Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Bull Soc Pathol Exot ; 107(3): 137-8, 135-6, 2014 Aug.
Article in English, French | MEDLINE | ID: mdl-24566884

ABSTRACT

The eradication of rinderpest virus was less celebrated than the eradication of smallpox virus. However, this is only the second campaign to eradicate a virus worldwide which is successful. This gives the opportunity to recall how important rinderpest had been these past centuries for farmers and for public health.


Subject(s)
Disease Eradication/methods , Rinderpest/prevention & control , Animals , Cattle , Cattle Diseases/prevention & control , Cattle Diseases/virology , Humans , Mass Vaccination/veterinary , Rinderpest virus/pathogenicity
2.
Emerg Infect Dis ; 19(1): 151-3, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23260811

ABSTRACT

After the 2011 declaration of rinderpest disease eradication, we surveyed 150 countries about rinderpest virus stocks. Forty-four laboratories in 35 countries held laboratory-attenuated strains, field strains, or diagnostic samples. Vaccine and reagent production and laboratory experiments continued. Rigorous standards are necessary to ensure that stocks are kept under safe conditions.


Subject(s)
Biohazard Release/prevention & control , Disease Eradication , Rinderpest virus/growth & development , Rinderpest/prevention & control , Viral Vaccines/supply & distribution , Animals , Biological Specimen Banks , Humans , Rinderpest/immunology , Rinderpest/virology , Rinderpest virus/pathogenicity , Surveys and Questionnaires , Vaccines, Attenuated , Viral Vaccines/biosynthesis , Viral Vaccines/immunology
3.
J Virol ; 86(23): 13115-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23118448

ABSTRACT

The genomes of three South Korean Rinderpest virus vaccine strains (L72, LA77, and LA96) were analyzed in order to investigate their genetic variability. These three vaccine strains were all derived from the same virus strain origin (Fusan) through repeated passages in different culture systems. The full genome length of the three strains was 15,882 nucleotides, and the sequence similarity between the three South Korean RPV strains at the nucleotide level was 98.1 to 98.9%. The genetic distance between Nakamura III, L72, LA77, LA96, and LATC06 and the Kabete strain was greater than that between the Fusan and Kabete strains for the P, V, and C genes. The difference in pathogenicity among these strains might be due to the V gene, which has a positive (>1) selection ratio based on the analysis of synonymous (dS) and nonsynonymous (dN) substitution rates (dN/dS ratio [ω]).


Subject(s)
Genetic Variation , Genome, Viral/genetics , Phylogeny , Rinderpest virus/genetics , Viral Vaccines/genetics , Base Sequence , Bayes Theorem , Evolution, Molecular , Models, Genetic , Molecular Sequence Data , Republic of Korea , Rinderpest virus/pathogenicity , Sequence Analysis, DNA , Serial Passage/methods
4.
Science ; 337(6100): 1309-12, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22984063

ABSTRACT

Rinderpest is only the second infectious disease to have been globally eradicated. In the final stages of eradication, the virus was entrenched in pastoral areas of the Greater Horn of Africa, a region with weak governance, poor security, and little infrastructure that presented profound challenges to conventional control methods. Although the eradication process was a development activity rather than scientific research, its success owed much to several seminal research efforts in vaccine development and epidemiology and showed what scientific decision-making and management could accomplish with limited resources. The keys to success were the development of a thermostable vaccine and the application of participatory epidemiological techniques that allowed veterinary personnel to interact at a grassroots level with cattle herders to more effectively target control measures.


Subject(s)
Disease Eradication/methods , Environmental Monitoring/methods , Rinderpest virus , Rinderpest/prevention & control , Vaccination/methods , Viral Vaccines/administration & dosage , Africa/epidemiology , Animals , Cattle , Epidemiological Monitoring , Hot Temperature , Immunization Programs , Rinderpest/epidemiology , Rinderpest/immunology , Rinderpest virus/immunology , Rinderpest virus/isolation & purification , Rinderpest virus/pathogenicity , Viral Vaccines/chemistry , Viral Vaccines/immunology
5.
Curr Opin Virol ; 2(3): 330-4, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22709518

ABSTRACT

In 2011 the Food and Agriculture Organization formally announced that rinderpest was eradicated from the globe. Rinderpest virus had long been associated with huge disease outbreaks among cattle. The disease not only had a devastating effect on cattle herds world-wide, but also on human populations that depended on them. Rinderpest virus - a member of the genus Morbillivirus of the family Paramyxoviridae - is a close relative of measles virus. Both viruses are highly infectious and share many other biological properties. Although no formal goal or timeframe has been set, plans are currently being developed to eradicate measles. Here, we discuss how lessons learned from the global eradication of rinderpest may help in the future eradication of measles.


Subject(s)
Disease Eradication/methods , Measles/epidemiology , Measles/prevention & control , Rinderpest/epidemiology , Rinderpest/prevention & control , Animals , Cattle , Global Health , Humans , Measles virus/immunology , Measles virus/pathogenicity , Rinderpest virus/immunology , Rinderpest virus/pathogenicity , United Nations
6.
Vaccine ; 30(11): 1927-35, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22265946

ABSTRACT

The eradication of rinderpest virus (RPV) from the globe was possible through the availability of a safe and effective live attenuated vaccine and a suitable companion diagnostic test. However, the inability to serologically 'Differentiate between naturally Infected and Vaccinated Animals' (DIVA) meant that both the time taken to complete the eradication programme and the economic burden on countries involved was significantly greater than if a vaccine and companion diagnostic test that fulfilled the DIVA concept had been available. During the RPV eradication campaign serosurveillance for RPV was primarily based on a competitive ELISA using a RPV specific (C1) monoclonal antibody (mAb) directed against the viral haemagglutinin (H) protein but this test was not able to meet DIVA requirements. To provide proof of concept for the generation of novel morbillivirus DIVA vaccines we have identified, by phage display, and mutated residues critical for C1 mAb binding and assessed the functionality of mutants in an in vitro fusion assay. Finally we have incorporated mutated epitopes into a full length clone and rescued recombinant RPV using reverse genetics techniques. Here we describe a novel mechanism of marking morbillivirus vaccines, using RPV as a proof of concept, and discuss the applicability of this method to the development of marked vaccines for peste des petits ruminants virus (PPRV).


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes/immunology , Hemagglutinins, Viral/immunology , Vaccines, DNA/biosynthesis , Viral Vaccines/biosynthesis , Amino Acid Sequence , Animals , Binding Sites, Antibody , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Hemagglutinins, Viral/genetics , Molecular Sequence Data , Mutation , Peptide Library , Peste-des-Petits-Ruminants/prevention & control , Peste-des-petits-ruminants virus/immunology , Peste-des-petits-ruminants virus/pathogenicity , Protein Structure, Tertiary , Rinderpest/prevention & control , Rinderpest virus/genetics , Rinderpest virus/immunology , Rinderpest virus/pathogenicity , Vaccines, DNA/immunology , Vero Cells , Viral Vaccines/immunology
7.
Uirusu ; 62(2): 175-82, 2012.
Article in Japanese | MEDLINE | ID: mdl-24153228

ABSTRACT

The genus Morbillivirus in the family Paramyxoviridae contains many pathogens, which are important for medicine or veterinary medicine. Because each morbillivirus has restricted host range and serologically monotypic, the virus infection and transmission is effectively controlled by vaccinations and surveillance. Rinderpest virus has been eradicated in 2011, and elimination of measles virus progresses worldwide. Recently, a new cell receptor for measles virus, nectin4 was identified. Both SLAM, a molecule expressing on immune cells, and nectin4, a molecule expressing on epithelial cells, are important to infectivity and pathogenicity of the virus.


Subject(s)
Cattle Diseases/virology , Distemper Virus, Canine , Dog Diseases/virology , Measles virus , Morbillivirus , Animals , Cattle , Distemper/virology , Distemper Virus, Canine/genetics , Distemper Virus, Canine/pathogenicity , Distemper Virus, Canine/physiology , Dogs , Epithelial Cells/virology , Genetic Structures , Genome, Viral , Humans , Measles/epidemiology , Measles/virology , Measles virus/genetics , Measles virus/pathogenicity , Measles virus/physiology , Morbillivirus/genetics , Morbillivirus/pathogenicity , Morbillivirus/physiology , Pneumovirinae , Protein Binding , Receptors, Virus , Rinderpest/virology , Rinderpest virus/pathogenicity , Virus Replication
9.
J Gen Virol ; 91(Pt 12): 2918-27, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20719989

ABSTRACT

A full-length DNA clone of a virulent strain of rinderpest virus was constructed with the gene for the enhanced green fluorescent protein (eGFP) inserted as a separate transcription unit between the P and M genes. Rescue of the virus from the modified clone using reverse genetics generated a virus that grew to the same levels as the virus rescued from the unmodified DNA clone in cell culture. The recombinant virus expressed eGFP to a high level and was used to follow virus replication in real-time using live-cell imaging. Cattle infected with both the recombinant wild-type virus and the recombinant eGFP expressing virus developed clinical disease similar to that of the wild-type natural virus isolate. Detection of virus in circulating peripheral blood leukocytes was equivalent to that of the animals infected with the wild-type virus. The high level of expression of soluble eGFP by this virus allowed us to detect viral replication in infected animals by confocal microscopy. Imaging vibrating microtome sections by confocal microscopy provided good preservation of tissue and cellular architecture as well as revealing the sites of replication of the virus in different tissues of infected animals.


Subject(s)
Cattle Diseases/virology , Genes, Reporter , Green Fluorescent Proteins/metabolism , Rinderpest virus/pathogenicity , Rinderpest/virology , Staining and Labeling/methods , Virology/methods , Animals , Cattle , Cattle Diseases/pathology , Cell Line , Chlorocebus aethiops , Green Fluorescent Proteins/genetics , Image Processing, Computer-Assisted/methods , Leukocytes/virology , Microscopy, Confocal , Rinderpest/pathology , Rinderpest virus/genetics , Rinderpest virus/growth & development , Transcription, Genetic , Virulence
10.
Arch Virol ; 155(4): 455-62, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20127373

ABSTRACT

ErbB3 binding protein Ebp1 has been shown to downregulate ErbB3 receptor-mediated signaling to inhibit cell proliferation. Rinderpest virus belongs to the family Paramyxoviridae and is characterized by the presence of a non-segmented negative-sense RNA genome. In this work, we show that rinderpest virus infection of Vero cells leads to the down-regulation of the host factor Ebp1, at both the mRNA and protein levels. Ebp1 protein has been shown to co-localize with viral inclusion bodies in infected cells, and it is packaged into virions, presumably through its interaction with the N protein or the N-RNA itself. Overexpression of Ebp1 inhibits viral transcription and multiplication in infected cells, suggesting that a mutual antagonism operates between host factor Ebp1 and the virus.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Down-Regulation , Gene Expression , Rinderpest virus/pathogenicity , Animals , Chlorocebus aethiops , Vero Cells
12.
J Gen Virol ; 86(Pt 12): 3349-3355, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16298981

ABSTRACT

Rinderpest, or cattle plague, is caused by Rinderpest virus (RPV), which is related most closely to human Measles virus (MV), both being members of the genus Morbillivirus, a group of viruses known to have strong immunosuppressive effects in vitro and in vivo. Here, it was shown that peripheral blood mononuclear cells (PBMCs) isolated from cattle experimentally infected with either wild-type or vaccine strains of RPV impaired the proliferation of PBMCs derived from uninfected animals; however, in contrast to either mild or virulent strains of wild-type virus, the inhibition induced by the vaccine was both weak and transient. Flow-cytometric analysis of PBMCs obtained from cattle infected with different strains of RPV showed that the proportion of infected cells was virus dose-dependent and correlated with lymphoproliferative suppression.


Subject(s)
Cell Proliferation , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Rinderpest virus/immunology , Rinderpest virus/pathogenicity , Animals , Cattle , Cell Separation , Cells, Cultured , Flow Cytometry , Formazans/metabolism , Models, Animal , Rinderpest/immunology , Rinderpest/virology , Tetrazolium Salts/metabolism , Viral Vaccines/immunology
13.
J Gen Virol ; 86(Pt 4): 1083-1092, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784902

ABSTRACT

Rinderpest virus (RPV) is a morbillivirus that causes cattle plague, a disease of large ruminants. The viral genome is flanked at the 3' and 5' genome termini by the genome promoter (GP) and antigenome promoter (AGP), respectively. These promoters play essential roles in directing replication and transcription as well as RNA encapsidation and packaging. It has previously been shown that individual changes to the GP of RPV greatly affect promoter activity in a minigenome assay and it was therefore proposed that individual nucleotide changes in the GP and AGP might also have significant effects on the ability of the virus to replicate and cause disease in cattle. The Plowright vaccine strain of RPV has been derived by tissue-culture passage from the virulent Kabete 'O' isolate (KO) and is highly attenuated for all ruminant species in which it has been used. Here, it was shown that swapping the GP and the first 76 nt of the AGP between virulent and avirulent strains affected disease progression. In particular, it was shown that flanking the virulent strain with the vaccine GP and AGP sequences, while not appreciably affecting virus growth in vitro, led to attenuation in vivo. The reverse was not true, since the KO promoters did not alter the vaccine's attenuated nature. The GP/AGP therefore play a role in attenuation, but are not the only determinants of attenuation in this vaccine.


Subject(s)
Gene Expression Regulation, Viral , Promoter Regions, Genetic , Rinderpest virus/pathogenicity , Rinderpest/physiopathology , Transcription, Genetic , Virus Replication , Animals , Base Sequence , Cattle , Cell Line , DNA, Complementary , Genome, Viral , Leukopenia/physiopathology , Leukopenia/veterinary , Leukopenia/virology , Molecular Sequence Data , RNA, Viral/biosynthesis , Recombination, Genetic , Rinderpest/virology , Rinderpest virus/classification , Rinderpest virus/genetics , Rinderpest virus/physiology
14.
J Gen Virol ; 86(Pt 4): 1093-1101, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784903

ABSTRACT

The currently used vaccine strain of Rinderpest virus was derived by serial passage of the highly virulent Kabete 'O' strain (KO). A full-length cDNA copy of the KO strain was made from which a virus identical in pathogenicity to the wild-type virus was rescued. A series of chimeric viruses was prepared in which the coding sequences for the N, P, F, H or L proteins were replaced with the corresponding sequences from the vaccine strain. The KO-based virus with the vaccine strain H gene and that with the carboxy-terminal half of the L gene replaced with the corresponding sequence from the vaccine strain retained all or almost all of the virulence of the original KO virus. Animals infected with the KO-based virus containing the vaccine strain N, P or F gene, or the amino-terminal half of the L gene, developed high and prolonged pyrexia and leukopenia, but with reduced or absent lesions and other clinical signs; although partially attenuated, none was nearly as attenuated as the vaccine strain itself. These data indicate that the high attenuation and stability of the current vaccine are due to the accumulation of a number of separate mutations, none of which is itself so sufficiently debilitating that there is strong selective pressure in favour of the revertant.


Subject(s)
Mutation , Rinderpest virus/pathogenicity , Viral Proteins/genetics , Viral Vaccines/genetics , Animals , Cattle , Cell Line , Genome, Viral , Molecular Sequence Data , Phenotype , Recombination, Genetic , Rinderpest/physiopathology , Rinderpest/virology , Rinderpest virus/classification , Rinderpest virus/genetics , Viral Proteins/metabolism
15.
Article in English | MEDLINE | ID: mdl-15228548

ABSTRACT

Peste des petits ruminants (PPR) is an emerging, economically important viral disease of goats and sheep in the Indian subcontinent. In the present investigation, 15 hill goats were experimentally infected with 2 ml of 10% splenic suspension of a virulent isolate of PPR virus (PPR/Izatnagar/94) that had caused heavy mortality (>75%) in goats during 1994 outbreaks in northern India. More than 86% (13 of 15) animals died between 9 and 13 days post inoculation at the height of temperature or when temperatures were declining. Necropsy findings included congestion of gastrointestinal tract (GIT), nasal sinuses, consolidation of antero-ventral lobes of lungs, engorged spleen, and occasionally oedematous lymph nodes. Histopathological examination of major organs of GIT revealed degeneration and necrosis of labial mucosa, severe mucosal and submucosal congestion, degeneration and necrosis of intestinal epithelium and lymphoid cell depletion from Peyer's patches along with presence of syncytia at times. Lungs showed broncho-interstitial changes and presence of intracytoplasmic and intranuclear eosinophilic inclusions in alveolar macrophages and syncytial cells. These changes in lungs were frequently complicated with serofibrinous pneumonia (57%, eight of 14). Lymphocytolysis and occasional syncytia formation were evident in the lymphoid tissues. Immunohistochemical (IHC) findings included presence of PPR virus antigen in the labial, intestinal, and bronchiolar epithelial cells, pneumocytes, macrophages and syncytial cells in lungs, and lymphoid (intact and necrotic) and reticular cells in lymphoid organs. The findings of the study indicated the highly virulent nature of the PPR virus isolate (PPR/Izatnagar/94), causing 100% mortality and characteristic pathological changes in the target organs such as lungs, intestines and lymphoid tissues. The results of the IHC study suggested that indirect immunoperoxidase could be an alternative method in the absence of more sophisticated methods of laboratory diagnosis of PPR virus infection in goats.


Subject(s)
Goat Diseases/virology , Rinderpest virus/pathogenicity , Rinderpest/virology , Animals , Disease Outbreaks/veterinary , Female , Goat Diseases/epidemiology , Goat Diseases/pathology , Goats , India/epidemiology , Intestine, Small/microbiology , Intestine, Small/pathology , Lung/microbiology , Lung/pathology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Male , Rinderpest/epidemiology , Rinderpest/pathology , Rinderpest virus/immunology
16.
J Virol ; 78(12): 6676-81, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15163758

ABSTRACT

We previously demonstrated that the rinderpest virus (RPV) hemagglutinin (H) protein plays an important role in determining host range but that other viral proteins are clearly required for full RPV pathogenicity to be manifest in different species. To examine the effects of the RPV nucleocapsid (N) protein and phosphoprotein (P) genes on RPV cross-species pathogenicity, we constructed two new recombinant viruses in which the H and P or the H, N, and P genes of the cattle-derived RPV RBOK vaccine were replaced with those from the rabbit-adapted RPV-Lv strain, which is highly pathogenic in rabbits. The viruses rescued were designated recombinant RPV-lapPH (rRPV-lapPH) and rRPV-lapNPH, respectively. Rabbits inoculated with RPV-Lv become feverish and show leukopenia and a decrease in body weight gain, while clinical signs of infection are never observed in rabbits inoculated with RPV-RBOK or with rRPV-lapH. However, rabbits inoculated with either rRPV-lapPH or rRPV-lapNPH became pyrexic and showed leukopenia. Further, histopathological lesions and high virus titers were clearly observed in the lymphoid tissues from animals infected with rRPV-lapPH or rRPV-lapNPH, although they were not observed in rabbits infected with RPV-RBOK or rRPV-lapH. The clinical, virological, and histopathological signs in rabbits infected with the two new recombinant viruses did not differ significantly; therefore, the RPV P gene was considered to be a key determinant of cross-species pathogenicity.


Subject(s)
Phosphoproteins/genetics , Phosphoproteins/metabolism , Rinderpest virus/pathogenicity , Rinderpest/physiopathology , Viral Proteins/genetics , Viral Proteins/metabolism , Animals , Cattle , Female , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Nucleocapsid Proteins/genetics , Rabbits , Recombination, Genetic , Rinderpest/pathology , Rinderpest/virology , Rinderpest virus/genetics , Species Specificity , Virulence
17.
J Comp Pathol ; 127(1): 14-21, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12354541

ABSTRACT

The presence, type, and extent of cellular death in lymphatic tissues of cattle experimentally infected with rinderpest virus strains of different virulence was investigated morphologically. Cells with DNA strand breaks were identified in histological sections of palatine tonsil, spleen, and mesenteric and mandibular lymph nodes by the TUNEL (terminal desoxynucleotidyl transferase-mediated dUTP nick end labelling) assay. In addition, representative samples of lymphatic tissues were examined by transmission electron microscopy. The results indicated that cellular disassembly in lymphatic tissues was caused by both apoptosis and oncosis. Cells with DNA strand breaks were observed in follicular and parafollicular areas of lymphatic tissues and their numbers were determined. A significant correlation was found between the number of TUNEL-positive cells and viral virulence. These results suggest that, in addition to oncosis, apoptotic cellular death in lymphatic tissues contributes substantially to the pathogenesis of rinderpest.


Subject(s)
Apoptosis , Cattle Diseases/pathology , Lymphoid Tissue/pathology , Rinderpest virus/physiology , Rinderpest/pathology , Animals , Cattle , Cattle Diseases/virology , Cell Count , Cellular Structures/ultrastructure , DNA/analysis , Female , Fluorescent Antibody Technique, Indirect/veterinary , Immunoenzyme Techniques/veterinary , In Situ Nick-End Labeling/veterinary , Lymphoid Tissue/virology , Rinderpest/etiology , Rinderpest/virology , Rinderpest virus/classification , Rinderpest virus/immunology , Rinderpest virus/pathogenicity , Species Specificity
18.
J Gen Virol ; 83(Pt 6): 1457-1463, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12029161

ABSTRACT

A major molecular determinant of virus host-range is thought to be the viral protein required for cell attachment. We used a recombinant strain of Rinderpest virus (RPV) to examine the role of this protein in determining the ability of RPV to replicate in rabbits. The recombinant was based on the RBOK vaccine strain, which is avirulent in rabbits, carrying the haemagglutinin (H) protein gene from the lapinized RPV (RPV-L) strain, which is pathogenic in rabbits. The recombinant virus (rRPV-lapH) was rescued from a cDNA of the RBOK strain in which the H gene was replaced with that from the RPV-L strain. The recombinant grew at a rate equivalent to the RPV-RBOK parental virus in B95a cells but at a lower rate than RPV-L. The H gene swap did not affect the ability of the RBOK virus to act as a vaccine to protect cattle against virulent RPV challenge. Rabbits inoculated with RPV-L became feverish, showed a decrease in body weight gain and leukopenia. High virus titres and histopathological lesions in the lymphoid tissues were also observed. Clinical signs of infection were never observed in rabbits inoculated with either RPV-RBOK or with rRPV-lapH; however, unlike RPV-RBOK, both RPV-L and rRPV-lapH induced a marked antibody response in rabbits. Therefore, the H protein plays an important role in allowing infection to occur in rabbits but other viral proteins are clearly required for full RPV pathogenicity to be manifest in this species.


Subject(s)
Glycoproteins/physiology , Hemagglutinins, Viral/physiology , Rabbits/virology , Rinderpest virus , Viral Proteins/physiology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/biosynthesis , Cattle , Cell Line , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Giant Cells , Glycoproteins/genetics , Hemagglutinins, Viral/genetics , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Necrosis , Recombination, Genetic , Rinderpest/immunology , Rinderpest/prevention & control , Rinderpest virus/chemistry , Rinderpest virus/pathogenicity , Rinderpest virus/physiology , Species Specificity , Vaccines, Synthetic/administration & dosage , Viral Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
19.
J Neurovirol ; 8(1): 45-52, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11847591

ABSTRACT

Members of the morbillivirus genus, canine distemper (CDV), phocine distemper virus (PDV), and the cetacean viruses of dolphins and porpoises exhibit high levels of CNS infection in their natural hosts. CNS complications are rare for measles virus (MV) and are not associated with rinderpest virus (RPV) and peste des petits ruminants virus (PPRV) infection. However, it is possible that all morbilliviruses infect the CNS but in some hosts are rapidly cleared by the immune response. In this study, we assessed whether RPV and PPRV have the potential to be neurovirulent. We describe the outcome of infection, of selected mouse strains, with isolates of RPV, PPRV, PDV, porpoise morbillivirus (PMV), dolphin morbillivirus (DMV), and a wild-type strain of MV. In the case of RPV virus, strains with different passage histories have been examined. The results of experiments with these viruses were compared with those using neuroadapted and vaccine strains of MV, which acted as positive and negative controls respectively. Intracerebral inoculation with RPV (Saudi/81) and PPRV (Nigeria75/1) strains produced infection in Balb/C and Cd1, but not C57 suckling mice, whereas the CAM/RB rodent-adapted strain of MV infected all three strains of mice. Weanling mice were only infected by CAM/RB. Intranasal and intraperitoneal inoculation failed to produce infection with any virus strains. We have shown that, both RPV and PPRV, in common with other morbilliviruses are neurovirulent in a permissive system. Transient infection of the CNS of cattle and goats with RPV and PPRV, respectively, remains a possibility, which could provide relevant models for the initial stages of MV infection in humans.


Subject(s)
Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/pathogenicity , Rinderpest virus/pathogenicity , Rinderpest/virology , Animals , Animals, Suckling , Brain/pathology , Brain/virology , Chlorocebus aethiops , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peste-des-Petits-Ruminants/pathology , Peste-des-petits-ruminants virus/genetics , RNA, Viral/analysis , Rinderpest/pathology , Rinderpest virus/genetics , Vero Cells , Virulence , Weaning
20.
Trop Anim Health Prod ; 34(6): 471-87, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12537386

ABSTRACT

In January 1997, Tanzania requested international assistance against rinderpest on the grounds that the virus had probably entered the country from southern Kenya. Over the next few months, a variety of attempts were made to determine the extent of the incursion by searching for serological and clinical evidence of the whereabouts of the virus. At the clinical level, these attempts were hampered by the low virulence of the strain, and at the serological level by the lack of a baseline against which contemporary interpretations could be made. Once it became apparent that neither surveillance tool was likely to produce a rapid result, an infected area was declared on common-sense grounds and emergency vaccination was initiated. The vaccination programme had two objectives, firstly to prevent any further entry across the international border, and secondly to contain and if possible eliminate rinderpest from those districts into which it had already entered. On the few occasions that clinical rinderpest was subsequently found, it was always within this provisional infected area. Emergency vaccination campaigns within the infected area ran from January to the end of March 1997 but were halted by the onset of the long rains. At this time, seromonitoring in two districts showed that viral persistence was still theoretically possible and therefore a second round of emergency vaccination was immediately organized. Further seromonitoring then indicated a large number of villages with population antibody prevalences of over 85%. These populations were considered to have been 'immunosterilized'. Although no clinical disease had been observed in them, it was decided to undertake additional vaccination in a group of districts to the south of the infected area. Serosurveillance indicated that rinderpest could have been present in a number of these districts prior to vaccination. Serosurveillance in 1998 suggested that numerous vaccinated animals had probably moved into districts outside the infected and additional vaccination areas, but did not rule out the continued presence of field infection.


Subject(s)
Antibodies, Viral/blood , Cattle Diseases/prevention & control , Rinderpest virus/immunology , Rinderpest/prevention & control , Vaccination/veterinary , Animals , Cattle , Cattle Diseases/blood , Cattle Diseases/epidemiology , Disease Outbreaks/veterinary , Rinderpest/blood , Rinderpest/epidemiology , Rinderpest virus/pathogenicity , Seroepidemiologic Studies , Tanzania/epidemiology , Viral Vaccines/immunology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...