Network


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

Hotspot


Dive into the research topics where Zongqiang Lai is active.

Publication


Featured researches published by Zongqiang Lai.


Nanoscale Research Letters | 2016

Aptamer-Functionalized Fluorescent Silica Nanoparticles for Highly Sensitive Detection of Leukemia Cells.

Juntao Tan; Nuo Yang; Zixi Hu; Jing Su; Jian-Hong Zhong; Yang Yang; Yating Yu; Jianmeng Zhu; Dabin Xue; Yingying Huang; Zongqiang Lai; Yong Huang; Xiaoling Lu; Yongxiang Zhao

A simple, highly sensitive method to detect leukemia cells has been developed based on aptamer-modified fluorescent silica nanoparticles (FSNPs). In this strategy, the amine-labeled Sgc8 aptamer was conjugated to carboxyl-modified FSNPs via amide coupling between amino and carboxyl groups. Sensitivity and specificity of Sgc8-FSNPs were assessed using flow cytometry and fluorescence microscopy. These results showed that Sgc8-FSNPs detected leukemia cells with high sensitivity and specificity. Aptamer-modified FSNPs hold promise for sensitive and specific detection of leukemia cells. Changing the aptamer may allow the FSNPs to detect other types of cancer cells.


Oncology Reports | 2017

An ‘activatable’ aptamer-based fluorescence probe for the detection of HepG2 cells

Zongqiang Lai; Juntao Tan; Ruirong Wan; Jie Tan; Zhenghua Zhang; Zixi Hu; Jieping Li; Wei Yang; Yiwei Wang; Yafeng Jiang; Jian He; Nuo Yang; Xiaoling Lu; Yongxiang Zhao

It is significant to develop a probe with sensitivity and specificity for the detection of cancer cells. The present study aimed to develop an ‘activatable’ aptamer-based fluorescence probe (AAFP) to detect cancer cells and frozen cancer tissue. This AAFP consisted of two fragments: aptamer TLS11a that targets HepG2 cells, and two short extending complementary DNA sequences with a 5′- and 3′-terminus that make the aptamer in hairpin structure a capable quencher to fluorophore. The ability of the AAFP to bind specifically to cancer cells was assessed using flow cytometry, fluorescence spectroscopy and fluorescence microscopy. Its ability to bind to frozen cancer tissue was assessed using fluorescence microscopy. As a result, in the absence of cancer cells, AAFP showed minimal fluorescence, reflecting auto-quenching. In the presence of cancer cells, however, AAFP showed a strong fluorescent signal. Therefore, this AAFP may be a promising tool for sensitive and specific detection of cancer.


Nanoscale Research Letters | 2017

Aptamer Combined with Fluorescent Silica Nanoparticles for Detection of Hepatoma Cells

Zixi Hu; Juntao Tan; Zongqiang Lai; Rong Zheng; Jian-Hong Zhong; Yiwei Wang; Xiaoxue Li; Nuo Yang; Jieping Li; Wei Yang; Yong Huang; Yongxiang Zhao; Xiaoling Lu

PurposeThe purpose of this study is to develop a simple, effective method to label hepatoma cells with aptamers and then detect them using fluorescent silica nanoparticles (FSNPs).MethodStreptavidin was conjugated to carboxyl-modified fluorescein isothiocyanate (FITC)-doped silica nanoparticles which were prepared by the reverse microemulsion method. The resulting streptavidin-conjugated fluorescent silica nanoparticles (SA-FSNPs) were mixed with hepatoma cells that had been labeled with biotin-conjugated aptamer TLS11a (Bio-TLS11a). The specificity and sensitivity of the nanoprobes were assessed using flow cytometry and fluorescence microscopy. Their toxicity was assessed in normal human liver cell cultures using the MTT assay, as well as in nude mice using immunohistochemistry.ResultsSA-FSNPs showed uniform size and shape, and fluorescence properties of them was similar to the free FITC dye. SA-FSNPs were able to detect aptamer-labeled hepatoma cells with excellent specificity and good sensitivity, and they emitted strong, photobleach-resistant fluorescent signal. SA-FSNPs showed no significant toxic effects in vitro or in vivo.ConclusionThe combination of biotin-conjugated aptamers and SA-FSNPs shows promise for sensitive detection of hepatoma cells, and potentially of other tumor cell types as well.


Journal of Nanoscience and Nanotechnology | 2018

Graphene-Based Multifunctional Nanomaterials in Cancer Detection and Therapeutics

Wenlin Gong; Zixi Hu; Ying Liang; Yiwei Wang; Rong Zheng; Juntao Tan; Zongqiang Lai; Xiaoxue Li; Jieping Li; Xiaoling Lu; Yongxiang Zhao

Nanotechnology for early diagnosis and treatment of malignant tumor is a forefront topic in the international field of biotechnology and medicine. In order to improve the effect of cancer therapy, the timely and accurate detection of the cancer is important and necessary. Graphene and its derivatives have various excellent characteristics. For example, biological sensors based on graphene are good at amplifying detection signals, and its derivatives play an important role in the early diagnosis and cancer therapy. In view of this, we discussed the biological sensor application based on graphene and its derivatives in the detection and therapy of cancer.


Oncology Reports | 2017

Screening and antitumor effect of an anti‑CTLA‑4 nanobody

Ruirong Wan; Aiqun Liu; Xiaoqiong Hou; Zongqiang Lai; Jieping Li; Nuo Yang; Juntao Tan; Fengzhen Mo; Zixi Hu; Xiaomei Yang; Yongxiang Zhao; Xiaoling Lu

Cytotoxic T-lymphocyte antigen-4 (CTLA-4) is a critical negative regulator of immune responses. CTLA-4 is rapidly upregulated following T-cell activation, and then binds to B7 molecules with a higher affinity than CD28. CTLA-4 may abolish the initiation of the responses of T cells by raising the threshold of signals required for full activation of T cells, and it also may terminate ongoing T-cell responses. This regulatory role has led to the development of monoclonal antibodies (mAbs) designed to block CTLA-4 activity for enhancing immune responses against cancer. mAbs have several disadvantages including high production cost and unstable behavior. Nanobodies (Nbs) are single-domain antigen-binding fragments derived from the camelid heavy-chain antibodies, which are highly attractive in cancer immunotherapy due to their small size, high specificity, and stability. We selected CTLA-4-specific Nbs from a high quality dromedary camel immune library by phage display technology. Four positive colonies were sequenced and classified based on the amino acids sequences in the CDR3 region. These Nbs recognized unique epitopes on CTLA-4 and displayed high binding rates when used on PHA-stimulated human T cells. Treatment of B16 melanoma-bearing C57BL/6 mice with anti-CTLA-4 nanobody 16 (Nb16) delayed melanoma growth and prolonged the survival time of mice. These data indicate that anti-CTLA-4 Nbs selected from a high quality phage display library may be effective for the treatment of patients with tumors.


Nanoscale Research Letters | 2018

Correction to: A Graphene Oxide-Based Fluorescent Aptasensor for the Turn-on Detection of CCRF-CEM

Jie Tan; Zongqiang Lai; Liping Zhong; Zhenghua Zhang; Rong Zheng; Jing Su; Yong Huang; Panpan Huang; Hui Song; Nuo Yang; Sufang Zhou; Yongxiang Zhao

In the original publication of this article [1] the author Liping Zhong was omitted. In this correction article the author and the corresponding details are provided. The publisher apologizes to the readers and authors for the inconvenience.


Journal of Nanoscience and Nanotechnology | 2018

Recent Progress of Wnt Pathway Inhibitor Dickkopf-1 in Liver Cancer

Jieping Li; Wenlin Gong; Xiaoxue Li; Ruirong Wan; Fengzhen Mo; Zhenghua Zhang; Panpan Huang; Zixi Hu; Zongqiang Lai; Xiaoling Lu; Yongxiang Zhao

Hepatocellular carcinoma (HCC) is one of the most common cancers around the world. Multiple etiologic factors such as virus and environment can lead to HCC. It is a challenge for us to successfully detect early HCC due to the lack of effective characterized and specific biomarkers. However, if the early diagnosis is successfully realized, it provides crucial chance for HCC patients to receive effective treatment as early as possible. Dickkopf-1 (DKK-1) is a secretary glycoprotein, which negatively regulates Wnt pathway through binding to surface receptors LRP5/6 and Kremen 1/2. The expression of DKK-1 is regulated by p53, V-Myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN), β-catenin, etc. Ectopic expression of DKK-1 can inhibit cell proliferation, or induce apoptosis with apoptosis enhancing factors. DKK-1 is low-expressed in many tumors, but overexpressed in others. Growing evidences show that DKK-1 plays complex and different roles in tumorigenesis, tumor progression and metastasis of different cancers. We herein review the recent progress in the expression and function of DKK-1 in hepatocellular carcinoma.


Journal of Biomedical Nanotechnology | 2018

TLS11a Aptamer/CD3 Antibody Anti-Tumor System for Liver Cancer

Zixi Hu; Jian He; Wenlin Gong; Nuo Zhou; Sufang Zhou; Zongqiang Lai; Rong Zheng; Yiwei Wang; Xiaomei Yang; Wei Yang; Liping Zhong; Xiaoling Lu; Yongxiang Zhao

New therapeutic approaches are needed for hepatocellular carcinoma (HCC), which is the most common primary malignancy of the liver. Bispecific T-cell engagers (BiTE) can effectively redirect T cells against tumors and show a strong anti-tumor effect. However, the potential immunogenicity, complexity, and high cost significantly limit their clinical application. In this paper, we used the hepatoma cells-specific aptamer TLS11a and anti-CD3 for to establish an aptamer/antibody bispecific system (AAbs), TLS11a/CD3, which showed advantages over BiTE and can specifically redirect T cells to lyse tumor cells. TLS11a-SH and anti-CD3-NH2 were crosslinked with sulfosuccinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (sulfo-SMCC). T cell activation, proliferation, and cytotoxicity of TLS11a/CD3 were analyzed by flow cytometry. Cytokine array was used to detect cytokine released from activated T cells. Hepatoma xenograft model was used to monitor the tumor volume and survival. TLS11a/CD3 could specifically bind hepatoma cells (H22) and T cells, activated T cells to mediate antigen-specific lysis of H22 cells in vitro, and effectively inhibited the growth of implanted H22 tumors as well as prolonged mice survival. TLS11a/CD3 could simultaneously target hepatoma cells and T cells, specifically guide T cells to kill tumor cells, and enhance the anti-tumor effect of T cells both in vitro and in vivo.


OncoImmunology | 2017

Collagen I enhances the efficiency and anti-tumor activity of dendritic-tumor fusion cells

Jian He; Rong Zheng; Zhenghua Zhang; Jie Tan; Chaofan Zhou; Guoqing Zhang; Xinglu Jiang; Qianyi Sun; Sufang Zhou; Duo Zheng; Yong Huang; Lige Wu; Zongqiang Lai; Jieping Li; Nuo Yang; Xiaoling Lu; Yongxiang Zhao

ABSTRACT Low fusion efficiency and nominal activity of fusion cells (FCs) restrict the clinical application of dendritic cell (DC)/tumor fusion cells. Collagen I (Col I) is an interstitial collagen with a closely-knit structure used to repair damaged cell membranes. This study evaluated whether Col I could improve the fusion efficiency of polyethylene glycol (PEG)-induction and enhance the immunogenicity of fusion vaccine. DC/B16 melanoma and controlled DC/H22 hepatoma cell fusions were induced by PEG with or without Col I. Col I/PEG treatment increased the levels of DC surface molecules and the secretion of lactate, pro- and anti-inflammatory cytokines in fusion cells. Col I/PEG-treated FCs enhanced T-cell proliferation and cytotoxic T lymphocyte activity. The Col I-prepared fusion vaccine obviously suppressed tumor growth and prolonged mice survival time. Thus Col I treatment could significantly improve the efficiency of PEG-induced DC/tumor fusion and enhance the anticancer activity of the fusion vaccine. This novel fusion strategy might promote the clinical application of DC/tumor fusion immunotherapy.


Theranostics | 2017

A new theranostic system based on endoglin aptamer conjugated fluorescent silica nanoparticles

Juntao Tan; Nuo Yang; Liping Zhong; Jie Tan; Zixi Hu; Qing Zhao; Wenlin Gong; Zhenghua Zhang; Rong Zheng; Zongqiang Lai; Yanmei Li; Chaofan Zhou; Guoqing Zhang; Duo Zheng; Ying Zhang; Siyu Wu; Xinglu Jiang; Jian-Hong Zhong; Yong Huang; Sufang Zhou; Yongxiang Zhao

Collaboration


Dive into the Zongqiang Lai's collaboration.

Top Co-Authors

Avatar

Yongxiang Zhao

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Nuo Yang

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaoling Lu

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Zixi Hu

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Rong Zheng

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Jieping Li

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Juntao Tan

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Yong Huang

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhenghua Zhang

Guangxi Medical University

View shared research outputs
Top Co-Authors

Avatar

Jie Tan

Guangxi Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge