JWPR  
Poultry Research  
J. World Poult. Res. 10(2S): 195-202, June 14, 2020  
Journal of World’s  
Research Paper, PII: S2322455X2000025-10  
License: CC BY 4.0  
Molecular Identification of a Velogenic Newcastle Disease Virus  
Strain Isolated from Egypt  
Shakal M.1*, Mira Maher2, Abdulrahman S. Metwally2, Mohammed A. AbdelSabour3, Yahia M. Madbbouly3  
and Gehan Safwat2.  
1Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt.  
2 Faculty of Biotechnology, October University for Modern Sciences and Arts, MSA, 6th of October City, Egypt.  
3Veterinary Serum and Vaccine Research Institute, VSVRI, ARC, Abbassia, Cairo 11381, Egypt.  
*Corresponding author’s Email: shakal2000@gmail.com; ORCID: 0000-0002-1625-7324  
Received: 14 Feb. 2020  
Accepted: 16 Mar. 2020  
ABSTRACT  
Newcastle Disease Virus (NDV) is still a major concern for the Egyptian poultry industry in spite of the mass  
vaccination programs implemented from a long years ago. The current study aimed to carry out the molecular  
identification of surface glycoprotein genes of NDV field strain isolated from the Giza governorate, Egypt.  
Tracheae were collected from 10 broilers NDV-vaccinated chicken flocks (at least three samples from each flock)  
suffering from mild to moderate respiratory symptoms; with mortalities varying from 10-40% during October 2019.  
Only five samples showed HA positive activity after propagation in specific pathogen-free embryonated chicken eggs  
and only one sample was positive for Avian avulavirus 1 by real-time reverse transcription-PCR. Sequencing for the  
cleavage site of the F protein gene of the positive isolate showed the typical known sequence of velogenic NDV  
strains (112RRQKRF117). Phylogenetic analysis of both F and HN genes showed high similarity and close relation to  
Chinese strains of Genotype VII and more specifically subtype VIId, suggesting the role of migratory wild birds in  
NDV evolution in Egypt. In conclusion, further epidemiological and surveillance studies are strongly recommended  
to define the exact role of migratory wild birds in NDV evolution in Egypt.  
Keywords: Broilers, Newcastle Disease, Poultry industry, Velogenic  
INTRODUCTION  
2015). NDV was recorded in Egypt since 1942 (Daubeny  
and Mancy, 1947) and has been reported ever since.  
reported in both vaccinated and non-vaccinated flocks  
subclinical infection manifested by respiratory, intestinal,  
and nervous symptoms, with mortalities up to 100% may  
be a result of NDV infection according to virus strain  
pathogenicity in infected birds. Based on the pathogenicity  
of the virus, NDV strains can be categorized into three  
main types; lentogenic, mesogenic, and velogenic strains.  
NDV can be classified into two classes; class I and class  
II. NDV isolates of class I are grouped into one genotype,  
whereas NDV isolates of class II are grouped into at least  
eighteen genotypes, some with subgenotypes. Genotype  
VII viruses are responsible for the fourth panzootic that  
has spread from Asia, Africa, Europe, and has even been  
isolated in South America, which continues today  
(Dimitrov et al., 2016). The NDV is an enveloped virus  
that has a linear, single-stranded RNA genome of negative  
Newcastle Disease (ND) is one of the most important viral  
diseases affecting poultry which is caused by Avian  
avulavirus 1 (APMV-1) (Abd El Aziz et al., 2016). The  
natural hosts of ND virus (NDV) include chickens,  
turkeys, ducks, geese, pigeons, quail, pheasants, guinea  
fowl, ostriches, and several species of wild birds (Wang et  
ND as an acute viral infectious disease affects  
domestic poultry regardless of gender and age (Saad et al.,  
2017) and causes great economic losses, especially in  
developing countries (Westbury, 2001). Production  
inefficiencies are considered as a greater concern  
compared to mortality losses in breeders and layers flocks  
while mortalities usually reported to be more significant in  
outbreaks were reported in the past years around the  
To cite this paper: Shakal M, Maher M, Metwally AS, AbdelSabour MA, Madbbouly YM, Safwat G (2020). Molecular Identification of a velogenic Newcastle Disease Virus Strain  
Isolated from Egypt. J. World Poult. Res., 10 (2S): 195-202. DOI: https://dx.doi.org/10.36380/jwpr.2020.25  
195  
Shakal et al., 2020  
polarity; with a genome length of about 15.2ꢀkb (Aldous et  
Giza governorate during October of 2019. Tracheae from  
the same flock kept together for isolation. Samples history  
mentioned in Table 1.  
al., 2003; Ashraf et al., 2016). The genome of NDV  
consists of 15,186, 15,192 nucleotides or 15,198  
nucleotides that contains six genes coding six structural  
and two non-structural proteins including an RNA-  
Table 1. History of flock sampled in the present study  
Age of  
directed  
RNA  
polymerase  
(L),  
hemagglutinin-  
Birds  
No/Flock  
Mortalities  
(%)  
Sample  
birds  
(day)  
35  
55  
24  
26  
22  
23  
NDV vaccination  
neuraminidase protein (HN), fusion protein (F), matrix  
protein (M), phosphoprotein (P), and nucleoprotein (N).  
Both F with HN proteins play a collective role in NDV  
infection prosses. The fusion protein is the most important  
key in the NDV virulence determining process (Peeters et  
al., 1999). Mutations affecting NDV viral genome which  
alter its biological properties and virulence, in addition to  
altered immunity, and improper vaccination processes can  
increase the incidence of NDV outbreaks in vaccinated  
flocks (Kattenbelt et al., 2006). Virulence of ND can be  
distinguished on the basis of the cleavage site sequence of  
the F protein (Selim et al., 2018). HN is one of the  
membrane glycoproteins, through its neuraminidase (NA)  
activity it mediates attachment to sialic acid-containing  
receptors (Wang et al., 2015). Recently, the molecular  
identification and phylogenetic analysis of any new NDV  
isolates become an important and usual approach to find  
out which of the applied control measures needs to be  
Sequence analysis of mainly F and of HN proteins genes -  
two surface glycoproteins- is wildly used for molecular  
identification of NDV isolates. Brevity, the current applied  
NDV vaccination programs consist of live and/or  
inactivated genotype I or II NDV or genetically modified  
vaccines depending on flock age and type.  
S1  
S2  
S3  
S4  
S5  
S6  
10,000  
8,000  
11,000  
4,000  
15,000  
2,000  
12.5 %.  
14%.  
37.6%.  
35.8%.  
22.4%.  
10.7%.  
Twice, live LaSota  
S7  
1,000  
34.5%.  
35  
Once, live LaSota  
Twice, live LaSota  
S8  
S9  
S10  
3,000  
4,000  
12,000  
21.3%.  
39.1%.  
35.7%.  
40  
41  
33  
Isolation  
Virus isolation was done from tracheal swabs after  
immersion in Phosphate-Buffered Saline (PBS) mixed  
with gentamycin antibiotic (50 μg/ml) and mycostatin  
(1000 units/mL). Swabs from different birds from the  
same flock were immersed in the same PBS solutions.  
Samples were named numerically as sample 1 (S1):  
sample 10 (S10).  
PBS-containing samples were clarified by  
centrifugation at 5000 rpm for 15 minutes. A 200 µl of  
supernatant fluid from each sample was inoculated into the  
allantoic cavity of five 10-day-old Specific Pathogen Free  
Embryonated Chicken Eggs (SPF-ECE). Allantoic fluid  
from each egg was harvested 3 to 5 days post-inoculation  
and was tested for hemagglutination (HA) activity by  
rapid slide HA test. HA negative samples were submitted  
for two blind passages of SPF-ECE. Collectively, samples  
that showed HA positive activity were kept for further  
molecular identification (OIE manual, 2018).  
In the present study, analysis of nucleotides  
sequences of F and HN genes were done for a recently  
isolated NDV strain obtained from samples collected from  
different chicken flocks showing mild to severe respiratory  
symptoms with variable mortality rates in Giza  
governorate, Egypt.  
Viral RNA extraction  
MATERIALS AND METHODS  
Viral RNA from HA positive allantoic fluid was  
extracted using Pure Link® (Invitrogen, USA) RNA Mini  
Kit following the manual’s instruction.  
Ethical approval  
Institutional, national, and international animal care  
guidelines were followed.  
Real-time reverse transcription-polymerase chain  
reaction  
Sampling and samples history  
Tracheae (at least three samples from each flock)  
from 10 freshly dead broilers, NDV vaccinated chicken  
flocks suffering from mild to moderate respiratory  
symptoms; with mortalities varying from 10 to 40 % as  
well as a range of NDV indicative postmortem lesions at  
Real-time Reverse Transcription PCR (RT-qPCR)  
was performed in one step. Using TOPreal™ One-step -  
SYBR Green with low ROX - RT qPCR Kit (Enzynomics,  
Korea) according to the manufacturer’s instructions and  
196  
J. World Poult. Res., 10(2S): 195-202, 2020  
using the CFX96 Touch real-time PCR detection system  
Table 3. Primers used for F and HN genes amplification  
and sequencing.  
(Bio-Rad Laboratories, USA). Primers used were designed  
according to Wise et al. (2004) which are specific for the  
matrix protein gene of APMV-1 viruses selected from a  
conserved region of the M gene (Table 2).  
Target  
gene  
Primer  
Sequence  
Name  
Fus-F  
Fus-R  
5’-ATGGGCTCCAAACTTTCT-3’  
F protein  
gene  
5’-CATGCTCTTGTAGTGGCTCTC-3’  
The thermal conditions were as follows; reverse  
transcription at 50°C for 30 mins followed by 10 mins at  
95 °C for reverse transcriptase inactivation and initial  
denaturation. Then, followed by 40 amplification cycles of  
95 °C denaturation for 5 s, 52°C annealing for 10 s, and 60  
°C extension for 30 s.  
HN  
protein  
gene  
Hae-F  
Hae-R  
5’-CATGGACCGCGCGGTTAAC -3’  
5’-CTAAACTCTATCATCCTTG-3’  
Sequencing  
Sequencing of the purified RT-PCR products was  
done by the Bigdye Terminator V3.1 cycle sequencing kit  
(Perkin- Elmer, Foster City, CA) and Applied Biosystems  
3130 genetic analyzer machine (ABI, USA).  
Melting curve analysis was performed to determine  
the specificity of amplification as follows: 95°C  
denaturation for 10 s, 65°C annealing for 5 s, and heating  
to 95 °C with an increment 0.5 °C for 0.05 s.  
Genetic alignment  
The quality of obtained F and HN genes sequences  
The melting temperature (Tm) of melting curves and  
Cp values were calculated using the Bio-Rad CFX  
manager 3.1 software (Figure 1).  
were checked, assembled, edited using Bioedit software  
version 7.0.4.1 (Hall, 1999), and submitted to GenBank  
using  
BankIt  
tool  
of  
the  
GenBank  
Table 2. Primers used for Newcastle disease virus  
with accession numbers MN905162 and MN905163,  
respectively.  
detection using RT-qPCR.  
Primer  
Sequence  
Name  
F Primer  
R primer  
5’-AGTGATGTGCTCGGACCTTC-3’  
5’-CCTGAGGAGAGGCATTTGCTA-3’  
Phylogenetic analysis  
The tree was constructed using the neighbor-joining  
method; bootstrapping at 500 repeats using Mega 6  
software version 7.0.26 (Tamura et al., 2013).  
F and HN genes amplification  
Positive NDV RNA samples (by RT-qPCR) samples  
were subjected to one-step RT-PCR using SuperScript™  
III One-Step RT-PCR System with Platinum™ Taq DNA  
Polymerase according to the manufacturer’s instructions to  
amplify full-length F protein gene and HN protein gene  
using two sets of primes kindly provided by Dr.  
Mohammed Rohaim, Virology Department, Cairo  
University, Egypt (Table 3) and using the ProFlex  
PCR thermal cycler (Applied biosystem, USA).  
RESULTS  
Hemagglutination activity  
After three blind passages only S3, S4, S7, S9, and  
S10 samples were positive for hemagglutination activity.  
S7 and S10 were positive for HA after the 1st egg passage,  
S3, and S4 were positive for HA after the 2nd egg passage,  
and S9 was positive for HA after the 3rd egg passage. RNA  
from 5 positive HA samples were sent for one-step RT-  
qPCR.  
Thermal amplification conditions were as follows;  
reverse transcription at 50 °C for 30 min followed by  
initial denaturation for 2 min at 94 °C. Then followed by  
40 cycles of denaturation at 94 °C for 15 s, annealing at 65  
°C for 30 s for F gene while 51 °C for 30 s for HN gene,  
and extension at 68 °C for 120 s followed by one cycle of  
final extension at 68 °C for 5 min.  
NDV detection by RT-qPCR  
Only S4 was positive for Avian avulavirus 1 by RT-  
qPCR with a threshold cycle (CT) of 29.34 with a starting  
quantity of 3.033 log10 in comparison with a standard  
curve (Figure 2) with melting peak at 79 °C (Figure 3 and  
4).  
PCR products were analyzed by agarose gel  
electrophoresis (1%) and then purified using a QIAquick  
Gel Extraction Kit (Qiagen) following the manufacturer’s  
instructions.  
197  
Shakal et al., 2020  
protein gene showed that S4 isolate is closely related to  
genotype VII subtype D (Figure 5).  
Amplification of full F and full HN proteins genes  
by RT-PCR  
RT PCR products gel electrophoresis revealed the  
expected and correct size bands for full-length F and HN  
proteins genes.  
HN protein gene  
Blasting of sequence results obtained for the full HN  
protein gene showed similarities with Chinese genotype  
VII strains with similarities varying from 95.69 % to  
98.72% and with some Egyptian isolates varying from  
94.9% to 95.45 %The phylogenetic tree of the full HN  
protein gene showed that S4 isolate is closely related to  
the Chinese genotype VII (Figure 6). Three-dimensional  
structure of F and HN monomer for S4 isolate was created  
by SWISS-Model modeling online server and visualized  
by PyMOL program version 2.3.4 (Figure 7 and 8).  
Genetic and phylogenetic analysis  
F protein gene  
Blasting of sequence results obtained for the full F  
protein gene showed similarities with Chinese genotype  
VII strains with similarities varying from 95.5 % to  
97.28% and with many Egyptian isolates varying from  
94.5% to 95.5 %. The phylogenetic tree of the full F  
Figure 1. Thermal conditions applied at RT-qPCR and for the melting curve.  
Figure 2. Threshold cycles of tested samples, green lines represent positive control samples (standard curve samples), the red line  
represents positive for Avian avulavirus sample (S4) appeared after 29.34 CT, and pink lines represent the negative for Avian  
avulavirus samples.  
198  
J. World Poult. Res., 10(2S): 195-202, 2020  
Figure 3. Melting curve of tested samples, green lines represent positive control samples (standard curve samples), the red line  
represents positive for Avian avulavirus sample (S4), and pink lines represent the negative for Avian avulavirus samples.  
Figure 4. Melting peak of tested samples, green lines represent positive control samples (standard curve samples), the red line  
represents positive for Avian avulavirus sample (S4) showed a different melting peak, and pink lines represent the negative for  
Avian avulavirus samples.  
199  
Shakal et al., 2020  
Figure 6. Neighbor-joining phylogenetic tree of the full-  
length HN gene of Egyptian isolate of Newcastle disease  
virus (NDV) (S4) in comparison to other NDV strains from  
GenBank. Bootstrap values are shown above the branches. S4  
isolate is indicated by a solid green circle.  
Figure 7. 3D structure for F protein of Newcastle disease  
virus (S4 isolate) created by SWISS-Model modeling online  
server and visualized by PyMOL program version 2.3.4, red  
color represents the cleavage site.  
Figure 5. Neighbor-joining phylogenetic tree of the full-  
length F gene of Egyptian isolate of Newcastle disease virus  
(NDV) (S4) in comparison to other NDV strains from  
GenBank. Bootstrap values are shown above the branches. S4  
isolate is indicated by a solid green circle.  
Figure 8. 3D structure for HN protein of Newcastle disease  
virus (S4 isolate) created by SWISS-Model modeling online  
server and visualized by PyMOL program version 2.3.4.  
200  
J. World Poult. Res., 10(2S): 195-202, 2020  
migratory wild birds in NDV evolution in Egypt with  
DISCUSSION  
defining the main causes of the inability of currently used  
vaccines to protect chickens against infection with  
Newcastle disease virus.  
In the current study, only five samples (50% samples)  
showed HA positive activity indicating infection with a  
hemagglutinating virus. To confirm NDV infection, RT-  
qPCR was performed using the HA positive samples.  
Only S4 isolate was positive for NDV using  
universal primers for APMV-1. Negative RT-qPCR results  
for S3, S7, S9, and S10 may indicate an infection with  
another hemagglutinating virus-like avian influenza H9 or  
H5; however, history of mortalities and symptoms severity  
indicated H9 infection mixed with other respiratory  
pathogens other than H5 (Hussein et al., 2014; Sedeik et  
al., 2018). The most important pathogenicity indicator for  
NDV is the F protein gene sequence analysis mainly for  
cleavage site in which velogenic strains have polybasic  
amino acid sequences; therefore, molecular identification  
and phylogenetic analysis of the F gene is a major  
determinant  
of NDV virulence instead of conventional methods  
be considered as a reliable way for NDV virulence  
evaluation when compared to traditional ways of  
Results of F protein gene sequencing revealed that  
the cleavage site motif of S4 isolate has the sequence of  
velogenic NDV strains 112RRQKRF117 in agreement with  
(Sedeik et al., 2019). Also, the neurological effects of  
NDV infections by is thought to be due to the presence of  
the phenylalanine (F) residue at position 117 (Collins et  
al., 1993). The full sequence of both F and HN protein  
genes were submitted to the GenBank database with  
accession number MN905162 for the full F protein gene  
sequence and MN905163 for the full HN protein gene  
sequence.  
DECLARATION  
Authors' contributions  
All authors reviewed the final manuscript. This work  
is a part of Mira Maher, and Abdulrahman S. Metwally  
thesis under the supervision of Shakal M, and Gehan  
Safwat. Shakal M. designed, supervised the experiments,  
and co-wrote the paper. Gehan Safwat co-designed the  
experiment and co-wrote the paper. Mohammed A. Abdel  
Sabour conducted samples pooling, virus isolation, and co-  
wrote the paper. Mira Maher and Abdulrahman S.  
Metwally conducted RNA extraction, genes amplification  
by PCR, and conducted genetic alignment. Yahia M.  
Madbouly conducted RNA extraction, real-time reverse  
transcription PCR, GenBank submission, phylogenetic  
analysis, and co-wrote the paper.  
Competing interests  
The authors declare that they have no competing  
interests.  
Acknowledgments  
Authors gratefully acknowledge Dr. Mohammed  
Rohaim, Virology Department, Cairo University, Egypt  
for his kind support and comments.  
REFERENCES  
Abd El Aziz M, Abd El-Hamid H, Ellkany H, Nasef S, Nasr S, and El  
Bestawy A (2016). Biological and molecular characterization of  
Newcastle disease virus circulating in chicken flocks, Egypt, during  
2014-2015. Zagazig Veterinary Journal, 44(1): 920. DOI:  
F and HN proteins genes genetic and phylogenetic  
analysis in the present study revealed high similarity of S4  
isolate with Chinese isolates and relatively fewer  
similarities with the Egyptian isolates which may strongly  
refer to the role of migratory wild birds in NDV evolution  
in Egypt.  
Aldous EW, Mynn JK, Banks J, and Alexander DJ (2003). A molecular  
epidemiological study of avian paramyxovirus type 1 (Newcastle  
disease virus) isolates by phylogenetic analysis of a partial  
nucleotide sequence of the fusion protein gene. Avian Pathology,  
Ashraf A, Shah MSU, Habib M, Hussain M, Mahboob S, and Al-Ghanim  
K (2016). Isolation, identification and molecular characterization of  
highly pathogenic Newcastle disease virus from field outbreaks.  
Brazilian Archives of Biology and Technology, 59: e16160301.  
CONCLUSION  
Collins MS, Bashiruddin JB, and Alexander DJ (1993). Deduced amino  
acid sequences at the fusion protein cleavage site of Newcastle  
disease viruses showing variation in antigenicity and pathogenicity.  
Newcastle disease still occurs in sporadic cases despite  
massive vaccination programs implemented in the  
Egyptian poultry field. Migratory wild birds are supposed  
to have a big role in the continuous evolution of NDV in  
Egypt. Further epidemiological and surveillance work is  
strongly recommended to define the exact role of  
128:  
363-370.  
DOI:  
Damena D, Fusaro A, Sombo M, Belaineh R, Heidari A, Kebede A,  
Kidane, M, and Chaka H (2016). Characterization of Newcastle  
disease virus isolates obtained from outbreak cases in commercial  
201  
 
Shakal et al., 2020  
chickens and wild pigeons in Ethiopia. Springer Plus, 5: 476-453.  
wintering site for migratory birds from Southern Brazil. Preventive  
Veterinary Medicine, 116: 197-202. DOI:  
Diel DG, Susta L, Cardenas Garcia S, Killian ML, Brown CC, Miller PJ,  
and Afonso CL (2012). Complete genome and clinicopathological  
characterization of a virulent Newcastle disease virus isolate from  
South America. Journal of Clinical Microbiology, 50:378387.  
Mase M, Imai K, Sanada Y, Sanada N, Yuasa N, Imada T, Tsukamoto K,  
and Yamaguchi S (2002). Phylogenetic analysis of Newcastle  
disease virus genotypes isolated in Japan. Journal of Clinical  
Microbiology,  
40:  
3826–  
3830.  
DOI:  
Dimitrov KM, Ramey AM, Qiu X, Bahl J, and Afonso CL (2016).  
Temporal, geographic, and host distribution of avian paramyxovirus  
1 (Newcastle disease virus). Infection, Genetics and Evolution, 39:  
Mohamed MHA, Kumar S, Paldurai A, and Samal SK (2011). Sequence  
analysis of fusion protein gene of Newcastle disease virus isolated  
from outbreaks in Egypt during 2006. Virology Journal, 8: 237-240.  
Daubeny R. and Mansy W (1947). The occurrence of Newcastle disease  
OIE Manual (2018). Newcastle Disease, Chapter 2.3.14. OIE, Paris.  
Available  
at:  
Ewies SS, Ali A, Tamam SM, and Madbouly HM (2017). Molecular  
characterization of Newcastle disease virus (genotype VII) from  
broiler chickens in Egypt. Beni-Suef University Journal of Basic  
Saad AM, Samy A, Soliman MA, Arafa A, Zanaty A, Hassan MK, and  
Hussein AH (2017). Genotypic and pathogenic characterization of  
genotype VII Newcastle disease viruses isolated from commercial  
farms in Egypt and evaluation of heterologous antibody responses.  
and  
Applied  
Sciences,  
6
(3):  
232-237.  
DOI:  
Fringe R, Bosman, AM, Ebersohn K, Bisschop S, Abolnik C, and Venter  
E (2012). Molecular characterisation of Newcastle disease virus  
isolates from different geographical regions in Mozambique in  
2005. Onderstepoort Journal of Veterinary Research, 79 (1):409-  
Archives  
of  
Virology,  
162(7):  
19851994.  
DOI:  
Sedeik ME, El-Shall, NA, Awad AM, and Kandil N (2018). Molecular  
survey and characterization of H5N8 isolates during 2016-2017 in  
Egypt. Journal of World’s Poultry Research, 8 (4):127-133.  
416.  
Available  
at:  
Available  
at  
Ganar, K., Das, M., Sinha, S., Kumar, S (2014). Newcastle disease virus:  
current status and our understanding. Virus Research, 184: 7181.  
Sedeik ME, Nahed A El-Shall, Ashraf M Awad, Mohamed A Saif, and  
Hall TA (1999). BioEdit: a user-friendly biological sequence alignment  
editor and analysis program for Windows 95/98/NT. Nucleic Acids  
Hadeer  
D
Elsayed (2019). Surveillance and molecular  
characterization of Newcastle disease virus from chickens in north  
Egypt during 2015-2017. Alexandria Journal of Veterinary  
Sciences, 62(2): 82-90. DOI: http://dx.doi.org/10.5455/ajvs.55709.  
Symposium  
Series,  
41:95-98.  
Available  
at:  
Koichiro T, Glen S, Daniel P, Alan F, and Sudhir K, MEGA6 (2013).  
Molecular Evolutionary Genetics Analysis Version 6.0. Molecular  
Hassan KE, Shany SA, Ali A, Dahshan AH, El-Sawah AA, and El-Kady  
MF (2016). Prevalence of avian respiratory viruses in broiler flocks  
Biology and Evolution,  
30 (12): 27252729. DOI:  
in  
Egypt.  
Poultry  
Science, 95 (6): 1271-1280.  
DOI:  
Peeters BP, de Leeuw OS, Koch G, and Gielkens AL (1999). Rescue of  
Newcastle disease virus from cloned cDNA: evidence that  
cleavability of the fusion protein is a major determinant for  
virulence. Journal of Virology, 73:50015009. Available at:  
Hussein HA, Emara MM, Samy AM, and Shalaby MA (2000). Antigenic  
diversity of Newcastle disease virus isolated from 52 breeder and  
broiler flocks in Egypt.The Egyptian .J .of Imm .Vet, 2: 3-15.  
Available at Egyptian Journal of Veterinary Sciences  
Selim KM, Selim A, Arafa A, Hussein HA, and Elsanousi AA (2018).  
Molecular characterization of full fusion protein (F) of Newcastle  
disease virus genotype VIId isolated from Egypt during 2012-  
Hussein AH, Emara MM, Rohaim MA, Ganapathy K, and Arafa AM  
(2014). Sequence analysis of infectious bronchitis virus IS/1494  
like strain isolated from broiler chicken coinfected with Newcastle  
disease virus in Egypt during 2012. International Journal of Poultry  
2016. Veterinary  
World, 11(7)  
930938.  
DOI:  
Science,  
13  
(9):  
530-536.  
DOI:  
Shahid M, Muhammad Q, Al-Ghanim KA, Al-Misned F, and Al-Mulhim  
N (2020). Isolation, identification and molecular characterization of  
Newcastle disease virus using SDS-PAGE. Journal of King Saud  
Jaganathan S, Ooi PT, Phang LY, Allaudin ZN, Yip LS, Choo PY, Lim  
BK, Lemiere S, and Audonnet JC (2015). Observation of risk  
factors, clinical manifestations and genetic characterization of  
recent Newcastle disease virus outbreak in West Malaysia. BMC  
University  
-
Science,  
32(1):  
1000-1003.  
DOI:  
Wang JY, Liu WH, Ren JJ, Tang P, Wu N, Wu HY, and Liu H J (2015).  
Characterization of emerging Newcastle disease virus isolates in  
Veterinary  
Research,  
11:  
219-227.  
DOI:  
China. Virology  
Journal, 12(1):  
119.  
DOI:  
Kang Y, Li Y, Yuan R, Li X, Sun M, Wang Z, Feng M, Jiao P, and Ren  
T (2014). Phylogenetic relationships and pathogenicity variation of  
two Newcastle disease viruses isolated from domestic ducks in  
Southern China. Virology Journal, 11: 147- 159. DOI:  
Westbury HA (2001). Newcastle disease virus: an evolving pathogen.  
Avian Pathology, 30: 511. DOI:  
Wise GM, David LS, Bruce SS, Janice CP, Dennis AS, Daniel  
JK, Darrell R K, and Erica S (2004). Development of a Real-Time  
reverse-transcription PCR for detection of Newcastle disease virus  
Kattenbelt JA, Meers J, and Gould AR (2006). Genome sequence of the  
thermostable Newcastle disease virus (strain I-2) reveals a possible  
phenotypic locus. Veterinary Microbiology, 114: 134-141. DOI:  
RNA  
Microbiology, 42 (1) 329-338.  
in  
clinical  
samples.  
Journal  
of  
Clinical  
Marks FS, Rodenbusch CR, Okino CH, Hein HE, Costa EF, Machado G,  
Canal CW, Brentano L, and Corbellini LG (2014). Targeted survey  
of Newcastle disease virus in backyard poultry flocks located in  
202