Network


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

Hotspot


Dive into the research topics where Ningyi Jin is active.

Publication


Featured researches published by Ningyi Jin.


BioMed Research International | 2013

Lactobacilli Reduce Chemokine IL-8 Production in Response to TNF-α and Salmonella Challenge of Caco-2 Cells

Dayong Ren; Chang Li; Yanqing Qin; Ronglan Yin; Shouwen Du; Fei Ye; Hongfeng Liu; Maopeng Wang; Yang Sun; Xiao Li; Mingyao Tian; Ningyi Jin

The probiotic properties of two selected lactobacilli strains were assessed. L. salivarius and L. plantarum displayed higher hydrophobicity (48% and 54%, resp.) and coaggregation ability with four pathogens (from 7.9% to 57.5%). L. salivarius and L. plantarum had good inhibitory effects on S. aureus (38.2% and 49.5%, resp.) attachment to Caco-2 cells. Live lactobacilli strains and their conditioned media effectively inhibited IL-8 production (<14.6 pg/mL) in TNF-α-induced Caco-2 cells. Antibiotic-treated and the sonicated lactobacilli also maintained inhibitory effects (IL-8 production from 5.0 to 36.3 pg/mL); however, the heat-treated lactobacilli lost their inhibitory effects (IL-8 production from 130.2 to 161.0 pg/mL). These results suggest that both the structural components and the soluble cellular content of lactobacilli have anti-inflammatory effects. We also found that pretreatment of Caco-2 cells with lactobacilli inhibited S. typhimurium-induced IL-8 production (<27.3 pg/mL). However, lactobacilli did not inhibit IL-8 production in Caco-2 cells pretreated with S. typhimurium. These results suggest that the tested lactobacilli strains are appropriate for preventing inflammatory diseases caused by enteric pathogens but not for therapy. In short, L. salivarius and L. plantarum are potential candidates for the development of microbial ecological agents and functional foods.


International Journal of Molecular Medicine | 2012

Therapeutic efficacy of an hTERT promoter-driven oncolytic adenovirus that expresses apoptin in gastric carcinoma

Lei Liu; Wenbin Wu; Guangze Zhu; Liming Liu; Guofang Guan; Xiao Li; Ningyi Jin; Baorong Chi

The efficacy and specificity of treatment are the major challenges for cancer gene therapy. Oncolytic virotherapy is an attractive drug delivery platform of cancer gene therapy. Previous studies have determined that apoptin is a p53-independent, Bcl-2-insensitive apoptotic protein that has the ability to induce apoptosis specifically in tumor cells. In this study, we show that the administration of a dual cancer-specific oncolytic adenovirus construct, Ad-hTERT-E1a-apoptin [in which the adenovirus early region 1a (E1a) gene is driven by the cancer-specific promoter of human telomerase reverse transcriptase (hTERT) and that expresses apoptin simultaneously], suppresses tumor growth in gastric carcinoma cells in vitro and reduces the tumor burden in vivo in xenografted nude mice. The observation that infection with the Ad-hTERT-E1a-apoptin construct significantly inhibited the growth of gastric cancer cells and protected normal human gastric epithelium from growth inhibition confirmed the induction of cancer cell-selective adenovirus replication, growth inhibition and apoptosis by this therapeutic approach. In vivo assays were performed using BALB/c nude mice that had established primary tumors. Subcutaneous primary tumor volume was reduced not only in the intratumoral injection group but also in the systemic delivery mice following treatment with Ad-hTERT-E1a-apoptin. Furthermore, treatment of primary models with Ad-hTERT-E1a-apoptin increased the mouse survival time. These data reinforce previous research and highlight the potential therapeutic application of Ad-hTERT-E1a-apoptin for the treatment of neoplastic diseases in clinical trials.


Journal of Virological Methods | 2011

Immune responses to the oral administration of recombinant Bacillus subtilis expressing multi-epitopes of foot-and-mouth disease virus and a cholera toxin B subunit

Bo Hu; Chang Li; Huijun Lu; Zhanbo Zhu; Shouwen Du; Ming Ye; Lei Tan; Dayong Ren; Jiali Han; Shifu Kan; Jing Wang; Ningyi Jin

Bacillus subtilis has been engineered successfully to express heterologous antigens for use as a vaccine vehicle that can elicit mucosal and systemic immunity response. In this study, a recombinant B. subtilis expressing the B subunit of cholera toxin (CT-B) and an epitope box constituted with antigen sites from foot-and-mouth disease virus (FMDV) type Asia 1 was constructed and named 1A751/CTB-TEpiAs. Its capability to induce mucosal, humoral, and cellular responses in mice and guinea pigs was evaluated after oral administration with vegetative cells of 1A751/CTB-TEpiAs. In addition, its capability to protect guinea pigs against homologous virus challenge was examined. All animals were given booster vaccination at day 21 after initial inoculation and guinea pigs were challenged 3 weeks after booster vaccination. The control groups were inoculated with a commercial vaccine or administered orally with 1A751/pBC38C or an oral buffer. All animals vaccinated with 1A751/CTB-TEpiAs developed specific anti-FMDV IgA in lung and gut lavage fluid, serum ELISA antibody, neutralizing antibody as well as T lymphocyte proliferation, and IFN-γ secretory responses. Three of the five guinea pigs vaccinated with 1A751/CTB-TEpiAs were protected completely from the viral challenge. The results demonstrate the potential viability of a B. subtilis-based recombinant vaccine for the control and prevention of FMDV infections.


Clinical & Developmental Immunology | 2012

Protection against SHIV-KB9 Infection by Combining rDNA and rFPV Vaccines Based on HIV Multiepitope and p24 Protein in Chinese Rhesus Macaques

Chang Li; Zhenwei Shen; Xiao Li; Jieying Bai; Lin Zeng; Mingyao Tian; Ying Jin Song; Ming Ye; Shouwen Du; Dayong Ren; Cunxia Liu; Na Zhu; Dandan Sun; Yi Li; Ningyi Jin

Developing an effective vaccine against HIV infection remains an urgent goal. We used a DNA prime/fowlpox virus boost regimen to immunize Chinese rhesus macaques. The animals were challenged intramuscularly with pathogenic molecularly cloned SHIV-KB9. Immunogenicity and protective efficacy of vaccines were investigated by measuring IFN-γ levels, monitoring HIV-specific binding antibodies, examining viral load, and analyzing CD4/CD8 ratio. Results show that, upon challenge, the vaccine group can induce a strong immune response in the body, represented by increased expression of IFN-γ, slow and steady elevated antibody production, reduced peak value of acute viral load, and increase in the average CD4/CD8 ratio. The current research suggests that rapid reaction speed, appropriate response strength, and long-lasting immune response time may be key protection factors for AIDS vaccine. The present study contributes significantly to AIDS vaccine and preclinical research.


Viruses | 2014

Anti-Tumor Effects of an Oncolytic Adenovirus Expressing Hemagglutinin-Neuraminidase of Newcastle Disease Virus in Vitro and in Vivo

Dongyun He; Lili Sun; Chang Li; Ningning Hu; Yuan Sheng; Zhifei Chen; Xiao Li; Baorong Chi; Ningyi Jin

Oncolytic virotherapy has been an attractive drug platform for targeted therapy of cancer over the past few years. Viral vectors can be used to target and lyse cancer cells, but achieving good efficacy and specificity with this treatment approach is a major challenge. Here, we assessed the ability of a novel dual-specific anti-tumor oncolytic adenovirus, expressing the hemagglutinin-neuraminidase (HN) gene from the Newcastle disease virus under the human telomerase reverse transcriptase (hTERT) promoter (Ad-hTERTp-E1a-HN), to inhibit esophageal cancer EC-109 cells in culture and to reduce tumor burden in xenografted BALB/c nude mice. In vitro, infection with Ad-hTERT-E1a-HN could inhibit the growth of EC-109 cells significantly and also protect normal human liver cell line L02 from growth suppression in 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays. Ad-hTERT-E1a-HN also effectively and selectively decreased the sialic acid level on EC-109 cells, but not on L02 cells. Furthermore, Ad-hTERT-E1a-HN was shown to induce the apoptosis pathway via acridine orange and ethidium bromide staining (AO/EB staining), increase reactive oxygen species (ROS), reduce mitochondrial membrane potential and release cytochrome c. In vivo, xenografted BALB/c nude mice were treated via intratumoral or intravenous injections of Ad-hTERT-E1a-HN. Although both treatments showed an obvious suppression in tumor volume, only Ad-hTERT-E1a-HN delivered via intratumoral injection elicited a complete response to treatment. These results reinforced previous findings and highlighted the potential therapeutic application of Ad-hTERT-E1a-HN for treatment of esophageal cancer in clinical trials.


Journal of Virological Methods | 2012

Construction and evaluation of a new triple-gene expression cassette vaccinia virus shuttle vector

Shouwen Du; Chang Li; Yuhang Wang; Cunxia Liu; Dayong Ren; Yi Li; Yanqing Qin; Maopeng Wang; Dandan Sun; Na Zhu; Ningyi Jin

A vaccinia virus shuttle vector pSTKE with a triple-gene expression cassette was designed, and the derived recombinant virus could express at least three different target genes. A vaccinia virus and its mutant as the original viruses and EGFP as the reporter gene were used to verify the three expression cassettes. Two recombinant viruses containing EGFP were obtained by homologous recombination and plaque screening. The expression and genetic stability of the recombinant virus and foreign genes were analyzed using PCR, real-time PCR, and Western blot. And then EGFP, RFP and BFP were inserted into MCS1, MCS2 and MCS3 of pSTKE respectively, resulting in the generation of recombinant expressing three fluorescent proteins mentioned above, and the recombinant was continuously passaged 20 times. The results showed that EGFP, RFP and BFP were highly expressed in vaccinia virus, and no interaction between the three expression cassettes was observed. Recombinant viruses were stable genetically. The shuttle vector pSTKE can be used for efficient and stable gene expression to address problems in recombinant vaccinia viruses, such as low expression efficiency, limited number of inserted genes. In addition, this study provides a solid foundation for the development of a new genetically engineered vaccine.


Antiviral Research | 2012

Characterization of an attenuated TE3L-deficient vaccinia virus Tian Tan strain

Yuhang Wang; Shifu Kan; Shouwen Du; Yanxin Qi; Jinhui Wang; Liming Liu; Huifan Ji; Dongyun He; Na Wu; Chang Li; Baorong Chi; Xiao Li; Ningyi Jin

An attenuated vaccinia virus (VACV), TE3L(-)VTT, was evaluated for virulence and safety to determine its potential use as a vaccine or as a recombinant virus vector to express foreign genes. The virulence of TE3L(-)VTT was compared with that of the wild-type VTT both in vivo and in vitro. The humoral and cellular immune responses were detected in a mouse model to test the vaccine efficacy of the TE3L mutant. The results suggested that deletion of the TE3L gene decreased the virulence and neurovirulence significantly in mice and rabbit models, yet retained the immunogenicity. Thus, the deletion of TE3L improved the safety of the VTT vector; this approach may yield a valuable resource for studies of recombinant VACV-vectored vaccines.


Virology Journal | 2010

Protection against H1N1 influenza challenge by a DNA vaccine expressing H3/H1 subtype hemagglutinin combined with MHC class II-restricted epitopes.

Lei Tan; Huijun Lu; Dan Zhang; Mingyao Tian; Bo Hu; Zhuoyue Wang; Ningyi Jin

BackgroundMultiple subtypes of avian influenza viruses have crossed the species barrier to infect humans and have the potential to cause a pandemic. Therefore, new influenza vaccines to prevent the co-existence of multiple subtypes within a host and cross-species transmission of influenza are urgently needed.MethodsHere we report a multi-epitope DNA vaccine targeted towards multiple subtypes of the influenza virus. The protective hemagglutinin (HA) antigens from H5/H7/H9 subtypes were screened for MHC II class-restricted epitopes overlapping with predicted B cell epitopes. We then constructed a DNA plasmid vaccine, pV-H3-EHA-H1, based on HA antigens from human influenza H3/H1 subtypes combined with the H5/H7/H9 subtype Th/B epitope box.ResultsEpitope-specific IFN-γ ELISpot responses were significantly higher in the multi-epitope DNA group than in other vaccine and control groups (P < 0.05). The multi-epitope group significantly enhanced Th2 cell responses as determined by cytokine assays. The survival rate of mice given the multi-epitope vaccine was the highest among the vaccine groups, but it was not significantly different compared to those given single antigen expressing pV-H1HA1 vaccine and dual antigen expressing pV-H3-H1 vaccine (P > 0.05). No measurable virus titers were detected in the lungs of the multi-epitope immunized group. The unique multi-epitope DNA vaccine enhanced virus-specific antibody and cellular immunity as well as conferred complete protection against lethal challenge with A/New Caledonia/20/99 (H1N1) influenza strain in mice.ConclusionsThis approach may be a promising strategy for developing a universal influenza vaccine to prevent multiple subtypes of influenza virus and to induce long-term protective immune against cross-species transmission.


Clinical and Experimental Immunology | 2005

Engineering and functional evaluation of a single-chain antibody against HIV-1 external glycoprotein gp120.

H. W. Wang; David K. Cole; Wenzheng Jiang; H. T. Jin; N. Fu; Z. L. Chen; Ningyi Jin

The HIV‐1 envelope glycoprotein surface subunit gp120 is an attractive target for molecular intervention. This is because anti‐HIV‐1 gp120 neutralizing antibodies display the potential ability to inhibit HIV‐1 infection. The present investigation describes the construction of a genetically engineered single chain antibody (scFv102) against HIV‐1 gp120, its expression and functional evaluation. The parental hybridoma cell line (102) produces an immunoglobulin directed against the conserved CD4‐binding region of gp120. cDNAs encoding the variable regions of the heavy (VH) and light (VL) chains were prepared by reverse transcription PCR and linked together with an oligonucleotide encoding a linker peptide (Gly4Ser)3 to produce a single chain antibody gene. The resulting DNA construct was cloned into a prokaryotic expression vector (pET28) and recombinant scFv102 was expressed in Eserichia coli as an insoluble protein. The denatured scFv102 was refolded and purified by immobilized metal ion affinity chromatography. Purified scFv102 had the same specificity as the intact IgG in immuno‐blotting assays and immuno‐fluorescence (IF) detection, but ELISA analyses demonstrated the affinity of scFv102 to be 5‐fold lower than that of the parental monoclonal antibody. In neutralization assays, scFv102 at concentrations lower than 40 µg/ml exhibited efficient interference with viral replication and inhibition of viral infection (90%) across a range of primary isolates of subtype B HIV‐1. These results suggest that the constructed anti‐HIV‐1 gp120 scFv102 has good biological activity and can potentially be used for in vitro diagnostic and in vivo therapeutic applications.


Toxicology and Applied Pharmacology | 2014

Preclinical pharmacology and toxicology study of Ad-hTERT-E1a-Apoptin, a novel dual cancer-specific oncolytic adenovirus.

Yanxin Qi; Huanhuan Guo; Ningning Hu; Dongyun He; Shi Zhang; Yunjie Chu; Yubin Huang; Xiao Li; LiLi Sun; Ningyi Jin

Clinical studies have demonstrated that conditionally replicating adenovirus is safe. We constructed an oncolytic adenovirus, Ad-hTERT-E1a-Apoptin, using a cancer-specific promoter (human telomerase reverse transcriptase promoter, hTERTp) and a cancer cell-selective apoptosis-inducing gene (Apoptin). Ad-hTERT-E1a-Apoptin was proven effective both in vitro and in vivo in our previous study. In this study, the preclinical safety profiles of Ad-hTERT-E1a-Apoptin in animal models were investigated. At doses of 5.0×10(8), 2.5×10(9), and 1.25×10(10) viral particles (VP)/kg, Ad-hTERT-E1a-Apoptin had no adverse effects on mouse behavior, muscle cooperation, sedative effect, digestive system, and nervous systems, or on beagle cardiovascular and respiratory systems at 5.0×10(8), 2.5×10(9), and 1.25×10(10) VP/kg doses. In acute toxicity tests in mice, the maximum tolerated dose>5×10(10) VP/kg. There was no inflammation or ulceration at the injection sites within two weeks. In repeat-dose toxicological studies, the no observable adverse effect levels of Ad-hTERT-E1a-Apoptin in rats (1.25×10(10) VP/kg) and beagles (2.5×10(9) VP/kg) were 62.5- and 12.5-fold of the proposed clinical dose, respectively. The anti-virus antibody was produced in animal sera. Bone marrow examination revealed no histopathological changes. Guinea pigs sensitized by three repeated intraperitoneal injections of 1.35×10(10) VP/mL Ad-hTERT-E1a-Apoptin each and challenged by one intravenous injection of 1.67×10(8) VP/kg Ad-hTERT-E1a-Apoptin did not exhibit any sign of systemic anaphylaxis. Our data from different animal models suggest that Ad-hTERT-E1a-Apoptin is a safe anti-tumor therapeutic agent.

Collaboration


Dive into the Ningyi Jin's collaboration.

Top Co-Authors

Avatar

Chang Li

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huijun Lu

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Kuoshi Jin

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mingyao Tian

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cunxia Liu

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Hongtao Jin

Academy of Military Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge