19 benv january 2015 en

50
BENV National Veterinary Epidemiological Bulletin January 2015 Number 19 - CESME National Reference Centre for the study and verification of Foreign Animal Diseases - COVEPI Operational Veterinary Centre for Epidemiology Programming and Information Photo by Mark Dumont

Upload: benv-bollettino-epidemiologico-nazionale-veterinario

Post on 07-Apr-2016

236 views

Category:

Documents


1 download

DESCRIPTION

 

TRANSCRIPT

Page 1: 19 benv january 2015 en

BENVNational Veterinary Epidemiological Bulletin

January 2015Number 19

-

CESMENational Reference Centrefor the study and verificationof Foreign Animal Diseases

-

COVEPIOperational VeterinaryCentre for EpidemiologyProgrammingand Information

Pho

to b

y M

ark

Dum

ont

Page 2: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

2 Index

INDEX

-EDITORIAL 3

-IN THESE MONTHSWest Nile Disease: epidemiological situation in Italy, in Europe and in the Countries of the Mediterranean Basin 4Epidemological situation of Aethina tumida in Italy 6The national information system for Salmonella control programs in Italy 10

-HAND ON DATANumber of outbreaks reported to SIMAN in 2014 19Number of outbreaks reported by Regions to SIMAN in 2014 20Animals involved in outbreaks reported to SIMAN in 2014 24

-A LOOK AT THE MAPS 25

-AROUND USHighly Pathogenic Avian Influenza H5N8 – Epidemiological situation in Europe 29Lumpy skin disease: still a neglected disease? 34Mathematical models for the study of microbial growth, survival and inactivation: predictive microbiology between past and future 41

-OFFICIALLY FREE TERRITORIES 46 -CONTACTS & EDITORIAL STAFF 50

Page 3: 19 benv january 2015 en

January 2015 Number 19

EDITORIALThe BENV as a tool for disseminating information

Dear readers,

in this first issue of 2015, the Benv is presenting a lot of interesting articles, ranging from animal health to food safety.

The outbreaks of Highly Pathogenic Avian Influenza (AI) H5N8 occurred in Europe in late 2014 is one of the main topics of this issue: H5N8 virus strain have been identified in poultry farms located in an European area encompassing United Kingdom, the Netherlands and Germany. In December, the virus was also identified in a fattening turkeys farm in north-eastern Italy. In the section Around us, an article updates the current epidemiological situation in Europe, with particular attention to the prompt measures applied to limit the spread of AI to neighbouring areas.

In the same section, a review on Lumpy skin disease (LSD) is also presented: given the recent and important spread of LSD throughout the Middle East, including Turkey, where it is now considered endemic, this review could be of interest for the readers.

In a World experiencing an unprecedented rate of change and globalization, it is often necessary to apply proactive strategies in order to rapidly react to the threats against consumer’s safety. Preventing instead of correcting is the preferable approach. To this aim, statistic and mathematical tools specifically designed to predict microbial behaviour in food processing can be indispensable tools. In this framework, an overview on Predictive microbiology is given in the section Around us. It is an interdisciplinary research area that has become more and more used to evaluate, through suitable mathematical models, the responses of pathogenic or spoilage microorganisms to different environmental conditions that could be find in foods during processing and storing.

In the section In recent months, you can find the usual update on the trend of the diseases occurred in the national territory and in the Mediterranean area. An article shows the epidemiological situation of West Nile Disease in Italy and in the neighbouring countries. Another article presents the updated situation of the outbreaks due to Aethina tumida in the South of Italy. This parasite of bees has been responsible of 60 confirmed cases in Calabria and one case of infestation in Sicily. It has been responsible for serious damages to the hives and honeycombs, causing fermentation of honey contained in them.

Finally, in the same section the Benv introduces the Italian Information System for the collection and management of data on the national Salmonella control programmes (SISalm) in poultry flocks of Gallus gallus and turkeys. This system collects data on official sampling carried out by the official veterinary services and on industry sampling carried out by the food business operator. Therefore, it is a comprehensive system which is integrated with the other national information systems and databases in place in Italy, such as the national information system for the notification of outbreaks of animal diseases (SIMAN) and the national database of animals and holdings (BDN).

Regarding the data on outbreaks, in the Hand on data section, you can consult the tables with the data on outbreaks of animal diseases reported to SIMAN in 2014, the health status of the territories and the animal species involved in the outbreaks.

The maps show the distribution of the main animal diseases occurred in Italy in 2014.

Finally, it our pleasure to inform you that from this year the bulletin is published on Issuu as well, a web service for uploading digital documents (such as books, magazines, newspapers). It is integrated with social networks to endorse the loaded material. Like most of the documents published on the internet, some can also be downloaded and saved. So you can find, display and download this and the previous issues also through the Issuu tool.

We wish you all to enjoy a happy new year together with the Benv.

Simona Iannetti COVEPI

3 Editorial

Page 4: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

4 In these months

IN THESE MONTHSThe main events of epidemiological interest in the lastmonths in Italy and in the European Union

West Nile Disease: epidemiological situation in Italy, in Europe and in the Countries of the Mediterranean Basin in 2014

Introduction

West Nile virus (WNV) is a Flavivirus belonging to the family Flaviviridae and is transmitted by mosquitoes.

The infection is maintained in nature through transmission cycles between birds and mosquitoes particularly Culex spp [Komar et al., 2001], however mammals including humans and equidae are also susceptible to the infection and, although considered accidental or dead-end hosts, can show clinical symptoms ranging from a flu-like syndrome to a meningoencephalitis which can cause the death of patient [Komar., 2000]. Furthermore in humans have been described cases of vertical transmission, from mother to foetus or through breast milk or even transmission through blood transfusions or organ transplants from infected donors

Epidemiological situation in Europe and in the Countries of the Mediterranean Basin

WNV is one of the most widespread arbovirus in the world, in the last 30 years the increasing incidence of West Nile Disease (WND) in horses and humans in Europe and in the Countries of the Mediterranean Basin, raised the attention on the WNV which nowadays is one of the major causes of viral meningoencephalitis in humans [Calistri et al., 2010; Di Sabatino et al., 2014]. It is difficult to determine if the increasing number of WND cases is due to a wider spread of the virus rather than the growing attention for this infection. Whatever the reasons for its enhanced spread, it is important to clarify the ecological and epidemiological models of infection, which are related to the endemisation of the infection as well as the possible ways of virus introduction.

In 2014 during the epidemic of WND that affected Europe and the Countries of the Mediterranean Basin, 210 cases of human infections have been notified in Bosnia Erzegovina, in Greece, in Russian Federation, in Serbia, in Austria, in Italy, in Romania, in Israel, in Palestina and in Hungary as well as 39 cases of infection in equidae in Greece, in Croatia, in Italy, in Spain and in Turkey (figure 1 and and table 1).

Page 5: 19 benv january 2015 en

January 2015 Number 19

5 In these months

Figura 1.Geographical distribution of WND human and equidae cases (probable

and confirmed) in Europe and in Countries of the Mediterranean

Basin in 2014 - last updated December 18th, 2014

Table 1. Details of the WND cases (probable and confirmed) in Europe and in the Countries of the Mediterranean Basin in 2014 - last updated December 18 th, 2014

State SpeciesNo.

Probable cases

No. Confirmed cases

Bosnia and Herzegovina Human 13 0

GreeceEquidae 4 4Human 15 13

Russian Federation Human 29 --Serbia Human 76 56Croatia Equidae 1 1Austria Human 1 1Israel Human 17 7Palestine Human 1 1

ItalyEquidae 27 27Human 24 24

Romania Human 23 22Hungary Human 11 3Spain Equidae 6 6Turkey Equidae 1 1

Total human 210 127Total equidae 39 39

Source of human date ecdc

Page 6: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

6 In these months

Epidemiological situation in Italy

In Italy, the first reported WND outbreak occurred during the late summer of 1998 in the area around the marshes of Fucecchio, in Tuscany Region, with some clinical cases in equidae.

Following this epidemic, the Italian Ministry of Health, since 2002, put in place the WND National Surveillance Plan aiming to monitor the introduction and circulation of WNV in the Country. The surveillance program allowed to identify in 2008, 10 years after the first outbreak, the circulation of WNV belonging to Lineage I, in Emilia Romagna, in Veneto and in Lombardy Regions in birds, in mammals and in vectors [Monaco et al., 2010]. Since then the infection has been reported every year in humans and animals involving both new areas of Central and Southern Italy and areas affected by the viral circulation in previous years demonstrating the endemisation of the infection. To complete the epidemiological scenario of the WND in Italy is essential to report the entrance and circulation of a new viral lineage, lineage II, in 2011 in the provinces of Udine and Treviso. During the following epidemic seasons, the new lineage spread also in the areas previously infected by the circulation of WNV lineage I, namely in Sardinia in 2012, in Emilia Romagna and Lombardy in 2013.

During the 2014 WND epidemic season, both human cases in Emilia Romagna, in Veneto, in Lombardy Regions and animal cases in Emilia Romagna, in Lombardy, in Veneto, in Friuli Venezia Giulia, in Piedmont, in Puglia, in Sicily and in Liguria Regions were reported. All the regions have been already affected by the viral circulation during the previous years with the exception of Liguria. The first positivity was documented on June 16th in chickens in Sicily Region; followed by the notification of WNV in early July in a pool of mosquitoes in Lombardy and in Veneto Regions and in a Carrion crow in Emilia Romagna Region, while the first clinical cases in equidae and in humans occurred in the same areas of Northern Italy at least 6 weeks later. Once again the veterinary surveillance activities were able to ensure an early warning essential for the rapid implementation of control measures to prevent the transmission of the infection to humans.

On 18/12/2014 the National Reference Centre for the study of Foreign Diseases of Animals (CESME) confirmed (Table 2):

• 27 WND cases in equidae distributed in 17 outbreaks in Emilia Romagna, in Lombardy, in Veneto, in Friuli Venezia Giulia, in Piedmont and in Puglia Regions. Nervous symptoms referable to WNV infection were reported in 6 horses in Reggio Emilia, in Piacenza, in Lodi, in Mantova and in Vicenza Provinces;

• 34 WNV PCR positive target birds from Emilia Romagna and Lombardy Regions. In particular the virus was detected in tissues of 16 Carrion crows (Corvus corone cornix), 17 Magpies (Pica pica) and one Eurasian jay (Garrulus glandarius);

• 8 WNV PCR positive in Emilia Romagna Region from tissues of 5 Collared doves (Streptopelia decaocto), one little owl (Athene noctua) and 2 Goshawk (Accipiter gentilis);

• 109 WNV PCR positive from 109 mosquitoes pools collected in Lombardy, in Emilia Romagna, in Veneto, in Friuli Venezia Giulia, in Sardinia, in Piedmont and in Liguria Regions;

• 7 serological positivity in poultry in Sicily Regions.

In 2014 WNV strains circulating in Italy were identified as belonging to the Lineage II with the exception of a viral strain responsible for clinical symptoms in a horse in Vicenza Province.

The nucleotide sequences analysis clustered the virus isolated from the tissues of the animal within the Lineage I. Figure 2 shows the geographical distribution of viral circulation in 2014 in Italy up to December 18th.

The information related to the evolution of the epidemiological situation in Europe and in the Countries of the Mediterranean Basin are collected and published in the WND Bulletin, constantly updated by the CESME and the National Reference Centre for Veterinary Epidemiology, Programming, Information and Risk Analysis (COVEPI) and freely accessible on the website of the Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “Giuseppe Caporale” (IZSAM).

Page 7: 19 benv january 2015 en

January 2015 Number 19

7 In these months

Figure 2.Geographical distribution of

the WNV circulation in Italy in 2014 - last updated December

18th, 2014 -

Page 8: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

8 In these months

Table 2. Details of the 2014 WND cases in animals according to the province - last updated December 18th, 2014 -

Region Province Animals No. Positive animals

Emilia Romagna

Parma

Carrion crow 7Magpie 1Mosquitoes pool 7Goshawk 2

Bologna Mosquitoes pool 10

ModenaMosquitoes pool 18Magpie 4

Reggio EmiliaMagpie 1Mosquitoes pool 19Horses 2

Ferrara

Mosquitoes pool 21Magpie 7Eurasian Jay 1Collared dove 5Little owl 1

PiacenzaHorses 1Mosquitoes pool 7Carrion crow 1

RavennaMosquitoes pool 1Magpie 1

Bologna Magpie 1

Lombardy

CremonaCarrion crow 2Mosquitoes pool 5

Mantova

Carrion crow 2Magpie 1Mosquitoes pool 3Horses 3

LodiMosquitoes pool 1Horses 7

BresciaMosquitoes pool 4Magpie 1

Bergamo Carrion crow 2Pavia Mosquitoes pool 1

MilanoMosquitoes pool 1Carrion crow 1

Lecco Carrion crow 1

Veneto

VicenzaMosquitoes pool 1Horses 1

VeronaMosquitoes pool 4Horses 6

Rovigo Horses 1Sicily Catania Chicken 7

Friuli Venezia GiuliaUdine

Mosquitoes pool 1Horses 2

Pordenone Mosquitoes pool 1Sardegna Olbia Tempio Mosquitoes pool 1

Piemonte AlessandriaMosquitoes pool 2Horses 1

Puglia Lecce Horses 1Brindisi Horses 2

Liguria Genova Mosquitoes pool 1Total 185

Fonte SIMAN, 29/09/2014

Page 9: 19 benv january 2015 en

January 2015 Number 19

9 In these months

References

1. Autorino GL, Battisti A, Deubel V, Ferrari G, Forletta R, Giovannini A, Lelli R, Murri S, Scicluna MT. West Nile virus epidemic in horses, Tuscany region, Italy. Emerg Infec Dis. 2002. 8, 1372-1378.

2. Monaco F, Lelli R, Teodori L, Pinoni C, Di Gennaro A, Polci A, Calistri P, Savini G. Re-emergence of West Nile virus in Italy. Zoonoses Public Health. 2010.;57(7-8):476-86.

3. Calistri P, Giovannini A, Hubalek Z, Ionescu A, Monaco F, Savini G, Lelli R. Epidemiology of West Nile in Europe and in the Mediterranean basin. Open Virol J. 2010 Apr 22;4:29-37.

4. Di Sabatino D, Bruno R, Sauro F, Danzetta ML, Cito F, Iannetti S, Narcisi V, De Massis F, Calistri P. Epidemiology of West Nile disease in Europe and in the Mediterranean Basin from 2009 to 2013. Biomed Res Int. 2014;2014:907852. Epub 2014 Sep 11.

5. Komar N, Panella NA, Burns JE, Dusza SW, Mascarenhas TM,Talbot TO. Serologic evidence for West Nile virus infection in birds in the New York City vicinity during an outbreak in 1999. Emerg Infect Dis. 2001; 7:621-5.

6. Komar N. West Nile viral encephalitis. Rev Sci Tech 2000; 19:166-76.

--Edited by:Rossana BrunoCOVEPIiIstituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale”

Page 10: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

10 In these months

Epidemological situation of Aethina tumida in Italy

Introduction

Aethina tumida, often reported with the acronym SHB (Small Hive Beetle) is a beetle of the family Nitidulidae. It ‘s a scavenger of Apis mellifera capensis and Apis mellifera scutellata, two bee populations in sub-Saharan Africa, that have behavioral defenses more marked than European subspecies (Neumann, 2008). Those characteristics make more difficult, in African bees, reproduction and spread of parasite, and make less damaging to hives the impact of the infestation. A. tumida can infest also colonies of insects belonging to the genus Bombus, it can also survive and reproduce on different varieties of ripe fruit. However, its reproduction on fruit occurs at orders of magnitude less than on bee products in hives (Buchholz et al., 2008). The main damages caused to the hive, depend on the presence of the larvae of the parasite that feeds brood, pollen and honey; they also destroy honeycombs, causing fermentation of honey contained in them. The final consequence is often the abandonment of the hive by the colony. The adult parasite is able to flight even several kilometers, facilitating the spread of the infestation in other apiaries in the same area (Cuthbertson, 2013). Through exchanges of bees and beekeeping equipment, A . tumida expanded its range of distribution to the American continent. The first report of A . tumida dates back to 1996 in South Carolina, then it was found in Florida in 1998, in 2000 in Egypt, in Canada and Australia in 2002 (De Guzman, 2010).

In 2004 in Portugal, eggs and larvae of the beetle were found in transport cages of queen bees from the US (Texas). In that circumstance were traced and destroyed all the queens belonging to that batch, as welle as the apiaries where the queens were allocated, and the infestation was eradicated (Valerio Da Silva, 2014)). The disease was exotic in EU until September 2014, and subject to notify as provided by Commission Decision (CE) No. 2004/216. It is included in the list OIE (2015). The importation of queen bees from third countries is regulated by strict rules laid down by Commission Regulation (EU) No. 206/2010.

Aethina tumida in southern Italy 2014

On September 5, 2014, in an apiary owned by the University of Reggio Calabria, located near the port of Gioia Tauro, Calabria region, were found three infested hives with numerous adults and larvae specimens of A.tumida. The identification was made by Prof. Palmeri of the University of Reggio Calabria (Palmeri et al., 2014). It was then identified by the National Reference Centre for Beekeeping based on morphology, and confirmed by morphological identification and through molecular techniques by the European Reference Centre of Sophia - Antipolis (Mutinelli et al., 2014). As a result of confirmation of the presence of A. tumida in Italy the Italian Ministry of Health, on September 12, issued a note in which was ordered the tracing and control of the apiaries that had carried out activities of nomadism in Calabria region during 2014 productive season, furthermore were indicated the measures to be taken in case of positivity, as seizure and destruction of bees and of all beekeeping equipment possible vehicles of infection. Treatments with insecticides on the surrounding land to the hives were recommended. On 19 September, by a decree of President of Regional Council of Calabria, was established a protection zone with a 20 km radius from the first outbreak site in the town of Gioia Tauro, and was circumscribed a surveillance zone with a 100 kilometers radius. Within the protection zone was forbidden any movement of colonies or beekeeping material, and in all apiaries present was checked a number of hives statistically appropriate to detect an expected percentage of infestation prevalence of 5% with a 95% confidence interval. Within the surveillance zone, inspected apiaries were chosen based on the presence of risk factors, or in the absence of these, according to a randomness criterion. In this zone were checked a statistically appropriate number of hives to detecting a percentage of infestation expected prevalence of 2% with a 95% confidence interval. Controls were performed through the execution of complete inspections of the hives, furthermore were collocated traps to catch adult beetles in all hives of the apiaries visited (Mutinelli et al., 2014).

Page 11: 19 benv january 2015 en

January 2015 Number 19

11 In these months

The last update of the epidemiological situation in Calabria region made available by the National Reference Centre for beekeeping, Istituto Zooprofilattico Sperimentale delle Venezie in date 12.01.2015, reports 60 confirmed cases of infestation A .tumida, all included in the protection zone (Figure 1). In six outbreaks were found, in addition to the presence of adults of A .tumida, larvae, and pupae too in one of them.

Figure 1.Epidemiological situation of confirmed cases in Calabria

(map edited by IZS Venezie)

!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!( !(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(!(!(

!(!(!(

!(

!(

!(!(!(

!(

!(

!(

!(

!(

!(!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(!(

!(

!(

!(

!(!(

!(!(

!(!(

!(

!(

!(

!(

!(!(!(!( !(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(!(!(

!(

!(!(

!(!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(!(!(

!(!(!(

!(

!(!(!(

!(

!(

!( !(

!(

!(

!(!(!(

!(!(!(

!(

!(

!(

!(!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(!(!(

!(!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(!( !(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(!(

!(

!(!(!(!( !(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!( !(!(

!(

!(!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!( !(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!( !(

!(!(

!(

!(!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(!(

!(

!(!(

!(

!( !(

!(

!(

!(

!(

!(

EE

E

E

EEEEE

EE

E

EE

EE

E

EE

EEE

E

E

E

EE

E

E

EE

EE

E

E

E

E

E

E

E

E

E

E

E

EE

E

EE

E

E

E

E

EE

E

E

E

E

E

Villa SanGiovanni

Seminara

Condofuri

Samo

Platì

Rosarno

Maropati

Limbadi

Ionadi Francica

Zambrone

Sant'Ilariodello Ionio

Portigliola

Dasà

MonterossoCalabro

Capistrano

Pazzano

Placanica

Jacurso

Camini

Messina

Calanna

Careri

SanFerdinando

Feroletodella

Chiesa

Candidoni

Caraffa delBianco

Siderno

Fabrizia

Pizzoni

Maierato

Nardodipace

Caulonia

Cardinale

Guardavalle

Vallefiorita

Gagliato

MontebelloIonico

Sant'Eufemiad'Aspromonte

Sinopoli

Melicuccà

Palmi

Ricadi

BruzzanoZeffirio

San GiorgioMorgeto

Cinquefrondi

Filandari

SanCostantino

Calabro

Laureana diBorrello

Parghelia

SanGregoriod'Ippona

Galatro

Mammola

Acquaro

Sorianello

RoccellaIonica

Brognaturo

Torre diRuggiero

ChiaravalleCentrale

Palermiti Montauro

Riace

Tropea

Cosoleto

SanProcopio

Scido

Molochio

Cittanova

Anoia

Mileto

Zaccanopoli

Bianco

Casignana

Bovalino

Ciminà

Ardore

Grotteria

Simbario

Pizzo

Francavil laAngitola

Polia

Filadelfia

Martone

Stignano

Sant'AndreaApostolo

dello Ionio

Olivadi

Stalettì

Reggio diCalabria

Roghudi

BagnaraCalabra

Drapia

Sant'Agatadel Bianco

SantaCristina

d'Aspromonte

Varapodio

Taurianova

Nicotera

Zungri

Giffone

Stefanaconi

Sant'Onofrio

StiloMongiana

San Vitosullo Ionio

Centrache

Argusto

Cardeto

Fiumara

Gioia Tauro

Spilinga

Bova

Delianuova

OppidoMamertina

Melicucco

Serrata

Briatico

ViboValentia

Ferruzzano

Benestare

Gerace

San Pietrodi Caridà

Gerocarne

SerraSan Bruno

Filogaso

Spadola

Petrizzi

Montepaone

Monasterace

Laganadi

SantoStefano in

Aspromonte

Scilla

Roccafortedel Greco

Staiti

San Luca

Africo

Rizziconi

Polistena

SanCalogero Dinami

Cessaniti

Antonimina

Canolo

Arena

Vazzano

GioiosaIonica

IscasulloIonio

Badolato

Cenadi

CortaleGirifalco

Gasperina

Satriano

Davoli

Soverato

CampoCalabro

Motta SanGiovanni

Sant'Alessioin Aspromonte

BagaladiSan

Lorenzo

SanRoberto

JoppoloRombiolo

AgnanaCalabra

Locri

SorianoCalabro

San Nicolada Crissa

Vallelonga

Marinadi Gioiosa

Ionica

San Giovannidi Gerace

Bivongi

SanSostene

Santa Caterinadello Ionio

CATANZARO

REGGIO DICALABRIA

VIBOVALENTIA

MESSINA

0 7,5 153,75km

¯12/01/2015

E

E

E

E

EE

EE

E

E E

E

E

E

E

E

E

E

E

E

E

E

E

E

E

EE

E

E

E

E

E

E

E

E

E

E

E

E

E

E

E

E

E

E

E

E

EE

E

E

E

E

E E

E

E

E

E

E

Seminara

Rosarno

Maropati

Limbadi

SanFerdinando Feroleto

della Chiesa

Candidoni

Sant'Eufemiad'Aspromonte

Melicuccà

Sinopoli

Palmi

SanGiorgioMorgeto

Cinquefrondi

Laureana diBorrello

Galatro

SanProcopio Cosoleto

Scido

Molochio

Cittanova

Anoia

Mileto

CiminàSantaCristinad'Aspromonte

Varapodio

Taurianova

Nicotera

Giffone

GioiaTauro

Delianuova

OppidoMamertina

Melicucco

Serrata

San Pietrodi Caridà

RizziconiPolistena

SanCalogero

Dinami

1 23

4

5

67

8

9

1011

12

1314

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

3031

3233

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

4950

51

52

54

5556

57

58

59

60

61

E Positive

!( Negative

20000 m.

Province Municipality

Figure 2.Epidemiological situation of

confirmed cases in Sicily (map edited by IZS Venezie)

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!( !(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(!( !(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(!(

!(

!(

!(

!(!(

!(

!(

!(

!(

!(!(

!(!(

!(!(

!(

!(

!(

!(

!(

!(!(

!(

!(

!(

!(!(

!(

!( !(

!(

!(

!(

!(

!(!(

!(

!(

!(!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

!(

E

LicodiaEubea

Scordia

Ferla

MottaSant'Anastasia

SantaMaria

di Licodia

Trecastagni

AciCastello

Acireale

Buccheri

Castel diIudica

Ramacca

Buscemi

Avola

Floridia

TremestieriEtneo

MonterossoAlmo

Mineo

Paternò

Misterbianco

CamporotondoEtneo

Siracusa

San Giovannila Punta

SanGregorio

di Catania

Militelloin Val diCatania

Agira

Centuripe

Rosolini

Cassaro

Nicolosi

Sant'Agatali Battiati

ChiaramonteGulfi

Ragusa

Modica

Sortino

Melilli

Carlentini

Belpasso

Ragalna

Mascalucia

CanicattiniBagni

Solarino

Augusta

AciSant'Antonio

Comiso

Giarratana

Vizzini

Grammichele

Adrano

San PietroClarenza

PrioloGargallo

Palagonia

Regalbuto

Pedara

AciCatena

Caltagirone

Francofonte

Lentini

Catenanuova

Biancavilla

Noto

PalazzoloAcreide

Catania

ViagrandeAci

Bonaccorsi

ENNA

SIRACUSA

RAGUSA

CATANIA

0 7,5 153,75km

¯12/01/2015

E

Sortino

Melilli

Carlentini

Augusta

PrioloGargallo

Lentini

Catania

53

E Positivo

!( Negativo

20000 m.

Provincia

Comune

Two hundred and nineteen apiaries within the protection zone, 419 within the surveillance zone, 338 outside the two areas, a total of 976 apiaries were found negative for the presence of A. tumida (Mutinelli et al., 2014).

On November 7th adults of A. tumida were found in a nomadic apiary in the municipality of Melilli, in the province of Syracuse, Sicily. The epidemiological investigation carried out by the veterinary services showed that these hives had been in the area of Gioia Tauro, site of the first discovery in Calabria, in the period from April to August 2014. On 12 January, 2015, as reported by the National Reference Centre for ‘beekeeping, no further outbreak of A. tumida in Sicily was reported. Were performed checks in 24 apiaries in the protection zone, 164 in the surveillance zone, 10 outside of the two zones for a total of 198 apiaries (Figure 2).

Page 12: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

12 In these months

The note of the Ministry of Health of the 1st of October 2015, established the criteria for the implementation of surveillance activities in the regions in which was not detected the presence of A. tumida. Data on the activity carried out so far on the Italian territory are not yet available, but up to now there are no reports of the presence of the parasite outside the regions of Calabria and Sicily.

Figure 3 resumes the surveillance activities carried out in Calabria and in Sicily regions within the two areas of surveillance zones of 100 Km radius areas.

Public health actions undertaken

In accordance with the legislation in force all infested apiaries were destroyed by incineration on the site, after killing the bees with vaporization of sulfur dioxide. The soil around the apiary has been plowed, and further treated with a 1% solution of cypermethrin and tetramethrin.

On 19 November, 2014, the Ministry of Health by a decree, reaffirms, pending the identification of the prevalence of the infestation and the availability of alternative measures of containment, the obligation of destruction of all hives present in infested apiary, as well nuclei, bee queens or any biological material capable of conveying eggs, larvae or adults of A. tumida. The same decree states that the owners of the apiaries destroyed will be paid the compensation provided by law June 2, 1988 n. 218.

Commission implementing Decision (EU) No. 2014/909 of 12 December 2014, forbids the shipping outside the territory of the regions of Calabria and Sicily to other areas of the Union of bees, bumble bees, bee-products unprocessed, beekeeping equipment and comb honey for human consumption.

The effects of this measure will remain in force until 31 May 2015.

Hypothesis on the introduction of A. tumida in Italy

The location of the first outbreak of A. tumida in the immediate vicinity of the cargo port of Gioia Tauro did focus initially on this possible source of introduction of the parasite, although the investigations performed have not been confirmed (Mutinelli et al., 2014).

Another hypotheses that can be reasonably be considered is through the illegal introduction of bees and of beekeeping material.

Figure 3.Surveillance activities carried out in the protection zones of Calabria and Sicily (map edited by IZS Venezie)

!

!!

!! !!

!

!

!

!

!

!

!

!

!!

!

! !!

!

!

!

!!!

!

!

!

!

!

!

!

!

!

!

!!

!

!

!

!

!

!

!

!

!

! !

!

!

!

!

!!

!

!!

!

!!

!!!!!!

!

!

!!!

!

!

!

!

!

!!!

!

!!

!

!!

!

!

!

!!

!!

!

!

!

!

!!!

!

!

!

!

!

!

!

!!!!

!!

!

!!

!!

!

!

!!!!

!!!!

!

!

!

!

!

!!!

!

!

!

!!

!

!

!

!

!!

!

!

!!!

!

!

!

!!

! !

!!

!

!!

!

!!!!!

!

!!

!

!!

!

!

!!

!

!!

!

!

!

!

!!

!!!

!

!

!

!!!

!

!!

!

!!

!!

!

!

!

!!!

!!

!

!

!

!

!

!

!

!!

!

!!

!

!!!

!

!

!

!

!

!

!!!

!

!

!

!!

!

!!

!

!

!

!

!!!

!!

!

!

!!

!!!

!!!

!!

!!!

!

!

!

!

!

!

!!

!!

!!

! !

!

!

!!

! !

!

!!

!!

!!

!

! !!

!

!

!

!

!

!!

!

!

!!

!

!!

!

!

!!

!

!

!

!!!!

!!!!

!!

!

!

!

!

!

!

!

!!

!

!

!!

!

!!

!!

!

!!

!

!

!

!

!!!

!!!

!

!

!

!

!

!

!

!

!!

!

!

!!

!

!

!

!

!

!!!

!

!!

!

!

!!

!

!!!!!!

!

!!

!

!!

!

!

!!!

!

!

! !

!

!

!

!

!

!!

!

!

!

!

!!

!

!

! !

!

!

!

!!

!

!

!

!

!!!

!!!

!!

!!

!

!! !

!!

!

!

!

!

!! !

!

!

!!!!

!

!!

!

!!!

!!!

!

!!

!

!

!

!

!!

!

!!

!!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!!

!

!

!

!

!

!

!

!!

!

!

!!

!!

!

!

!

!

!

!!

!

!

!

!

!

!!!

!

!

! !

!

!

!

!

!

!

!

!

!!

!!

!

!

!!

!

!!!

!!!

!

!

!

!!!

!

!

!

!

!!

!

!

!!

!

!

!!

!

!

!

!

!

!!!

!!!

!!

!

!!

!

!!

!!

!!

!

!

!

!!!

!

!!

!

!

!

!!! !

!!

!

!

!

!

!!!

!

!

!

!

!

!

!

!

!!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!!

!!

!

!

!!

!

!

!

!

!

!

!

!

!

!

!

!

!!

!

!!

!!

!

!

!

!

!!

!

!

!

!

!

!

!

!

!

!!

!

!

!

!!

!

!

! !

!!

!!

!

!

!

!!

!

!!!!!

!

!

!!

!

!!

!

!

!

!

!!!

!

!

!

!

!

!

!

!

!!

!

!

!

!!!

!

!!

!

!

!

!

!

!!

!

!

!

!

!

!

!

!

!!

!

!

!!!!

!!!

!

!

!

!

!

!

!!

!! !

!

!

!

!

!

!

!

!

!!

!

!

!

!

!!

!

!

!

!

!

!

!

!

!

!

!

!!

!

!

!

!

!!!!

!

!

!

!

!!!

!

!!

!

!!!

!

!!

!

!

!

!

!! !

!

!

!

!

!!!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!

!

!

!

!

!

!!

!

! !!

!

!

!

!

!!

!

!

!

!

!

!

!

!! !

!

!!

!

!

!

!!!

!

!

!

!

!

!

!

!!

!

!

!

!

! !

!

!!

!

!

!!

!

! !

! !

!

!

!

!

!

!

!

!!

!

!

!

!

! !

!

!!

!!

!!

!

!

!

!

!

!

!

!!

!

!

!

!!

!!

!

! !

!

!

!

!

!!

!

!

! !

!

!!

!

!!

!!

!!! !

!

!!!

!

!

!

!

!!

!

!

!

!

!!!

!!

!!

!

!

!

!! !!!

!

!

!!!

!

!!

!!!

!

!

!

!

!!

!

!

!

!

!

!

!

!

!

!!

!

!!!

!!

!

!!

!

!!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!

!!

!

!

!

!

!!

!

!

!!!

!

!!

!

!

!

!

!! !!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!

!!

!!

!

!

!!

!!

!

!

!

!

!

!

!

!!

! !

!

!

!!

!

!

!

!

!

!

!

!

!

!!

!!

!

!

!

!!

!

!!

!

!!

!!

!

!!!

!!

! !

!

!!

!

!!

!!

!

!

!

!!!

!

!

!

!

!

EEEEEEEE E

EE

EEE

EE

EEE

EEE

E

EEEE

E

EEE

E EEE

E

E

E

EE

E

EE

EEE

E

EE E

E

E

E

E

EEEEE

EE

AGRIGENTO

ENNA

SIRACUSA

COSENZA

CROTONE

TRAPANI

CATANZARO

RAGUSA

REGGIO DICALABRIA

CALTANISSETTA

VIBOVALENTIA

PALERMOCATANIA

MESSINA

0 30 6015km

¯12/01/2015

E Positive! Negative

100000 m.

Province Municipality

Page 13: 19 benv january 2015 en

January 2015 Number 19

13 In these months

Unfortunately, there are no certainties over the origin nor the date of introduction, but probably it should not be before the beginning of the production season of 2014 (February-March).

Possible future scenarios

In the plain of Gioia Tauro in the months of April and May, coinciding with the flowering period of citrus plants, thousands of hives converge from all parts of Italy. At the end of the harvest these hives return to their territories of origin, often performing even intermediate steps, for purposes of exploitation of blooms in other geographical locations. In light of these considerations it is evident that it cannot be underestimated the risk that the parasites may have spread to other areas of the Italian peninsula. The outbreak in Sicily is the proof . Is therefore essential that the monitoring activities be extended to the whole country by the veterinary services, through the adoption of a surveillance plan dedicated to this parasite. During winter control activities on hives, due to adverse weather conditions, undergoes a sharp slowdown, therefore it is likely that a clearer picture of the prevalence of infestation we will have during the production season 2015. Under the framework that emerge, we will have the elements to decide whether the action of eradication will be brought to completion, or if we have to choose a strategy that points to the containment of the infestation.

References

1. Buchholz S.; Schäfer M.O.; Spiewok S.; Pettis J.S.; Duncan M.; Ritter W.; Spooner-Hart R.; Neumann P. (2008) Alternative food sources of Aethina tumida (Coleoptera: Nitidulidae). Journal of Apicultural Research and Bee World, 47(3): 202–209.

2. COMMISSIONE DELLE COMUNITÀ EUROPEE 2004 DECISIONE n. 2004/216/CE del 1o marzo 2004 che modifica la direttiva 82/894/CEE del Consiglio concernente la notifica delle malattie degli animali nella Comunità al fine di includere talune malattie degli equidi e talune malattie delle api nell’elenco delle malattie soggette a denuncia. Gazz Uff, L 67, 27-30.

3. COMMISSIONE DELLE COMUNITÀ EUROPEE 2010 REGOLAMENTO (UE) N. 206/2010 del 12 marzo 2010 che istituisce elenchi di paesi terzi, territori o loro parti autorizzati a introdurre nell’Unione europea determinati animali e carni fresche e che definisce le condizioni di certificazione veterinaria. Gazz Uff, L 73, 120 pp.

4. COMMISSIONE DELLE COMUNITÀ EUROPEE 2014 DECISIONE DI ESECUZIONE DELLA COMMISSIONE n. 2014/909/UE del 12 dicembre 2014 relativa ad alcune misure di protezione a seguito della presenza confermata del piccolo scarabeo dell’alveare in Italia. Gazz Uff, L 359, 161-163.

5. CUTHBERTSON, A G S; WAKEFIELD, M E; POWELL, M E; MARRIS, G; ANDERSON, H; BUDGE, G E; MATHERS, J J; BLACKBURN, L F; BROWN, M A (2013) The small hive beetle Aethina tumida: A review of its biology and control measures. Current Zoology, 59(5): 644–653.

6. VALÉRIO DA SILVA, M J (2014) The first report of Aethina tumida in the European Union, Portugal 2004. Bee World, 91(4): 90-91.

7. MUTINELLI F. et al. (2014) Detection of Aethina tumida Murray (Coleoptera: Nitidulidae.) in Italy: outbreaks and early reaction measures. Journal of Apicultural Research 53(5): 569-575.

8. NEUMANN P.; ELLIS J.D. (2008) The small hive beetle (Aethina tumidaMurray, Coleoptera: Nitidulidae): distribution, biology and control of an invasive species. Journal of Apicultural Research and Bee World 47(3): 181–183.

9. PALMERI V.; SCIRTÒ G.; MALACRINÒ A.; LAUDANI F.; CAMPOLO O. (2014) A new pest for European honey bees: first report of Aethina tumida Murray (Coleoptera Nitidulidae) in Europe. Apidologie: http://dx.doi.org/10.1007/s13592-014-0343-9.

--Edited by:Luciano Ricchiuti1 and Franco Mutinelli21Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale” 2Centro di referenza Nazionale per l’Apicoltura, Istituto Zooprofilattico Sperimentale delle Venezie

Page 14: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

14 In these months

The national information system for Salmonella control programs in Italy

The framework

Salmonella infections are the main cause of food-borne outbreaks in Italy as well as in other industrialized countries; food of poultry origin is the main source of infection. Salmonella infection continue to be an important public health issue in the European Union (EU) even if it continued to decrease in 2012, with a total of 91,034 confirmed cases reported. This represented a 4.7 % decrease in confirmed cases compared with 2011 (EFSA and ECDC, 2014): in the last published EU Summary Report on zoonoses, zoonotic agents and food-borne outbreaks of the European Food Safety Authority (EFSA), a statistically significant decreasing trend has been observed in the period 2008-2012 in the EU, partly due to the combined and “from farm to fork” approach adopted by the EU, and much due to the successful application of the Salmonella control program in poultry sector, that led the majority of Member states to reach the reduction targets set for 2012. The EU legislation on food safety (White Paper, Commission Regulation (EC) no. 178/2002) identifies in the control of the food chain the more effective approach to ensure food safety for humans, indicating in the primary production a pivotal point for surveillance and control of Salmonella infections. EU Member States, European Commission, European Parliament, EFSA and the European Centre for Disease Prevention and Control (ECDC) are involved each for their expertise in risk management and assessment. This combined approach helped to almost halve in five years (2004-2009) the cases of salmonellosis in humans in the EU.

In 2003 the EU, considering the risk of Salmonella a priority, established measures for a broad control of zoonoses. The EU set for the Member States the objectives of reducing the prevalence of Salmonella and of other food-borne zoonotic agents on the basis of specific control programs approved by the Commission (Commission Regulation (EC) no. 2160/2003). Under this Regulation, eradication programs, control and monitoring of salmonellosis (serotypes relevant to public health) in breeding flocks of Gallus gallus, laying hens, broilers, turkeys, breeding pigs and pigs for meat production were approved (table 1). With the subsequent Regulation (EC) No. 1003/2005, implementation of Regulation (EC) No. 2160/2003, the requirement for farmers to implement an industry sampling program was also introduced, thus contributing to the verification of the achievement of the Community objective of reducing the prevalence of infection.

In this framework, an information system has become indispensable to collect and manage information and data derived from the application of the Salmonella Control programs in Italy (PNS), in agreement with the community legislation, to standardize data collection and information flows, thus avoiding redundancy and errors.

This system, named SISalm, is available at the unique portal of the Italian veterinary information systems and collects detailed data on the activities foreseen by the PNS.

Page 15: 19 benv january 2015 en

January 2015 Number 19

15 In these months

Organization of the information system

SISalm collects data on official and industry sampling. Data on industry sampling derives directly from the farmer: in fact, the use of the system became mandatory in 2009 for the collection of data on official and industry sampling in broiler flocks; in 2010 the use of the system became mandatory also for turkey flocks. From 2012, data on industry sampling shall be collected for all the categories of poultry included in the PNS. SISalm is fed by the regional veterinary services and by the Local health authorities, which access the system also for consulting detailed data on their activities, both official and industry, of their territorial competence.

Industry sampling is an integral part of the official control: where the food business operator (OSA) is required to apply the relevant legislation, the competent authority has the obligation to ensure its conformity (Regulation (EC) no. 882 / 2004). This principle is valid for all OSA and for all the stages of production, from farm to the sale of food to the consumer, in line with European policy “from farm to fork” (Regulation (EC) no. 178/2002, Regulation (EC) no. 852/2004). The farmers must make a specific request for accessing SISalm. Once logged in through appropriate credentials, the farmer can feed the system, view and download data for its own farm as well as it has been reported in the BDN (Note of the Ministry of Health DGSAF Prot. 12682-P of 08/07/2009).

Functionalities

SISalm allows to entry and visualize data of territorial competence on the activities performed in the context of the PNS by form on line, Upload and Web services. The veterinary services can download in Excel format data of their own competence. The epidemiological unit in the PNS is the flock defined as “all poultry of the same health status of the same breeding cycle, with the same date of placement kept on the same premises or in the same enclosure and constituting a single epidemiological unit”. From July

Table 1. Specified zoonoses and zoonotic agents for which Community targets for the reduction of prevalence are to be established pursuant to Article 4

1. Zoonosis or z zoonotic agent

2. Animal population

3. Stage of food chain

4. Date by which target must be established (*)

5. Date from which testing must take place

All salmonella serotypes with public health significance

Breeding flocks of Gallus gallus

Primary production

12 months after the date of entry into force of this

Regulation

18 months after the date referred to in column 4

All salmonella serotypes with public health significance

Laying hens Primary

production

24 months after the date of entry into force of this

Regulation

18 months after the date referred to in column 4

All salmonella serotypes with public health significance

Broilers Primary

production

36 months after the date of entry into force of this

Regulation

18 months after the date referred to in column 4

All salmonella serotypes with public health significance

TurkeysPrimary

production

48 months after the date of entry into force of this

Regulation

18 months after the date referred to in column 4

All salmonella serotypes with public health significance

Herds of slaughter pigs

Slaughter48 months after the date of entry into force of this

Regulation

18 months after the date referred to in column 4

All salmonella serotypes with public health significance

Breeding herds of pigs

Primary production60 months after the date of entry into force of this

Regulation

18 months after the date referred to in column 4

(*) These dates are based on the assumption that comparable data on prevalence will be available at least six months before the establishment of the target. If such data were not available, the date for the establishment of the target would be postponed accordingly.

Page 16: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

16 In these months

Integration with other information systems

In line with the strategies of application cooperation aiming for data sharing, avoiding duplications and inefficiencies due to the need to maintain aligned different archives, SISalm is designed to be integrated as with the BDN as with the Italian Information System for the notification of outbreaks of animal diseases (SIMAN) for the automatic notification of the suspected outbreak (figure 2).

The integration with the BDN allows printing sampling models with the registry section already pre-filled with data of the farm as recorded in the BDN. In this way models with handwritten and often illegible information can be avoided and it is possible to the read the barcodes in the models to avoid manual entry of the information, while encoding it in the computer systems.

SISalm cooperates with SIMAN querying a web service for the notification of the suspected outbreak to automatically load the suspected outbreak in case of a positive result in one or more of the Salmonella serotypes relevant to the plan (either in case of official and industry sampling).

Figure 1.Reports with statistics on official control activities carried out by the Competent Authorities (local health units) and on industry sampling carried out by the farmer

2014, the new national poultry computerized registry has been implemented, and this topic will be examined in depth in one of the next issues of the BENV. Under the new registry, the shed is uniquely identified and recorded. This aspect is crucial because the flock is the unit of reference for the evaluation of the reduction target of the prevalence of Salmonella.

SISalm provides users with statistics on its feeding by the users: which Regions feed the system and how, how many samples are entered in the period compared to those expected, how many positive results over the total of the data entered and compared to Salmonella serotypes. All reports are available in Excel format (Figure 1).

Recently, SISalm has been implemented with the functionality of managing the list of the private laboratories compliant to the legislation requirements.

Private laboratories must sign up by filling out the form available in SISalm indicating the possession of the requirements foreseen by the legislation.

To verify the maintenance of these requirements during time, SISalm interfaces with the database of Accredia and with the National Reference Centre for Salmonella, which organizes an annual inter-laboratory circuit, in order to verify the ability of the laboratory to isolate Salmonella at different concentrations from samples of faeces and / or dust.

Page 17: 19 benv january 2015 en

January 2015 Number 19

17 In these months

Figure 2.SISALM integration with other

information systems

In case of the notification of the suspected outbreak, SIMAN send an email alert to the veterinary service of the local health unit (ASL), of the Region and to the Ministry of Health. Each week the email alert is renewed in case of the absence of confirmation/deletion of the suspected outbreak.

SISalm feeds also the information system “Rendicontazioni”, that is responsible for collecting and sending official data on the national co-financed programs to fulfil the information debt towards the European Commission, in application of Decision (EC) No. 2008/940 and Decision (EC) No. 2003/886.

Data collected by SISalm also feed the Italian information system on zoonoses data collection (SINZoo), which collects data on the monitoring of zoonoses and zoonotic agents in animal health, food and feed, in order to fulfil the information debt towards the EFSA, as required by Directive (EC) no. 2003/99.

In particular, data collected by SISalm is aggregated and processed according to the EFSA’s specifications for the evaluation of the reduction target set by the EU legislation.

The integration of all the information systems allows a comprehensive reporting with statistics on sampling activities planned and carried out, on the results of the samples taken and on the reported outbreaks.

The integration also allows optimizing the fulfilment of the information debts towards the European Commission, the EFSA and the OIE, avoiding double reporting of the same information with different timelines and different levels of aggregation. In this way, data is always uniform and unique because it has been derived from a single source.

Page 18: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

18 In these months

Conclusions

Through the use of SISalm, the information flow is unique and trace the entire process, from the sampling to the analytical results (both for official and industry sampling).

Competent Authorities have always available the updated information on farms and flocks, on industry sampling, on the tested samples, on the analytical results, on the laboratories involved, etc. The data collected also allows the programming of activities, their periodic verification and risk analysis.

Finally, thanks to SISalm the number of official and industry sampling has been increased, as well as the state of implementation of the computerized registry of poultry farms and the reporting to SISalm of data related to industry sampling (Figure 3). Moreover, SISalm has played a pivotal role to achieve the reduction target of the prevalence of Salmonella in poultry.

References

1. European Commission, 2005. Regulation (EC) No. 1003/2005 of the Commission of 30 June 2005 implementing Regulation (EC) No. 2160/2003 of the European Parliament and of the Council as regards a Community target for the reduction of the prevalence of certain Salmonella serotypes in breeding flocks of Gallus gallus and amending Regulation (EC) No. 2160/2003. OJ L 170, 1.7.2005, p. 12-17

2. EFSA (European Food Safety Authority) and ECDC (European Centre for Disease Prevention and Control), 2014. The European Union Summary Report on Trends and Sources of Zoonoses, Zoonotic Agents and Food-borne Outbreaks in 2012. EFSA Journal 2014;12(2):3547, 312 pp. doi:10.2903/j.efsa.2014.3547

3. Italian Ministry of Health. Note of the DGSAF Prot. 12682-P, 8th of July 20094. European Parliament, Council of the European Union, 2003. Council Regulation

(EC) No. 2160/2003 of the European Parliament and of the Council of 17 November 2003 on the control of Salmonella and other specified in foods. OJ L 325, 12.12.2003, p. 1-15

--Edited by:Simona Iannetti (COVEPI), Elio Malizia, Patrizia Colangeli Data Processing Center, Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale”

Figure 3.SISALM integration with other

information systems

Page 19: 19 benv january 2015 en

January 2015 Number 19

19 Hand on data

HAND ON DATA Processing date: 19th January 2015Number of outbreaks reported to SIMAN in 2014

Disease Jan Feb Mar Apr May Jun Jul Aug Sep Oct Nov Dec Total outbreaks

Aethina tumida 14 30 8 1 53

African swine fever 21 9 2 5 13 12 3 2 24 6 97

American foulbrood of honey bees 2 11 7 19 14 14 10 7 3 87

Antrax 1 2 3

Avian typhosis 1 1 2

Bluetongue 16 7 3 1 3 10 162 295 533 232 143 57 1462

Bovine leucosis 5 1 4 2 4 6 3 1 1 1 1 1 30

Bovine tuberculosis 32 31 28 31 52 57 24 17 19 13 15 9 328

Brucellosis of cattle, buffalo, sheep, goats and pigs 33 24 58 64 76 65 50 24 40 43 46 34 557

Caprine arthritis/encephalitis 1 2 1 3 1 1 2 4 15

Chlamydiosis 1 1

Clostridiosis (Enterotoxemia) 1 1

Contagious agalactia 6 5 7 10 2 4 4 1 1 1 41

Contagious bovine mastitis 1 1

Crayfish plague (Aphanomyces astaci) 1 1

Echinococcosis/Idatidosis 1 1 2

Equine infectious anaemia 2 1 2 1 3 3 3 1 3 2 21

Equine rhinopneumonitis 2 2

Erysipelas 1 1 3 1 1 7

European foulbrood of honey bees 1 3 4

Fowl pox 1 1

High patogenicity Avian influenza in poultry 1 1

Infectious hematopoietic necrosis 1 1 2

Koi herpesvirus disease 1 1 2

Leishmaniosis 1 1 2

Leptospirosis 3 5 2 5 4 3 2 3 3 2 1 33

Low patogenicity Avian influenza in poultry 1 1 1 1 1 5

Maedi-visna 1 1 2

Mixomatosis 1 1 1 1 1 3 1 9

Non-typhoidal avian salmonellosis 6 2 4 4 2 1 7 3 3 32

Paratuberculosis 1 4 2 2 1 1 1 12

Pasteurellosis of cattle, buffalo, sheep, goats and pigs 1 1 1 3

Q fever 1 2 1 4

Rabbit haemorrhagic disease 1 1 1 1 2 1 1 1 9

Salmonellae equine abortion 1 1

Salmonellosis (S. abortusovis) 5 10 4 1 2 1 5 2 2 32

Salmonellosis of animals 1 2 3 1 7

Scrapie 1 3 3 2 4 1 1 3 3 1 22

Swine vescicular disease 2 1 2 5

Trichinellosis 3 1 1 5

Viral haemorrhagic septicaemia (VHS) 1 1

West Nile Disease 1 20 48 30 8 1 108

Page 20: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

20 Hand on data

Number of outbreaks reported by Regions to SIMAN in 2014

Region DiseaseTrimester Total

outbreaksI II III IV

ABRUZZO

Avian typhosis 1 1

Bluetongue 130 14 144

Bovine leucosis 1 1 2

Bovine tuberculosis 1 1 2

Brucellosis of cattle, buffalo, sheep, goats and pigs 6 3 9

Equine infectious anaemia 1 1

Rabbit haemorrhagic disease 1 1

Salmonellae equine abortion 1 1

AOSTA VALLEY Anemia infettiva degli equini 2 2

BOLZANOMal rossino 2 2

Peste americana 1 2 2 5

APULIA

American foulbrood of honey bees 1 1

Bluetongue 8 65 73

Bovine leucosis 7 8 1 1 17

Bovine tuberculosis 7 3 3 13

Brucellosis of cattle, buffalo, sheep, goats and pigs 21 18 14 20 73

Echinococcosis/Idatidosis 1 1

Equine infectious anaemia 1 1 2

Fowl pox 1 1

Non-typhoidal avian salmonellosis 1 1

Rabbit haemorrhagic disease 1 1 2

West Nile Disease 2 2

BASILICATA

Antrax 1 1 2

Bluetongue 25 10 35

Bovine tuberculosis 1 1 1 3

Brucellosis of cattle, buffalo, sheep, goats and pigs 9 10 6 3 28

Equine infectious anaemia 2 2

Swine vescicular disease 2 2

BOLZANOAmerican foulbrood of honey bees 1 2 2 5

Erysipelas 2 2

CALABRIA

Aethina tumida 14 38 52

Bluetongue 5 4 288 27 324

Bovine tuberculosis 1 9 7 17

Brucellosis of cattle, buffalo, sheep, goats and pigs 24 49 18 27 118

Echinococcosis/Idatidosis 1 1

Salmonellosis of animals 1 1

Scrapie 1 1

Swine vescicular disease 2 2

CAMPANIA

Bluetongue 3 1 74 33 111

Bovine leucosis 1 2 3

Bovine tuberculosis 12 27 1 5 45

Brucellosis of cattle, buffalo, sheep, goats and pigs 19 55 22 20 116

Chlamydiosis 1 1

Equine infectious anaemia 2 1 3

Leptospirosis 1 1

Non-typhoidal avian salmonellosis 1 1 2

Rabbit haemorrhagic disease 1 1

Scrapie 2 2

Swine vescicular disease 1 1

Page 21: 19 benv january 2015 en

January 2015 Number 19

21 Hand on data

Region DiseaseTrimester Total

outbreaksI II III IV

EMILIA ROMAGNA

American foulbrood of honey bees 6 3 1 10

Bluetongue 19 19

Erysipelas 1 2 3

European foulbrood of honey bees 1 1

Leptospirosis 1 1

Low patogenicity Avian influenza in poultry 1 1 2

Mixomatosis 1 1 1 3

Non-typhoidal avian salmonellosis 2 1 3

Salmonellosis (S. abortusovis) 1 1

Scrapie 1 1

West Nile Disease 60 60

FRIULI VENEZIA GIULIA

Crayfish plague (Aphanomyces astaci) 1 1

Leishmaniosis 2 2

Leptospirosis 7 5 7 4 23

Low patogenicity Avian influenza in poultry 1 1

Mixomatosis 1 1

Non-typhoidal avian salmonellosis 1 1

West Nile Disease 3 1 4

LAZIO

American foulbrood of honey bees 1 1

Bluetongue 5 2 274 58 339

Bovine leucosis 3 1 1 5

Bovine tuberculosis 3 5 4 1 13

Brucellosis of cattle, buffalo, sheep, goats and pigs 4 1 5

Equine infectious anaemia 1 4 1 4 10

Equine rhinopneumonitis 2 2

Mixomatosis 1 1

Non-typhoidal avian salmonellosis 1 1 3 5

Q fever 1 1 2

Salmonellosis (S. abortusovis) 2 1 3

Scrapie 2 1 1 4

LIGURIA

Bovine tuberculosis 1 1

Leptospirosis 1 1 2

West Nile Disease 1 1

LOMBARDY

Bovine tuberculosis 1 1

Equine infectious anaemia 1 1

Low patogenicity Avian influenza in poultry 1 1

Non-typhoidal avian salmonellosis 3 1 1 3 8

West Nile Disease 22 1 23

MARCHE

Bluetongue 56 23 79

Clostridiosis (Enterotoxemia) 1 1

Mixomatosis 1 1

Non-typhoidal avian salmonellosis 2 2 4

Scrapie 2 1 1 4

MOLISE

Bluetongue 14 21 35

Bovine tuberculosis 2 2

Brucellosis of cattle, buffalo, sheep, goats and pigs 2 2 1 2 7

Equine infectious anaemia 1 1

Trichinellosis 2 2

Page 22: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

22 Hand on data

Region DiseaseTrimester Total

outbreaksI II III IV

PIEDMONT

Bovine tuberculosis 1 2 3

Infectious hematopoietic necrosis 1 1

Low patogenicity Avian influenza in poultry 1 1

Paratuberculosis 3 3

West Nile Disease 3 3

SARDINIA

African swine fever 32 30 5 30 97

Avian typhosis 1 1

Bluetongue 5 5 6 4 20

Bovine tuberculosis 1 1

Brucellosis of cattle, buffalo, sheep, goats and pigs 1 1 2

Caprine arthritis/encephalitis 3 1 5 6 15

Contagious agalactia 13 14 6 1 34

Contagious bovine mastitis 1 1

Erysipelas 1 1

Leptospirosis 2 2 1 5

Maedi-visna 1 1 2

Mixomatosis 1 1

Non-typhoidal avian salmonellosis 2 1 3

Paratuberculosis 2 2

Pasteurellosis of cattle, buffalo, sheep, goats and pigs 1 1 2

Q fever 2 2

Salmonellosis (S. abortusovis) 16 1 2 9 28

Salmonellosis of animals 2 2 4

Scrapie 3 2 2 1 8

Trichinellosis 2 1 3

West Nile Disease 1 1

SICILY

Aethina tumida 1 1

Antrax 1 1

Bluetongue 8 2 19 44 73

Bovine leucosis 2 1 3

Bovine tuberculosis 72 88 41 23 224

Brucellosis of cattle, buffalo, sheep, goats and pigs 39 61 49 50 199

Contagious agalactia 1 1

Leptospirosis 1 1

Non-typhoidal avian salmonellosis 1 1

Pasteurellosis of cattle, buffalo, sheep, goats and pigs 1 1

Rabbit haemorrhagic disease 1 1

Scrapie 1 1 2

West Nile Disease 1 2 2 5

TRENTO

American foulbrood of honey bees 1 19 32 5 57

Contagious agalactia 4 2 6

European foulbrood of honey bees 3 3

Paratuberculosis 5 1 6

TUSCANY

American foulbrood of honey bees 6 1 4 11

Bluetongue 83 83

Bovine tuberculosis 1 1

Koi herpesvirus disease 2 2

Mixomatosis 1 1

Non-typhoidal avian salmonellosis 1 1

Paratuberculosis 1 1

Salmonellosis of animals 1 1

Page 23: 19 benv january 2015 en

January 2015 Number 19

23 Hand on data

Region DiseaseTrimester Total

outbreaksI II III IV

UMBRIA

Bluetongue 96 31 127

Bovine tuberculosis 1 1

Equine infectious anaemia 1 1

Salmonellosis of animals 1 1

VENETO

American foulbrood of honey bees 2 2

Erysipelas 1 1

High patogenicity Avian influenza in poultry 1 1

Infectious hematopoietic necrosis 1 1

Mixomatosis 1 1

Non-typhoidal avian salmonellosis 1 1 1 3

Rabbit haemorrhagic disease 3 1 4

Viral haemorrhagic septicaemia (VHS) 1 1

West Nile Disease 7 2 9

Page 24: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

24 Hand on data

Animals involved in outbreaks reported to SIMAN in 2014

Disease name Animals involvedNo. Of

animal in the holding

No. Of diseased animals

No. Of died

animals

No. Of culled

animals

No. Of destroyed

animas

Aethina tumida Bees 440 423 84 421 421

African swine fever Suidae 1204 209 118 1078 711

American foulbrood of honey bees Bees 250929 250205 150006 250037 250202

Antrax Ruminants 11 2 2 0 2

Avian typhosisBirds 1 1 1 0 1

Poultry 25120 25120 12500 12620 25120

BluetongueRuminants 189171 17022 5866 4 4351

Wild animals 18 18 0 0 0

Bovine leucosis Ruminants 2716 140 1 91 1

Bovine tuberculosisRuminants 20580 2335 19 2463 20

Wild animals 1 1 1 0 1

Brucellosis of cattle, buffalo, sheep, goats and pigsRuminants 52870 8031 21 7587 12

Suidae 34 4 2 0 2

Caprine arthritis/encephalitis Ruminants 1049 382 1 0 1

Contagious agalactia Ruminants 12022 1757 7 6 7

Contagious bovine mastitis Ruminants 7 1 0 0 0

Crayfish plague (Aphanomyces astaci) Acquatic animals 1 1 0 0

Echinococcosis/Idatidosis Ruminants 61 4 0 0 0

Equine infectious anaemia Equidae 145 19 1 9 1

Equine rhinopneumonitis Equidae 204 4 0 0 0

Erysipelas Suidae 6374 44 28 35 0

European foulbrood of honey bees Bees 214 85 80 0 3

Fowl pox Poultry 10 10 10 0 0

Infectious hematopoietic necrosis Acquatic animals 43001 0 0 0

Koi herpesvirus disease Acquatic animals 2807 2807 0 2840

Leishmaniosis Domestic carnviores 9 5 1 0 0

Leptospirosis

Domestic carnviores 616 26 18 0 8

Equidae 1 1 0 0 0

Ruminants 151 6 1 0 0

Suidae 730 31 0 30 0

Low patogenicity Avian influenza in poultryBirds 7731 103 0 7085 7070

Poultry 2141 67 1 1765 1765

Maedi-visna Ruminants 129 15 0 0 0

Mixomatosis Lagomorphs 480 122 102 378 478

Non-typhoidal avian salmonellosisBirds 108814 96737 0 0 0

Poultry 1078245 777856 37 60344 51886

Paratuberculosis Ruminants 2860 59 3 10 0

Pasteurellosis of cattle, buffalo, sheep, goats and pigs Ruminants 310 16 11 0 11

Q fever Ruminants 548 1 1 0 0

Rabbit haemorrhagic disease Lagomorphs 52877 12136 11936 21836 24141

Salmonellae equine abortion Equidae 14 3 0 0 0

Salmonellosis (S. abortusovis) Ruminants 9600 541 14 0 12

Salmonellosis of animalsRuminants 1284 17 2 0 0

Suidae 16 1 16 16 1

Scrapie Ruminants 6599 275 14 427 55

Swine vescicular disease Suidae 690 687 0 678 678

TrichinellosisSuidae 9 2 0 0 1

Wild animals 2 2 2 0 2

Viral haemorrhagic septicaemia (VHS) Acquatic animals 200 200 200 200

West Nile Disease

Birds 56 44 11 2 1

Equidae 187 19 0 0 0

Insects 395 121 4 0 0

Poultry 32 4 0 4 1

Page 25: 19 benv january 2015 en

January 2015 Number 19

25 A look at the maps

A LOOK AT THE MAPSThe geographical distribution of the main animal diseasesreported to SIMAN in 2014

Equine Infectious Anaemia

Bluetongue

--Geographical distribution of the outbreaks

--Geographical distribution of the outbreaks

Page 26: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

26 A look at the maps

Avian Influenza, low patogenicity

--Geographical distribution of the outbreaks

Avian Influenza, high patogenicity

--Geographical distribution of the outbreaks

Page 27: 19 benv january 2015 en

January 2015 Number 19

African Swine Fever

--Geographical distribution of the outbreaks

27 A look at the maps

Swine vesicular disease

--Geographical distribution of the outbreaks

Page 28: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

28 A look at the maps

Aethina tumida

--Geographical distribution of the outbreaks

West Nile Disease

--Geographical distribution of the outbreaks

Page 29: 19 benv january 2015 en

January 2015 Number 19

29 Around us

AROUND USThe main events of epidemiological interest in the last months in the European Union and in the neighbour countries

Highly Pathogenic Avian Influenza H5N8 – Epidemiological situation in Europe

Background

In late 2014 several outbreaks of Highly Pathogenic Avian Influenza (HPAI) caused by a H5N8 virus strain have been identified in poultry farms located in an European area encompassing United Kingdom, The Netherlands and Germany. In December 2014 a H5N8 HPAI virus was also identified in a fattening turkeys farm in north-eastern Italy, located in a zone highly frequented by wild waterfowl and on the flyway of many migrant bird species (Bodini, 2000). As of 15 January 2015, HPAIV H5N8 has been reported in nine domestic poultry holdings and in a zoo in Europe (Figure 1, Table 1)

Figure 1.Location of H5N8 HPAI outbreaks in

poultry farms in Europe

Page 30: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

30 Around us

H5N8 HPAI Situation in Europe

GermanyA total of three H5N8 HPAI outbreaks in poultry farms had been detected in Germany between early November and late December; positivity to H5N8 virus has been identified in white storks kept in zoo in January 2015, and in wild waterfowl.

The first case was confirmed by the National Reference Laboratory (NRL) at Friedrich Löffler Institut on 6 November 2014, in a fattening turkeys farm located in the state of Mecklenburg- Vorpommern. Depopulation was promptly enforced, and control and restriction measures provided in the Council Directive 2005/94/EC were put into force (European Union, 2005). Epidemiological investigations showed no evidence further virus spread from the outbreak to contact farms. Additionally, an active surveillance plan was put into force on wild birds, including sampling of droppings from the environment in the protection zone (ECDC, 2014). The H5N8 virus was identified in a clinically healthy wild duck, shot on 17 November 2014 in the Mecklenburg- Vorpommern state, approximately 100 km from the infected farm.

One month later, on 17 December 2014, H5N8 HPAI virus was detected in another fattening turkeys farm, in Lower Saxony. Local authorities promptly applied containment measures to avoid the spread of the disease to neighboring areas, including depopulation of three rural laying hen farms within 1 km. Nevertheless, on 20 December, another H5N8 HPAI outbreak was notified in Lower Saxony, involving a fattening duck farm; on the same date the presence of H5N8 was also confirmed in a mallard duck (Anas platyrhynchos) collected in Saxony-Anhalt.

All of the clinical and laboratory tests performed on poultry farms within the restriction and surveillance zones, and no poultry products had been exported toward other European and Third Countries.

Table 1. Chronological list of H5N8 HPAI outbreaks in Europe)

Notification Date Country Province/Region/County Farm Type

6 Nov 2014 Germany Mecklenburg- Vorpommern Fattening turkeys

15 Nov 2014 Netherlands Utrecht Laying hens

16 Nov 2014 United Kindom East Yorkshire Fattening ducks

17 Nov 2014 Germany Mecklenburg- Vorpommern Anas spp.

20 Nov 2014 Netherlands Zuid-Holland Laying hens

21 Nov 2014 Netherlands Overijssel Breeders

21 Nov 2014 Netherlands Overijssel (1)

30 Nov 2014 Netherlands Zuid-Holland Laying hens

16 Dec 2014 Italy Rovigo (Veneto) Fattening turkeys

17 Dec 2014 Germany Lower Saxony Fattening turkeys

20 Dec 2014 Germany Lower Saxony Fattening ducks

20 Dec 2014 Germany Saxony-Anhalt Mallard

7 Jan 2015 Germany Mecklenburg- Vorpommern White Storks(2)

(1) Farm located within 1 km from the outrbreak in the poultry breeder farm (2) The positive birds were kept in a zoo

Page 31: 19 benv january 2015 en

January 2015 Number 19

31 Around us

On 7 January 2015, the H5N8 was confirmed in three white storks (Ciconia ciconia) kept in a zoo in Rostock (Mecklenburg- Vorpommern). A total of 496 birds belonging to 85 different species were present in the zoo. Thirty-nine birds were culled, including the three positive storks and the birds exposed to a higher risk of having had contact with them.

The NetherlandsThe first outbreak in the Netherlands was confirmed on 15 November 2014, when the Health Authorities declared the presence of a H5N8 virus in a laying hens farm in Utrecht province. Control measures were promptly enforced, including depopulation of the farm and restriction and control zones were defined. Laboratory tests indicated high similarity with the virus detected in Germany on 6 November. A second laying hens farm was confirmed positive for HPAI H5N8 virus on 20 November; the farm was located in the Zuid-Holland province, at about 24 km south of the first outbreak. Following this occurrence, the Dutch Government proclaimed a ban on poultry and poultry products movement on the entire National territory. On 21 November another poultry farm with 10,000 birds tested positive for HPAIV H5N8. The farm was located in Kamperveen village (Overijssel province), at about 80 km from the previous two outbreaks. Pre-emptive slaughtering was applied in two farms within 1 km, one of which showing symptoms referable to avian influenza.

To contain the HPAI spreading and quickly eradicate the disease, the Dutch Government introduced a new series of control measures. The Netherlands were divided into 4 areas and poultry and poultry products movements were possible only within the same area, to reduce the possible contact between farms. A general ban on poultry restocking was also enforced in the whole National territory.

On 30 November, a H5 virus was detected in a laying hens located in the Zoeterwoude municipality (Zuid-Holland province); on 2 December the virus was confirmed belonging to the H5N8 strain.

Following improved surveillance measures to test the actual role of wild waterfowl in the introduction and spread of the H5N8 HPAI in the Netherlands, on 3 December the Agriculture Ministry proclaimed that some of the samples collected from Eurasian widgeons (Anas penelope) in the Utrecht province, tested positive for H5N8. Moreover, epidemiological investigations and phylogenetic analyses indicated that 4 out of the 5 outbreaks identified resulted being related to different H5N8 virus and only the two in Kamperveen were linked, strongly suggesting a key role of wild birds in the epidemic (European Food Safety Authority, 2014).

United KingdomUnited Kingdom experienced only one H5N8 HPAI outbreak, confirmed on 16 November 2014. The outbreak involved a fattening ducks farm with 6,000 birds, located in East Yorkshire. The farm was depopulated and restriction and surveillance zones were defined. On 21 November 2014, European Laboratory of Reference at the Animal Plant Health Agency (APHA) confirmed that the isolated H5N8 virus showed high similarity with those detected in the fattening turkey farm in Germany and in laying hens farm in the Netherlands, corroborating the hypothesis of the involvement of wild migratory waterfowl in the introduction and spread of the disease in Europe.

ItalyOn 10 December 2014 an unspecific gastro-enteric syndrome was observed in a fattening turkeys farm in Rovigo province (Veneto Region, north-eastern Italy); on 12 December an increase in the mortality rates was also detected in the farm. On 15 December 2014, the National Reference Laboratory (NRL) for Avian Influenza at the Istituto Zooprofilattico Sperimentale delle Venezie confirmed the presence of a H5N8 HP virus in the premise. The infected farm was located in a relatively low density poultry populated area, in close proximity to large wetlands with highly populated resting sites for migratory birds and wild waterfowls.

Phylogenetic analysis showed a high level clustering of the Italian virus with the H5N8 viruses collected in The Netherlands, Germany and UK in November 2014. Moreover a high nucleotide similarity (99.8%) with the strain isolated in UK and in Eurasian widgeons in the Netherlands was also observed, supporting again the hypothesis of

Page 32: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

32 Around us

the involvement of wild waterfowl in the disease spreading. No additional glycosilation sites or molecular markers were identified, indicating no adaptation to mammalian hosts.

Following the confirmation of the H5N8 HPAI outbreak in Italy, on 16 December 2014, a series of control measures were implemented on the territory of the Veneto Region, as provided for in Council Directive 2005/94/EC (European Union, 2005). Starting on 22 December 2014, further measures were adopted on a National scale. All of the fattening turkeys farms belonging to the poultry company involved in the outbreak were subjected to weekly mandatory submission of virological samples; virological controls were also enforced in contact farms together with enhancement in biosecurity measures to avoid contacts with wild birds, and on restriction of movements of birds and eggs. Turkeys farms located in more densely poultry populated Italian regions had to be virologically tested every 10 days. At a National level, mandatory virological tests have to be performed on pullets and laying hens before loading for transportation. Moreover, any bird fair, bird exhibition, and live bird market has been forbidden up to a date to be defined and no live decoy bird can be used for hunting.

Final remarks

The actual route of introduction of the H5N8 HPAI virus into Europe is still not completely ascertained. The virus was already detected in East Asia earlier in 2014, being isolated from wild waterfowl and causing outbreaks in domestic poultry in Korea, China, and Japan. Contacts with migratory birds represent the most likely route of introduction of the virus into Europe; the hypothesis is strongly supported by the results of phylogenetic analyses which show a high similarity (99.5%) between the European viruses and the Korean H5N8 isolates (European Food Safety Authority, 2014). Nevertheless the direct introduction from south-eastern Asia into Europe is quite unlikely, and led to the hypothesis that migratory birds from the East Atlantic Flyway and East Asian-Australasia Flyways had mingled in common breeding grounds such as Siberia (European Food Safety Authority, 2014).

The multiple introduction of H5N8 viruses in Europe, and their similarity with those circulating earlier in Asia, indicate the possibility that the disease is spread at very large distances within a limited timeframe. This indicates the threat represented by avian influenza on a global scale, both for the animal health and the poultry industry with potentially huge economical effects.

The infection with the virus has been associated with mortality in several wild bird species; nevertheless the H5N8 virus was also detected in wild waterfowl with mild or no symptoms, as in the case of the mallard in Germany and widgeon in the Netherlands (Jeong et al., 2014). Also anti-H5 antibodies were detected in various wild species in south Korea, indicating recovery from the disease (Jeong et al., 2014). This may impair surveillance activities in the wild host, as it is usually related to passive measures. The recent outbreaks of H5N8 HPAI indicate the need for preparedness. Measures, including strict control activities, well defined biosecurity procedures and improved surveillance, represent the only way to early detect any new introduction and possible spill-over of the disease to domestic poultry sector.

References

1. Bodini, A., 2000. A Multimethodological Approach for the Sustainable Management of Perifluvial Wetlands of the Po River (Italy). Environ. Manage. 26, 59–72. doi:10.1007/s002670010071

2. ECDC, 2014. Outbreaks of highly pathogenic avian influenza A ( H5N8 ) in Europe - 20 November 2014.

3. European Food Safety Authority, 2014. Highly pathogenic avian influenza A subtype H5N8. EFSA J. 12, 3941. doi:10.2903/j.efsa.2014.3941

4. European Union, 2005. Council Directive 2005/94/EC on Community measures for the control of avian influenza and repealing Directive 92/40/EEC.

5. Jeong J, et al., 2014. Highly pathogenic avian influenza virus (H5N8) in domestic poultry and its relationship with migratory birds in South Korea during 2014. Vet. Microbiol. 173, 249–57. doi:10.1016/j.vetmic.2014.08.002

Page 33: 19 benv january 2015 en

January 2015 Number 19

33 Around us

6. Lee YJ, 2014. Novel reassortant influenza A(H5N8) viruses, South Korea, 2014. Emerg. Infect. Dis. 20, 1087–9. doi:10.3201/eid2006.140233

7. ProMed, 2014a. Archive Number: 20141222.3049031 (accessed 1.14.15).8. ProMed, 2014b. Archive Number: 20141218.3040607 (accessed 1.14.15).9. Wu H, 2014. Novel reassortant influenza A(H5N8) viruses in domestic ducks,

eastern China. Emerg. Infect. Dis. 20, 1315–8. doi:10.3201/eid2008.140339

--Edited by:Centro di Referenza Nazionale per Influenza Aviaria e Malattia di New Castle,Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro (Padova) - Italy

Page 34: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

34 Around us

Lumpy skin disease: still a neglected disease?

For many years Lumpy skin disease (LSD) has been considered a “neglected disease” because present in developing countries alone and not of major interest for the international trade. Historically, in fact, LSD of cattle has been confined to the African continent, with sporadic outbreaks occurring in the Middle East. More recently, LSD has attracted the interest of the European Union and Mediterranean countries after its spread throughout the Near East, Turkey and Cyprus at a scale that has never been seen before (Tuppurainen E. et al., 2014). It is important, therefore, to review and make the point about the current knowledge on LSD. This short review mainly results from a very recent 2015 EFSA report (EFSA, 2015).

What is LSD ?

LSD is a viral disease of cattle characterised by severe losses, especially in naive animals. LSD virus (LSDV) belongs to the family Poxviridae which is divided into two subfamilies—poxviruses affecting insects (Entomopoxvirinae) and vertebrates (Chordopoxvirinae)—and several genera. The genus Capripoxvirus (CaPV) comprises LSDV, sheep pox virus (SPPV) and goat pox virus (GTPV). The prototype of LSDV, Neethling strain, was isolated in South Africa (Alexander et al., 1957). The replication of LSDV occurs in the cytoplasm of the host cell, in intracytoplasmic eosinophilic inclusion bodies (Weiss, 1968; Prozesky and Barnard, 1982).

Which is the host range ?

In addition to cattle, natural infections have been reported in Asian water buffalo (Bubalus bubalis) in Egypt, but with significantly lower prevalence rate (1.6 %) than in cattle (30.8 %) (Ali et al., 1990; El-Nahas et al., 2011). LSDV replicates in cell cultures of sheep and goat origin and experimentally infected sheep and goats show a local reaction at the inoculation site but no reports exist on clinical disease in small ruminants caused by LSDV. Thin-skinned, high-producing dairy Bos taurus breeds are highly susceptible against LSDV, whereas indigenous (Bos indicus) breeds such as zebu and zebu hybrids are likely to have some natural resistance against the virus (Davies, 1991; Gari et al., 2011). It is not known which genetic factors influence the disease severity (Babiuk et al. 2008). High air temperatures, coupled with farming practices to produce high milk yields, could be deemed to stress the animals and contribute to the severity of the disease in Holstein–Friesian cattle (Tageldin et al., 2014).

WildlifeClinical signs of LSD have been observed in impala (Aepyceros melampus) and giraffe (Giraffa camelopardalis) after experimental inoculation with LSDV (Young et al., 1970). LSD was reported in an Arabian oryx (Oryx leucoryx) in Saudi Arabia (Greth et al., 1992). Antibodies against SPPV and GTPV and LSDV cannot be differentiated from each other by serological tests, and this is an important limitation. The presence of antibodies in an animal species indicates its susceptibility to the virus and its potential involvement in the epidemiology of the disease (Barnard, 1997). There is evidence that African wild ruminant species may have a role in the epidemiology of LSD. Although certain wildlife species can be experimentally infected by LSDV, information on the susceptibility of wildlife to LSD is scarce and further limited by the inability to distinguish antibodies evoked by LSDV from those evoked by sheep and goat pox.

Page 35: 19 benv january 2015 en

January 2015 Number 19

35 Around us

Which clinical signs ?

The course of the disease may be acute, subacute or chronic. Only 40 to 50 % of experimentally infected animals develops generalised skin lesions; many cases are subclinical but can be viraemic and can transmit the virus (Weiss, 1968). The incubation period of LSD is two to four weeks in field conditions (Haig, 1957). In animals that develop clinical disease, there is a biphasic febrile reaction that may exceed 41°C. They remain febrile for 4 up to 14 days. This is accompanied by depression, disinclination to move, inappetence, salivation, lachrymation and a nasal discharge, which may be mucoid or mucopurulent. Lachrymation may be followed by conjunctivitis and, in some cases, by corneal opacity and blindness. The superficial lymph nodes, especially prescapular, precrural and subparotid, are usually markedly enlarged (Thomas and Marè, 1945; Haig, 1957; Weiss, 1968; Prozesky and Barnard, 1982; Barnard et al., 1994; Carn and Kitching, 1995a). The eruption of nodular skin lesions usually occurs within 48 hours of onset of the febrile reaction.

Where is LSD present ?

LSD is endemic in most African countries, and outbreaks outside the African continent occurred in the Middle East in 2006 and 2007 and in Mauritius in 2008 (OIE, 2014). The American continent and Australia are free of CaPV infections. LSD was first reported in Zambia in 1929, then 15 years later was observed in Botswana and in South Africa, where eight million cattle were affected. Until the 1970s, LSD had spread northwards to Kenya and Sudan, westwards to Nigeria and then reported in Mauritania, Mali, Ghana and Liberia (OIE, 2014). The global occurrence of LSD from 2005 to 2013 is reported in figure 1.

Page 36: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

36 Around us

Figure 1.Global occurrence of LSD from 2005 to 2013 (Source: EFSA AHAW Panel, 2015. Scientific Opinion on lumpy skin disease. EFSA Journal 2015;13(1):3986, 73 pp. doi:10.2903/j.efsa.2015.3986)

 

Until 1989, LSD occurrence was restricted to sub-Saharan Africa, but Egypt reported its first LSD outbreak in 1988 (House et al., 1990) and Israel in 1989 (Yeruham et al., 1995).

In subsequent years, Bahrain, Kuwait, Oman, Yemen, Israel and the West Bank also reported LSD incursions. From July 2012 to August 2013, 293 outbreaks were reported in Israel; in 2012, 34 outbreaks were reported to OIE in Lebanon; in May 2013, two outbreaks were reported in Jordan; in September 2013, 28 outbreaks were reported in Iraq (still on-going); since August 2013, 236 outbreaks have been reported in Turkey; in May 2014, four outbreaks were reported in Iran; and, in July 2014, two outbreaks were reported in Azerbaijan. LSD may have now become endemic in Turkey. Although LSD has not been reported in the Syrian Arab Republic—most likely owing to the current armed conflict—the disease is present (FAO, 2013b). LSD probably spread from Syria and Iraq to Turkey, as most outbreaks in Turkey occurred along the eastern part of the southern border with Syria and Iraq.

Page 37: 19 benv january 2015 en

January 2015 Number 19

37 Around us

How can be diagnosed ?

Clinical diagnosisA presumptive diagnosis of the disease can be made based on highly characteristic clinical signs of LSD. However, mild and asymptomatic disease may be difficult to diagnose and rapid laboratory methods are needed to confirm the diagnosis. The skin lesions of pseudo LSD (bovine herpesvirus-2, BHV-2), insect bites, Demodex infection, onchocerciasis, besnoitiosis and dermatophilosis can be confused with LSD (Barnard et al., 1994). Generally, BHV-2 infection causes more superficial skin lesions, has a shorter course and is a milder disease than LSD. In addition, the presence of histopathologically demonstrable intranuclear inclusion bodies in BHV-2 infection, as opposed to intracytoplasmic inclusions in LSD, is characteristic. In contrast to the rapid molecular diagnostic tools available for LSD, the detection of BHV-2 in negatively stained biopsy specimens by electron microscopy or the isolation of the virus is only possible approximately one week after the development of skin lesions. In some cases, diseases causing mucosal lesions, can be confused with LSD, such as bovine viral diarrhoea/mucosal disease and bovine malignant catarrhal fever (Barnard et al., 1994).

Laboratory techniquesThe antigen detection through a general CaPV real-time PCR method is more sensitive than other diagnostic assays for the detection of the virus; the test detects CaPV viral DNA, but does not differentiate between the different members of the genus, which may be required if characteristic clinical signs for LSD are not clear. Species-specific real-time PCR methods for differentiation between SPPV, GTPV and LSDV have been published. The general CaPV gel-based PCR method has been validated and published in the OIE Manual of Diagnostic Tests and Vaccines for Terrestrial Animals for LSD but this method is not as fast or as sensitive as real-time PCR. Portable pen-side PCR was recently developed, and it has promising preliminary results: it is easy to use in field conditions and results are obtained within one hour (EFSA, 2015). Electron microscopy: CaPV is morphologically indistinguishable from Orthopoxvirus, but, apart from vaccinia virus, no Orthopoxvirus causes lesions in sheep and goats. Virus isolation is not suitable for primary diagnostics but is needed to confirm the infectivity of the virus. If tissue culture systems are available, then the virus can be isolated on a variety of cells, such as primary lamb kidney or testis cell cultures.

The detection of antibodies against LSDV through Serum/virus neutralisation tests are gold standard tests for serology. They are very specific but not sufficiently sensitive for the detection of low antibody titres in animals with mild clinical disease and vaccinated animals. The capacity of Indirect fluorescent antibody test (IFAT) allows testing of larger numbers of samples than with the neutralisation test. Tests may cross-react with other poxviruses. The agar gel immune diffusion test is a very simple test requiring the bare minimum laboratory facilities. However, it lacks sensitivity (EFSA, 2015) and can cross-react with Parapoxvirus (Mangana-Vougiouka et al., 2000), which is one of the differential diagnoses. Positive test results must be confirmed with another test. The performance of enzyme-linked immunosorbent assays (ELISA) for sheep pox/goat pox screened from the literature ranges from 70 to 100 % for sensitivity and from 84 to 100 % for specificity. Western blotting is difficult to perform and requires purified antigens, and it cannot be used as a primary assay but can be used if inconclusive or positive SNT/ELISA results need to be confirmed.

Which is/are the route/s of transmission?

Transmission of LSDV is mainly through blood-feeding insects and ticks although indirect or direct contact and seminal transmission of the virus can also occur. Transmission of LSDV by Aedes aegypti mosquitos (Chihota and others 2001) and transmission of the closely related sheep pox (SPP) virus by stable flies (Stomoxys calcitrans) (Kitching and Mellor 1986) have been demonstrated, although it is not known if insects can be also biological vectors for the virus. In recent years, the ability of ixodid ticks to transmit LSDV has been investigated in some detail. It has been shown that LSDV can be transmitted by ticks mechanically (Tuppurainen and others 2013a) and the virus can survive from one tick life cycle stage to the next.

Page 38: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

38 Around us

The virus has also been detected in tick eggs, indicating transovarial transmission (Tuppurainen and others 2013b), and in different tick life cycle stages (Lubinga and others 2013, 2014) that are developing off the host in soil or vegetation. This results in the environment being contaminated with LSDV-infected ticks, thus hampering the eradication of the virus without the use of vaccination.

Are vaccines available ?

Only live attenuated vaccines against LSD are currently commercially available. The attenuated Neethling strain (LSDV) vaccine is used to vaccinate cattle in Africa. It is possible to use the sheep pox/goat pox vaccine for cattle (Capstick and Coackley, 1961) but the cross-protection is not satisfactory and the use of this vaccine has been restricted to those countries where sheep and goat pox are endemic. Neutralising antibodies to LSDV persist for at least two to three years after vaccination. In some animals, the antibody levels are too low to demonstrate, but they are, nevertheless, still resistant to challenge (Weiss, 1968). Antibodies appear 10 days after vaccination and reach the highest level 30 days post inoculation. Calves born to immunised cows will have passive immunity that persists for about six months (Weiss, 1968).

The following excerpts may be of interest to readers :

1. The eradication of LSDV is notoriously difficult, if not impossible, without the use of vaccination. Early detection of the index cases is required in order to stand any chance of controlling the disease through the stamping out of infected and in-contact animals.

2. The field diagnosis of LSD, particularly in the early stages, is not easy. The disease can be confused with many other diseases and conditions, including pseudo-LSD and insect bites. In addition, beef cattle may not be monitored daily, and mild cases of LSD are easily missed. The situation is further complicated by the fact that only about half of infected viraemic animals show clinical signs of the disease, and those apparently healthy animals are able to transmit the virus mechanically to blood-feeding vectors. The likely delay in diagnosing the disease in the field and in the lab may allow sufficient time to pass for the virus to contaminate vector populations, making the control of the disease through the culling of infected and in-contact animals ineffective.

3. According to the current OIE Terrestrial Code (OIE 2014), 3 disease-free years are required for a country to regain the official disease-free status following an LSD outbreak. If vaccination of cattle against LSD is implemented, international trade restrictions or a total ban of live cattle and their products will automatically follow.

4. Currently, only attenuated live vaccines are commercially available for use against LSD. Live vaccines are thought to provide good protection; however, the use of live vaccines in countries that are free of disease is considered risky because of potential safety issues, and also costly because of the associated trade restrictions that would ensue. Recently, novel inactivated LSDV and SPP virus vaccines have been developed. A safety and efficacy study in which sheep and goats were vaccinated with an inactivated SPP vaccine and then challenged with a virulent field strain has been conducted with promising results (EFSA, 2015). Similar experiments are currently in progress in which cattle are being vaccinated with an inactivated LSDV vaccine and then challenged with virulent (Tuppurainen E. and Oura C., 2014).

5. The spread of LSD is likely to have been a result of multiple factors; however, between 2012 and 2014, LSDV outbreaks are likely to have been associated with the severe political unrest and conflicts in Syria and Iraq, which have resulted in the collapse of veterinary services, the associated lack of effective vaccinaton actons in these countries, and the movement of hundreds of thousands of refugees and unvaccinated domestic ruminants from infected areas to neighbouring territories.

6. Disease outbreaks have recently been reported in countries neighbouring Syria including Israel, Lebanon, Jordan, Turkey and Iraq, and for the first time in Iran in July 2014. In addition, LSD is capable of spreading over long distances, as demonstrated by the latest LSD outbreak in Azerbaijan in July 2014. The long-distance spread of the disease from Iraq, Iran and/or Turkey to Azerbaijan is more likely to have occurred via legal or illegal animal movements rather than through arthropod vectors (Tuppurainen E. and Oura C., 2014).

Page 39: 19 benv january 2015 en

January 2015 Number 19

39 Around us

7. As stamping out of infected and in-contact animals is not feasible in developing countries and the implementation of total restrictions of cattle movements is next to impossible because of transhumance and nomadic cattle farming practices, the control of the disease relies heavily on the immunisation of sufficient numbers of cattle. By using enforced compulsory vaccination campaigns, the spread of the disease has so far been curbed in Israel, Lebanon, and partially in Jordan.

8. LSD is a high impact but largely neglected cattle disease that is on the move and is posing a real and present threat to the cattle populations in European countries.

Bibliografia

1. Ali AA, Esmat M, Attia H, Selim A and Abdel-Hamid YM, 1990. Clinical and pathological studies on lumpy skin disease in Egypt. Veterinary Record, 127, 549-550.

2. Alexander RA, Plowright W and Haig DA, 1957. Cytopathic agents associated with LSD of cattle. Bulletin of epizootic diseases of Africa, 5, 489–492

3. Barnard B, Munz E, Dumbell K and Prozesky L, 1994. Lumpy skin disease. Infectious diseases of livestock with special reference to Southern Africa, 1, 604-612

4. Barnard BJ, 1997. Antibodies against some viruses of domestic animals in southern African wild animals. Onderstepoort Journal of Veterinary Research, 64, 95-110.

5. Capstick P and Coackley W, 1961. Protection of cattle against lumpy skin disease. Research in Veterinary Science, 2, 362-375

6. Carn VM and Kitching RP, 1995. The clinical response of cattle experimentally infected with lumpy skin disease (Neethling) virus. Archives of virology, 140, 503-513 OIE (Office International des Epizooties), 2014. Lumpy skin disease - Technical disease card. Available online

7. Davies FG, 1991. Lumpy skin disease, an African capripox virus disease of cattle. The British veterinary journal, 147, 489-503.

8. El-Nahas E, El-Habbaa A, El-bagoury G and Radwan ME, 2011. Isolation and Identification of Lumpy Skin Disease Virus from Naturally Infected Buffaloes at Kaluobia, Egypt. Global Veterinaria 7, 234-237.

9. European Food Safety Authority (EFSA). 2015. Scientific Opinion on lumpy skin disease. EFSA Journal 2015;13(1):3986

10. Gari G, Bonnet P, Roger F and Waret-Szkuta A, 2011. Epidemiological aspects and financial impact of lumpy skin disease in Ethiopia. Preventive Veterinary Medicine, 102, 274-283.

11. Greth A, Gourreau JM, Vassart M, Nguyen Ba V, Wyers M and Lefevre PC, 1992. Capripoxvirus disease in an Arabian oryx (Oryx leucoryx) from Saudi Arabia. Journal of Wildlife Diseases, 28, 295-300.

12. Haig DA, 1957. Lumpy skin disease. Bulletin of epizootic diseases of Africa, 5, 421-430

13. House JA, Wilson TM, el Nakashly S, Karim IA, Ismail I, el Danaf N, Moussa AM and Ayoub NN, 1990. The isolation of lumpy skin disease virus and bovine herpesvirus-4 from cattle in Egypt. Journal of Veterinary Diagnostic Investigation, 2, 111-115.

14. Lubinga JC, Clift SJ, Tuppurainen ES, Stoltsz WH, Babiuk S, Coetzer JA and Venter EH, 2014a. Demonstration of lumpy skin disease virus infection in Amblyomma hebraeum and Rhipicephalus appendiculatus ticks using immunohistochemistry. Ticks and tick borne diseases, 5, 113-120.

15. Lubinga JC, Tuppurainen ES, Coetzer JA, Stoltsz WH and Venter EH, 2014b. Transovarial passage and transmission of LSDV by Amblyomma hebraeum, Rhipicephalus appendiculatus and Rhipicephalus decoloratus. Experimental & applied acarology, 62, 67-75.

16. Lubinga JC, Tuppurainen ES, Mahlare R, Coetzer JA, Stoltsz WH and Venter EH, 2013a. Evidence of transstadial and mechanical transmission of Lumpy Skin Disease Virus by Amblyomma hebraeum ticks. Transboundary and Emerging Diseases.

17. Lubinga JC, Tuppurainen ES, Stoltsz WH, Ebersohn K, Coetzer JA and Venter EH, 2013b. Detection of lumpy skin disease virus in saliva of ticks fed on lumpy skin disease virus-infected cattle. Experimental & applied acarology, 61, 129-138.

18. Lubinga JC, Tuppurainen ESM, Coetzer JAW, Stoltsz WH and Venter EH, 2014c. Transovarial passage and transmission of LSDV by Amblyomma hebraeum, Rhipicephalus appendiculatus Tuppurainen ESM, Lubinga JC, Stoltsz WH, Troskie

Page 40: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

40 Around us

M, Carpenter ST, Coetzer JAW, Venter EH and Oura CAL, 2013b. Evidence of vertical transmission of lumpy skin disease virus in Rhipicephalus decoloratus ticks. Ticks and tick borne diseases, 4, 329-333.

19. Prozesky L and Barnard BJ, 1982. A study of the pathology of lumpy skin disease in cattle. Onderstepoort Journal of Veterinary Research, 49, 167-175.

20. Tageldin MH, Wallace DB, Gerdes GH, Putterill JF, Greyling RR, Phosiwa MN, Al Busaidy RM and Al Ismaaily SI, 2014. Lumpy skin disease of cattle: an emerging problem in the Sultanate of Oman. Tropical Animal Health and Production, 46, 241-246.

21. Thomas A and Marè C, 1945. Knopvelsiekte. Journal of the South African Veterinary Association, 16, 36–43.

22. Tuppurainen ES, Lubinga JC, Stoltsz WH, Troskie M, Carpenter ST, Coetzer JA, Venter EH and Oura CA, 2013a. Mechanical transmission of lumpy skin disease virus by Rhipicephalus appendiculatus male ticks. Epidemiology and infection, 141, 425-430

23. Tuppurainen E., Oura C., 2014 Lumpy skin disease: an African cattle disease getting closer to the EU. Veterinary Record. September 27, 2014 pp. 300-301

24. Weiss KE, 1968. Lumpy skin disease. In: Virology Monographs. Vienna-New York, Springer Verlag, 111-131.

25. Yeruham I, Nir O, Braverman Y, Davidson M, Grinstein H, Haymovitch M and Zamir O, 1995. Spread of lumpy skin disease in Israeli dairy herds. Veterinary Record, 137, 91-93.

26. Young E, Basson PA and Weiss KE, 1970. Experimental infection of game animals with lumpy skin disease virus (prototype strain Neethling). Onderstepoort Journal of Veterinary Research, 37, 79-87.

--Edited by:Maria Luisa DanzettaCOVEPIIstituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale”

Page 41: 19 benv january 2015 en

January 2015 Number 19

41 Around us

Mathematical models for the study of microbial growth, survival and inactivation: predictive microbiology between past and future

Ensuring healthy foods is a priority for food industries and competent authorities in charge of control activities. In a world experiencing an unprecedented rate of change and globalisation, it is often necessary to apply proactive strategies in order to rapidly react to the threats against consumer’s safety. Preventing instead of correcting is the preferable approach. Nowadays food microbiology needs new tools able to address critical knowledge gaps and trigger quick responses for food risk management, also in the absence of comprehensive laboratory data, in the past considered crucial to apply efficacy control measures. Moreover, laboratory activities can be expensive, and costs cannot always be covered by food industries. Important elements of a proactive approach are the accumulation of quantitative information on microbial behaviour in foods and an increased understanding of microbial physiology (McMeekin et al. 2002). In fact, these are the elements on which predictive microbiology is based. It is an interdisciplinary research area that has become more and more used to evaluate, through suitable mathematical models, the responses of pathogenic or spoilage microorganisms to different environmental conditions that could be find in foods during processing and storing.

A great boost to predictive microbiology has been given by EC regulation 2073/2005 on microbiological criteria, currently in force, that has clearly given to food business operators the possibility of using “predictive mathematical modelling established for the food in question, using critical growth or survival factors for the micro-organisms of concern in the product”. Moreover, Codex Alimentarius recommended to move food control activities from a hazard- based and final testing approach to a more risk-based management approach (Codex Alimentarius 1999, Tenenhaus-Aziza & Ellouze 2015). This has made more and more necessary statistic and mathematical tools specifically designed to predict microbial behaviour in food processing. Predictive models cannot completely replace lab testing nor the judgement of an expert food microbiologist. However, they can yield information very useful to take food safety decisions (Whiting & Buchanan, 2002).

What is the exact definition of “predictive microbiology”? McMeekin et al. (1993) described it as a quantitative science that enables users to evaluate objectively the effect of processing, distribution and storage operations on the microbiological safety and quality of foods. The same authors also used the expression “Quantitative Microbial Ecology of Food”, while more recently the need to describe the microbial responses to the food environments by mathematical models was also highlighted (McKellar & Xu, 2003, Ceron-Carrillo et al. 2014). Baranyi and Roberts, who designed the most popular primary predictive model, see in this progression a sign of the evolution of predictive microbiology into a more and more exact science (Baranyi & Roberts 2004).

Predictive microbiology history begin in 1922 thanks to Esty and Meyer, who described how the thermal inactivation of Clostridium botulinum spores happens according to a log-linear model, still used to estimate heat processing of canned foods: at a given temperature, the specific death rate of a microorganism is constant with time. Another step was made in 1936 by Scott, who studied the direct relation between bacterial growth and available water, that today is called water-activity (aw ) and measured from 0 (no water) to 1 (water). Moreover, Scott established that the specific growth rate of microorganisms in fresh meat was influenced by temperature and that the knowledge of growth kinetic of spoilage bacteria could be useful to define shelf-life of foodstuffs at different storing conditions. The basis of mathematical predictive models had been laid down, in particular for primary models (describe changes in bacterial concentrations in foods in function of time – figure 1) and secondary models (describe parameters of the primary models as a function of environmental conditions as temperature, pH and aw – figure 2).

Page 42: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

42 Around us

Figure 1.Primary predictive model. After the lag phase, the logarithm of the population size increases linearly with time (exponential phase), until the stationary phase

Figure 2.Secondary predictive model. The

specific rate (sqr: square root) increases linearly with temperature

After these “pioneers”, there was a long time of silence in scientific literature about predictive microbiology until 1960’s and 1970’s, when predictive models were used to manage botulism problems (Spencer and Baines, 1967; Roberts et al. 1981). It was during the 1980’s that the interest toward predictive microbiology noticeably increased, after many foodborne outbreaks from Listeria monocytogenes and Salmonella in USA, UK, Australia and New Zealand and the consequent need of new tools to guarantee food safety (Ceron-Carrillo et al. 2014). A number of different approaches to predictive microbiology were experimented during that period. However, particularly in the 1990’s, the use of mechanistic models according to a two-step approach was preferred and currently it is the most used one (“classical” approach): the first step is to establish the growth/death primary model in constant environment, the second step is to produce a secondary model to establish how the parameters of the primary model are affected by environmental factors.

But as bacteria are living organisms, they do not always follow simple mathematical rules. Therefore, different factors that could influence bacterial growth had to be considered in order to develop these mechanistic models. Even if it is recognized that in normal conditions bacterial populations grow or die at constant specific rate, complicating factor could arise and modify the most probable standard model.

Page 43: 19 benv january 2015 en

January 2015 Number 19

43 Around us

Figure 3.The four stages of microbic growth: lag phase (lag), exponential phase (log), stationary phase and death phase

Mathematical foundations necessary to design predictive models were defined by Gibson et al. (1988), who proposed the use of the sigmoid function of Gompertz for the primary models and a quadratic polynomial for the secondary models. As it was not originally developed for modelling bacterial growth, the Gompertz model had important limitations although it had been modified so that parameters had biological interpretation (Zwietering et al 1990, Grijspeerdt & Vanrolleghem 1999). Therefore, a truly dynamic model able to deal with time varying environmental conditions that can occur during food shelf-life was specifically developed by Baranyi and Roberts (1993, 1994, 1995): it is still the most used primary model. As concerns secondary models, despite of some efforts for alternative methods made in 1990’s (Rosso et al. 1995), the use of simple empirical multivariate polynomials is still the most popular approach.

Other than classic predictive microbiology models, during the 2000’s tertiary models became more and more important. They are the integration of primary and secondary models into databases and software tools. These programs collect lab data and yield predictions of bacterial behaviour at different environmental conditions. Some examples are the ComBase, managed by an international consortium funded also by European Union, the Pathogen Modeling Program (PMP), that is a tool from the USA Government, the Danish Food Spoilage and Safety Predictor (FSSP). Recently, predictive microbiology software have been also integrated in programs for risk analysis, just as Dairy Product Safety Predictor, FDA-iRISK, MicroHibro (Tenenhaus-Aziza & Ellouze 2015).

Other than tertiary models, researches during the last decade have been aimed to introduce variability in predictive models. In particular a mathematical theory was prosed and then validated in order to conclude the behaviour of the population from observing many individual cells and design statistical distributions including the natural variability of the whole population (Baranyi & Pin 2001, Elfwing et al. 2004). The introduction of variability is crucial for the use of predictive microbiology in the framework of quantitative microbial risk assessment (QMRA) models, and it needs further research. In fact, predictive microbiology is a science in constant evolution, aiming to predict with more and more precision bacterial behaviour, taking into considerations factors that could influence microbial growth or death. Among future

Particularly for growth, the prior history of the cells is important, because it could affect their capability of coping with the new environment and therefore the duration of the transitional period (lag phase), during which bacteria arrive at the exponential phase. Another stage difficult to be predicted is the post-exponential (stationary phase), when bacteria get to the maximum carrying capacity of their environment. Next, when the nutrients have been consumed, cells die (death phase). The four stages of bacterial growth in function of time are reported in figure 3. In the case of primary death models there is a shoulder instead of a lag phase, and a tail in the place of the stationary phase.

Page 44: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

44 Around us

developments could be the study of bacterial behaviour at the growth-no growth boundary region, which is difficult to be predicted and should be included in new types of models. New models should also consider “persister” cells, which have been reported in bacterial populations during recent year and that could survive after inactivation treatments (Balaban 2011). Finally, it could be useful to relate predictive microbiology and molecular microbiology, considering data on how genes are switched on as function of changing environmental conditions.

However, it is clear that the use of predictive microbiology will increase during next years. Currently, it is more and more applied in order to estimate the risk associated to the consumption of different foodstuffs, particularly when laboratory data are scarce. An example of this tendency is given by two recent EFSA opinions, which based proposals of important changes to CE regulations mostly on results from predictive mathematical models (EFSA 2014a, 2014b). This is the most evident demonstration of the overwhelming evolution of this research area, that only a few decades ago was still considered by food microbiologists not reliable enough to be used in food industry.

References

1. Balaban N.Q. 2011. Persistence: mechanisms for triggering and enhancing phenotypic variability. Curr Opin Genet Dev 21: 768–775.

2. Baranyi J. and Pin C. 2001. A parallel study on modelling bacterial growth and survival curves. J Theor Biol 210: 327-336.

3. Baranyi J. and Roberts T.A. 1994. A dynamic approach to predicting bacterial growth in food. Int J Food Microbiol 23: 277-294.

4. Baranyi J. and Roberts T.A. 1995. Mathematics of predictive food microbiology. Int J Food Microbiol 26: 199-218.

5. Baranyi J. and Roberts T.A. 2004. Predictive Microbiology – Quantitative Microbial Ecology. Culture – March 2004. http://www.ifr.ac.uk/safety/comicro/Culture_25.pdf accessed on 07/01/2015 accessed on 12/01/2015.

6. Baranyi J., Roberts T.A., McClure P.J. 1993. A non-autonomous differential equation to model bacterial growth. Food Microbiol 10: 43-59.

7. Ceron-Carrillo T.G., Luna-Villa M., Munguia- Perez R., Santiesteban- Lopez N.A. 2014. Description and importance of some predictive models that are used as tools in food conservation. Ann Biol Res 5: 18-25.

8. Codex Alimentarius 1999. Principles and Guidelines for the Conduct of Microbiological Risk Assessment. CAC/GL-30, p. 6.

9. Commissione Europea 2005. Regolamento 2073/2005 della Commissione del 15 novembre 2005 sui criteri microbiologici applicabili ai prodotti alimentari. Gazzetta Ufficiale dell’Unione Europea L 338 del 22.12.2005, pag. 1.

10. Elfwing, A., Le Marc, Y., Baranyi, J., Ballagi A. 2004. Observing the growth and division of large number of individual bacteria using image analysis. Appl Environ Microbiol 70: 675-678.

11. Esty J.R. and Meyer K.F. 1922. The heat resistance of the spore of Bacillus botulinus and allied anaerobes, J Infect Dis 31: 650-663.

12. European Food Safety Authority (EFSA) 2014a. Scientific Opinion on the public health risks related to the maintenance of the cold chain during storage and transport of meat. Part 1 (meat of domestic ungulates). EFSA Journal 12: 3601.

13. European Food Safety Authority (EFSA) 2014b. Scientific Opinion on the public health risks related to the maintenance of the cold chain during storage and transport of meat. Part 2 (minced meat from all species). EFSA Journal 12: 3783.

14. Gibson A.M., Bratchell N, Roberts T.A. 1988. Predicting microbial growth: growth responses of salmonellae in a laboratory medium as affected by pH, sodium chloride and storage temperature. Int J Food Microbiol 6: 155-178.

15. Grijspeerdt K. and Vanrolleghem P. 1999. Estimating the parameters of the Baranyi model for bacterial growth. Food Microbiol 16: 593-605.

16. McKellar R.C., and Lu X. 2003. Modelling Microbial Responses in Food. CRC Press, Boca Raton, FL.

17. McMeekin T.A., Olley J.N., Ross. T, Ratkowsky D.A. 1993. Predictive Microbiology. John Wiley & Sons Ltd. Chichester UK.

18. McMeekin T.A., Olley J., Ratkowsky D.A., Ross T. 2002. Predictive microbiology: towards the interface and beyond. Int J Food Microbiol 73: 395-605.

Page 45: 19 benv january 2015 en

January 2015 Number 19

45 Around us

19. Roberts T.A., Gibson A. M., Robinson A. 1981. Prediction of toxin production by Clostridium botulinum in pasteurized pork slurry. J Food Technol 16: 337-355.

20. Rosso L., Lobry J.R., Bajard S., Flandrois J.P. 1995. A convenient model to describe the combined effects of temperature and pH on microbial growth. Appl Env Microbiol 61: 610-616.

21. Scott W.J. (1928). The growth of microorganisms on ox muscle. I. The influence of water content of substrate on rate of growth at -1°C. J Counc Sci Ind Res 9: 177-182.

22. Spencer R. and Baines C.R. 1964. The effect of temperature on the spoilage of wet fish: I. Storage at constant temperature between -1 °C and 25 °C. Food Technol Champaign 18: 769-772.

23. Tenenhaus-Aziza F., Ellouze M. 2015. Software for predictive microbiology and risk assessment: A description and comparison of tools presented at the ICPMF8 Software Fair. Food Microbiol 45: 290-299.

24. Whiting R.C. and Buchanan R.L. 2001. Predictive microbiology and risk assessment. In Food Microbiology. Fundamentals and Frontiers. American Society for Microbiology Press, Washington DC.

25. Zwietering M.H., Jongenburger I., Rombouts F.M., van’t Riet K. 1990. Modeling of the bacterial growth curve. Appl Environ Microbiol 56: 1875-1881.

--Edited by:Luigi Iannetti1 & Romolo Salini2

Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale” 1 National Reference Laboratory for Listeria monocytogenes2 COVEPI - Statistica e GIS

Page 46: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

46 Officially free territories

OFFICIALLY FREE TERRITORIES

Bovine tuberculosis: provinces and regions officially free according to the community legislation up to 14/02/2014

Bovine tuberculosis

Decision Province Region

2003/467/CE

Bergamo

LombardiaLecco

Sondrio

Ascoli Piceno Marche

BolzanoTrentino Alto Adige

Trento

2004/230/CE Grosseto Toscana

2005/28/CEComo Lombardia

Prato Toscana

2006/169/CEPescara Abruzzo

All the region Friuli Venezia Giulia

2007/174/CE

All the region Emilia Romagna

NovaraPiemonte

Verbania

Livorno

ToscanaLucca

Siena

BellunoVeneto

Padova

2008/97/CE

Vercelli Piemonte

PisaToscana

Pistoia

2008/404/CE All the region Veneto

2009/342/CE Oristano Sardegna

2010/391/CE

All the region Lombardia

All the region Toscana

Cagliari

SardegnaMedio-Campidano

Ogliastra

Olbia-Tempio

2011/277/CERieti

LazioViterbo

2012/204/UE

AstiPiemonte

Biella

Fermo Marche

BENV National Veterinary Epidemiological Bulletin

Page 47: 19 benv january 2015 en

January 2015 Number 19

Bovine tuberculosis

Officially free territories 47

Bovine leukosis: Provinces and Regions Officially Free according to the EU legislation up to 14/02/2014

Decision Province Region

2003/467/CE

Bergamo

Lombardia

Brescia

Como

Lecco

Mantova

Sondrio

Varese

Ascoli Piceno Marche

Bolzano Trentino Alto Adige

Bologna

Emilia Romagna

Ferrara

Forlì

Cesena

Modena

Parma

Piacenza

Ravenna

Reggio Emilia

Rimini

Aosta Valle D'Aosta

2004/63/CE

Cremona

LombardiaLodi

Milano

Arezzo

Toscana

Firenze

Grosseto

Livorno

Lucca

Pisa

Pistoia

Prato

Siena

2005/28/CE

Pavia Lombardia

Massa-Carrara Toscana

PerugiaUmbria

Terni

2005/604/CE

Alessandria

Piemonte

Asti

Biella

Cuneo

Novara

Torino

Verbania

Vercelli

Decision Province Region

2006/169/CE

Pescara Abruzzo

Tutta la regione Friuli Venezia Giulia

FrosinoneLazio

Rieti

Imperia Liguria

Ancona

MarcheMacerata

Pesaro

2006/290/CE Tutta la regione Molise

2007/174/CE

Savona Liguria

Oristano in Sardegna; Sardegna

Tutta la regione Veneto

2009/342/CE Tutta la regione Sardegna

2010/391/CE

Napoli Campania

Brindisi Puglia

Agrigento Sicilia

Caltanissetta

Siracusa

Trapani

2011/277/CE Viterbo Lazio

2012/204/UE

Catania

SiciliaEnna

Palermo

Ragusa

2013/177/UE Benevento Campania

2014/91/UE

Latina Lazio

Tutta la regione Liguria

Avellino Campania

Bovine leukosis

Page 48: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

48 Officially free territories

Decision Province Region

2003/467/CE

Bergamo

Lombardia

Como

Lecco

Mantova

Sondrio

Varese

Ascoli Piceno Marche

BolzanoTrentino Alto Adige

Trento

Bologna

Emilia Romagna

Ferrara

Forlì

Cesena

Modena

Parma

Piacenza

Ravenna

Reggio Emilia

Rimini

Cagliari

SardegnaNuoro

Oristano

Sassari

2004/63/CE

Cremona

LombardiaLodi

Pavia

2005/28/CE

Pavia Lombardia

Massa-Carrara Toscana

PerugiaUmbria

Terni

2005/604/CE

Alessandria

Piemonte

Asti

Biella

Novara

Verbania

Vercelli

2006/169/CE

Pescara Abruzzo

Tutta la regione Friuli Venezia Giulia

Rieti Lazio

ImperiaLiguria

Savona

Milano Lombardia

PistoiaToscana

Siena

Bovine brucellosis: Provinces and Regions Officially Free according to the EU legislation up to 14/02/2014

Decision Province Region

2007/174/CE

Torino Piemonte

Firenze Toscana

Tutta la regione Veneto

2008/97/CEBrindisi Puglia

Tutta la regione Toscana

2009/342/CE

Ancona

MarcheMacerata

Pesaro

Cuneo Piemonte

2010/391/CE Campobasso Molise

2011/277/CE

Frosinone

LazioLatina

Viterbo

2012/204/UE Tutta la regione Valle d’Aosta

2014/91/UE Tutta la regione Liguria

Bovine brucellosis

BENV National Veterinary Epidemiological Bulletin

Page 49: 19 benv january 2015 en

January 2015 Number 19

Bovine brucellosis

Officially free territories 49

Decision Province Region

2002/482/CE Bolzano Trentino Alto Adige

2003/237/CE

Arezzo Toscana

Cagliari

SardegnaNuoro

Sassari

Oristano

2003/732/CE

Bergamo

Lombardia

Brescia

Como

Cremona

Lecco

Lodi

Mantova

Milano

Pavia

Sondrio

Varese

Trento Trentino Alto Adige

2004/199/CERieti

LazioViterbo

2005/28/CE

Firenze

Toscana

Livorno

Lucca

Massa-Carrara

Pisa

Pistoia

Prato

Siena

Perugia

Terni Umbria

2005/764/CE Grosseto Toscana

2005/604/CE

Ancona

Marche

Ascoli Piceno

Macerata

Pesaro

Urbino

Alessandria

Piemonte

Asti

Biella

Cuneo

Novara

Torino

Verbania

Vercelli

2006/169/CE

Pescara Abruzzo

Tutta la regione Friuli Venezia Giulia

Savona Liguria

Isernia Molise

Decision Province Region

2008/97/CE

RomaLazio

Latina

Tutta la regione Veneto

2010/391/CE Tutta la regione Molise

2011/277/CETutta la regione Emilia Romagna

Tutta la regione Valle d’Aosta

2014/91/UETutta la regione Lazio

Tutta la regione Liguria

Ovine and caprine brucellosis: Provinces and Regions Officially Free according to the EU legislation up to 14/02/2014

Ovine and caprine brucellosis

July 2014 Number 17

Page 50: 19 benv january 2015 en

BENV National Veterinary Epidemiological Bulletin

50 Contacts & Editorial staff

-National Reference Centre for Veterinary Epidemiology, Planning, Information and Risk Analysis (COVEPI)

EpidemiologyDr. Paolo Calistriph +39 0861 332241

Statistics and GISDr. Annamaria Conteph +39 0861 332246

-CoordinatorSimona Iannetti (COVEPI)

Editorial boardBarbara AlessandriniPaolo CalistriFabrizio De Massis Gianfranco DilettiNicola FerriArmando Giovannini Federica MonacoDaniela MorelliFrancesco PomilioGiovanni Savini

Istructional designerAlessandro De Luca

Web masterand desktop publishingSandro Santarelli

mail [email protected] +39 0861 332251www.izs.it

-National Reference Centre for the study and verification of Foreign Animal Diseases (CESME)

Diagnostics and Monitoring of Exotic Viral DiseasesDr. Federica Monacoph +39 0861 332446

Diagnostic and surveillance of exotic diseases, Virology laboratory. Windhoek, Namibia Dr. Massimo Scacchiaph +39 0861 332405

CONTACTS& EDITORIAL STAFF