• Aucun résultat trouvé

Genotyping Canine Distemper Virus (CDV) by a hemi-nested multiplex PCR provides a rapid approach for investigation of CDV outbreaks

N/A
N/A
Protected

Academic year: 2021

Partager "Genotyping Canine Distemper Virus (CDV) by a hemi-nested multiplex PCR provides a rapid approach for investigation of CDV outbreaks"

Copied!
35
0
0

Texte intégral

(1)

HAL Id: hal-00532193

https://hal.archives-ouvertes.fr/hal-00532193

Submitted on 4 Nov 2010

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci- entific research documents, whether they are pub- lished or not. The documents may come from teaching and research institutions in France or

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires

hemi-nested multiplex PCR provides a rapid approach for investigation of CDV outbreaks

Martella Vito, Elia Gabriella, Lucente Maria Stella, Decaro Nicola, Lorusso Eleonora, Banyai Krisztian, Blixenkrone-Møller Merete, Lan Nguyen Thi,

Yamaguchi Ryoji, Cirone Francesco, et al.

To cite this version:

Martella Vito, Elia Gabriella, Lucente Maria Stella, Decaro Nicola, Lorusso Eleonora, et al.. Geno- typing Canine Distemper Virus (CDV) by a hemi-nested multiplex PCR provides a rapid approach for investigation of CDV outbreaks. Veterinary Microbiology, Elsevier, 2007, 122 (1-2), pp.32.

�10.1016/j.vetmic.2007.01.005�. �hal-00532193�

(2)

Title: Genotyping Canine Distemper Virus (CDV) by a hemi-nested multiplex PCR provides a rapid approach for investigation of CDV outbreaks

Authors: Martella Vito, Elia Gabriella, Lucente Maria Stella, Decaro Nicola, Lorusso Eleonora, Banyai Krisztian,

Blixenkrone-Møller Merete, Lan Nguyen Thi, Yamaguchi Ryoji, Cirone Francesco, Carmichael Leland Eugene, Buonavoglia Canio

PII: S0378-1135(07)00013-2

DOI: doi:10.1016/j.vetmic.2007.01.005

Reference: VETMIC 3559

To appear in: VETMIC Received date: 13-10-2006 Revised date: 22-12-2006 Accepted date: 2-1-2007

Please cite this article as: Vito, M., Gabriella, E., Stella, L.M., Nicola, D., Eleonora, L., Krisztian, B., Merete, B.-M., Thi, L.N., Ryoji, Y., Francesco, C., Eugene, C.L., Canio, B., Genotyping Canine Distemper Virus (CDV) by a hemi-nested multiplex PCR provides a rapid approach for investigation of CDV outbreaks,Veterinary Microbiology (2007), doi:10.1016/j.vetmic.2007.01.005

This is a PDF file of an unedited manuscript that has been accepted for publication.

As a service to our customers we are providing this early version of the manuscript.

The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

(3)

Accepted Manuscript

1

Genotyping Canine Distemper Virus (CDV) by a hemi-nested multiplex

1

PCR provides a rapid approach for investigation of CDV outbreaks

2

3

Martella Vito*,1 Elia Gabriella,1 Lucente Maria Stella,1 Decaro Nicola, 1 Lorusso

4

Eleonora,1 Banyai Krisztian,2 Blixenkrone-Møller Merete,3 Lan Nguyen Thi, 4 Yamaguchi

5

Ryoji, 4 Cirone Francesco,1 Carmichael Leland Eugene,5 Buonavoglia Canio.1

6 7

1 Department of Animal Health and Well-being, University of Bari, Valenzano, Bari, Italy 8

2 Regional Laboratory of Virology, Baranya County Institute of State Public Health Service, Pecs, Hungary 9

3 Laboratory of Virology, Department of Veterinary Pathobiology, The Royal Veterinary and Agricultural 10

University, Stigbojlen 7, 1870 Frederiksberg C, Copenhagen, Denmark.

11

4 Department of Veterinary Pathology, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan, 889-2192 12

5 James A. Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 13

14853, US 14

15

Running title: genotyping of vaccine and field CDV strains.

16 17

*Corresponding author:

18

Vito Martella - Dipartimento di Sanità e Benessere Animale, Facoltà di Medicina Veterinaria

19

di Bari, S.p. per Casamassima km 3, 70010 Valenzano, Bari, Italia

20

Tel: 39 080 4679805

21

Fax: 39 080 4679843

22

E-mail: v.martella@veterinaria.uniba.it

23

(4)

Accepted Manuscript

ABSTRACT

1

CDV is a highly contagious viral pathogen causing a lethal systemic disease in dogs

2

and other carnivores. Several lineages or genotypes of CDV exist that are variously distributed

3

throughout several continents. Legal or uncontrolled trading of animals may modify the

4

epidemiology of CDV, introducing novel strains in CDV-naïve areas or accounting for the

5

resurgence of CDV in areas where vaccine prophylaxis was effective and successful to control

6

the disease. A hemi-nested PCR system was developed to genotype strains of the major CDV

7

lineages, America-1, Europe, Asia-1, Asia-2 and Arctic. The assay was tested using a

8

collection of 27 laboratory and vaccine strains and of 36 field CDV strains. Distinct lineages

9

could be differentiated by specific primers targeted to the H gene. The method could be useful

10

for molecular epidemiological studies of CDV, providing a tool for large-scale studies, and for

11

the diagnosis of vaccine-related disease.

12 13

Keywords: Canine distemper virus, characterization, protein H, RT-PCR

14 15 16 17 18 19 20 21 22 23

(5)

Accepted Manuscript

1. Introduction

1

Canine distemper virus (CDV) belongs to genus Morbillivirus in the Paramyxoviridae

2

family. The virus possesses a single-stranded negative RNA that encodes for a single envelope-

3

associated protein (M), two glycoproteins, [hemagglutinin/attachment protein (H) and fusion

4

protein (F)], two transcriptase-associated proteins (the phosphoprotein P and the large protein

5

L), and the nucleo-capsid protein (N), that encapsidates the viral RNA (van Regenmortel et al.,

6

2000).

7 8

CD is an acute to subacute contagious systemic disease with high mortality rates in

9

dogs and wild carnivores. The disease is characterized by pyrexia, nasal and ocular discharge,

10

gastrointestinal and respiratory symptoms and nervous signs (Appel, 1987). The infection and

11

the disease are not age-restricted and may be prevented by passive or active immunization

12

(Appel, 1987).

13 14

The introduction of modified live (ML) CDV vaccines in the 1950s and their extensive

15

use has greatly helped to keep the disease under control (Appel, 1987; Appel and Summers,

16

1995; Greene and Appel, 1998). Notwithstanding, the incidence of CDV-related disease in

17

canine population throughout the world seems to have increased in the last decades and several

18

episodes of CDV disease in vaccinated animals have been reported (Blixenkrone-Møller et al.,

19

1992; Decaro et al., 2004; Frolich et al., 2000; Gemma et al., 1996a and 1996b; Kai et al.,

20

1993; Patronek et al., 1995; Scagliarini et al., 2003).

21 22 23

(6)

Accepted Manuscript

Molecular epidemiology is useful to trace the origin of CDV strains and to investigate

1

the dynamics of circulation of the various strains in susceptible animals. Unfortunately, the

2

lack of common standards and protocols in epidemiologic studies of CDV does not allow

3

precise evaluation of the epidemiologic features of this important pathogen, since previous

4

studies have targeted different genome regions. Sequence analysis of CDV strains identified in

5

different geographical settings and from various animal species has revealed that the H

6

gene/glycoprotein of CDV undergoes a genetic/antigenic drift, according to geographic

7

patterns related to the geographic location of the circulating strains. In addition, the H gene has

8

been shown to be a reliable target to reconstruct and investigate the genetic relationships

9

among CDV strains. The majority of CDV field strains cluster into six major genetic lineages,

10

designated America-1 and –2, Asia-1 and –2, European and Arctic (Bolt et al., 1997; Carpenter

11

et al., 1998; Haas et al., 1997; Harder et al., 1996; Hashimoto et al., 2001; Iwatsuki et al., 1997;

12

Lan et al., 2005b and 2006; Lednicky et al., 2004; Martella et al., 2002 and 2006; Mochizuki

13

et al., 1999; Pardo et al., 2005) (Figure 1). Current CDV ML vaccines have been produced

14

using CDV isolates within the America-1 lineage. The Snyder Hill strain was isolated in Ithaca,

15

N.Y., USA in the 1950's from the brain of a dog and passaged in vivo in dogs before being

16

adapted to cell growth in NL-DKC cells (Brown et al., 1972). The Onderstepoort strain, used

17

worldwide as an attenuated live vaccine, dates back to a disease outbreak among North

18

American ranched foxes in the 1930’s (Haig, 1956). America-1 CDVs have not been detected

19

over the last 5 decades and it is not known whether they are still circulating in the field.

20

Recently, vaccine-like CVDs have been detected from outbreaks in wildlife animals but they

21

have been reported only occasionally in dogs (Harder and Osterhaus, 1997; Lednicky et al.,

22

2004). In addition, the detection of Arctic CDV strains, formerly believed to circulate

23

(7)

Accepted Manuscript

exclusively in the Arctic ecosystem, has been reported in United States (Pardo et al., 2005) and

1

Europe (Martella et al., 2006) raising questions on the origin of these unusual strains.

2

Uncontrolled trading of animals has been conjectured as one reason for the observed changes

3

in CDV epidemiology as a consequence of the introduction of novel strains into CDV-naïve

4

areas or into countries where CDV spread had been effectively controlled by vaccination.

5 6

Based on the genetic heterogeneity in the H gene, an RT-PCR genotyping assay was

7

developed that can differentiate CDV strains of vaccine origin from field strains as well as

8

predict the genotype of field strains. The system described here was validated in a multicentric

9

study involving three laboratories in Japan and Europe. Tests were done on strains representing

10

all CDV genotypes identified thus far that are considered epidemiologically relevant.

11 12

2. Materials and methods

13

2.1 Viruses and clinical specimens

14

Thirty-eight CDV-positive cases were identified in our laboratories from 2002 through

15

April 2006 by screening animals affected with CDV-associated symptoms, e.g. neurological

16

signs, enteritis, respiratory distress, nasal and ocular discharge, fever. Samples were submitted

17

by animal hospitals in several regions of Central and Southern Italy and CDV RNA was

18

detected by reverse transcription (RT)-PCR, using primer pair P2-P7 that amplifies a 478-bp-

19

long fragment of the N gene (Shin et al., 1995) and by a real-time PCR assay (Elia et al., 2006).

20 21

The canine strains DK91B and DK91C and the mink strain DK86, isolated in Denmark

22

(Blixenkrone-Møller et al., 1992; Blixenkrone-Møller et al., 1993) were used as prototypes of

23

(8)

Accepted Manuscript

the European CDV genotype. Strain GR88 was detected from a sledge dog in remote Inuit

1

settlement in Northern Greenland (Blixenkrone-Møller et al., 1992) and was used as prototype

2

of the Arctic genotype. Italian strains 179/04 and 48/04 were detected in 2004 and 2005 in

3

Rome and Messina, respectively, and also were used as reference strains of the Arctic lineage

4

(Martella et al., 2006). Strains KDK-1, S124C, Ac96I and P94S (Japan) were used as reference

5

strains of lineage Asia-1, and strains 007Lm, 009L and 011C (Japan) as reference strains of

6

lineage Asia-2 (Lan et al., 2005a and 2005b and 2006; Mochizuki et al., 1999). The vaccine

7

strains Onderstepoort (five different vaccine products, A to E), Snyder Hill and “Distemperoid”

8

were obtained from vaccines obtained from the market. In addition, three strains of the

9

reference Onderstepoort virus and a Snyder Hill strain, maintained in our laboratories, were

10

included in the analysis as prototypes of the genotype America-1. The strain Javelina/US89

11

(Appel et al., 1991), Dog/US89 (Blixenkrone-Møller et al., 1992) and Leopard/US91 (Appel et

12

al., 1994) were used as prototypes of the genotype America-2. The vaccine and reference

13

strains used in this study are listed in Table 1.

14 15

2.2 Genotyping strategy and primer selection

16

Primers were synthesized in Tib Molbiol laboratories (Genoa, Italy). The typing

17

primers were selected taking advantage of nucleotide polymorphisms conserved within strains

18

of each lineage, but divergent among strains of the various lineages. Three specific typing

19

primers (minus-sense) were prepared for gene H sequences of strains of lineage Europe (primer

20

200, prototype strain DK91A), lineage America-1 (primer 201b, prototype strain

21

Onderstepoort), and lineage Arctic-like (primer 202b, prototype strain GR88). An additional

22

two specific typing primers (minus-sense) were prepared for gene H sequences of strains of

23

(9)

Accepted Manuscript

lineage Asia-1 (primer 199b, prototype strain KDK-1) and Asia-2 (primer 203b, prototype

1

strain HM-6). Due to the heterogeneity of strains and to lack of conserved nucleotide

2

polymorphism, it was not possible to design primers specific for the lineage America-2. One

3

pair of primers, 204-232b, made against the consensus sequence of 55 gene H sequences

4

available in the databases, was chosen for the first RT-PCR amplification, in regions that are

5

highly conserved among CDV strains and include all the lineage-specific polymorphisms

6

useful for the genotyping strategy. The nucleotide (nt) positions and sequences of the primers

7

are shown in Table 2. The genotyping strategy is outlined in Figure 2.

8 9

Reverse transcription and PCR amplification of the H gene of CDV were achieved in a

10

single step procedure. Total RNA was obtained from 25 mg samples of tissue homogenates or

11

swab washes. The RNA was extracted using the RNeasy Kit (Qiagen, Gmbh, Germany)

12

according to the manufacturer's instructions. The RNA was eluted in 50 µl of RNase-free

13

water. To obtain a PCR product of the H gene, 0.5 µl each of primers 231 and 232 (50 pmol/µl)

14

was added to a total of 24 µl of the reaction mixture containing 0.2 mM of each dNTP, 1.2 mM

15

MgSO4 and 0.5 µl of a mix of SuperScript II H- Reverse Transcriptase and Platinum Taq HiFi

16

(Invitrogen - Life Technologies, Milan, Italy). The RNA was reverse transcribed and

17

immediately subjected to PCR amplification in a single-step protocol, using SuperScript One-

18

Step RT-PCR kit (Invitrogen - Life Technologies, Milan, Italy). Reverse transcription was

19

carried out at 48°C for 60 min, followed by denaturation of the reverse transcriptase at 95°C

20

for 2 min. Amplification was conducted by a temperature cycling protocol consisting of 40

21

cycles of 30 s for denaturation at 94°C, 30 s of primer annealing at 50°C, and 1.5 min of

22

extension at 68°C, followed by 10 min of final extension at 68°C. With this RT-PCR a

23

(10)

Accepted Manuscript

fragment of 1160 bp of the gene H of CDV was amplified. The cDNA was diluted 1: 200 in

1

distilled water and used as template for the second-round PCR. In this PCR, portions of the

2

1160-bp cDNA were amplified using a cocktail of primers that includes the plus-sense primer

3

204 and the minus-sense lineage-specific primers 199, 200, 201, 202, 203b. Selection of

4

specific primers that gave products of different size permitted identification of the gene H

5

genetic lineage group by agarose gel analysis. The use of a cocktail of primers has the added

6

advantage of identifying in one PCR the lineage of any CDV isolate. For the typing reaction

7

(second-round PCR), 1 µl of the diluted cDNA was added to a total of 49 µl of reaction

8

mixture consisting of 0.25 µl of AmpliTaq Gold (1.25 U) (Applera, Milan, Italy), 5 µl of 10×

9

PCR buffer, 2 µl of 2.5 mM dNTP mixture, 35.25 µl of distilled H2O, 0.25 µl of each primer

10

(50 pM). After 10 min at 94°C for activation of DNA polymerase, amplification was conducted

11

by a temperature cycling protocol consisting of 25 cycles of 1 min of denaturation at 94°C, 1

12

min of primer annealing at 50°C, and 1 min of extension at 72°C, followed by 10 min of final

13

extension phase at 72°C.

14 15

2.3 Sequence analysis of the full-length gene H

16

To confirm the results of the genotyping strategy, eight CDV strains were selected to

17

obtain sequence information on the H gene. Reverse transcription and PCR amplification of the

18

full-length H gene of CDV were achieved with primers RH-3 and RH-4 as previously

19

described, with minor modifications (Mochizuki et al., 1999). The RH3-RH4 PCR products

20

were purified with Ultrafree-DA Columns (Amicon, Millipore). The DNA was then used as

21

template for direct sequencing (Genome Express, Meylan France). The DNA was sequenced

22

using the conserved primers RH3 and RH4 and specific primers designed according to an

23

(11)

Accepted Manuscript

overlapping strategy. The sequences were then assembled using Bioedit software package

1

version 2.1 (Hall, 1999).

2 3

3. Results

4

3.1 Gene H typing and sequence analysis of the H gene

5

Analysis of seven vaccine CDV strains (five Onderstepoort-based vaccines, one Snyder

6

Hill-based vaccine and one ‘Distemperoid’ vaccine), of three Onderstepoort strains and of one

7

Snyder Hill strain available in our laboratory, of 16 reference CDV strains from our laboratory

8

collection and of 36 field strains was carried out by two PCR amplifications. In the first PCR,

9

the primers 204 and 232b yielded intense DNA products of the predicted size (1160 bp) for all

10

63/63 strains tested (Figure 3). Some nonspecific bands were observed sporadically. The nature

11

of nonspecific but discrete products was not characterized but it probably resulted from

12

mispriming events during amplification of the nucleic acids. Those products did not interfere

13

with gene H typing.

14 15

RNA concentrations were determined by using a real time RT-PCR specific for CDV

16

(Elia et al., 2006). The RT-PCR sensitivity (primers 204/232b) was evaluated by using samples

17

containing known amounts of viral RNA. Samples containing 107 to 102 copies of viral RNA,

18

negative tissue samples and distilled water were tested. CDV-specific bands were observed

19

with samples containing 107, 106, 105, 104 and 103 copies of CDV RNA.

20 21

In the second-round PCR, representatives of the various genetic lineages were correctly

22

characterized, yielding amplicons of the expected size (Figure 4). The reference strains

23

(12)

Accepted Manuscript

DK91B, DK91C and DK86 were correctly characterized as CDVs of the European lineage by

1

primer 200, while strains GR88, 179/04 and 48/05 were correctly recognized as Arctic-like

2

CDVs by the lineage-specific primer 202b (Figure 5). Strains KDK-1, S124C, Ac96I and P94S

3

were correctly characterized as Asia-1, and strains 007Lm, 009L and 011C were correctly

4

characterized as Asia-2 (Figure 6). The Onderstepoort-based vaccine strains (vaccines A to F

5

and our laboratory strain), as well as the Snyder Hill and Distemperoid vaccine were correctly

6

recognized by primer 201b. Similarly, the Onderstepoort and Snyder Hill strains were correctly

7

characterized by primer 201b (Figure 7). No cross-reactions were observed among heterotypic

8

primers and the various CDV strains. Fourteen field isolates were characterized as Arctic-like

9

and 22 as European.

10 11

Partial or full-length sequence analysis of the H gene of eight strains confirmed the

12

results obtained by the RT-PCR genotyping assay. Three strains were confirmed as European

13

CDVs and five were confirmed as Arctic strains (Figure 1).

14 15

4. Discussion

16

In this study, an RT-PCR genotyping system was developed, that took advantage of the

17

genetic divergence observed in the H gene among the major CDV lineages. The RT-PCR

18

genotyping system was designed on the basis of lineage-specific nucleotide polymorphisms

19

scattered over the H gene. The system was designed to allow prediction of the main genetic

20

lineages of CDV and it proved useful for the correct genotypic characterization of the genotype

21

of all vaccine strains, 17/20 laboratory reference strains representative of the various genotypes

22

and all the CDV strains collected by our laboratories over a 6-year period.

23

(13)

Accepted Manuscript

1

As expected, the America-2 CDVs were not recognized by any of the genotype-specific

2

primers. Indeed, due to the genetic heterogeneity of the America-2 strains, we were unable to

3

design a primer that would recognize all the strains within this lineage. The lack of primer

4

reactivity was a pattern observed only for the America-2-like strains. Nevertheless, the

5

adoption of a negative marker is intrinsically affected by the possibility that unidentified CDV

6

strains may be mischaracterized and therefore such pattern must be interpreted with caution.

7

Accordingly, the system may be adopted to recognize unequivocally five out of the six main

8

genetic lineages of CDV, and, potentially, it could be adopted in laboratories in Europe, Asia

9

and America.

10 11

The RT-PCR sensitivity (primers 204/232b) was evaluated by using samples containing

12

known amounts of viral RNA (107 to 102 copies) and the assay was able to detect up 103 copies

13

of viral RNA. Quantification of viral loads by real-time PCR revealed that CDV RNA copies in

14

infected animals ranged between 5.91 x 105 and 4.93 x 107/µl of template in ocular swabs and

15

between 6.26 x 104 to 7.58 x 106 RNA copies/µl of template in blood samples. Amounts in

16

urine reached 2.35 x 109 RNA copies/µl of template(Elia et al., 2006). Therefore, the quantity

17

of viral RNA in biological fluids and tissues during the period of viraemia is largely beyond the

18

limits of detection of the H-gene based RT-PCR assay. Accordingly, the 204/232b RT-PCR

19

proved to be sensitive and applicable to the majority of samples collected for diagnostic

20

purpose, including ocular swabs and urine. Therefore, it would be suitable for the in vivo

21

diagnosis of CDV infection. In addition, the possibility to alleviate time-consuming

22

(14)

Accepted Manuscript

procedures, such as gene cloning or direct sequencing, for precise genotype characterization

1

makes this genotyping system highly suitable for diagnostic laboratories.

2 3

Molecular characterization of measles virus (MV), another well-characterized member

4

of the genus Morbillivirus, highly related to CDV, is a key component of measles surveillance

5

and epidemiological investigations to identify the source and trace the transmission pathways

6

of the virus. In MV surveillance studies, determination of the full-length haemagglutinin gene

7

sequence is required to define correctly the genetic features of novel genotypes. According to

8

current classification scheme for MVs, the strains which differ at least by 2% nt sequence from

9

other strains in their haemagglutinin gene qualifies as a new candidate of genotype. Thus far at

10

least 23 MV genotypes have been recognized by WHO and their distribution varies

11

geographically and temporarily (Weekly Epidemiological Report, WHO, 40, 2005, 80, 347-

12

351, http:/www.who.int/wer). The initial proposal of the term ‘genotype’ to distinguish among

13

the various genetic lineages of CDV was made by Bolt et al., (1997) and it is now widely

14

accepted for defining novel CDV lineages (Mochizuki et al., 1999). However, such CDV

15

genotype definition does not correspond well to the definition of MV genotype, since the

16

genetic distance among the various CDV genotypes is greater than that observed for MV

17

genotypes. Amino acid variation between the various lineages of CDV is > 4% (32), while the

18

highest variation (> 8%) is observed between the America-1 strains, used to develop the

19

modified live CDV vaccines currently available in the market, and all the other CDV

20

genotypes (Bolt et al., 1997; Carpenter et al., 1998; Haas et al., 1997; Harder et al., 1996;

21

Iwatsuki et al., 1997; Lednicky et al., 2004; Martella et al., 2002; Martella et al., 2006;

22

Mochizuchi et al., 1999). Therefore, CDV strains appear to be more heterogeneous and to

23

(15)

Accepted Manuscript

differ more from currently used vaccine than comparable MV field and vaccine strains, since

1

the H protein divergence for MV isolates and vaccine strains only reaches 3% (Rota et al.,

2

1992).

3 4

The existence of several CDV genetic lineages raises a number of questions with regard

5

to the reasons and the effects of such observed diversification. Antigenic heterogeneity has

6

been observed between America-1 and new CDVs, by using either monoclonal antibodies

7

(Blixenkrone-Møller et al., 1992; Blixenkrone-Møller et al., 1993; Harder et al., 1993; Iwatzuki

8

et al., 2000; Örvell et al., 1990) or polyclonal immune sera (Gemma et al., 1996a; Harder et al.,

9

1993; 1996; Mochizuki et al., 2002). Sera raised against wild-type CDV isolates may have

10

neutralizing titers up to tenfold higher against the homologous virus than against vaccine

11

strains (Harder et al., 1996). While it is unlikely that slight antigenic variation may

12

compromise the effectiveness of the canine vaccines in dogs, that are protected by a strong

13

active immune response elicited by repeated vaccine administrations, it is possible, however,

14

that critical amino acid substitutions in key epitopes of the H protein among circulating strains

15

may allow escape from the limited antibody repertoire of maternal origin of young

16

unvaccinated pups.

17 18

Differentiation between vaccine and field strains of CDV have relied on the use of

19

monoclonal antibodies (Iwatzuki et al., 2000; Örvell et al., 1990) and restriction or sequence

20

analysis of the full-length or partial gene H sequence (Lan et al., 2006; Mochizuki et al. 1999;

21

Ohashi et al., 1998; Uema et al. 2005). In this multicentric study we developed a genotyping

22

protocol that successfully characterised a large collection of field, laboratory and vaccine

23

(16)

Accepted Manuscript

strains. Using the method described here, CDV was detected in 10/36 pups, less than 3 months

1

old, that had been recently vaccinated against CDV with ML-CDV vaccines. The pups were

2

suspected to have developed vaccine-induced disease. Samples from the pups were sent to our

3

laboratories for diagnostic analysis and all tested positive for CDV. Since the pups became ill

4

within 2 weeks after vaccine administration, the owners and some veterinarians believed that

5

the illnesses were associated with the CDV vaccines used. These resulted in disputes between

6

the veterinary practitioners and the pet owners. In each of those cases, field CDV strains were

7

found to be the cause of disease and the deaths were not related to the vaccine strains.

8 9

Interestingly, analysis of the CDV strains collected during our 2005-2006 surveillance

10

revealed the spread of Arctic-like CDVs in Italian dogs. In Italy, the Arctic strains were first

11

described in 2004 (Martella et al., 2006) and it was not clear whether this finding was

12

occasional. However, 7/14 strains in 2005 and 8/17 strains in 2006 were characterised as

13

Arctic, suggesting dissemination of this novel genotype in Italy. This pattern is similar to that

14

observed with MV endemic strains that may be subjected to rapid replacement over

15

consecutive years (Santibanez et al., 2002). Due to the paucity of the epidemiological surveys

16

and the use of different genes targeted in various studies, the global distribution of the major

17

CDV lineages is not clear. Analysis of CDV strains detected globally and from a variety of

18

host species would provide a more comprehensive understanding of the global ecology of

19

CDV.

20

21

Acknowledgments

22

The Authors are extremely grateful to Mr Donato Narcisi for his expert technical assistance.

23

(17)

Accepted Manuscript

The study was supported by a grant from the Danish Fur Breeders Research Fund and by a

1

grant of the Italian Ministry of University and Research (MIUR).

2 3

References

4 5

Appel, M., 1987. Canine distemper virus. In: M. J. Appel (ed.), Virus infections of

6

carnivores. Elsevier Science Publishers B.V., Amsterdam, The Netherlands, p.133-159.

7

Appel, M.J.G., Summers, B.A., 1995. Pathogenicity of morbilliviruses for terrestrial

8

carnivores. Vet. Microbiol. 44, 187-191.

9 10

Appel, M.J., Reggiardo, C., Summers, B.A., Pearce-Kelling, S., Mare, C.J., Noon, T.H.,

11

Reed, R.E., Shively, J.N., Orvell. C., 1991. Canine distemper virus infection and

12

encephalitis in javelinas (collared peccaries). Arch. Virol. 119, 147-152.

13

14

Appel, M.J., Yates, R.A., Foley, G.L., Bernstein, J.J., Santinelli, S., Spelman, L.H.L.,

15

Miller, D., Arp, L.H., Anderson, M., Barr, M., et al., 1994. Canine distemper epizootic in

16

lions, tiger, and leopards in North America. J. Vet. Diagn. Invest. 6, 277-288.

17 18

Blixenkrone-Møller, M., Svansson, V., Appel, M., Krogsrud, J., Have, P., Orvell, C.,

19

1992. Antigenic relationship between field isolates of morbilliviruses from different

20

carnivores. Arch. Virol. 123, 279-294.

21 22

(18)

Accepted Manuscript

Blixenkrone-Møller, M., Svansson, V., Have, P., Orvell, C., Appel, M., Pedersen, I. R.,

1

Dietz, H.H., Henriksen, P., 1993. Studies on manifestations of canine distemper virus

2

infection in an urban dog population. Vet. Microbiol. 37, 163-173.

3 4

Bolt, G., Jensen, T.D., Gottschalck, E., Arctander, P., Appel, M.J.G., Buckland, R.,

5

Blixenkrone-Møller, M., 1997. Genetic diversity of the attachment (H) protein gene of

6

current field isolates of canine distemper virus. J. Gen. Virol. 78, 367-372.

7 8

Brown, A.L., Vitamvas, J.A., Merry, D.L.Jr, Beckenhauer, W.H., 1972. Immune response

9

of pups to modified live-virus canine distemper-measles vaccine. Am. J. Vet. Res. 33,

10

1447-1456.

11 12

Carpenter, M.A., Appel, M.J.G., Roelke Parker, M.E., Munson, L., Hofer, H., East, M.,

13

O'Brien, S. J., 1998. Genetic characterization of canine distemper virus in Serengeti

14

carnivores. Vet. Immunol. Immunopathol. 65, 259-266.

15 16

Decaro, N., Camero, M., Greco, G., Zizzo, N., Tinelli, A., Campolo, M., Pratelli, A.,

17

Buonavoglia, C., 2004. Canine distemper and related diseases: report of a severe outbreak

18

in a kennel. The new Microbiologica 27, 177-182.

19 20

Elia, G., Decaro, N., Martella, V., Cirone, F., Lucente, M.S., Lorusso, E., Di Trani, L.,

21

Buonavoglia, C., 2006. Canine distemper virus detection in dogs by real time PCR. J.

22

Virol. Methods. 136, 171-176.

23

(19)

Accepted Manuscript

1

Frolich, K., Czupalla, O., Haas, L., Hentschke, J., Dedek, J., Fickel, J., 2000.

2

Epizootiological investigations of canine distemper virus in free-ranging carnivores from

3

Germany. Vet. Microbiol. 74, 283-292.

4 5

Gemma, T., Iwatsuki, K., Shin, Y.S., Yoshida, E., Kai, C., Mikami, T., 1996a. Serological

6

analysis of canine distemper virus using an immunocapture ELISA. J. Vet. Med. Sci. 58,

7

791-794.

8 9

Gemma, T., Watari, T., Akiyama, K., Miyashita, N., Shin, Y.S., Iwatsuki, K., Kai, C.,

10

Mikami, T., 1996b. Epidemiological observations on recent outbreaks of canine distemper

11

in Tokyo area. J. Vet. Med. Sci. 58, 547-550.

12 13

Greene, C.E., Appel, M.J., 1998. Canine distemper,. In: C.E. Greene (ed.), Infectious

14

diseases of the dog and cat, 2nd ed. The W. B. Saunders Co., Philadelphia, Pa., pp. 9-22.

15 16

Haas, L., Martens, W., Greiser-Wilke, I., Mamaev, L., Butina, T., Maack, D., Barrett, T.,

17

1997. Analysis of the haemagglutinin gene of current wild-type canine distemper virus

18

isolates from Germany. Virus Res. 48, 165-171.

19 20

Haig, D.A., 1956. Canine distemper: immunization with avianized virus. Onderstepoort J.

21

Vet. Res. 17,19-53.

22 23

(20)

Accepted Manuscript

Hall, T.A., 1999. BioEdit: a user-friendly biological sequence alignment and analysis

1

program for Windows 95/98/NT. Nucl. Acids Symp. Ser. 41, 95-98.

2 3

Harder, T.C., Osterhaus, A.D., 1997. Canine distemper virus: a morbillivirus in search of

4

new hosts?. Trends Microbiol. 5, 120-124.

5 6

Harder, T.C., Klusmeyer, K., Frey, H.R., Örvell, C., Liess, B., 1993. Intertypic

7

differentiation and detection of intratypic variants among canine and phocid distemper

8

morbillivirus isolates by kinetic neutralization using a novel immunoplaque assay. J. Virol.

9

Methods 41, 77-92.

10 11

Harder, T.C., Kenter, M., Vos, H., Siebelink, K., Huisman, W., van Amerongen, G., Örvell,

12

C., Barrett, T., Appel, M.J.G., Osterhaus, A.D.M.E., 1996. Canine distemper virus from

13

diseased large felids: biological properties and phylogenetic relationships. J. Gen. Virol. 77,

14

397-405.

15 16

Hashimoto, M., Une, Y., Mochizuki, M., 2001. Hemagglutinin genotype profiles of canine

17

distemper virus from domestic dogs in Japan. Arch. Virol. 146, 149-155.

18 19

Iwatsuki, K., Miyashita, N., Yoshida, E., Gemma, T., Shin, Y.S., Mori, T., Hirayama, N.,

20

Kai, C., Mikami, T., 1997. Molecular and phylogenetic analyses of the haemagglutinin (H)

21

proteins of field isolates of canine distemper virus from naturally infected dogs. J. Gen.

22

Virol. 78, 373-380.

23

(21)

Accepted Manuscript

1

Iwatsuki, K., Tokiyoshi, S., Hirayama, N., Nakamura, K., Ohashi, K., Wasaka, C., Mikami,

2

T., Kai, C., 2000. Antigenic difference in the H proteins of canine distemper viruses. Vet.

3

Microbiol. 71, 281-286.

4 5

Kai, C., Ochikubo, F., Okita, M., Iinuma, T., Mikami, T., Kobune, F., Yamanouchi, K.,

6

1993. Use of B95a cells for isolation of canine distemper virus from clinical cases. J. Vet.

7

Med. Sci. 55, 1067-1070.

8 9

Lan, N.T., Yamaguchi, R., Uchida, K., Sugano, S., Tateyama, S., 2005a. Growth Profiles

10

of Recent Canine Distemper Isolates on Vero Cells Expressing Canine Signalling

11

Lymphocyte Activation Molecule (SLAM). J. Com. Path. 133, 77-81.

12 13

Lan, N.T., Yamaguchi, R., Furuya, Y., Inomata, A., Ngamkala, S., Naganobu, K., Kai, K.,

14

Mochizuki, M., Kobayashi, Y., Uchida, K., Tateyama. S., 2005b. Pathogenesis and

15

phylogenetic analyses of canine distemper virus strain 007Lm, a new isolate in dogs. Vet.

16

Microbiol. 110, 197-207.

17 18

Lan, N.T., Yamaguchi, R., Inomata, A., Furuya, Y., Uchida, K., Sugano, S., Tateyama, S.,

19

2006. Comparative analyses of canine distemper viral isolates from clinical cases of canine

20

distemper in vaccinated dogs. Vet. Microbiol. 115, 32-42.

21 22

(22)

Accepted Manuscript

Lednicky, J.A., Dubach, J., Kinsel, M.J., Meehan, T.P., Bocchetta, M., Hungerford, L.L.,

1

Sarich, N.A., Witecki, K.E., Braid, M.D., Pedrak, C., Houde, C.M., 2004. Genetically

2

distant American Canine distemper virus lineages have recently caused epizootics with

3

somewhat different characteristics in raccoons living around a large suburban zoo in the

4

USA. Virol. J. 1, doi: 10.1186/1743-422X-1-2.

5 6

Martella, V., Pratelli, A., Cirone, F., Zizzo, N., Decaro, N., Tinelli, A., Foti, M.,

7

Buonavoglia, C., 2002. Detection and genetic characterization of canine distemper virus

8

(CDV) from free-ranging red foxes in Italy. Molecular and Cellular Probes 16, 77-83.

9 10

Martella, V., Cirone, F., Elia, G., Lorusso, E., Decaro, N., Campolo, M., Desario, C.,

11

Lucente, M.S., Bellacicco, A.L., Blixenkrone-Møller, M., Carmichael, L.E., Buonavoglia,

12

C., 2006. Heterogeneity within the hemagglutinin genes of canine distemper virus (CDV)

13

strains detected in Italy. Vet Microbiol. 116, 301-309.

14 15

Mochizuki, M., Hashimoto, M., Hagiwara, S., Yoshida, Y., Ishiguro, S., 1999. Genotypes

16

of canine distemper virus determined by analysis of the hemagglutinin genes of recent

17

isolates from dogs in Japan. J. Clin. Microbiol. 37, 2936-2942.

18 19

Mochizuki, M., Motoyoshi, M., Maeda, K., Kai, K., 2002. Complement-mediated

20

neutralization of canine distemper virus in vitro: cross-reaction between vaccine

21

Onderstepoort and field KDK-1 strains with different hemagglutinin gene characteristics.

22

Clin. Diagn. Lab. Immunol. 9, 921-924.

23

(23)

Accepted Manuscript

1

Ohashi, K., Iwatsuki, K., Nakamura, K., Mikami, T., Kai, C., 1998. Molecular

2

identification of a recent type of canine distemper virus in Japan by restriction fragment

3

length polymorphism. J. Vet. Med. Sci. 60, 1209-1212.

4 5

Örvell, C., Blixenkrone-Møller, M., Svansson, V., Have, P., 1990. Immunological

6

relationships between phocid and canine distemper virus studied with monoclonal

7

antibodies. J. Gen. Virol. 71, 2085-2092.

8 9

Pardo, I.D., Johnson, G.C., Kleiboeker, S.B., 2005. Phylogenetic characterization of canine

10

distemper viruses detected in naturally infected dogs in North America. J. Clin. Microbiol.

11

43, 5009-5017.

12 13

Patronek, G.J., Glickman, L.T., Johnson, R., Emerick, T.J., 1995. Canine distemper

14

infection in pet dogs: II. A case-control study of risk factors during a suspected outbreak in

15

Indiana. J. Am. Anim. Hosp. Assoc. 31, 230-235.

16 17

Rota, J.S., Hummel, K.B., Rota, P.A., Bellini, W.J., 1992. Genetic variability of the

18

glycoprotein genes of current wild-type measles isolates. Virology. 188, 135-142.

19 20

Santibanez, S., Tischer, A., Heider, A., Siedler, A., Hengel, H., 2002. Rapid replacement of

21

endemic measles virus genotypes. J. Gen. Virol. 83, 2699-2708.

22 23

(24)

Accepted Manuscript

Scagliarini, A., Battilani, M., Ciulli, S., Prosperi, S., Moranti, L., 2003. Molecular analysis

1

of the NP gene of Italian CDV isolates. Vet. Res. Commun. 27, 355-357.

2 3

Shin, Y.S., Mori, T., Okita, M., Gemma, T., Kai, C., Mikami, T., 1995. Detection of

4

canine distemper virus nucleocapsid protein gene in canine peripheral blood mononuclear

5

cells by RT-PCR. J. Vet. Med .Sci. 57, 439-445.

6 7

Uema, M., Ohashi, K., Wakasa, C., Kai. C., 2005. Phylogenetic and restriction fragment

8

length polymorphism analyses of hemagglutinin (H) protein of canine distemper virus

9

isolates from domestic dogs in Japan. Virus Res. 109, 59-63.

10 11

van Regenmortel, H.V.M., Fauquet, C.M., Bishop, D.H.L., Carstens, E., Estes, M.K.,

12

Lemon, S., Maniloff, J., Mayo, M.A., McGeoch, D., 2000. In C. R. Pringle, and R. B.

13

Wickner (ed.). Virus taxonomy. Seventh Report of the International Committee on

14

Taxonomy of Viruses. Academic Press, New York, N.Y.

15 16 17 18 19 20 21 22 23

(25)

Accepted Manuscript

Table 1. List of the vaccine and reference CDV strains used in this study. n.r.: non reactive

1 2 3

Strain Origin Lineage/

genotype Primer reactivity Onderstepoort Vaccine A America-1 201b Onderstepoort Vaccine B America-1 201b Onderstepoort Vaccine C America-1 201b Onderstepoort Vaccine D America-1 201b Onderstepoort Vaccine E America-1 201b Snyder Hill Vaccine America-1 201b Distemperoid Vaccine America-1 201b Onderstepoort Laboratory America-1 201b Onderstepoort Laboratory America-1 201b Onderstepoort Laboratory America-1 201b Snyder Hill Laboratory America-1 201b

Dog/US89 US America-2 n.r.

Javelina/US89 US America-2 n.r.

Leopard/US91 US America-2 n.r.

Dog/GR88 Greenland Arctic 202b

48/05 Italy Arctic 202b

179/04 Italy Arctic 202b

Dog/DK91B Denmark European 200 Dog/DK91C Denmark European 200 Mink/DK86 Denmark European 200

KDK-1 Japan Asia-1 199b

S124C Japan Asia-1 199b

Ac96I Japan Asia-1 199b

P94S Japan Asia-1 199b

007Lm Japan Asia-2 203b

009L Japan Asia-2 203b

011C Japan Asia-2 203b

4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

(26)

Accepted Manuscript

Table 2: Sequence and position of the primers used in this study. Sense (+) and antisense (-)

1

direction is also indicated.

2 3

Primer Position (nt) Sequence 5’ to 3’

204 (+) 388-409 GAA TTC GAC TTC CGC GAT CTC C 232b (-) 1543-1519 TAG GCA ACA CCA CTA ATT TRG ACT C 199b (-) 613-554 TAG ATA CGG ATA GGG GGA AT

200 (-) 670-650 TAG CAG TTA GCA TAT TGG 201b (-) 1010-990 CAG GTA TCA CTT CCT CCA TG 202b (-) 1127-1110 TTT TTT TGT TCC TCT AGG 203b (-) 1474-1455 GAT AAC AAT TTC CAC TCG AC

4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

(27)

Accepted Manuscript

FIGURE LEGENDS

1 2

Figure 1: Phylogenetic relationships between CDV strains on the basis of the nucleotide

3

alignment of the H protein. Abbreviations: It, Italy; US, United States; Ch, China; Ger,

4

Germany; Dk, Denmark, Tw, Taiwan; Thai, Thailand; Tk, Turkey; Jp, Japan.

5 6

Figure 2: Scheme of the genotyping strategy used for CDV characterization.

7 8

Figure 3: RT-PCR with primer 204-232b for amplification of the 1160 bp fragment of the H

9

gene of CDV. M: 1 kb DNA ladder (Gene RulerTM, Fermentas Gmbh, Germany); lane 1:

10

Onderstepoort; lane 2: DK91B; lane 3: GR88; lane 4: KDK-1; lane 5: 007Lm; lane 6: negative

11

control.

12 13

Figure 4: Second-round PCR for prediction of CDV genotype. M: 1 kb DNA ladder (Gene

14

RulerTM, Fermentas Gmbh, Germany); lane 1: Onderstepoort (America-1); lane 2: DK91B

15

(European); lane 3: GR88 (Arctic); lane 4: KDK-1 (Asia-1); lane 5: 007Lm (Asia-2); lane 6:

16

negative control.

17 18

Figure 5: Second-round PCR for prediction of CDV genotype. Analysis of European and

19

Arctic strains. M: 100 bp DNA ladder (Gene RulerTM, Fermentas Gmbh, Germany); lane 1:

20

Onderstepoort (America-1); lane 2: DK91B (European); lane 3: DK91A (European); lane 4:

21

DK86 (European); lane 5: GR88 (Arctic); lane 6: 48/05 (Arctic); lane 7: 179/04 (Arctic).

22 23

(28)

Accepted Manuscript

Figure 6: Second-round PCR for prediction of CDV genotype. Analysis of Asia-1 and Asia-2

1

strains. 100 bp DNA ladder (Gene RulerTM, Fermentas Gmbh, Germany); lane 1:

2

Onderstepoort (America-1); lane 2: DK91B (European); lane 3: GR88 (Arctic); line 4: KDK-1

3

(Asia-1); lane 5: 007Lm (Asia-2); lane 6: S124C (Asia-1); lane 7: 009L (Asia-2); lane 8: 011C

4

(Asia-2); lane 9: Ac96I (Asia-1); line 10: P94S (Asia-1); lane 11: Snyder Hill vaccine

5

(America-1).

6 7

Figure 7: Second-round PCR for prediction of CDV genotype. Analysis of America-1

8

(vaccine) and -2 strains. 100 bp DNA ladder (Gene RulerTM, Fermentas Gmbh, Germany); lane

9

1: Onderstepoort, laboratory 1 strain (America-1); lane 2 to 6: Onderstepoort-based vaccine

10

strains, A to E, respectively; lane 7: Snyder Hill (America-1); lane 8: Distemperoid (America-

11

1); lane 9: Dog/US89 (America-2).

12

13 14

(29)

Accepted Manuscript

gi|5870826|LesserPanda/Ch gi|5870824|GiantPanda/Ch

gi|2315144|Yanaka/Jp gi|1944378|Ueno/Jp gi|6594288|KDK-1/Jp gi|3721905|TANU96/Jp

gi|1944380|Hamamatsu/Jp AB212965/S124C/Jp gi|37626037|TN/Ch gi|35187511|CDTaichung/Tw DQ887547/NTU-1/Thai/05

DQ887547/NTU/Thai/05 DQ191767/NTU-4/Thai/03 AB250943/Ac96I-p20/Jp

AB212964/P94S/Jp AB212963/Ac96I-p1/Jp

Asia-1

gi|38230168|00-2601/Raccon/Us gi|1296494|A92-27/4/Lopard/US

gi|40738381|01-2676/Raccon/Us gi|51172727|01-2689/Raccon/US gi|5733642|A75/17

gi|1296492|A92-6/Black leopard/US gi|1149601|US91/Leopard/US gi|42555995|01-2641/Raccon/US

gi|1149613|US89/Dog/US gi|1149603|javelina/US

gi|1149605|US89/Raccon/US

America-2

It-3 It-5

DQ494317/324/03/It gi|1657252|2544/Ger

gi|1657256|4513/Ger gi|1657254|404/Ger

gi|20152159|Tk 111/03A2/It 111/03B1/It 312/04/It 265/02-3

It-6

gi|1149611|DK91C/Dk gi|33320171|DK91A/Dk gi|33320183|DK91D/Dk gi|37654450|5804P/Ger gi|37654441|5804/Ger

Europe

gi|1149607|DK86/Mink/Dk DQ228166|207/00/Fox gi|693950|PDV-2Seal/Siberia gi|1149609|GR88/Dog/Greenland

gi|5733086|Liud/Dog/Ch It-4

179/04/It 48/05/It

It-7 It-1

It-2 It-8

Arctic

gi|13365646|HM-3/Jp gi|13365648|HM-6/Jp gi|5869513|98-002/Jp gi|13365644|26D/Jp gi|34328599|5VD/Jp

gi|34328597|5B/Jp AB212730/007Lm/Jp

AB252717/011C/Jp AB252718/009L/Jp

Asia-2

gi|10281214|Snyder Hill/US Snyder Hill/Vaccine gi|49182263|98-2646/Raccon/US

gi|49182261|98-2654Raccon/US gi|517242|Convac/US

Onderstepoort/VaccineA gi|14150871|Onderstepoort/US Onderstepoort/Lab3

Onderstepoort/VaccineB Onderstepoort/VaccineE Onderstepoort/VaccineD Onderstepoort/Lab2

America-1 (vaccines)

2 6 3 9 1 0 0

1 0 0

1 0 0 1 0 0

1 0 0

8 8 9 9

1 0 0 9 9 6 0 1 0 0

9 2 1 0 0

9 6 8 7 1 0 0

9 9 6 3 9 9

6 4 9 7 5 3 3 9

9 5

1 0 0

1 0 0

9 9

7 4 9 8 9 5

7 3 9 0 8 8

8 2 9 9 9 9

9 6 1 0 0

8 8 9 9 9 2

8 5 5 0

4 2 2 7

2 4 8 2

8 1 5 6 5 6

7 8

3 6

1 0 0

6 6 9 2

9 8 8 0

8 3 8 9

9 0

(30)

Accepted Manuscript

Gene H (1824 bp)

204 232b

203b 202b

201b 200

199b 1160 bp

1086 bp 739 bp 622 bp 282 bp 225 bp

(Asia 2)

(Asia 1)

(America 1, vaccines) (Europe)

(Arctic)

Figure 2

(31)

Accepted Manuscript

Figure 3

M 1 2 3 4 5 6

(32)

Accepted Manuscript

Figure 4

M 1 2 3 4 5 6

(33)

Accepted Manuscript

Figure 5

M 1 2 3 4 5 6 7

(34)

Accepted Manuscript

Figure 6

M 1 2 3 4 5 6 7 8 9 10 11

(35)

Accepted Manuscript

Figure 7

M 1 2 3 4 5 6 7 8 9

Références

Documents relatifs

Prevalence of antibodies against canine distemper virus and canine parvovirus among foxes and wolves from Spain... Title: Prevalence of antibodies against canine distemper virus

Since CDV is a highly infectious disease that can spill over to wildlife, particularly wild canids, we recommend that dog popu- lation control and management through responsible

Morbillivirus Measles  virus Rinderpest virus Peste des petits ruminants virus Phocin distemper  virus Canine  distemper  virus Dolphin  morbillivirus Porpoise 

Être en couple ne veut pas forcément dire qu’ils vécurent heureux et eurent beaucoup d’enfants Il y a toujours des hauts, des bas, des gens, des lois qui feront qu’être en

prochées, dont celles des côtés sont divergentes, comme dans les conques, à moins qu’il n’y ait une longue dent latérale,- et la plupart de ces cardiacées sont des coquilles

Loci that gave a PCR product in any of the tropical deer species were then screened for a higher number of samples : 8 Swamp deer from Zoo, 8 Rusa deer (four from Mauricia island

For 175375, the DNA product obtained after one-tube nested RT-PCR using panpes- tivirus primers (324 × 326 and A11 × A14) was identified only with a TaqMan BVDV2- specific

We found that (1) the emergence of H3N8 CIV from H3N8 EIV occurred in approximately 2002; (2) this interspecies transmission was by a reassortant virus of the circulating