Network


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

Hotspot


Dive into the research topics where Chunping Jia is active.

Publication


Featured researches published by Chunping Jia.


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 | 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 Lung Cancer | 2015

Early Detection of Lung Cancer in Serum by a Panel of MicroRNA Biomarkers

Ping Wang; Dawei Yang; Honglian Zhang; Xuyu Wei; Tianle Ma; Zule Cheng; Qunying Hong; Jie Hu; Hanjing Zhuo; Yuanlin Song; Chunping Jia; Fengxiang Jing; Qinghui Jin; Chunxue Bai; Hongju Mao; Jianlong Zhao

INTRODUCTION The objective of the study was to develop a panel of microRNAs (miRNAs) as highly sensitive and specific biomarkers for lung cancer early detection. MATERIALS AND METHODS The study contained 2 phases: first, preliminary marker selection based on previous reports on the serum of 24 early stage lung cancer patients and 24 healthy control subjects by TaqMan probe-based real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR); and second, validation of miRNA markers on 94 early stage lung cancer, 48 stage III to IV lung cancer, and 111 healthy control serum samples. RESULTS A total of 3 miRNAs (miR-125a-5p, miR-25, and miR-126) were selected for further analysis in this study. The combination of the 3 miRNAs could produce 0.936 area under the receiver operating characteristic curve value in distinguishing early stage lung cancer patients from control subjects with 87.5% sensitivity and 87.5% specificity, respectively. The diagnostic value of the miRNA panel in an independent set of lung cancer patients confirmed the sensitivity and specificity. CONCLUSION The results demonstrated that the panel of miRNA biomarkers had the potential for the early detection of lung cancer.


Biosensors and Bioelectronics | 2016

Bead-based microarray immunoassay for lung cancer biomarkers using quantum dots as labels.

Lifen Liu; Simin Wu; Fengxiang Jing; Hongbo Zhou; Chunping Jia; Gang Li; Hui Cong; Qinghui Jin; Jianlong Zhao

In this study, we developed a multiplex immunoassay system that combines the suspension and planar microarray formats within a single layer of polydimethylsiloxane (PDMS) using soft lithography technology. The suspension format was based on the target proteins forming a sandwich structure between the magnetic beads and the quantum dot (QD) probes through specific antibody-antigen interactions. The planar microarray format was produced by fabricating an array of micro-wells in PDMS. Each micro-well was designed to trap a single microbead and eventually generated a microbead array within the PDMS chamber. The resultant bead-based on-chip assay could be used for simultaneously detecting three lung cancer biomarkers-carcinoembryonic antigen (CEA), fragments of cytokeratin 19 (CYFRA21-1) and neuron-specific enolase (NSE)-in 10 μl of human serum, with a wide linear dynamic range (1.03-111 ng/mL for CEA and CYFRA21-1; 9.26-1000 ng/ml for NSE) and a low detection limit (CEA: 0.19 ng/ml; CYFRA21-1: 0.97 ng/ml; NSE: 0.37 ng/ml; S/N=3). Our micro-well chip does not require complex e-beam lithography or the reactive ion etching process as with existing micro-well systems, which rely on expensive focused ion beam (FIB) milling or optical fiber bundles. Furthermore, the current approach is easy to operate without extra driving equipment such as pumps, and can make parallel detection for multiplexing with rapid binding kinetics, small reagent consumption and low cost. This work has demonstrated the importance of the successful application of on-chip multiplexing sandwich assays for the detection of biomarker proteins.


Talanta | 2016

Multiplexed detection of lung cancer biomarkers based on quantum dots and microbeads

Simin Wu; Lifen Liu; Gong Li; Fengxiang Jing; Hongju Mao; Qinghui Jin; Wanyin Zhai; Hongfeng Zhang; Jianlong Zhao; Chunping Jia

We have developed a multiplexed fluoroimmunoassay of three lung cancer biomarkers based on multicolor quantum dots (QDs) as detection elements and micro-magnetic beads as immune carriers. QDs have the ability to simplify multiplexed analysis. In our method, the fluorescent signals derived from three cross-talk-free QD conjugated probes with emission maxima at 525, 585 and 625nm could be analyzed to determine the concentrations of the target proteins. With this system, fragments of cytokeratin 19 (CYRFA 21-1), carcinoembryonic antigen (CEA), and neuron-specific enolase (NSE), were simultaneously detected in a single sample with a low detection limit down to the 1.0ng/mL level (364pg/mL for CYRFA 21-1, 38pg/mL for CEA, 370pg/mL for NSE in a single detection). Additional advantages of the presented method include ease of operation, low cost, and a very low sample volume (20µL).


Biosensors and Bioelectronics | 2017

Label-free graphene biosensor targeting cancer molecules based on non-covalent modification.

Lin Zhou; Hongju Mao; Chunyan Wu; Lin Tang; Zhenhua Wu; Hao Sun; Honglian Zhang; Hongbo Zhou; Chunping Jia; Qinghui Jin; Xianfeng F. Chen; Jianlong Zhao

A label-free immunosensor based on antibody-modified graphene field effect transistor (GFET) was presented. Antibodies targeting carcinoembryonic antigen (Anti-CEA) were immobilized to the graphene surface via non-covalent modification. The bifunctional molecule, 1-pyrenebutanoic acid succinimidyl ester, which is composed of a pyrene and a reactive succinimide ester group, interacts with graphene non-covalently via π-stacking. The succinimide ester group reacts with the amine group to initiate antibody surface immobilization, which was confirmed by X-ray Photoelectron Spectroscopy, Atomic Force Microscopy and Electrochemical Impedance Spectroscopy. The resulting anti-CEA modified GFET sufficiently monitored the reaction between CEA protein and anti-CEA in real-time with high specificity, which revealed selective electrical detection of CEA with a limit of detection (LOD) of less than 100pg/ml. The dissociation constant between CEA protein and anti-CEA was estimated to be 6.35×10-11M, indicating the high affinity and sensitivity of anti-CEA-GFET. Taken together, the graphene biosensors provide an effective tool for clinical application and point-of-care medical diagnostics.


PLOS ONE | 2013

High-Performance Size-Based Microdevice for the Detection Of Circulating Tumor Cells from Peripheral Blood in Rectal Cancer Patients

Wenjie Sun; Chunping Jia; Ting Huang; Weiqi Sheng; Guichao Li; Honglian Zhang; Fengxiang Jing; Qinghui Jin; Jianlong Zhao; Gang Li; Zhen Zhang

Since individualized therapy becomes more and more important in the treatment of rectal cancer, an accurate and effective approach should be established in the clinical settings to help physicians to make their decisions. Circulating tumor cells (CTCs), originated from either primary or metastatic cancer, could provide important information for diagnosis and monitoring of cancer. However, the implication and development of CTCs are limited due to the extreme rarity of these tumor cells. In this study we fabricated a simple and high-performance microfluidic device, which exploited numerous filtered microchannels in it to enrich the large-sized target tumor cells from whole blood. A very high CTC capture efficiency (average recovery rate: 94%) was obtained in this device at the optimum flow rate of 0.5 mL/h and channel height of 5 µm. Additionally, we used this device for detecting CTCs in 60 patients with rectal cancer. The CTC counts of rectal cancer patients were significantly higher than those in healthy subjects. Furthermore, the CTC counts detected by this device were significantly higher than those by EpCAM bead-based method for rectal cancer patients with various stage. Especially, for localized rectal cancer patients, the positive rates of samples with more than 3 CTCs per 5 mL blood by use of microdevice vs. EpCAM-based ones were 100% vs. 47%, respectively. Thus, this device provides a new and effective tool for accurate identification and measurement of CTCs in patients with rectal cancer, and has broad potential in clinical practice.


Talanta | 2016

Highly sensitive and selective lateral flow immunoassay based on magnetic nanoparticles for quantitative detection of carcinoembryonic antigen

Fangming Liu; Honglian Zhang; Zhenhua Wu; Haidao Dong; Lin Zhou; Dawei Yang; Yuqing Ge; Chunping Jia; Huiying Liu; Qinghui Jin; Jianlong Zhao; Qiqing Zhang; Hongju Mao

Carcinoembryonic antigen (CEA) is an important biomarker in cancer diagnosis. Here, we present an efficient, selective lateral-flow immunoassay (LFIA) based on magnetic nanoparticles (MNPs) for in situ sensitive and accurate point-of-care detection of CEA. Signal amplification mechanism involved linking of detection MNPs with signal MNPs through biotin-modified single-stranded DNA (ssDNA) and streptavidin. To verify the effectiveness of this modified LFIA system, the sensitivity and specificity were evaluated. Sensitivity evaluation showed a broad detection range of 0.25-1000ng/ml for CEA protein by the modified LFIA, and the limit of detection (LOD) of the modified LFIA was 0.25ng/ml, thus producing significant increase in detection threshold compared with the traditional LFIA. The modified LFIA could selectively recognize CEA in presence of several interfering proteins. In addition, this newly developed assay was applied for quantitative detection of CEA in human serum specimens collected from 10 randomly selected patients. The modified LFIA system detected minimum 0.27ng/ml of CEA concentration in serum samples. The results were consistent with the clinical data obtained using commercial electrochemiluminescence immunoassay (ECLIA) (p<0.01). In conclusion, the MNPs based LFIA system not only demonstrated enhanced signal to noise ratio, it also detected CEA with higher sensitivity and selectivity, and thus has great potential to be commercially applied as a sensitive tumor marker filtration system.

Collaboration


Dive into the Chunping Jia's collaboration.

Top Co-Authors

Avatar

Qinghui Jin

Chinese Academy of Sciences

View shared research outputs
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

Fengxiang Jing

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

Honglian Zhang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Hongbo Zhou

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhenhua Wu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge