fbpx
Wikipedia

Bovine viral diarrhea

Bovine viral diarrhea (BVD), bovine viral diarrhoea (UK English) or mucosal disease, previously referred to as bovine virus diarrhea (BVD), is an economically significant disease of cattle that is found in the majority of countries throughout the world.[1] Worldwide reviews of the economically assessed production losses and intervention programs (e.g. eradication programs, vaccination strategies and biosecurity measures) incurred by BVD infection have been published.[2][3] The causative agent, bovine viral diarrhea virus (BVDV), is a member of the genus Pestivirus of the family Flaviviridae.[1]

Bovine viral diarrhea
Immunofluorescence image of BVDV (CP7 type). Nuclei are stained blue with DAPI. The replication complexes of the viruses are marked red by NS3 protein binding antibodies
Scientific classification
(unranked): Virus
Realm: Riboviria
Kingdom: Orthornavirae
Phylum: Kitrinoviricota
Class: Flasuviricetes
Order: Amarillovirales
Family: Flaviviridae
Genus: Pestivirus
Groups included
  • Pestivirus A (formerly Bovine viral diarrhea virus 1)
  • Pestivirus B (formerly Bovine viral diarrhea virus 2)
Cladistically included but traditionally excluded taxa
Tongue lesions on confirmed BVD/MD case (mucosal disease form)

BVD infection results in a wide variety of clinical signs, due to its immunosuppressive effects,[4] as well as having a direct effect on respiratory disease and fertility.[5] In addition, BVD infection of a susceptible dam during a certain period of gestation can result in the production of a persistently infected (PI) fetus.[6]

PI animals recognise intra-cellular BVD viral particles as ‘self’ and shed virus in large quantities throughout life; they represent the cornerstone of the success of BVD as a disease.

Currently, it was shown in a worldwide review study that the PI prevalence at animal level ranged from low (≤0.8% Europe, North America, Australia), medium (>0.8% to 1.6% East Asia) to high (>1.6% West Asia). Countries that had failed to implement any BVDV control and/or eradication programmes (including vaccination) had the highest PI prevalence.[7]

Virus classification and structure edit

BVDVs are members of the genus Pestivirus, belonging to the family Flaviviridae. Other members of this genus cause Border disease (sheep) and classical swine fever (pigs) which cause significant financial loss to the livestock industry.[8]

Pestiviruses are small, spherical, single-stranded, enveloped RNA viruses of 40 to 60 nm in diameter.[9]

The genome consists of a single, linear, positive-sense, single-stranded RNA molecule of approximately 12.3 kb.[10] RNA synthesis is catalyzed by the BVDV RNA-dependent RNA polymerase (RdRp). This RdRp can undergo template strand switching allowing RNA-RNA copy choice recombination during elongative RNA synthesis.[11]

Two BVDV genotypes are recognised, based on the nucleotide sequence of the 5’untranslated (UTR) region; BVDV-1 and BVDV-2.[12] BVDV-1 isolates have been grouped into 16 subtypes (a –p) and BVDV-2 has currently been grouped into 3 subtypes (a – c).[13]

BVDV strains can be further divided into distinct biotypes (cytopathic or non-cytopathic) according to their effects on tissue cell culture; cytopathic (cp) biotypes, formed via mutation of non-cytopathic (ncp) biotypes, induce apoptosis in cultured cells.[14] Ncp viruses can induce persistent infection in cells and have an intact NS2/3 protein. In cp viruses the NS2/3 protein is either cleaved to NS2 and NS3 or there is a duplication of viral RNA containing an additional NS3 region.[15] The majority of BVDV infections in the field are caused by the ncp biotype.[1]

Epidemiology edit

BVD is considered one of the most significant infectious diseases in the livestock industry worldwide due to its high prevalence, persistence and clinical consequences.[16]

In Europe the prevalence of antibody positive animals in countries without systematic BVD control is between 60 and 80%.[17] Prevalence has been determined in individual countries and tends to be positively associated with stocking density of cattle.[citation needed]

BVDV-1 strains are predominant in most parts of the world, whereas BVDV-2 represents 50% of cases in North America.[16] In Europe, BVDV-2 was first isolated in the UK in 2000 and currently represents up to 11% of BVD cases in Europe.[18]

Transmission of BVDV occurs both horizontally and vertically with both persistently and transiently infected animals excreting infectious virus. Virus is transmitted via direct contact, bodily secretions and contaminated fomites, with the virus being able to persist in the environment for more than two weeks. Persistently infected animals are the most important source of the virus, continuously excreting a viral load one thousand times that shed by acutely infected animals.[19]

Pathogenesis edit

 
Turbinate cells infected with BVDV

Acute, transient infection edit

Following viral entry and contact with the mucosal lining of the mouth or nose, replication occurs in epithelial cells. BVDV replication has a predilection for the palatine tonsils, lymphoid tissues and epithelium of the oropharynx.

Phagocytes take up BVDV or virus-infected cells and transport them to peripheral lymphoid tissues; the virus can also spread systemically through the bloodstream. Viraemia occurs 2–4 days after exposure and virus isolation from serum or leukocytes is generally possible between 3–10 days post infection.[20]

During systemic spread the virus is able to gain entry into most tissues with a preference for lymphoid tissues. Neutralising antibodies can be detected from 10 to 14 days post infection with titres continuing to increase slowly for 8–10 weeks. After 2–3 weeks, antibodies effectively neutralise viral particles, promote clearance of virus and prevent seeding of target organs.[21]

Intrauterine infections edit

Fetal infection is of most consequence as this can result in the birth of a persistently infected neonate. The effects of fetal infection with BVDV are dependent upon the stage of gestation at which the dam suffers acute infection.

BVDV infection of the dam prior to conception, and during the first 18 days of gestation, results in delayed conception and an increased calving to conception interval. Once the embryo is attached, infection from days 29–41 can result in embryonic infection and resultant embryonic death.

Infection of the dam from approximately day 30 of gestation until day 120 can result in immunotolerance and the birth of calves persistently infected with the virus.

BVDV infection between 80 and 150 days of gestation may be teratogenic, with the type of birth defect dependent upon the stage of fetal development at infection. Abortion may occur at any time during gestation. Infection after approximately day 120 can result in the birth of a normal fetus which is BVD antigen-negative and BVD antibody-positive. This occurs because the fetal immune system has developed, by this stage of gestation, and has the ability to recognise and fight off the invading virus, producing anti-BVD antibodies.

Chronic infections edit

BVD virus can be maintained as a chronic infection within some immunoprivileged sites following transient infection. These sites include ovarian follicles, testicular tissues, central nervous system and white blood cells. Cattle with chronic infections elicit a significant immune response, exhibited by extremely high antibody titres.

Clinical signs edit

BVDV infection has a wide manifestation of clinical signs including fertility issues, milk drop, pyrexia, diarrhea, and fetal infection.[9] Occasionally, a severe acute form of BVD may occur. These outbreaks are characterized by thrombocytopenia with high morbidity and mortality. However, clinical signs are frequently mild and infection insidious, recognized only by BVDV's immunosuppressive effects perpetuating other circulating infectious diseases (particularly scours and pneumonias).

PI animals edit

Persistently infected animals did not have a competent immune system at the time of BVDV transplacental infection. The virus, therefore, entered the fetal cells and, during immune system development, was accepted as self. In PIs the virus remains present in a large number of the animal's body cells throughout its life and is continuously shed. PIs are often ill-thrifty and smaller than their peers, however, they can appear normal. PIs are more susceptible to disease, with only 20% of PIs surviving to two years of age.[22] If a PI dam is able to reproduce they always give birth to PI calves.[23]

Mucosal disease edit

The PI cattle that do survive ill-thrift are susceptible to mucosal disease. Mucosal disease only develops in PI animals and is invariably fatal.[5] Disease results when a PI animal is superinfected with a cytopathic biotype arising from mutation of the non-cytopathic strain of BVDV already circulating in that animal.[24] The cp BVDV spreads to the gastro-intestinal epithelium, and necrosis of keratinocytes results in erosion and ulceration. Fluid leaks from the epithelial surface of the gastro-intestinal tract causing diarrhoea and dehydration. In addition, bacterial infection of the damaged epithelium results in secondary septicaemia. Death occurs in the ensuing days or weeks.

Diagnosis edit

Various diagnostic tests are available for the detection of either active infection or evidence of historical infection. The method of diagnosis used also depends upon whether the vet is investigating at an individual or a herd level.

Virus or antigen detection edit

Antigen ELISA and rtPCR are currently the most frequently performed tests to detect virus or viral antigen. Individual testing of ear tissue tag samples or serum samples is performed. It is vital that repeat testing is performed on positive samples to distinguish between acute, transiently infected cattle and PIs. A second positive result, acquired at least three weeks after the primary result, indicates a PI animal. rtPCR can also be used on bulk tank milk (BTM) samples to detect any PI cows contributing to the tank. It is reported that the maximum number of contributing cows from which a PI can be detected is 300.

BVD antibody detection edit

Antibody (Ig) ELISAs are used to detect historical BVDV infection; these tests have been validated in serum, milk and bulk milk samples. Ig ELISAs do not diagnose active infection but detect the presence of antibodies produced by the animal in response to viral infection. Vaccination also induces an antibody response, which can result in false positive results, therefore it is important to know the vaccination status of the herd or individual when interpreting results. A standard test to assess whether virus has been circulating recently is to perform an Ig ELISA on blood from 5–10 young stock that have not been vaccinated, aged between 9 and 18 months. A positive result indicates exposure to BVDV, but also that any positive animals are very unlikely to be PI animals themselves. A positive result in a pregnant female indicates that she has previously been either vaccinated or infected with BVDV and could possibly be carrying a PI fetus, so antigen testing of the newborn is vital to rule this out.[5] A negative antibody result, at the discretion of the responsible veterinarian, may require further confirmation that the animal is not in fact a PI.

At a herd level, a positive Ig result suggests that BVD virus has been circulating or the herd is vaccinated. Negative results suggest that a PI is unlikely however this naïve herd is in danger of severe consequences should an infected animal be introduced. Antibodies from wild infection or vaccination persist for several years therefore Ig ELISA testing is more valuable when used as a surveillance tool in seronegative herds.

Eradication and control edit

The mainstay of eradication is the identification and removal of persistently infected animals. Re-infection is then prevented by vaccination and high levels of biosecurity, supported by continuing surveillance. PIs act as viral reservoirs and are the principal source of viral infection but transiently infected animals and contaminated fomites also play a significant role in transmission.[1]

Leading the way in BVD eradication, almost 20 years ago, were the Scandinavian countries. Despite different conditions at the start of the projects in terms of legal support, and regardless of initial prevalence of herds with PI animals, it took all countries approximately 10 years to reach their final stages.[25][26]

Once proven that BVD eradication could be achieved in a cost efficient way, a number of regional programmes followed in Europe, some of which have developed into national schemes.[27]

Vaccination is an essential part of both control and eradication. While BVD virus is still circulating within the national herd, breeding cattle are at risk of producing PI neonates and the economic consequences of BVD are still relevant. Once eradication has been achieved, unvaccinated animals will represent a naïve and susceptible herd. Infection from imported animals or contaminated fomites brought into the farm, or via transiently infected in-contacts will have devastating consequences.

Vaccination edit

Modern vaccination programmes aim not only to provide a high level of protection from clinical disease for the dam, but, crucially, to protect against viraemia and prevent the production of PIs.[28] While the immune mechanisms involved are the same, the level of immune protection required for foetal protection is much higher than for prevention of clinical disease.[29]

While challenge studies indicate that killed, as well as live, vaccines prevent foetal infection under experimental conditions, the efficacy of vaccines under field conditions has been questioned.[30] The birth of PI calves into vaccinated herds suggests that killed vaccines do not stand up to the challenge presented by the viral load excreted by a PI in the field.[31]

See also edit

References edit

  1. ^ a b c d Fray, M.D; Paton, D.J; Alenius, S.; et al. (2000). "The effects of bovine viral diarrhoea virus on cattle reproduction in relation to disease control". Animal Reproduction Science. 60–61: 615–27. doi:10.1016/s0378-4320(00)00082-8. PMID 10844229.
  2. ^ Richter, V; Lebl, K; Baumgartner, W; Obritzhauser, W; Käsbohrer, A; Pinior, B (2017). "A systematic worldwide review of the direct monetary losses due to bovine viral diarrhea virus infection". The Veterinary Journal. 220: 80–87. doi:10.1016/j.tvjl.2017.01.005. PMID 28190502.
  3. ^ Pinior, B; Firth, C; Richter, V; Lebl, K; Trauffler, M; Dzieciol, M; Hutter, S; Burgstaller, J; Obritzhauser, W; Winter, P; Käsbohrer, A (2017). "A systematic review of financial and economic assessments of bovine viral diarrhea virus (BVDV) prevention and mitigation activities worldwide". Preventive Veterinary Medicine. 137 (Pt A): 77–92. doi:10.1016/j.prevetmed.2016.12.014. PMID 28040270.
  4. ^ Schaut, Robert G.; McGill, Jodi L.; Neill, John D.; Ridpath, Julia F.; Sacco, Randy E. (2015-10-02). "Bovine viral diarrhea virus type 2 in vivo infection modulates TLR4 responsiveness in differentiated myeloid cells which is associated with decreased MyD88 expression". Virus Research. 208: 44–55. doi:10.1016/j.virusres.2015.05.017. ISSN 1872-7492. PMID 26043978.
  5. ^ a b c Lanyon, Sasha R.; Hill, Fraser I.; Reichel, Michael P.; Brownlie, Joe; et al. (2014). "Bovine Viral Diarrhoea: Pathogenesis and diagnosis" (PDF). Veterinary Journal. 199 (2): 201–9. doi:10.1016/j.tvjl.2013.07.024. PMID 24053990.
  6. ^ Grooms, Daniel L. (2004). "Reproductive consequences of infection with bovine viral diarrhea virus". Veterinary Clinics of North America: Food Animal Practice. 20 (1): 5–19. doi:10.1016/j.cvfa.2003.11.006. PMID 15062471.
  7. ^ Scharnböck, B; Roch, Franz-Ferdinand; Richter, V; Funke, C; Firth, C; Obritzhauser, W; Baumgartner, W; Käsbohrer, A; Pinior, B (2018). "A meta-analysis of bovine viral diarrhoea virus (BVDV) prevalences in the global cattle population". Scientific Reports. 8 (1): 14420. Bibcode:2018NatSR...814420S. doi:10.1038/s41598-018-32831-2. PMC 6158279. PMID 30258185.
  8. ^ Hornberg, Andrea; Fernández, Sandra Revilla; Vogl, Claus; Vilcek, Stefan; Matt, Monika; Fink, Maria; Köfer, Josef; Schöpf, Karl (2009). "Genetic diversity of pestivirus isolates in cattle from Western Austria" (PDF). Veterinary Microbiology. 135 (3–4): 205–213. doi:10.1016/j.vetmic.2008.09.068. PMID 19019571. S2CID 46378359.
  9. ^ a b N. James MacLachlan; Edward J. Dubovi, eds. (2011). Fenner's Veterinary Virology (4th ed.). Elsevier.
  10. ^ Brett D. Lindenbach; Heinz-Jürgen Thiel; Charles M. Rice (2007). "Flaviviridae: The viruses and their replication" (PDF). In D. M. Knipe; P. M. Howley (eds.). Fields Virology (5th ed.). Philadelphia: Lippincott-Raven Publishers. pp. 1101–1133.
  11. ^ Kim, M.- J.; Kao, C. (2001). "Factors regulating template switch in vitro by viral RNA-dependent RNA polymerases: Implications for RNA-RNA recombination". Proceedings of the National Academy of Sciences. 98 (9): 4972–4977. Bibcode:2001PNAS...98.4972K. doi:10.1073/pnas.081077198. PMC 33148. PMID 11309487.
  12. ^ Ridpath, J.F.; Bolin, S.R.; Dubovi, E.J. (1994). "Segregation of bovine viral diarrhoea virus into genotypes". Virology. 205 (1): 66–74. doi:10.1006/viro.1994.1620. PMID 7975238.
  13. ^ Peterhans, Ernst; Bachofen, Claudia; Stalder, Hanspeter; Schweizer, Matthias (2010). "Cytopathic bovine viral diarrhea viruses (BVDV): emerging pestiviruses doomed to extinction". Veterinary Research. 41 (6): 44. doi:10.1051/vetres/2010016. PMC 2850149. PMID 20197026.
  14. ^ Gillespie, J. H.; Madin, S. H.; Darby, N. B. (1962). "Cellular resistance in tissue culture, induced by noncytopathogenic strains, to a cytopathogenic strain of virus diarrhea virus of cattle". Proceedings of the Society for Experimental Biology and Medicine. 110 (2): 248–50. doi:10.3181/00379727-110-27481. PMID 13898635. S2CID 12198102.
  15. ^ Qi, Fengxia; Ridpath, Julia F.; Berry, Eugene S. (1998). "Insertion of a bovine SMT3B gene in NS4B and duplication of NS3 in a bovine viral diarrhea virus genome correlate with the cytopathogenicity of the virus". Virus Research. 57 (1): 1–9. doi:10.1016/s0168-1702(98)00073-2. PMID 9833880.
  16. ^ a b Moennig, Volker; Houe, Hans; Lindberg, Ann (2005). "BVD control in Europe: current status and perspectives". Animal Health Research Reviews. 6 (1): 63–74. doi:10.1079/ahr2005102. PMID 16164009. S2CID 10581576.
  17. ^ Anon (2005). EU Thematic network on control of bovine viral diarrhoea virus (BVDV). Position Paper.
  18. ^ Wolfmeyer, A.; Wolf, G.; Beer, M.; Strube, W.; Hehnen, H. R.; Schmeer, N.; Kaaden, O. R. (1997). "Genomic (50-UTR) and serological differences among German BVDV field isolates". Archives of Virology. 142 (10): 2049–2057. doi:10.1007/s007050050222. PMID 9413513. S2CID 20365815.
  19. ^ Brownlie, J.; Clarke, M. C.; Howard, C. J.; Pocock, D. H. (1987). "Pathogenesis and epidemiology of bovine virus diarrhoea virus infection of cattle". Annales de Recherches Vétérinaires. 18 (2): 157–66. PMID 3619343.
  20. ^ Fray, M. D.; Clarke, M. C.; Thomas, L. H.; McCauley, J. W.; Charleston, B. (1998). "Prolonged nasal shedding and viraemia of cytopathogenic bovine virus diarrhoea virus in experimental late-onset mucosal disease". Veterinary Record. 143 (22): 608–11. doi:10.1136/vr.143.22.608. PMID 9871955. S2CID 26025942.
  21. ^ Chase, Christopher C.L; Elmowalid, Gamal; Yousif, Ausama A.A (2004). "The immune response to bovine viral diarrhea virus: a constantly changing picture". The Veterinary Clinics of North America. Food Animal Practice. 20 (1): 95–114. doi:10.1016/j.cvfa.2003.11.004. PMID 15062477.
  22. ^ Voges, H; Young, S; Nash, M (2006). "Direct adverse effects of persistent BVDV infection in dairy heifers – A retrospective case control study". VetScript. 19 (8): 22–25.
  23. ^ Moennig, Volker; Liess, Bernd (1995). "Pathogenesis of intrauterine infections with bovine viral diarrhoea virus". Veterinary Clinics of North America: Food Animal Practice. 11 (3): 477–488. doi:10.1016/S0749-0720(15)30462-X. PMID 8581858.
  24. ^ Brownlie, J.; Clarke, M.; Howard, C. (1984). "Experimental production of fatal mucosal disease in cattle". The Veterinary Record. 114 (22): 535–6. doi:10.1136/vr.114.22.535. PMID 6087539. S2CID 19523700.
  25. ^ Hult and Lindberg (2005) Prev Vet Med 72: 143–148
  26. ^ Rikula et al. (2005) Prev Vet Med 72: 139–142
  27. ^ Rossmanith et al. (2005) Prev Vet Med 72: 133–137
  28. ^ Stahl and Alenius (2012) Japanese Journal of Veterinary Research 60 (Supplement) S31–39.
  29. ^ Ridpath (2013) Biologicals 41: 14–19.
  30. ^ O’Rourke (2002) Journal of the American Veterinary Medical Association 220(12): 1770–1772
  31. ^ Graham et al. (2004) Revista Portuguesa de ciencias veterinarias 127: 38.

External links edit

  • New York State Cattle Health Assurance Program BVD Module
  • Description of the entity on the Merck Veterinary Manual
  • Animal viruses
  • Bovine Viral Diarrhea Resource Page
  • Specialist BVD site, Royal Veterinary College, London

bovine, viral, diarrhea, bovine, viral, diarrhoea, english, mucosal, disease, previously, referred, bovine, virus, diarrhea, economically, significant, disease, cattle, that, found, majority, countries, throughout, world, worldwide, reviews, economically, asse. Bovine viral diarrhea BVD bovine viral diarrhoea UK English or mucosal disease previously referred to as bovine virus diarrhea BVD is an economically significant disease of cattle that is found in the majority of countries throughout the world 1 Worldwide reviews of the economically assessed production losses and intervention programs e g eradication programs vaccination strategies and biosecurity measures incurred by BVD infection have been published 2 3 The causative agent bovine viral diarrhea virus BVDV is a member of the genus Pestivirus of the family Flaviviridae 1 Bovine viral diarrheaImmunofluorescence image of BVDV CP7 type Nuclei are stained blue with DAPI The replication complexes of the viruses are marked red by NS3 protein binding antibodiesScientific classification unranked VirusRealm RiboviriaKingdom OrthornaviraePhylum KitrinoviricotaClass FlasuviricetesOrder AmarilloviralesFamily FlaviviridaeGenus PestivirusGroups includedPestivirus A formerly Bovine viral diarrhea virus 1 Pestivirus B formerly Bovine viral diarrhea virus 2 Cladistically included but traditionally excluded taxaPestivirus C Pestivirus D Pestivirus E Pestivirus F Pestivirus G Pestivirus H Pestivirus I Pestivirus J Pestivirus KTongue lesions on confirmed BVD MD case mucosal disease form BVD infection results in a wide variety of clinical signs due to its immunosuppressive effects 4 as well as having a direct effect on respiratory disease and fertility 5 In addition BVD infection of a susceptible dam during a certain period of gestation can result in the production of a persistently infected PI fetus 6 PI animals recognise intra cellular BVD viral particles as self and shed virus in large quantities throughout life they represent the cornerstone of the success of BVD as a disease Currently it was shown in a worldwide review study that the PI prevalence at animal level ranged from low 0 8 Europe North America Australia medium gt 0 8 to 1 6 East Asia to high gt 1 6 West Asia Countries that had failed to implement any BVDV control and or eradication programmes including vaccination had the highest PI prevalence 7 Contents 1 Virus classification and structure 2 Epidemiology 3 Pathogenesis 3 1 Acute transient infection 3 2 Intrauterine infections 3 3 Chronic infections 4 Clinical signs 4 1 PI animals 4 2 Mucosal disease 5 Diagnosis 5 1 Virus or antigen detection 5 2 BVD antibody detection 6 Eradication and control 7 Vaccination 8 See also 9 References 10 External linksVirus classification and structure editBVDVs are members of the genus Pestivirus belonging to the family Flaviviridae Other members of this genus cause Border disease sheep and classical swine fever pigs which cause significant financial loss to the livestock industry 8 Pestiviruses are small spherical single stranded enveloped RNA viruses of 40 to 60 nm in diameter 9 The genome consists of a single linear positive sense single stranded RNA molecule of approximately 12 3 kb 10 RNA synthesis is catalyzed by the BVDV RNA dependent RNA polymerase RdRp This RdRp can undergo template strand switching allowing RNA RNA copy choice recombination during elongative RNA synthesis 11 Two BVDV genotypes are recognised based on the nucleotide sequence of the 5 untranslated UTR region BVDV 1 and BVDV 2 12 BVDV 1 isolates have been grouped into 16 subtypes a p and BVDV 2 has currently been grouped into 3 subtypes a c 13 BVDV strains can be further divided into distinct biotypes cytopathic or non cytopathic according to their effects on tissue cell culture cytopathic cp biotypes formed via mutation of non cytopathic ncp biotypes induce apoptosis in cultured cells 14 Ncp viruses can induce persistent infection in cells and have an intact NS2 3 protein In cp viruses the NS2 3 protein is either cleaved to NS2 and NS3 or there is a duplication of viral RNA containing an additional NS3 region 15 The majority of BVDV infections in the field are caused by the ncp biotype 1 Epidemiology editBVD is considered one of the most significant infectious diseases in the livestock industry worldwide due to its high prevalence persistence and clinical consequences 16 In Europe the prevalence of antibody positive animals in countries without systematic BVD control is between 60 and 80 17 Prevalence has been determined in individual countries and tends to be positively associated with stocking density of cattle citation needed BVDV 1 strains are predominant in most parts of the world whereas BVDV 2 represents 50 of cases in North America 16 In Europe BVDV 2 was first isolated in the UK in 2000 and currently represents up to 11 of BVD cases in Europe 18 Transmission of BVDV occurs both horizontally and vertically with both persistently and transiently infected animals excreting infectious virus Virus is transmitted via direct contact bodily secretions and contaminated fomites with the virus being able to persist in the environment for more than two weeks Persistently infected animals are the most important source of the virus continuously excreting a viral load one thousand times that shed by acutely infected animals 19 Pathogenesis edit nbsp Turbinate cells infected with BVDVAcute transient infection edit Following viral entry and contact with the mucosal lining of the mouth or nose replication occurs in epithelial cells BVDV replication has a predilection for the palatine tonsils lymphoid tissues and epithelium of the oropharynx Phagocytes take up BVDV or virus infected cells and transport them to peripheral lymphoid tissues the virus can also spread systemically through the bloodstream Viraemia occurs 2 4 days after exposure and virus isolation from serum or leukocytes is generally possible between 3 10 days post infection 20 During systemic spread the virus is able to gain entry into most tissues with a preference for lymphoid tissues Neutralising antibodies can be detected from 10 to 14 days post infection with titres continuing to increase slowly for 8 10 weeks After 2 3 weeks antibodies effectively neutralise viral particles promote clearance of virus and prevent seeding of target organs 21 Intrauterine infections edit Fetal infection is of most consequence as this can result in the birth of a persistently infected neonate The effects of fetal infection with BVDV are dependent upon the stage of gestation at which the dam suffers acute infection BVDV infection of the dam prior to conception and during the first 18 days of gestation results in delayed conception and an increased calving to conception interval Once the embryo is attached infection from days 29 41 can result in embryonic infection and resultant embryonic death Infection of the dam from approximately day 30 of gestation until day 120 can result in immunotolerance and the birth of calves persistently infected with the virus BVDV infection between 80 and 150 days of gestation may be teratogenic with the type of birth defect dependent upon the stage of fetal development at infection Abortion may occur at any time during gestation Infection after approximately day 120 can result in the birth of a normal fetus which is BVD antigen negative and BVD antibody positive This occurs because the fetal immune system has developed by this stage of gestation and has the ability to recognise and fight off the invading virus producing anti BVD antibodies Chronic infections edit BVD virus can be maintained as a chronic infection within some immunoprivileged sites following transient infection These sites include ovarian follicles testicular tissues central nervous system and white blood cells Cattle with chronic infections elicit a significant immune response exhibited by extremely high antibody titres Clinical signs editBVDV infection has a wide manifestation of clinical signs including fertility issues milk drop pyrexia diarrhea and fetal infection 9 Occasionally a severe acute form of BVD may occur These outbreaks are characterized by thrombocytopenia with high morbidity and mortality However clinical signs are frequently mild and infection insidious recognized only by BVDV s immunosuppressive effects perpetuating other circulating infectious diseases particularly scours and pneumonias PI animals edit Persistently infected animals did not have a competent immune system at the time of BVDV transplacental infection The virus therefore entered the fetal cells and during immune system development was accepted as self In PIs the virus remains present in a large number of the animal s body cells throughout its life and is continuously shed PIs are often ill thrifty and smaller than their peers however they can appear normal PIs are more susceptible to disease with only 20 of PIs surviving to two years of age 22 If a PI dam is able to reproduce they always give birth to PI calves 23 Mucosal disease edit The PI cattle that do survive ill thrift are susceptible to mucosal disease Mucosal disease only develops in PI animals and is invariably fatal 5 Disease results when a PI animal is superinfected with a cytopathic biotype arising from mutation of the non cytopathic strain of BVDV already circulating in that animal 24 The cp BVDV spreads to the gastro intestinal epithelium and necrosis of keratinocytes results in erosion and ulceration Fluid leaks from the epithelial surface of the gastro intestinal tract causing diarrhoea and dehydration In addition bacterial infection of the damaged epithelium results in secondary septicaemia Death occurs in the ensuing days or weeks Diagnosis editVarious diagnostic tests are available for the detection of either active infection or evidence of historical infection The method of diagnosis used also depends upon whether the vet is investigating at an individual or a herd level Virus or antigen detection edit Antigen ELISA and rtPCR are currently the most frequently performed tests to detect virus or viral antigen Individual testing of ear tissue tag samples or serum samples is performed It is vital that repeat testing is performed on positive samples to distinguish between acute transiently infected cattle and PIs A second positive result acquired at least three weeks after the primary result indicates a PI animal rtPCR can also be used on bulk tank milk BTM samples to detect any PI cows contributing to the tank It is reported that the maximum number of contributing cows from which a PI can be detected is 300 BVD antibody detection edit Antibody Ig ELISAs are used to detect historical BVDV infection these tests have been validated in serum milk and bulk milk samples Ig ELISAs do not diagnose active infection but detect the presence of antibodies produced by the animal in response to viral infection Vaccination also induces an antibody response which can result in false positive results therefore it is important to know the vaccination status of the herd or individual when interpreting results A standard test to assess whether virus has been circulating recently is to perform an Ig ELISA on blood from 5 10 young stock that have not been vaccinated aged between 9 and 18 months A positive result indicates exposure to BVDV but also that any positive animals are very unlikely to be PI animals themselves A positive result in a pregnant female indicates that she has previously been either vaccinated or infected with BVDV and could possibly be carrying a PI fetus so antigen testing of the newborn is vital to rule this out 5 A negative antibody result at the discretion of the responsible veterinarian may require further confirmation that the animal is not in fact a PI At a herd level a positive Ig result suggests that BVD virus has been circulating or the herd is vaccinated Negative results suggest that a PI is unlikely however this naive herd is in danger of severe consequences should an infected animal be introduced Antibodies from wild infection or vaccination persist for several years therefore Ig ELISA testing is more valuable when used as a surveillance tool in seronegative herds Eradication and control editThe mainstay of eradication is the identification and removal of persistently infected animals Re infection is then prevented by vaccination and high levels of biosecurity supported by continuing surveillance PIs act as viral reservoirs and are the principal source of viral infection but transiently infected animals and contaminated fomites also play a significant role in transmission 1 Leading the way in BVD eradication almost 20 years ago were the Scandinavian countries Despite different conditions at the start of the projects in terms of legal support and regardless of initial prevalence of herds with PI animals it took all countries approximately 10 years to reach their final stages 25 26 Once proven that BVD eradication could be achieved in a cost efficient way a number of regional programmes followed in Europe some of which have developed into national schemes 27 Vaccination is an essential part of both control and eradication While BVD virus is still circulating within the national herd breeding cattle are at risk of producing PI neonates and the economic consequences of BVD are still relevant Once eradication has been achieved unvaccinated animals will represent a naive and susceptible herd Infection from imported animals or contaminated fomites brought into the farm or via transiently infected in contacts will have devastating consequences Vaccination editModern vaccination programmes aim not only to provide a high level of protection from clinical disease for the dam but crucially to protect against viraemia and prevent the production of PIs 28 While the immune mechanisms involved are the same the level of immune protection required for foetal protection is much higher than for prevention of clinical disease 29 While challenge studies indicate that killed as well as live vaccines prevent foetal infection under experimental conditions the efficacy of vaccines under field conditions has been questioned 30 The birth of PI calves into vaccinated herds suggests that killed vaccines do not stand up to the challenge presented by the viral load excreted by a PI in the field 31 See also editAnimal virusesReferences edit a b c d Fray M D Paton D J Alenius S et al 2000 The effects of bovine viral diarrhoea virus on cattle reproduction in relation to disease control Animal Reproduction Science 60 61 615 27 doi 10 1016 s0378 4320 00 00082 8 PMID 10844229 Richter V Lebl K Baumgartner W Obritzhauser W Kasbohrer A Pinior B 2017 A systematic worldwide review of the direct monetary losses due to bovine viral diarrhea virus infection The Veterinary Journal 220 80 87 doi 10 1016 j tvjl 2017 01 005 PMID 28190502 Pinior B Firth C Richter V Lebl K Trauffler M Dzieciol M Hutter S Burgstaller J Obritzhauser W Winter P Kasbohrer A 2017 A systematic review of financial and economic assessments of bovine viral diarrhea virus BVDV prevention and mitigation activities worldwide Preventive Veterinary Medicine 137 Pt A 77 92 doi 10 1016 j prevetmed 2016 12 014 PMID 28040270 Schaut Robert G McGill Jodi L Neill John D Ridpath Julia F Sacco Randy E 2015 10 02 Bovine viral diarrhea virus type 2 in vivo infection modulates TLR4 responsiveness in differentiated myeloid cells which is associated with decreased MyD88 expression Virus Research 208 44 55 doi 10 1016 j virusres 2015 05 017 ISSN 1872 7492 PMID 26043978 a b c Lanyon Sasha R Hill Fraser I Reichel Michael P Brownlie Joe et al 2014 Bovine Viral Diarrhoea Pathogenesis and diagnosis PDF Veterinary Journal 199 2 201 9 doi 10 1016 j tvjl 2013 07 024 PMID 24053990 Grooms Daniel L 2004 Reproductive consequences of infection with bovine viral diarrhea virus Veterinary Clinics of North America Food Animal Practice 20 1 5 19 doi 10 1016 j cvfa 2003 11 006 PMID 15062471 Scharnbock B Roch Franz Ferdinand Richter V Funke C Firth C Obritzhauser W Baumgartner W Kasbohrer A Pinior B 2018 A meta analysis of bovine viral diarrhoea virus BVDV prevalences in the global cattle population Scientific Reports 8 1 14420 Bibcode 2018NatSR 814420S doi 10 1038 s41598 018 32831 2 PMC 6158279 PMID 30258185 Hornberg Andrea Fernandez Sandra Revilla Vogl Claus Vilcek Stefan Matt Monika Fink Maria Kofer Josef Schopf Karl 2009 Genetic diversity of pestivirus isolates in cattle from Western Austria PDF Veterinary Microbiology 135 3 4 205 213 doi 10 1016 j vetmic 2008 09 068 PMID 19019571 S2CID 46378359 a b N James MacLachlan Edward J Dubovi eds 2011 Fenner s Veterinary Virology 4th ed Elsevier Brett D Lindenbach Heinz Jurgen Thiel Charles M Rice 2007 Flaviviridae The viruses and their replication PDF In D M Knipe P M Howley eds Fields Virology 5th ed Philadelphia Lippincott Raven Publishers pp 1101 1133 Kim M J Kao C 2001 Factors regulating template switch in vitro by viral RNA dependent RNA polymerases Implications for RNA RNA recombination Proceedings of the National Academy of Sciences 98 9 4972 4977 Bibcode 2001PNAS 98 4972K doi 10 1073 pnas 081077198 PMC 33148 PMID 11309487 Ridpath J F Bolin S R Dubovi E J 1994 Segregation of bovine viral diarrhoea virus into genotypes Virology 205 1 66 74 doi 10 1006 viro 1994 1620 PMID 7975238 Peterhans Ernst Bachofen Claudia Stalder Hanspeter Schweizer Matthias 2010 Cytopathic bovine viral diarrhea viruses BVDV emerging pestiviruses doomed to extinction Veterinary Research 41 6 44 doi 10 1051 vetres 2010016 PMC 2850149 PMID 20197026 Gillespie J H Madin S H Darby N B 1962 Cellular resistance in tissue culture induced by noncytopathogenic strains to a cytopathogenic strain of virus diarrhea virus of cattle Proceedings of the Society for Experimental Biology and Medicine 110 2 248 50 doi 10 3181 00379727 110 27481 PMID 13898635 S2CID 12198102 Qi Fengxia Ridpath Julia F Berry Eugene S 1998 Insertion of a bovine SMT3B gene in NS4B and duplication of NS3 in a bovine viral diarrhea virus genome correlate with the cytopathogenicity of the virus Virus Research 57 1 1 9 doi 10 1016 s0168 1702 98 00073 2 PMID 9833880 a b Moennig Volker Houe Hans Lindberg Ann 2005 BVD control in Europe current status and perspectives Animal Health Research Reviews 6 1 63 74 doi 10 1079 ahr2005102 PMID 16164009 S2CID 10581576 Anon 2005 EU Thematic network on control of bovine viral diarrhoea virus BVDV Position Paper Wolfmeyer A Wolf G Beer M Strube W Hehnen H R Schmeer N Kaaden O R 1997 Genomic 50 UTR and serological differences among German BVDV field isolates Archives of Virology 142 10 2049 2057 doi 10 1007 s007050050222 PMID 9413513 S2CID 20365815 Brownlie J Clarke M C Howard C J Pocock D H 1987 Pathogenesis and epidemiology of bovine virus diarrhoea virus infection of cattle Annales de Recherches Veterinaires 18 2 157 66 PMID 3619343 Fray M D Clarke M C Thomas L H McCauley J W Charleston B 1998 Prolonged nasal shedding and viraemia of cytopathogenic bovine virus diarrhoea virus in experimental late onset mucosal disease Veterinary Record 143 22 608 11 doi 10 1136 vr 143 22 608 PMID 9871955 S2CID 26025942 Chase Christopher C L Elmowalid Gamal Yousif Ausama A A 2004 The immune response to bovine viral diarrhea virus a constantly changing picture The Veterinary Clinics of North America Food Animal Practice 20 1 95 114 doi 10 1016 j cvfa 2003 11 004 PMID 15062477 Voges H Young S Nash M 2006 Direct adverse effects of persistent BVDV infection in dairy heifers A retrospective case control study VetScript 19 8 22 25 Moennig Volker Liess Bernd 1995 Pathogenesis of intrauterine infections with bovine viral diarrhoea virus Veterinary Clinics of North America Food Animal Practice 11 3 477 488 doi 10 1016 S0749 0720 15 30462 X PMID 8581858 Brownlie J Clarke M Howard C 1984 Experimental production of fatal mucosal disease in cattle The Veterinary Record 114 22 535 6 doi 10 1136 vr 114 22 535 PMID 6087539 S2CID 19523700 Hult and Lindberg 2005 Prev Vet Med 72 143 148 Rikula et al 2005 Prev Vet Med 72 139 142 Rossmanith et al 2005 Prev Vet Med 72 133 137 Stahl and Alenius 2012 Japanese Journal of Veterinary Research 60 Supplement S31 39 Ridpath 2013 Biologicals 41 14 19 O Rourke 2002 Journal of the American Veterinary Medical Association 220 12 1770 1772 Graham et al 2004 Revista Portuguesa de ciencias veterinarias 127 38 Bovine Viral Diarrhoea Virus expert reviewed and published by Wikivet at http en wikivet net Bovine Viral Diarrhoea Virus accessed 21 07 2011External links editNew York State Cattle Health Assurance Program BVD Module Description of the entity on the Merck Veterinary Manual Animal viruses Bovine Viral Diarrhea Resource Page Specialist BVD site Royal Veterinary College London Retrieved from https en wikipedia org w index php title Bovine viral diarrhea amp oldid 1188449075, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.