Network


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

Hotspot


Dive into the research topics where Qinghui Jin is active.

Publication


Featured researches published by Qinghui Jin.


Biosensors and Bioelectronics | 2010

QDs-DNA nanosensor for the detection of hepatitis B virus DNA and the single-base mutants

Xiang Wang; Xinhui Lou; Yi Wang; Qingchuan Guo; Zheng Fang; Xinhua Zhong; Hongju Mao; Qinghui Jin; Lei Wu; Hui Zhao; Jianlong Zhao

We report here a quantum dots-DNA (QDs-DNA) nanosensor based on fluorescence resonance energy transfer (FRET) for the detection of the target DNA and single mismatch in hepatitis B virus (HBV) gene. The proposed one-pot DNA detection method is simple, rapid and efficient due to the elimination of the washing and separation steps. In this study, the water-soluble CdSe/ZnS QDs were prepared by replacing the trioctylphosphine oxide (TOPO) on the surface of QDs with 3-mercaptopropionic acid (MPA). Subsequently, oligonucleotides were attached to the QDs surface to form functional QDs-DNA conjugates. Along with the addition of DNA targets and Cy5-modified signal DNAs into the QDs-DNA conjugates, sandwiched hybrids were formed. The resulting assembly brings the Cy5 fluorophore, the acceptor, and the QDs, the donor, into proximity, leading to fluorescence emission from the acceptor by means of FRET on illumination of the donor. In order to efficiently detect single-base mutants in HBV gene, oligonucleotide ligation assay was employed. If there existed a single-base mismatch, which could be recognized by the ligase, the detection probe was not ligated and no Cy5 emission was produced due to the lack of FRET. The feasibility of the proposed method was also demonstrated in the detection of synthetic 30-mer oliginucleotide targets derived from the HBV with a sensitivity of 4.0nM by using a multilabel counter. The method enables a simple and efficient detection that could be potentially used for high throughput and multiplex detections of target DNA and the mutants.


Biosensors and Bioelectronics | 2009

Nano-ELISA for highly sensitive protein detection.

Chun Ping Jia; Xiao Qin Zhong; Bao Hua; Mei Ying Liu; Feng Xiang Jing; Xin Hui Lou; Shi Hua Yao; Jia Qing Xiang; Qinghui Jin; Jian Long Zhao

Highly sensitive protein detection method based on nanoparticles and enzyme-linked immunosorbent assays (ELISAs), named Nano-ELISA, was introduced. In this method, the micro-magnetic beads were modified with monoclonal antibody of the target protein p53. Gold nanoparticles (AuNPs) were modified with another monoclonal detector antibody and Horseradish peroxidase (HRP, for signal amplification). The presence of target protein p53 causes the formation of the sandwich structures (magnetic beads-target protein-AuNP probes) through the interaction between the antibodies and the antigen p53. The HRP at the surface of AuNPs catalytically oxidize the substrate and generate optical signals that reflected the quantity of the target protein. Down to 5 pg mL(-1) of protein was detected in less than 2 h with this method. The detection sensitivity of p53 classic ELISA kit is 0.125 ng mL(-1). This method is as simple as ELISA and has higher sensitivity than ELISA, which can potentially be exploited in clinic. This method can be used to detect protein markers of tumors, nervous system or other diseases for early diagnostics.


Lab on a Chip | 2009

Integrated microfluidic chip for endothelial cells culture and analysis exposed to a pulsatile and oscillatory shear stress

Jianbo Shao; Lei Wu; Jianzhang Wu; Yunhuan Zheng; Hui Zhao; Qinghui Jin; Jianlong Zhao

For a comprehensive understanding of cells or tissues, it is important to enable multiple studies under the controllable microenvironment of a chip. In this report, we present an integrated microfluidic cell culture platform in which endothelial cells (ECs) are under static conditions or exposed to a pulsatile and oscillatory shear stress. Through the integration of a microgap, self-contained flow loop, pneumatic pumps, and valves, the novel microfluidic chip achieved multiple functions: pulsatile and oscillatory fluid circulation, cell trapping, cell culture, the formation of ECs barrier, and adding shear stress on cells. After being introduced into the chip by gravity, the ECs arranged along the microgap with the help of hydrodynamic forces and grew in the microchannel for more than 7 days. The cells proliferated and migrated to form a barrier at the microgap to mimic the vessel wall, which separated the microenvironment into two compartments, microchannel and microchamber. An optimized pneumatic micropump was embedded to actuate flow circulation in a self-contained loop that induced a pulsatile and oscillatory shear stress at physiological levels on the ECs in the microchannel. All the analyses were performed under either static or dynamic conditions. The performance of the barrier was evaluated by the diffusion and distribution behaviors of fluorescently labeled albumin. The permeability of the barrier was comparable to that in traditional in vitro assays. The concentration gradients of the tracer formed in the microchamber can potentially be used to study cell polarization, migration and communications in the future. Additionally, the morphology and cytoskeleton of the ECs response to the pulsatile and oscillatory shear stress were analyzed. The microfluidic chip provided a multifunctional platform to enable comprehensive studies of blood vessels at the cell or tissue level.


Biosensors and Bioelectronics | 2014

Multi-nanomaterial electrochemical biosensor based on label-free graphene for detecting cancer biomarkers

Bing Jin; Ping Wang; Hongju Mao; Bing Hu; Honglian Zhang; Zule Cheng; Zhenhua Wu; Xiaojun Bian; Chunping Jia; Fengxiang Jing; Qinghui Jin; Jianlong Zhao

Developing a rapid, accurate and sensitive electrochemical biosensor for detecting cancer biomarkers is important for early detection and diagnosis. This work reports an electrochemical biosensor based on a graphene (GR) platform which is made by CVD, combined with magnetic beads (MBs) and enzyme-labeled antibody-gold nanoparticle bioconjugate. MBs coated with capture antibodies (Ab1) were attached to GR sheets by an external magnetic field, to avoid reducing the conductivity of graphene. Sensitivity was also enhanced by modifying the gold nanoparticles (AuNPs) with horseradish peroxidase (HRP) and the detection antibody (Ab2), to form the conjugate Ab2-AuNPs-HRP. Electron transport between the electrode and analyte target was accelerated by the multi-nanomaterial, and the limit of detection (LOD) for carcinoembryonic antigen (CEA) reached 5 ng mL(-1). The multi-nanomaterial electrode GR/MBs-Ab1/CEA/Ab2-AuNPs-HRP can be used to detect biomolecules such as CEA. The EC biosensor is sensitive and specific, and has potential in the detection of disease markers.


Biosensors and Bioelectronics | 2014

Highly sensitive enumeration of circulating tumor cells in lung cancer patients using a size-based filtration microfluidic chip

Ting Huang; Chun Ping Jia; Jun-Yang; Wen Jie Sun; Wen Tao Wang; Hong lian Zhang; Hui Cong; Feng Xiang Jing; Hong Ju Mao; Qinghui Jin; Zhen Zhang; Ying Jie Chen; Gang Li; Guo Xin Mao; Jian Long Zhao

Circulating tumor cells (CTCs) in the peripheral blood could serve as a surrogate marker for the diagnosis of cancer metastasis and for therapeutic evaluation. However, the separation and characterization of CTCs is technically challenging owing to the extremely low number of CTCs present. Here we developed a size-based and high-throughput microfluidic chip, which exploits filtration microchannels to isolate the relatively larger CTCs from the rest of the blood constituents. High isolation efficiency of our microfluidic chip was demonstrated with three lung cancer cell lines spiked in blood samples at an optimal flow rate of 0.4 mL/h. The average recovery rates of 96%, 95% and 92% were obtained for A549, SK-MES-1, and H446, respectively. To clinically validate the chip, we also employed it to isolate CTCs from 59 lung cancer patients. CTCs were detected in 96.7% of patients with the mean number of 18.6 cells/mL, which was significantly higher than normal controls (P<0.05). The work here indicates that the size-based microfluidic platform with the advantage of capturing tumor cells without reliance on cell surface expression markers can provide a novel, inexpensive and effective tool for CTC detection and evaluation of cancer status.


Biosensors and Bioelectronics | 2015

A microfluidic chip integrated with a high-density PDMS-based microfiltration membrane for rapid isolation and detection of circulating tumor cells.

Xiaoyun Fan; Chunping Jia; Jun Yang; Gang Li; Hongju Mao; Qinghui Jin; Jianlong Zhao

Isolation of circulating tumor cells (CTCs) by size exclusion is a widely researched technique that offers the advantage of capturing tumor cells without reliance on cell surface expression markers. In this work, we report the development of a novel polydimethylsiloxane (PDMS) membrane filter-based microdevice for rapid and highly efficient isolation of CTCs from peripheral blood. A precise and highly porous PDMS microfilter was fabricated and integrated into the microfiltration chip by combining a sacrificial transferring film with a sandwich molding method. We achieved >90% recovery when isolating lung cancer cells from spiked blood samples, with a relatively high processing throughput of 10 mL/h. In contrast to existing CTC filtration systems, which rely on low-porosity track-etch filters or expensive lithography-based filters, our microfiltration chip does not require complex e-beam lithography or the reactive ion etching process, therefore it offers a low-cost alternative tool for highly efficient CTC enrichment and in situ analysis. Thus, this new microdevice has the potential for use in routine monitoring of cancer development and cancer therapy in a clinical setting.


Biosensors and Bioelectronics | 2015

A microfluidic droplet digital PCR for simultaneous detection of pathogenic Escherichia coli O157 and Listeria monocytogenes

Xiaojun Bian; Fengxiang Jing; Gang Li; Xiaoyun Fan; Chunping Jia; Hongbo Zhou; Qinghui Jin; Jianlong Zhao

Sensitive and rapid identification of pathogenic bacterial is extremely important due to the serious threat of pathogens to human health. In this study, we demonstrate the simultaneous and sensitive detection of pathogenic Escherichia coli O157 and Listeria monocytogenes using a novel duplex droplet digital PCR (ddPCR) platform. The ddPCR platform, which uses a mineral oil-saturated polydimethylsiloxane (OSP) chip to overcome the problem of droplet evaporation, integrates the functions of droplet generation, on-chip amplification and end-point fluorescence readout. Simultaneous detection of two kinds of bacterial is achieved by the design of differentially labeled TaqMan-MGB fluorescent probes. Compared with a quantitative real-time PCR approach, the OSP chip-based duplex ddPCR platform exhibits high sensitivity, which is at the level of single molecule resolution without significant cross-assay interference. Moreover, the applicability of the proposed method is also evaluated in artificially contaminated drinking water sample, which displays a low detection limit down to 10 CFU/mL for both pathogenic bacterial within 2 h.


Biosensors and Bioelectronics | 2015

Absolute quantification of lung cancer related microRNA by droplet digital PCR.

Ping Wang; Fengxiang Jing; Gang Li; Zhenhua Wu; Zule Cheng; Jishen Zhang; Honglian Zhang; Chunping Jia; Qinghui Jin; Hongju Mao; Jianlong Zhao

Digital polymerase chain reaction (digital PCR) enables the absolute quantification of nucleic acids through the counting of single molecules, thus eliminating the need for standard curves or endogenous controls. In this study, we developed a droplet digital PCR (ddPCR) system based on an oil saturated PDMS (OSP) microfluidic chip platform for quantification of lung cancer related microRNA (miRNA). The OSP chip was made with PDMS and was oil saturated to constrain oil swallow and maintain the stability of droplets. Two inlets were designed for oil and sample injection with a syringe pump at the outlet. Highly uniform monodisperse water-in-oil emulsion droplets to be used for subsequent detection and analysis were generated at the cross section of the channel. We compared miRNA quantification by the ddPCR system and quantitative real-time PCR (qPCR) to demonstrate that the ddPCR system was superior to qPCR both in its detection limit and smaller fold changes measurement. This droplet PCR system provides new possibilities for highly sensitive and efficient detection of cancer-related genes.


Clinical Chemistry | 2011

Simultaneous Detection of High-Sensitivity Cardiac Troponin I and Myoglobin by Modified Sandwich Lateral Flow Immunoassay: Proof of Principle

Jimin Zhu; Nengli Zou; Danian Zhu; Jin Wang; Qinghui Jin; Jianlong Zhao; Hongju Mao

BACKGROUND Although numerous lateral flow immunoassays (LFIAs) have been developed and widely used, inadequate analytical sensitivity and the lack of multiple protein detection applications have limited their clinical utility. We developed a new LFIA device for the simultaneous detection of high-sensitivity cardiac troponin I (hs-cTnI) and myoglobin (Myo). METHODS We used a gold nanoparticle (AuNP) doubly labeled complex, in which biotinylated single-stranded DNA was used as a linkage to integrate 2 AuNPs and streptavidin-labeled AuNP, as an amplifier to magnify extremely low signals. RESULTS The detection limit of 1 ng/L achieved for hs-cTnI was 1000 times lower than that obtained in a conventional LFIA. The detection limit for simultaneously measured Myo was 1 μg/L. The linear measurement ranges for hs-cTnI and Myo were 1-10 000 ng/L and 1-10 000 μg/L, respectively. We observed concordant results between the LFIA and clinical assays in sera from 12 patients with acute myocardial infarction (hs-cTnI r = 0.96; Myo r = 0.98). Assay imprecision was <11% for both hs-TnI and myo. CONCLUSIONS The described proof-of-principle LFIA method could be used as a point-of-care device in multiple protein quantification and semiquantitative analysis.


IEEE\/ASME Journal of Microelectromechanical Systems | 2007

A Rapid and Low-Cost Procedure for Fabrication of Glass Microfluidic Devices

Qiang Chen; Gang Li; Qinghui Jin; Jianlong Zhao; Qiu-Shi Ren; Yuan-Sen Xu

In this paper, we present a simple, rapid, and low-cost procedure for fabricating glass microfluidic chips. This procedure uses commercially available microscopic slides as substrates and a thin layer of AZ 4620 positive photoresist (PR) as an etch mask for fabricating glass microfluidic components, rather than using expensive quartz glasses or Pyrex glasses as substrates and depositing an expensive metal or polysilicon/amorphous silicon layer as etch masks in conventional method. A long hard-baking process is proposed to realize the durable PR mask capable of withstanding a long etching process. In order to remove precipitated particles generated during the etching process, a new recipe of buffered oxide etching with addition of 20% HCl is also reported. A smooth surface microchannel with a depth of more than 110 mum is achieved after 2 h of etching. Meanwhile, a simple, fast, but reliable bonding process based on UV-curable glue has been developed which takes only 10 min to accomplish the efficient sealing of glass chips. The result shows that a high bonding yield (~ 100%) can be easily achieved without the requirement of clean room facilities and programmed high-temperature furnaces. The presented simple fabrication process is suitable for fast prototyping and manufacturing disposable microfluidic devices.

Collaboration


Dive into the Qinghui Jin's collaboration.

Top Co-Authors

Avatar

Jianlong Zhao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Hongju Mao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

B. Xu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Gang Li

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Chunping Jia

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Honglian Zhang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Fengxiang Jing

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Ping Wang

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge