Network


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

Hotspot


Dive into the research topics where Hongli Jin is active.

Publication


Featured researches published by Hongli Jin.


Scientific Reports | 2016

Treatment with hyperimmune equine immunoglobulin or immunoglobulin fragments completely protects rodents from Ebola virus infection

Xuexing Zheng; Gary Wong; Yongkun Zhao; Hualei Wang; Shihua He; Yuhai Bi; Weijin Chen; Hongli Jin; Weiwei Gai; Di Chu; Zengguo Cao; Chong Wang; Quanshui Fan; Hang Chi; Yuwei Gao; Tiecheng Wang; Na Feng; Feihu Yan; Geng Huang; Ying Zheng; Nan Li; Yuetao Li; Jun Qian; Yong Zou; Gary P. Kobinger; George F. Gao; Xiangguo Qiu; Songtao Yang; Xianzhu Xia

Recent successes with monoclonal antibody cocktails ZMappTM and MIL77 against Ebola virus (EBOV) infections have reignited interest in antibody-based therapeutics. Since the production process for monoclonal antibodies can be prolonged and costly, alternative treatments should be investigated. We produced purified equine antisera from horses hyperimmunized with EBOV virus-like particles, and tested the post-exposure efficacy of the antisera in a mouse model of infection. BALB/c mice were given up to 2 mg of purified equine antisera per animal, at 30 minutes, 1 or 2 days post-infection (dpi), in which all animals survived. To decrease the possibility of serum sickness, the equine antisera was digested with pepsin to generate F(ab′)2 fragments, with in vitro neutralizing activity comparable to whole immunoglobulin. Full protection was achieved with when treatment was initiated at 1 dpi, but the suboptimal protection observed with the 30 minute and 2 dpi groups demonstrate that in addition to virus neutralization, other Fc-dependent antibody mechanisms may also contribute to survival. Guinea pigs given 20 mg of antisera or F(ab′)2 at or starting at 1 or 2 dpi were also fully protected from EBOV infection. These results justify future efficacy studies for purified equine products in NHPs.


Archives of Virology | 2016

Isolation and sequence analysis of the complete NS1 and VP2 genes of canine parvovirus from domestic dogs in 2013 and 2014 in China

Hualei Wang; Hongli Jin; Qian Li; Guoxing Zhao; Nan Cheng; Na Feng; Xuexing Zheng; Jianzhong Wang; Yongkun Zhao; Ling Li; Zengguo Cao; Feihu Yan; Lina Wang; Tiecheng Wang; Yuwei Gao; Songtao Yang; Xianzhu Xia

Canine parvovirus (CPV) can cause severe disease in animals and continuously generates new variant and recombinant strains in dogs that have a strong impact on sanitation. It is therefore necessary to investigate epidemic CPV strains to improve our understanding of CPV transmission and epidemic behavior. However, most studies have focused on the analysis of VP2, and therefore, information about recombination and relationships between strains is still lacking. Here, 14 strains of CPV were isolated from domestic dogs suspected of hosting CPV between 2013 and 2014 in China. The complete NS1 and VP2 genes were sequenced and analyzed. The results suggest that the new CPV-2a and new CPV-2b types are the prevalent strains in China. In addition to a few mutations (residues 19, 544, 545, 572 and 583 of NS1 and residues 267, 370, 377 and 440 of VP2) that were preserved during transmission, new mutations (residues 60, 630 of NS1, and residues 21, 310 of VP2) were found in the isolated strains. A phylogenetic tree based on VP2 sequences illustrated that the new CPV-2a and new CPV-2b strains from China form single clusters that are distinct from lineages from other countries. Moreover, recombination between the new CPV-2a and new CPV-2b types was also identified in the isolated strains. Due to differences in selection pressures or recombination, there were a small number of inconsistencies between the phylogenetic trees for VP2 and NS1, which indicated that phylogenetic relationships based on VP2 might not be representative of those based on NS1. The data indicated that mutations and recombination are constantly occurring along with the spread of CPV in China.


Antiviral Research | 2017

Novel chimeric virus-like particles vaccine displaying MERS-CoV receptor-binding domain induce specific humoral and cellular immune response in mice.

Chong Wang; Xuexing Zheng; Wei-Wei Gai; Gary Wong; Hualei Wang; Hongli Jin; Na Feng; Yongkun Zhao; Weijiao Zhang; Nan Li; Guoxing Zhao; Junfu Li; Jinghua Yan; Yuwei Gao; Guixue Hu; Songtao Yang; Xianzhu Xia

ABSTRACT Middle East respiratory syndrome coronavirus (MERS‐CoV) has continued spreading since its emergence in 2012 with a mortality rate of 35.6%, and is a potential pandemic threat. Prophylactics and therapies are urgently needed to address this public health problem. We report here the efficacy of a vaccine consisting of chimeric virus‐like particles (VLP) expressing the receptor binding domain (RBD) of MERS‐CoV. In this study, a fusion of the canine parvovirus (CPV) VP2 structural protein gene with the RBD of MERS‐CoV can self‐assemble into chimeric, spherical VLP (sVLP). sVLP retained certain parvovirus characteristics, such as the ability to agglutinate pig erythrocytes, and structural morphology similar to CPV virions. Immunization with sVLP induced RBD‐specific humoral and cellular immune responses in mice. sVLP‐specific antisera from these animals were able to prevent pseudotyped MERS‐CoV entry into susceptible cells, with neutralizing antibody titers reaching 1: 320. IFN‐&ggr;, IL‐4 and IL‐2 secreting cells induced by the RBD were detected in the splenocytes of vaccinated mice by ELISpot. Furthermore, mice inoculated with sVLP or an adjuvanted sVLP vaccine elicited T‐helper 1 (Th1) and T‐helper 2 (Th2) cell‐mediated immunity. Our study demonstrates that sVLP displaying the RBD of MERS‐CoV are promising prophylactic candidates against MERS‐CoV in a potential outbreak situation. HIGHLIGHTSWe constructed a chimeric parvovirus virus‐like particles displaying the MERS‐CoV receptor‐binding domain on the surface.We found that the chimeric virus‐like particles induced RBD‐specific, neutralizing antibody responses in mice.Splenocytes from immunized mice had considerably higher secretion of both Th1‐ and Th2‐type cytokines.Our results reveal that chimeric virus‐like particles induce both specific humoral and cell‐mediated immunity.


Frontiers in Microbiology | 2016

Visual Detection of West Nile Virus Using Reverse Transcription Loop-Mediated Isothermal Amplification Combined with a Vertical Flow Visualization Strip.

Zengguo Cao; Hualei Wang; Lina Wang; Ling Li; Hongli Jin; Changping Xu; Na Feng; Jianzhong Wang; Qian Li; Yongkun Zhao; Tiecheng Wang; Yuwei Gao; Yiyu Lu; Songtao Yang; Xianzhu Xia

West Nile virus (WNV) causes a severe zoonosis, which can lead to a large number of casualties and considerable economic losses. A rapid and accurate identification method for WNV for use in field laboratories is urgently needed. Here, a method utilizing reverse transcription loop-mediated isothermal amplification combined with a vertical flow visualization strip (RT-LAMP-VF) was developed to detect the envelope (E) gene of WNV. The RT-LAMP-VF assay could detect 102 copies/μl of an WNV RNA standard using a 40 min amplification reaction followed by a 2 min incubation of the amplification product on the visualization strip, and no cross-reaction with other closely related members of the Flavivirus genus was observed. The assay was further evaluated using cells and mouse brain tissues infected with a recombinant rabies virus expressing the E protein of WNV. The assay produced sensitivities of 101.5 TCID50/ml and 101.33 TCID50/ml for detection of the recombinant virus in the cells and brain tissues, respectively. Overall, the RT-LAMP-VF assay developed in this study is rapid, simple and effective, and it is therefore suitable for clinical application in the field.


Viruses | 2016

Intranasal Immunization with Influenza Virus-Like Particles Containing Membrane-Anchored Cholera Toxin B or Ricin Toxin B Enhances Adaptive Immune Responses and Protection against an Antigenically Distinct Virus

Xianliang Ji; Zhiguang Ren; Na Xu; Lingnan Meng; Zhijun Yu; Na Feng; Xiaoyu Sang; Shengnan Li; Yuanguo Li; Tiecheng Wang; Yongkun Zhao; Hualei Wang; Xuexing Zheng; Hongli Jin; Nan Li; Songtao Yang; Jinshan Cao; Wensen Liu; Yuwei Gao; Xianzhu Xia

Vaccination is the most effective means to prevent influenza virus infection, although current approaches are associated with suboptimal efficacy. Here, we generated virus-like particles (VLPs) composed of the hemagglutinin (HA), neuraminidase (NA) and matrix protein (M1) of A/Changchun/01/2009 (H1N1) with or without either membrane-anchored cholera toxin B (CTB) or ricin toxin B (RTB) as molecular adjuvants. The intranasal immunization of mice with VLPs containing membrane-anchored CTB or RTB elicited stronger humoral and cellular immune responses when compared to mice immunized with VLPs alone. Administration of VLPs containing CTB or RTB significantly enhanced virus-specific systemic and mucosal antibody responses, hemagglutination inhibiting antibody titers, virus neutralizing antibody titers, and the frequency of virus-specific IFN-γ and IL-4 secreting splenocytes. VLPs with and without CTB or RTB conferred complete protection against lethal challenge with a mouse-adapted homologous virus. When challenged with an antigenically distinct H1N1 virus, all mice immunized with VLPs containing CTB or RTB survived whereas mice immunized with VLPs alone showed only partial protection (80% survival). Our results suggest that membrane-anchored CTB and RTB possess strong adjuvant properties when incorporated into an intranasally-delivered influenza VLP vaccine. Chimeric influenza VLPs containing CTB or RTB may represent promising vaccine candidates for improved immunological protection against homologous and antigenically distinct influenza viruses.


Oncotarget | 2017

Autophagy is highly targeted among host comparative proteomes during infection with different virulent RABV strains

Ling Li; Hongli Jin; Hualei Wang; Zengguo Cao; Na Feng; Jianzhong Wang; Yongkun Zhao; Xuexing Zheng; Pengfei Hou; Nan Li; Hang Chi; Pei Huang; Cuicui Jiao; Qian Li; Lina Wang; Tiecheng Wang; Weiyang Sun; Yuwei Gao; Changchun Tu; Guixue Hu; Songtao Yang; Xianzhu Xia

Rabies virus (RABV) is a neurotropic virus that causes serious disease in humans and animals worldwide. It has been reported that different RABV strains can result in divergent prognoses in animal model. To identify host factors that affect different infection processes, a kinetic analysis of host proteome alterations in mouse brains infected with different virulent RABV strains was performed using isobaric tags for a relative and absolute quantification (iTRAQ)-liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomics approach, and this analysis identified 147 differentially expressed proteins (DEPs) between the pathogenic challenge virus standard (CVS)-11 strain and the attenuated SRV9 strain. Bioinformatics analyses of these DEPs revealed that autophagy and several pathways associated with autophagy, such as mammalian target of rapamycin (mTOR) signaling, p70S6K signaling, nuclear factor erythroid 2-related factor 2 (NRF2)-mediated oxidative stress and superoxide radical degradation, were dysregulated. Validation of the proteomic data showed that attenuated SRV9 induced more autophagosome accumulation than CVS-11 in an in vitro model. Our findings provide new insights into the pathogenesis of RABV and encourage further studies on this topic.


Journal of Veterinary Science | 2018

Packaging of Rift Valley fever virus pseudoviruses and establishment of a neutralization assay method

Yuetao Li; Yongkun Zhao; Cuiling Wang; Xuexing Zheng; Hualei Wang; Weiwei Gai; Hongli Jin; Feihu Yan; Boning Qiu; Yuwei Gao; Nan Li; Songtao Yang; Xianzhu Xia

Rift Valley fever (RVF) is an acute, febrile zoonotic disease that is caused by the RVF virus (RVFV). RVF is mainly prevalent on the Arabian Peninsula, the African continent, and several islands in the Indian Ocean near southeast Africa. RVFV has been classified by the World Organisation for Animal Health (OIE) as a category A pathogen. To avoid biological safety concerns associated with use of the pathogen in RVFV neutralization assays, the present study investigated and established an RVFV pseudovirus-based neutralization assay. This study used the human immunodeficiency virus (HIV) lentiviral packaging system and RVFV structural proteins to successfully construct RVFV pseudoviruses. Electron microscopy observation and western blotting indicated that the size, structure, and shape of the packaged pseudoviruses were notably similar to those of HIV lentiviral vectors. Infection inhibition assay results showed that an antibody against RVFV inhibited the infective ability of the RVFV pseudoviruses, and an antibody neutralization assay for RVFV detection was then established. This study has successfully established a neutralization assay based on RVFV pseudoviruses and demonstrated that this method can be used to effectively evaluate antibody neutralization.


Viruses | 2016

Equine Immunoglobulin and Equine Neutralizing F(ab′)2 Protect Mice from West Nile Virus Infection

Jiannan Cui; Yongkun Zhao; Hualei Wang; Boning Qiu; Zengguo Cao; Qian Li; Yanbo Zhang; Feihu Yan; Hongli Jin; Tiecheng Wang; Weiyang Sun; Na Feng; Yuwei Gao; Jing Sun; Yanqun Wang; Stanley Perlman; Jincun Zhao; Songtao Yang; Xianzhu Xia

West Nile virus (WNV) is prevalent in Africa, Europe, the Middle East, West Asia, and North America, and causes epidemic encephalitis. To date, no effective therapy for WNV infection has been developed; therefore, there is urgent need to find an efficient method to prevent WNV disease. In this study, we prepared and evaluated the protective efficacy of immune serum IgG and pepsin-digested F(ab′)2 fragments from horses immunized with the WNV virus-like particles (VLP) expressing the WNV M and E proteins. Immune equine F(ab′)2 fragments and immune horse sera efficiently neutralized WNV infection in tissue culture. The passive transfer of equine immune antibodies significantly accelerated the virus clearance in the spleens and brains of WNV infected mice, and reduced mortality. Thus, equine immunoglobulin or equine neutralizing F(ab′)2 passive immunotherapy is a potential strategy for the prophylactic or therapeutic treatment of patients infected with WNV.


Archives of Virology | 2016

Visual detection of Ebola virus using reverse transcription loop-mediated isothermal amplification combined with nucleic acid strip detection

Changping Xu; Hualei Wang; Hongli Jin; Na Feng; Xuexing Zheng; Zengguo Cao; Ling Li; Jianzhong Wang; Feihu Yan; Lina Wang; Hang Chi; Wei-Wei Gai; Chong Wang; Yongkun Zhao; Yan Feng; Tiecheng Wang; Yuwei Gao; Yiyu Lu; Songtao Yang; Xianzhu Xia


Archives of Virology | 2016

Interferon-inducible GTPase: a novel viral response protein involved in rabies virus infection

Ling Li; Hualei Wang; Hongli Jin; Zengguo Cao; Na Feng; Yongkun Zhao; Xuexing Zheng; Jianzhong Wang; Qian Li; Guoxing Zhao; Feihu Yan; Lina Wang; Tiecheng Wang; Yuwei Gao; Changchun Tu; Songtao Yang; Xianzhu Xia

Collaboration


Dive into the Hongli Jin's collaboration.

Top Co-Authors

Avatar

Hualei Wang

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Songtao Yang

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Xianzhu Xia

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Yongkun Zhao

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Yuwei Gao

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Na Feng

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Tiecheng Wang

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Xuexing Zheng

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Feihu Yan

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Zengguo Cao

Academy of Military Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge