Network


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

Hotspot


Dive into the research topics where Linfeng Cheng is active.

Publication


Featured researches published by Linfeng Cheng.


Journal of Virology | 2017

The Long Noncoding RNA NEAT1 Exerts Anti-Hantaviral Effects by Acting as a Positive Feedback for RIG-I Signaling.

Hongwei Ma; Peijun Han; Wei Ye; Hesong Chen; Xuyang Zheng; Linfeng Cheng; Liang Zhang; Lan Yu; Xingan Wu; Zhikai Xu; Yingfeng Lei; Fanglin Zhang

ABSTRACT Hantavirus infection, which causes zoonotic diseases with a high mortality rate in humans, has long been a global public health concern. Over the past decades, accumulating evidence suggests that long noncoding RNAs (lncRNAs) play key regulatory roles in innate immunity. However, the involvement of host lncRNAs in hantaviral control remains uncharacterized. In this study, we identified the lncRNA NEAT1 as a vital antiviral modulator. NEAT1 was dramatically upregulated after Hantaan virus (HTNV) infection, whereas its downregulation in vitro or in vivo delayed host innate immune responses and aggravated HTNV replication. Ectopic expression of NEAT1 enhanced beta interferon (IFN-β) production and suppressed HTNV infection. Further investigation suggested that NEAT1 served as positive feedback for RIG-I signaling. HTNV infection activated NEAT1 transcription through the RIG-I–IRF7 pathway, whereas NEAT1 removed the transcriptional inhibitory effects of the splicing factor proline- and glutamine-rich protein (SFPQ) by relocating SFPQ to paraspeckles, thus promoting the expression of RIG-I and DDX60. RIG-I and DDX60 had synergic effects on IFN production. Taken together, our findings demonstrate that NEAT1 modulates the innate immune response against HTNV infection, providing another layer of information about the role of lncRNAs in controlling viral infections. IMPORTANCE Hantaviruses have attracted worldwide attention as archetypal emerging pathogens. Recently, increasing evidence has highlighted long noncoding RNAs (lncRNAs) as key regulators of innate immunity; however, their roles in hantavirus infection remain unknown. In the present work, a new unexplored function of lncRNA NEAT1 in controlling HTNV replication was found. NEAT1 promoted interferon (IFN) responses by acting as positive feedback for RIG-I signaling. This lncRNA was induced by HTNV through the RIG-I–IRF7 pathway in a time- and dose-dependent manner and promoted HTNV-induced IFN production by facilitating RIG-I and DDX60 expression. Intriguingly, NEAT1 relocated SFPQ and formed paraspeckles after HTNV infection, which might reverse inhibitive effects of SFPQ on the transcription of RIG-I and DDX60. To the best of our knowledge, this is the first study to address the regulatory role of the lncRNA NEAT1 in host innate immunity after HTNV infection. In summary, our findings provide additional insights regarding the role of lncRNAs in controlling viral infections.


PLOS ONE | 2014

Induction of Specific Humoral and Cellular Immune Responses in a Mouse Model following Gene Fusion of HSP70C and Hantaan Virus Gn and S0.7 in an Adenoviral Vector

Linfeng Cheng; Lan Yu; Xingan Wu; Kai Li; Fang Wang; Liang Zhang; Wei Ye; Puyuan Li; Fanglin Zhang; Zhikai Xu

Heat shock proteins (HSPs) display adjuvant functions when given as fusion proteins to enhance vaccination efficiency. To evaluate enhanced potency of Hantaan virus (HTNV) glycoprotein (GP) and nucleocapsid protein (NP) immunogenicity by heat shock protein 70 (HSP70), a recombinant adenovirus rAd-GnS0.7-pCAG-HSP70C expression vector was developed by genetically linking the HSP70 C-terminal gene (HSP70 359–610 aa, HSP70C) to the Gn and 0.7 kb fragment of the NP (aa1–274-S0.7). C57BL/6 mice were immunized with these recombinant adenoviral vectors. A series of immunological assays determined the immunogenicity of the recombinant adenoviral vectors. The results showed that rAd-GnS0.7-pCAG-HSP70C induced a stronger humoral and cellular immune response than other recombinant adenoviruses (rAd-GnS0.7-pCAG and rAd-GnS0.7) and the HFRS vaccine control. Animal protection experiments showed that rAd-GnS0.7-pCAG-HSP70C was effective at protecting C57BL/6 mice from HTNV infection. The results of the immunological experiments showed that HSP70C lead to enhanced vaccine potency, and suggested significant potential in the development of genetically engineered vaccines against HTNV.


Infection, Genetics and Evolution | 2015

Hantaan virus can infect human keratinocytes and activate an interferon response through the nuclear translocation of IRF-3.

Wei Ye; Yongni Xu; Yuan Wang; Yangchao Dong; Qianqian Xi; Mengyuan Cao; Lan Yu; Liang Zhang; Linfeng Cheng; Xingan Wu; Zhikai Xu; Yingfeng Lei; Fanglin Zhang

Hantaan virus (HTNV) is a rodent-borne virus that causes hemorrhagic fever with renal syndrome (HFRS) in Asia and can be transmitted to humans through bites or the inhalation of aerosolized urine, droppings, or saliva of infected rodents. Keratinocytes predominate in the epidermis and reportedly serve as a replication site for multiple vector-borne viruses, little is known about the susceptibility of human skin cells to HTNV infection. Therefore, we aimed to evaluate whether human keratinocytes support HTNV replication and elicit an immune response against HTNV infection. We found that a human keratinocyte cell line, HaCaT, supports HTNV replication. In addition, retinoic acid inducible gene-I (RIG-I) and melanoma differentiation associated gene-5 (MDA5) play key roles in the detection of HTNV infection in HaCaT cells and in the up-regulation of interferon (IFN)-β expression, which subsequently leads to the production of a large amount of antiviral interferon-stimulated genes (ISGs) and other chemokines used for immune cell recruitment. Furthermore, we suggest that interferon regulatory factor (IRF)-3, as opposed to NF-κB/p65 or IRF-7, is translocated to the nucleus to induce IFN-β. However, the early induction of chemokine CXCL10 was a direct result of HaCaT cells counteracting HTNV infection and was not due to the induction of IFN. Overall, our data demonstrate, for the first time, the permissiveness of human keratinocytes to HTNV infection.


International Journal of Molecular Medicine | 2013

Induction of Hantaan virus-specific immune responses in C57BL/6 mice by immunization with a modified recombinant adenovirus containing the chimeric gene, GcS0.7.

Kai Li; Puyuan Li; Xingan Wu; Liang Zhang; Ziyu Liu; Lan Yu; Lei Zhang; Linfeng Cheng; Wentao Bai; Fanglin Zhang; Zhikai Xu

Hantavirus glycoprotein Gc is one of the main components that contribute to the generation of humoral immune responses, while the nucleocapsid protein (NP) is involved in cellular immune responses through the induction of antibody-dependent cytotoxic T cells. In this study, a chimeric gene, GcS0.7, which encodes a fusion protein containing Gc and truncated NP, was constructed as a candidate for Hantaan virus (HTNV) vaccine development. The chimeric gene was cloned into an adenoviral vector in conjunction with the powerful hybrid cytomegalovirus (CMV) enhancer/chicken β-actin (CAG) promoter or the woodchuck hepatitis virus (WHV) post-transcriptional regulatory element (WPRE), or both. Both elements increased the expression level of the fusion protein. The rAd-GcS0.7-pCAG group demonstrated the highest fusion protein expression level, with a 2.3-fold increase compared with the unmodified adenoviral vector. To further evaluate the humoral and cellular immunity induced by the recombinant adenovirus, the antibody titers, interferon (IFN)-γ secretion level and cytotoxic T cell ratio were detected in immunized mice. The strongest HTNV‑specific humoral and cellular immune responses were detected in the rAd-GcS0.7‑pCAG group. The immunogenicity of these recombinant adenoviruses was compared with that of the inactivated vaccine through a series of immunological assays. In terms of the cellular immune responses, the rAd-GcS0.7-pCAG group even exceeded those induced by the vaccine control. The CAG hybrid promoter improved not only the expression level, but also the immunogenicity of the fusion protein, and may thus provide a promising strategy for HTNV vaccine research.


Frontiers in Cellular and Infection Microbiology | 2016

Incorporation of GM-CSF or CD40L Enhances the Immunogenicity of Hantaan Virus-Like Particles

Linfeng Cheng; Fang Wang; Liang Zhang; Lan Yu; Wei Ye; Ziyu Liu; Qikang Ying; Xingan Wu; Zhikai Xu; Fanglin Zhang

A safe and effective Hantaan virus (HTNV) vaccine is highly desirable because HTNV causes an acute and often fatal disease (hemorrhagic fever with renal syndrome, HFRS). Since the immunity of the inactivated vaccine is weak and the safety is poor, HTNV virus-like particles (VLPs) offer an attractive and safe alternative. These particles lack the viral genome but are perceived by the immune system as virus particles. We hypothesized that adding immunostimulatory signals to VLPs would enhance their efficacy. To accomplish this enhancement, we generated chimeric HTNV VLPs containing glycosylphosphatidylinositol (GPI)-anchored granulocyte macrophage colony-stimulating factor (GM-CSF) or CD40 ligand (CD40L) and investigated their biological activity in vitro. The immunization of mice with chimeric HTNV VLPs containing GM-CSF or CD40L induced stronger humoral immune responses and cellular immune responses compared to the HTNV VLPs and Chinese commercial inactivated hantavirus vaccine. Chimeric HTNV VLPs containing GM-CSF or CD40L also protected mice from an HTNV challenge. Altogether, our results suggest that anchoring immunostimulatory molecules into HTNV VLPs can be a potential approach for the control and prevention of HFRS.


Oncotarget | 2016

Construction and immunological characterization of CD40L or GM-CSF incorporated Hantaan virus like particle

Qikang Ying; Tiejun Ma; Linfeng Cheng; Xiaoxiao Zhang; Agnieszka D. Truax; Ruixue Ma; Ziyu Liu; Yingfeng Lei; Liang Zhang; Wei Ye; Fanglin Zhang; Zhikai Xu; Lei Shang; Rongrong Liu; Fang Wang; Xingan Wu

Infection of Hantaan virus (HTNV) usually causes hemorrhagic fever with renal syndrome (HFRS). China has the worst epidemic incidence of HFRS as well as high fatality. Inactivated whole virus has been used for HFRS vaccination, however there are still problems such as safety concerns. CD40 ligand (CD40L) and granulocyte macrophage colony-stimulating factor (GM-CSF) are well-known immune stimulating molecules that can enhance antigen presenting, lymphocytes activation and maturation, incorporation of CD40L and GM-CSF to the surface of virus like particles (VLPs) can greatly improve the vaccination effect. We constructed eukaryotic vectors expressing HTNV M segment and S segment, as well as vectors expressing HTNV M segment with CD40L or GM-CSF, our results showed successful production of CD40L or GM-CSF incorporated HTNV VLPs. In vitro stimulation with CD40L or GM-CSF anchored HTNV VLP showed enhanced activation of macrophages and DCs. CD40L/GM-CSF incorporated VLP can induce higher level of HTNV specific antibody and neutralizing antibody in mice. Immunized mice splenocytes showed higher ability of secreting IFN-γ and IL-2, as well as enhancing CTL activity. These results suggest CD40L/GM-CSF incorporated VLP can serve as prospective vaccine candidate.


Frontiers in Cellular and Infection Microbiology | 2016

Screening and Identification of an H-2Kb-Restricted CTL Epitope within the Glycoprotein of Hantaan Virus

Ruixue Ma; Linfeng Cheng; Qikang Ying; Rong-rong Liu; Tiejun Ma; Xiaoxiao Zhang; Ziyu Liu; Liang Zhang; Wei Ye; Fanglin Zhang; Zhikai Xu; Fang Wang; Xingan Wu

The cytotoxic T lymphocyte (CTL) response plays a key role in controlling viral infection, but only a few epitopes within the HTNV glycoprotein (GP) that are recognized by CTLs have been reported. In this study, we identified one murine HTNV GP-derived H2-Kb-restricted CTL epitope in C57BL/6 mice, which could be used to design preclinical studies of vaccines for HTNV infection. First, 15 8-mer peptides were selected from the HTNV GP amino acid sequence based on a percentile rank of <=1% by IEDB which is the most comprehensive collection of epitope prediction and analysis tool. A lower percentile rank indicates higher affinity and higher immune response. In the case of the consensus method, we also evaluated the binding score of peptide-binding affinity by the BIMAS software to confirm that all peptides were able to bind H2-Kb. Second, one novel GP-derived CTL epitope, GP6 aa456-aa463 (ITSLFSLL), was identified in the splenocytes of HTNV-infected mice using the IFN-γ ELISPOT assay. Third, a single peptide vaccine was administered to C57BL/6 mice to evaluate the immunogenic potential of the identified peptides. ELISPOT and cell-mediated cytotoxicity assays showed that this peptide vaccine induced a strong IFN-γ response and potent cytotoxicity in immunized mice. Last, we demonstrated that the peptide-vaccinated mice had partial protection from challenge with HTNV. In conclusion, we identified an H2-Kb-restricted CTL epitope with involvement in the host immune response to HTNV infection.


Human Vaccines & Immunotherapeutics | 2017

The assessment of Hantaan virus-specific antibody responses after the immunization program for hemorrhagic fever with renal syndrome in northwest China

Zhuo Li; Hanyu Zeng; Ying Wang; Yusi Zhang; Linfeng Cheng; Fanglin Zhang; Yingfeng Lei; Boquan Jin; Ying Ma; Lihua Chen

ABSTRACT Xianyang city is one of the main hemorrhagic fever with renal syndrome (HFRS) epidemic areas in northwest China. Although the HFRS immunity program has been provided in this city, HFRS is still occurred every year. In order to implement the vaccination program effectively and to control HFRS, the analysis of antibody responses specific to Hantaan virus (HTNV) in individuals after vaccination is essential. In this study, a total of 100 subjects were divided into 5 groups: unvaccinated, 1, 3, 29 and 33 months after boost vaccination. The levels and the positive rates of HTNV-NP-specific IgM and IgG antibodies as well as HTNV neutralizing antibodies were significantly increased in the serum of the vaccinated individuals. The positive rates and levels of HTNV-NP-specific IgG and HTNV neutralizing antibody reached their highest values at 3 months respectively and could be sustained up to 33 months after vaccination. Moreover, the titres of HTNV-NP-specific IgM or IgG antibody and the titres of HTNV neutralizing antibody at 1 month after vaccination have a positive correlation. The level of HTNV-NP-specific IgG antibody was much higher than that of HTNV-NP-specific IgM antibody or HTNV neutralizing antibody. In addition, the strongest responses of antibody-secreting cells were observed at 3 months after vaccination, which was consistent with the serum results. Therefore, the HFRS immunization program is effective to induce humoral immunity in the population of northwest China.


Frontiers in Cellular and Infection Microbiology | 2017

In-Cell Western Assays to Evaluate Hantaan Virus Replication as a Novel Approach to Screen Antiviral Molecules and Detect Neutralizing Antibody Titers.

Hongwei Ma; Wei Ye; Hesong Chen; Tie-Jian Nie; Linfeng Cheng; Liang Zhang; Peijun Han; Xingan Wu; Zhikai Xu; Yingfeng Lei; Fanglin Zhang

Hantaviruses encompass rodent-borne zoonotic pathogens that cause severe hemorrhagic fever disease with high mortality rates in humans. Detection of infectious virus titer lays a solid foundation for virology and immunology researches. Canonical methods to assess viral titers rely on visible cytopathic effects (CPE), but Hantaan virus (HTNV, the prototype hantavirus) maintains a relatively sluggish life cycle and does not produce CPE in cell culture. Here, an in-cell Western (ICW) assay was utilized to rapidly measure the expression of viral proteins in infected cells and to establish a novel approach to detect viral titers. Compared with classical approaches, the ICW assay is accurate and time- and cost-effective. Furthermore, the ICW assay provided a high-throughput platform to screen and identify antiviral molecules. Potential antiviral roles of several DExD/H box helicase family members were investigated using the ICW assay, and the results indicated that DDX21 and DDX60 reinforced IFN responses and exerted anti-hantaviral effects, whereas DDX50 probably promoted HTNV replication. Additionally, the ICW assay was also applied to assess NAb titers in patients and vaccine recipients. Patients with prompt production of NAbs tended to have favorable disease outcomes. Modest NAb titers were found in vaccinees, indicating that current vaccines still require improvements as they cannot prime host humoral immunity with high efficiency. Taken together, our results indicate that the use of the ICW assay to evaluate non-CPE Hantaan virus titer demonstrates a significant improvement over current infectivity approaches and a novel technique to screen antiviral molecules and detect NAb efficacies.


Archives of Virology | 2017

DDX50 inhibits the replication of dengue virus 2 by upregulating IFN-β production

Peijun Han; Wei Ye; Xin Lv; Hongwei Ma; Daihui Weng; Yangchao Dong; Linfeng Cheng; Hesong Chen; Liang Zhang; Zhikai Xu; Yingfeng Lei; Fanglin Zhang

Dengue virus (DENV) infects approximately 390 million people per year, and each of the four DENV serotypes (DENV-1, DENV-2, DENV-3, and DENV-4) is capable of causing infection. At present, there is no antiviral drug available for the treatment of DENV. Several DExD/H-box helicases have been shown to be involved in the antiviral immune response or viral replication. In the present study, we investigated the role of DDX50 in DENV-2 RNA replication. Our data showed that the level of DENV-2 RNA increased in DDX50 knockdown cells during an early stage of viral infection and decreased in DDX50-overexpressing cells. DDX50, in conjunction with RIG-I and MDA5, upregulated the production of IFN-β in infected cells through an additive effect on the IFN-β promoter. Furthermore, transcription of several IFN-stimulated genes was increased in DDX50-overexpressing cells infected with DENV-2. These results provide evidence that DDX50 negatively regulates DENV-2 replication during the early stages of infection by inducing IFN-β production.

Collaboration


Dive into the Linfeng Cheng's collaboration.

Top Co-Authors

Avatar

Fanglin Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Liang Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xingan Wu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhikai Xu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Lan Yu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Ye

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yingfeng Lei

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ziyu Liu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Fang Wang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Kai Li

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge