (multiple regions)
84% at herd level
90.16%
1998–1999
2007
2011
[56,86,87]
Bolivia
(multiple regions)
30.7% at individual level
2008[56]
Brazil
17.1% at herd level
60.8% at herd level
23.4% at herd level
1980–1989
1992–1995
2021
[88,89,90]
Chile (southern regions)
27.9% at individual level
2009[56]
Colombia
62% at individual level
2020[52]
Ecuador
5.6% at individual level
2012[91]
Paraguay (Asuncion)
54.7% at individual level
2008[56]
Peru (multiple regions)
31% at individual level,
42.3% at individual level
1983
2008
[56,92]
Venezuela
33.3% at individual level
60.83% at individual level
1978
2011
[93,94]
Uruguay
10.4% at individual level
2013[95]
AsiaChina
Dairy 49.1%, beef 1.6%
2013–2014[96]
Cambodia
Draught cattle 5.3%
2000[97]
Iran (nationwide)
22.1% to 25.4%
2012–2014[98]
Myanmar
9.1% at individual level
2016[56]
Japan (nationwide)
Dairy 49.1%, beef 1.6%
79.1% at dairy herd level
73.3% at individual level
2009–2011
2007
2012–2014
[6,99]
Mongolia
Dairy 3.9%
2014[100]
The Philippines
4.8% to 9.7%
2010–2012[101]
Pakistan
Dairy 20%
2019[102]
Taiwan
81.8% at animal level, 99.1% at herd level
2019[103]
Thailand
58.7% at individual level
2013–2014[60]
Middle EastIsrael
5% at individual level
2005[104]
Saudi Arabia
Dairy 20.2%
1990[105]
Turkey
Dairy 48.3%
2005[106]
Iraq
Dairy 7%
2015[107]
Egypt
Dairy 17.7%
2020[108]
Table 2. Serological techniques used for diagnosis of BLV.
Table 2. Serological techniques used for diagnosis of BLV.
Test AssaySample TypeTargetAdvantagesDisadvantagesReferences
AGIDSerum Antibodies
(p24, gp51)
Specific, simple, rapid, and low screening costLess sensitive, inconclusive, and fails to evaluate disease states[137,147,148,149,152]
ELISASerum, milk, bulk milkAntibodies
(p24, gp51)
Sensitive, specific, large-scale screening, and rapidFalse negatives
(particularly in cattle during the early stages of infection)
False positives
(maternally derived antibodies)
Cannot evaluate disease states of infected cattle
[142,143,148,149,153]
RIASerum Antibodies
(p24)
Sensitive in detecting BLV at an early stage of infectionCannot be used for mass screening[154]
PHAVirus particleBLV glycoproteinSensitive, specific, rapid, and low screening costAffected by pH and temperature
Hemagglutination activity reduced by trypsin and neuraminidase
[150]
Table 4. Control tools for EBL, modified from [76].
Table 4. Control tools for EBL, modified from [76].
Goal/MethodTool/ComponentsApplicability
Eradication
(elimination of infected animals)
Test and slaughter
Regular testing and promptly culling infected animals
Culling of the offspring of infected animals
Safe management practices should be followed to prevent the spread of the virus among animals
Small heard, with low herd BLV prevalence, supported for restocking
Low or moderate within-herd prevalence
Freedom from the infection can be achieved if the rate of removal of positive animals exceeds the annual incidence rate of infection
Support of national governments through economic compensation policies
Test and separate
Crucial to physically separate infected cattle from uninfected cattle
Gradual elimination of infected animals can be achieved by increasing the frequency of culling in the infected group.
Regular testing should be conducted on the seronegative group, and any positive animals should be promptly separated or eliminated
In the final stage of the eradication program, a ‘test and slaughter’ strategy is implemented
Safe management practices should be followed to prevent the spread of the virus among animals
Control
(reduction of the rate of effective contacts)
Safe herd management practices
Milk from cows that have tested BLV-negative or milk replacer to feed calves; milk from BLV-infected cows should be treated through freezing or heat treatment
Chemical dehorning or cautery
Disposable needles or needles sterilized by boiling between animals
Clean and disinfect ear tattoo implements
Separate gloves for rectal exploration
Separate calving paddocks for BLV-infected and uninfected cattle
Removal of calves from cows within 24 h of birth but after intake of colostrum
Fly control program
All herds
Prevention
(avoiding introduction)
Biosecurity measures
Introduction of animals from certified BLV-infection-free herds
Avoid any contact with infected animals, such as sharing common pastures
Iatrogenic introduction should be prevented
All herds
Surveillance
(maintaining disease-/infection free-status)
Serological surveillance at the herd level involves regular testing of individual or pooled milk or serum samples to detect BLV antibodies
At the regional or country level, surveillance for tumors is conducted during post-mortem inspection of slaughtered animals
Regular testing of representative samples of herds for BLV antibodies from bulk milk samples or individual milk or serum samples
Free herds/territories
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lv, G.; Wang, J.; Lian, S.; Wang, H.; Wu, R. The Global Epidemiology of Bovine Leukemia Virus: Current Trends and Future Implications. Animals 2024, 14, 297. https://doi.org/10.3390/ani14020297

AMA Style

Lv G, Wang J, Lian S, Wang H, Wu R. The Global Epidemiology of Bovine Leukemia Virus: Current Trends and Future Implications. Animals. 2024; 14(2):297. https://doi.org/10.3390/ani14020297

Chicago/Turabian Style

Lv, Guanxin, Jianfa Wang, Shuai Lian, Hai Wang, and Rui Wu. 2024. "The Global Epidemiology of Bovine Leukemia Virus: Current Trends and Future Implications" Animals 14, no. 2: 297. https://doi.org/10.3390/ani14020297

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