Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yuelong Shu is active.

Publication


Featured researches published by Yuelong Shu.


The Journal of Infectious Diseases | 2007

Surveillance of Resistance to Adamantanes among Influenza A(H3N2) and A(H1N1) Viruses Isolated Worldwide

Varough Deyde; Xiyan Xu; Rick A. Bright; Michael Shaw; Catherine B. Smith; Ye Zhang; Yuelong Shu; Larisa V. Gubareva; Nancy J. Cox; Alexander Klimov

Our previous reports demonstrated an alarming increase in resistance to adamantanes among influenza A(H3N2) viruses isolated in 2001-2005. To continue monitoring drug resistance, we conducted a comprehensive analysis of influenza A(H3N2) and A(H1N1) viruses isolated globally in 2005-2006. The results obtained by pyrosequencing indicate that 96.4% (n=761) of A(H3N2) viruses circulating in the United States were adamantane resistant. Drug resistance has reached 100% among isolates from some Asian countries. Analysis of correlation between the appearance of drug resistance and the evolutionary pathway of the hemagglutinin (HA) gene suggests at least 2 separate introductions of resistance into circulating populations that gave rise to identifiable subclades. It also indicates that resistant A(H3N2) viruses may have emerged in Asia in late 2001. Among A(H1N1) viruses isolated worldwide, resistance reached 15.5% in 2005-2006; in the United States alone, it was 4.0%. Phylogenetic analysis of the HA and M genes indicates that the acquisition of resistance in A(H1N1) viruses can be linked to a specific genetic group and was not a result of reassortment between A(H3N2) and A(H1N1) viruses. The results of the study highlight the necessity of close monitoring of resistance to existing antivirals as wells as the need for new therapeutics.


The Lancet | 2014

Clinical and epidemiological characteristics of a fatal case of avian influenza A H10N8 virus infection: a descriptive study

Haiying Chen; Hui Yuan; Rongbao Gao; Jinxiang Zhang; Dayan Wang; Ying Xiong; Guoyin Fan; Fan Yang; Xiaodan Li; Jianfang Zhou; Shumei Zou; Lei Yang; Tao Chen; Libo Dong; Hong Bo; Xiang Zhao; Ye Zhang; Yu Lan; Tian Bai; Jie Dong; Qun Li; Shiwen Wang; Zhang Y; Hui Li; Tian Gong; Yong Shi; Xiansheng Ni; Jianxiong Li; Jun Zhou; Jiyi Fan

BACKGROUND Human infections with different avian influenza viruses--eg, H5N1, H9N2, and H7N9--have raised concerns about pandemic potential worldwide. We report the first human infection with a novel reassortant avian influenza A H10N8 virus. METHODS We obtained and analysed clinical, epidemiological, and virological data from a patient from Nanchang City, China. Tracheal aspirate specimens were tested for influenza virus and other possible pathogens by RT-PCR, viral culture, and sequence analyses. A maximum likelihood phylogenetic tree was constructed. FINDINGS A woman aged 73 years presented with fever and was admitted to hospital on Nov 30, 2013. She developed multiple organ failure and died 9 days after illness onset. A novel reassortant avian influenza A H10N8 virus was isolated from the tracheal aspirate specimen obtained from the patient 7 days after onset of illness. Sequence analyses revealed that all the genes of the virus were of avian origin, with six internal genes from avian influenza A H9N2 viruses. The aminoacid motif GlnSerGly at residues 226-228 of the haemagglutinin protein indicated avian-like receptor binding preference. A mixture of glutamic acid and lysine at residue 627 in PB2 protein--which is associated with mammalian adaptation--was detected in the original tracheal aspirate samples. The virus was sensitive to neuraminidase inhibitors. Sputum and blood cultures and deep sequencing analysis indicated no co-infection with bacteria or fungi. Epidemiological investigation established that the patient had visited a live poultry market 4 days before illness onset. INTERPRETATION The novel reassortant H10N8 virus obtained is distinct from previously reported H10N8 viruses. The virus caused human infection and could have been associated with the death of a patient. FUNDING Emergency Research Project on human infection with avian influenza H7N9 virus, the National Basic Research Program of China, and the National Mega-projects for Infectious Diseases.


Emerging Infectious Diseases | 2006

Human Streptococcus suis Outbreak, Sichuan, China

Hongjie Yu; Huaiqi Jing; Zhihai Chen; Han Zheng; Xiaoping Zhu; Hua Wang; Shiwen Wang; Lunguang Liu; Rongqiang Zu; Longze Luo; Nijuan Xiang; Honglu Liu; Xuecheng Liu; Yuelong Shu; Shui Shan Lee; Shuk Kwan Chuang; Wang Y; Jianguo Xu; Weizhong Yang

Streptococcus suis outbreak was associated with exposure to sick or dead pigs.


Nature | 2013

The genesis and source of the H7N9 influenza viruses causing human infections in China.

Tommy Tsan-Yuk Lam; Jia Wang; Yongyi Shen; Boping Zhou; Lian Duan; C. L. Cheung; Chi Ma; Samantha Lycett; Connie Leung; Xinchun Chen; L Li; Wenshan Hong; Yujuan Chai; Linlin Zhou; Huyi Liang; Zhihua Ou; Yongmei Liu; Amber Farooqui; David J. Kelvin; Leo L.M. Poon; David K. Smith; Oliver G. Pybus; Gabriel M. Leung; Yuelong Shu; Robert G. Webster; Richard J. Webby; J. S. M. Peiris; Andrew Rambaut; Huachen Zhu; Yi Guan

A novel H7N9 influenza A virus first detected in March 2013 has since caused more than 130 human infections in China, resulting in 40 deaths. Preliminary analyses suggest that the virus is a reassortant of H7, N9 and H9N2 avian influenza viruses, and carries some amino acids associated with mammalian receptor binding, raising concerns of a new pandemic. However, neither the source populations of the H7N9 outbreak lineage nor the conditions for its genesis are fully known. Using a combination of active surveillance, screening of virus archives, and evolutionary analyses, here we show that H7 viruses probably transferred from domestic duck to chicken populations in China on at least two independent occasions. We show that the H7 viruses subsequently reassorted with enzootic H9N2 viruses to generate the H7N9 outbreak lineage, and a related previously unrecognized H7N7 lineage. The H7N9 outbreak lineage has spread over a large geographic region and is prevalent in chickens at live poultry markets, which are thought to be the immediate source of human infections. Whether the H7N9 outbreak lineage has, or will, become enzootic in China and neighbouring regions requires further investigation. The discovery here of a related H7N7 influenza virus in chickens that has the ability to infect mammals experimentally, suggests that H7 viruses may pose threats beyond the current outbreak. The continuing prevalence of H7 viruses in poultry could lead to the generation of highly pathogenic variants and further sporadic human infections, with a continued risk of the virus acquiring human-to-human transmissibility.


Science | 2013

Infectivity, Transmission, and Pathology of Human-Isolated H7N9 Influenza Virus in Ferrets and Pigs

Huachen Zhu; Dayan Wang; David J. Kelvin; L Li; Z. Zheng; S.-W. Yoon; S.-S. Wong; A. Farooqui; Jun Wang; D. Banner; R. Chen; R. Zheng; Jianfang Zhou; Yu Zhang; W. Hong; W. Dong; Q. Cai; M. H. A. Roehrl; S. S. H. Huang; A. A. Kelvin; T. Yao; Boping Zhou; Xinchun Chen; Gabriel M. Leung; L. L. M. Poon; Robert G. Webster; Richard J. Webby; J. S. M. Peiris; Yi Guan; Yuelong Shu

Avian Flu in Ferrets A recent outbreak of avian H7N9 influenza in humans in eastern China has been closely monitored for any evidence of human-to-human transmission and its potential for sparking a pandemic. Zhu et al. (p. 183, published online 23 May) examined the behavior of the avian virus in the ferret, a mammalian model for human influenza. The virus was excreted by the ferrets and could be transmitted readily by contact but displayed limited capacity for airborne infectivity. The pathology of H7N9 is similar to H1N1, and it seems that factors other than the intrinsic pathogenicity of the virus contribute to the reported high fatality rate. An emergent avian influenza virus shows high contact transmissibility but limited capacity for airborne spread between mammals. The emergence of the H7N9 influenza virus in humans in Eastern China has raised concerns that a new influenza pandemic could occur. Here, we used a ferret model to evaluate the infectivity and transmissibility of A/Shanghai/2/2013 (SH2), a human H7N9 virus isolate. This virus replicated in the upper and lower respiratory tracts of the ferrets and was shed at high titers for 6 to 7 days, with ferrets showing relatively mild clinical signs. SH2 was efficiently transmitted between ferrets via direct contact, but less efficiently by airborne exposure. Pigs were productively infected by SH2 and shed virus for 6 days but were unable to transmit the virus to naïve pigs or ferrets. Under appropriate conditions, human-to-human transmission of the H7N9 virus may be possible.


The Lancet | 2008

Probable limited person-to-person transmission of highly pathogenic avian influenza A (H5N1) virus in China.

Hua Wang; Zijian Feng; Yuelong Shu; Hongjie Yu; Lei Zhou; Rongqiang Zu; Yang Huai; Jie Dong; Changjun Bao; Leying Wen; Hong Wang; Peng Yang; Wei Zhao; Libo Dong; Minghao Zhou; Qiaohong Liao; Haitao Yang; Min Wang; Xiaojun Lu; Zhiyang Shi; Wei Wang; Ling Gu; Fengcai Zhu; Qun Li; Weidong Yin; Weizhong Yang; Dexin Li; Timothy M. Uyeki; Wang Y

BACKGROUND In December, 2007, a family cluster of two individuals infected with highly pathogenic avian influenza A (H5N1) virus was identified in Jiangsu Province, China. Field and laboratory investigations were implemented immediately by public-health authorities. METHODS Epidemiological, clinical, and virological data were collected and analysed. Respiratory specimens from the patients were tested by reverse transcriptase (RT) PCR and by viral culture for the presence of H5N1 virus. Contacts of cases were monitored for symptoms of illness for 10 days. Any contacts who became ill had respiratory specimens collected for H5N1 testing by RT PCR. Sera were obtained from contacts for H5N1 serological testing by microneutralisation and horse red-blood-cell haemagglutinin inhibition assays. FINDINGS The 24-year-old index case died, and the second case, his 52-year-old father, survived after receiving early antiviral treatment and post-vaccination plasma from a participant in an H5N1 vaccine trial. The index cases only plausible exposure to H5N1 virus was a poultry market visit 6 days before the onset of illness. The second case had substantial unprotected close exposure to his ill son. 91 contacts with close exposure to one or both cases without adequate protective equipment provided consent for serological investigation. Of these individuals, 78 (86%) received oseltamivir chemoprophylaxis and two had mild illness. Both ill contacts tested negative for H5N1 by RT PCR. All 91 close contacts tested negative for H5N1 antibodies. H5N1 viruses isolated from the two cases were genetically identical except for one non-synonymous nucleotide substitution. INTERPRETATION Limited, non-sustained person-to-person transmission of H5N1 virus probably occurred in this family cluster.


Science | 2013

Structures and receptor binding of hemagglutinins from human-infecting H7N9 influenza viruses.

Yi Shi; Wei Zhang; Fei Wang; Jianxun Qi; Ying Wu; Hao Song; Feng Gao; Yuhai Bi; Yanfang Zhang; Zheng Fan; Cheng-Feng Qin; Honglei Sun; Jinhua Liu; Joel Haywood; Wenjun Liu; Weimin Gong; Dayan Wang; Yuelong Shu; Wang Y; Jinghua Yan; George F. Gao

Two Viruses to Bind Structural studies of two different H7N9 influenza viruses isolated from humans—A/Shanghai/1/2013 and A/Anhui/1/2013—which have different amino acid sequences in the receptor binding site, provide data indicating that the virus is in transition with respect to host adaptation. The Shanghai virus was one of the first isolated in humans that binds avian receptor glycans with high affinity, but binds poorly to human receptors. However, the later Anhui isolates can bind both avian and human receptors at high affinity. Shi et al. (p. 243, published online 5 September) show that four hydrophobic mutations contribute to acquisition of affinity for the human receptor by the virus hemagglutinin (HA) and confirm this effect in binding studies with virus particles. Further comparison of a mutant H7N9 A/Anhui/1/2013 HA with the bird flu H5N1 virus revealed the significance of some of the naturally occurring changes observed in circulating H7N9 viruses, which helps to explain how these viruses have been able to cause many severe human infections in a short time. Four amino acids in the H7N9 influenza virus binding site provide a hydrophobic environment for human receptors. An avian-origin human-infecting influenza (H7N9) virus was recently identified in China. We have evaluated the viral hemagglutinin (HA) receptor–binding properties of two human H7N9 isolates, A/Shanghai/1/2013 (SH-H7N9) (containing the avian-signature residue Gln226) and A/Anhui/1/2013 (AH-H7N9) (containing the mammalian-signature residue Leu226). We found that SH-H7N9 HA preferentially binds the avian receptor analog, whereas AH-H7N9 HA binds both avian and human receptor analogs. Furthermore, an AH-H7N9 mutant HA (Leu226 → Gln) was found to exhibit dual receptor-binding property, indicating that other amino acid substitutions contribute to the receptor-binding switch. The structures of SH-H7N9 HA, AH-H7N9 HA, and its mutant in complex with either avian or human receptor analogs show how AH-H7N9 can bind human receptors while still retaining the avian receptor–binding property.


Cell Research | 2010

H5N1 influenza viruses: outbreaks and biological properties

Gabriele Neumann; Hualan Chen; George F. Gao; Yuelong Shu; Yoshihiro Kawaoka

All known subtypes of influenza A viruses are maintained in wild waterfowl, the natural reservoir of these viruses. Influenza A viruses are isolated from a variety of animal species with varying morbidity and mortality rates. More importantly, influenza A viruses cause respiratory disease in humans with potentially fatal outcome. Local or global outbreaks in humans are typically characterized by excess hospitalizations and deaths. In 1997, highly pathogenic avian influenza viruses of the H5N1 subtype emerged in Hong Kong that transmitted to humans, resulting in the first documented cases of human death by avian influenza virus infection. A new outbreak started in July 2003 in poultry in Vietnam, Indonesia, and Thailand, and highly pathogenic avian H5N1 influenza viruses have since spread throughout Asia and into Europe and Africa. These viruses continue to infect humans with a high mortality rate and cause worldwide concern of a looming pandemic. Moreover, H5N1 virus outbreaks have had devastating effects on the poultry industries throughout Asia. Since H5N1 virus outbreaks appear to originate from Southern China, we here examine H5N1 influenza viruses in China, with an emphasis on their biological properties.


Nature Communications | 2013

Interferon-induced transmembrane protein-3 genetic variant rs12252-C is associated with severe influenza in Chinese individuals.

Yonghong Zhang; Yan Zhao; Ning Li; Yanchun Peng; Eleni Giannoulatou; Ronghua Jin; Huiping Yan; Hao Wu; Jin-Hua Liu; Ning Liu; Dayan Wang; Yuelong Shu; Ling-Pei Ho; Paul Kellam; Andrew J. McMichael; Tao Dong

The SNP rs12252-C allele alters the function of interferon-induced transmembrane protein-3 increasing the disease severity of influenza virus infection in Caucasians, but the allele is rare. However, rs12252-C is much more common in Han Chinese. Here we report that the CC genotype is found in 69% of Chinese patients with severe pandemic influenza A H1N1/09 virus infection compared with 25% in those with mild infection. Specifically, the CC genotype was estimated to confer a sixfold greater risk for severe infection than the CT and TT genotypes. More importantly, because the risk genotype occurs with such a high frequency, its effect translates to a large population-attributable risk of 54.3% for severe infection in the Chinese population studied compared with 5.4% in Northern Europeans. Interferon-induced transmembrane protein-3 genetic variants could, therefore, have a strong effect of the epidemiology of influenza in China and in people of Chinese descent.


Proceedings of the National Academy of Sciences of the United States of America | 2011

High genetic compatibility and increased pathogenicity of reassortants derived from avian H9N2 and pandemic H1N1/2009 influenza viruses

Yipeng Sun; Kun Qin; Jingjing Wang; Juan Pu; Qingdong Tang; Yanxin Hu; Yuhai Bi; Xueli Zhao; Hanchun Yang; Yuelong Shu; Jinhua Liu

H9N2 influenza viruses have been circulating worldwide in multiple avian species and repeatedly infecting mammals, including pigs and humans, posing a significant threat to public health. The coexistence of H9N2 and pandemic influenza H1N1/2009 viruses in pigs and humans provides an opportunity for these viruses to reassort. To evaluate the potential public risk of the reassortant viruses derived from these viruses, we used reverse genetics to generate 127 H9 reassortants derived from an avian H9N2 and a pandemic H1N1 virus, and evaluated their compatibility, replication ability, and virulence in mice. These hybrid viruses showed high genetic compatibility and more than half replicated to a high titer in vitro. In vivo studies of 73 of 127 reassortants revealed that all viruses were able to infect mice without prior adaptation and 8 reassortants exhibited higher pathogenicity than both parental viruses. All reassortants with higher virulence than parental viruses contained the PA gene from the 2009 pandemic virus, revealing the important role of the PA gene from the H1N1/2009 virus in generating a reassortant virus with high public health risk. Analyses of the polymerase activity of the 16 ribonucleoprotein combinations in vitro suggested that the PA of H1N1/2009 origin also enhanced polymerase activity. Our results indicate that some avian H9-pandemic reassortants could emerge with a potentially higher threat for humans and also highlight the importance of monitoring the H9-pandemic reassortant viruses that may arise, especially those that possess the PA gene of H1N1/2009 origin.

Collaboration


Dive into the Yuelong Shu's collaboration.

Top Co-Authors

Avatar

Dayan Wang

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Rongbao Gao

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Xiang Zhao

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Lei Yang

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Ye Zhang

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Wenfei Zhu

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Dexin Li

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Hongjie Yu

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Jianfang Zhou

Chinese Center for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Libo Dong

Chinese Center for Disease Control and Prevention

View shared research outputs
Researchain Logo
Decentralizing Knowledge