Zhenhui Lu
Guangxi Medical University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Zhenhui Lu.
Materials Science and Engineering: C | 2018
Xingchen Yang; Zhenhui Lu; Huayu Wu; Wei Li; Li Zheng; Jinmin Zhao
Articular cartilage repair is still a huge challenge for researchers and clinicians. 3D bioprinting could be an innovative technology for cartilage tissue engineering. In this study, we used collagen type I (COL) or agarose (AG) mixed with sodium alginate (SA) to serve as 3D bioprinting bioinks and incorporated chondrocytes to construct in vitro 3D printed cartilage tissue. Swelling ratio, mechanical properties, scanning electron microscopy (SEM), cell viability and cytoskeleton, biochemistry analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were performed to investigate the function of different bioinks in 3D printing cartilage tissue engineering applications. The results showed that the mechanical strength was improved in both SA/COL and SA/AG groups compared to SA alone. Besides, the addition of COL or AG has little impact on gelling behavior, demonstrating the advantage as bioinks for 3D printing. Among the three scaffolds, SA/COL could distinctly facilitated cell adhesion, accelerated cell proliferation and enhanced the expression of cartilage specific genes such as Acan, Col2al and Sox9 than the other two groups. Lower expression of Col1a1, the fibrocartilage marker, was present in SA/COL group than that in both of SA and SA/AG groups. The results indicated that SA/COL effectively suppressed dedifferentiation of chondrocytes and preserved the phenotype. In summary, 3D bioprinted SA/COL with favorable mechanical strength and biological functionality is promising in cartilage tissue engineering.
Tohoku Journal of Experimental Medicine | 2015
Qin Liu; Zhenhui Lu; Huayu Wu; Li Zheng
Articular cartilage is characterized by the lack of blood vessels and has a poor self-healing potential. Limited cell numbers and dedifferentiation of chondrocytes when expanded in vitro are the major obstacles of autologous chondrocyte implantation. Autologous chondrocyte implantation is a cell-based treatment that can be used as a second-line measure to regenerate chondral or osteochondral defects in younger, active patients. There is an urgent need to find an effective chondrogenic protection agent alleviating or inhibiting chondrocyte dedifferentiation. In this study, we explored the effect of taurine (2-aminoethane sulfonic acid) on proliferation and phenotype maintenance of human articular chondrocytes by analyzing the cell proliferation, morphology, viability, and expression of cartilage specific mRNAs and proteins. Primary chondrocytes were isolated from human articular cartilage tissues. Results showed that taurine effectively promoted chondrocyte growth and enhanced accumulation of glycosaminoglycans and collagens in the conditioned media of chondrocytes. Moreover, taurine exposure caused significant increases in the relative expression levels of mRNAs for cartilage specific markers, including aggrecan, collagen type II and SOX9. Aggrecan is a cartilage-specific proteoglycan, and SOX9 is a chondrogenic transcription factor. In contrast, the mRNA expression of collagen type I, a marker for chondrocyte dedifferentiation, was significantly decreased in cells treated with taurine, indicating that taurine inhibits the chondrocyte dedifferentiation. This study reveals that taurine is effective in proliferation promotion and phenotype maintenance of chondrocytes. Thus, taurine may be a useful pro-chondrogenic agent for autologous chondrocyte implantation in the treatment of cartilage repair.
Brazilian Journal of Medical and Biological Research | 2014
Guojie Xu; Zhenhui Lu; Xiao Lin; Cuiwu Lin; Li Zheng; Jinmin Zhao
Tissue engineering encapsulated cells such as chondrocytes in the carrier matrix have been widely used to repair cartilage defects. However, chondrocyte phenotype is easily lost when chondrocytes are expanded in vitro by a process defined as “dedifferentiation”. To ensure successful therapy, an effective pro-chondrogenic agent is necessary to overcome the obstacle of limited cell numbers in the restoration process, and dedifferentiation is a prerequisite. Gallic acid (GA) has been used in the treatment of arthritis, but its biocompatibility is inferior to that of other compounds. In this study, we modified GA by incorporating sulfamonomethoxine sodium and synthesized a sulfonamido-based gallate, JJYMD-C, and evaluated its effect on chondrocyte metabolism. Our results showed that JJYMD-C could effectively increase the levels of the collagen II, Sox9, and aggrecan genes, promote chondrocyte growth, and enhance secretion and synthesis of cartilage extracellular matrix. On the other hand, expression of the collagen I gene was effectively down-regulated, demonstrating inhibition of chondrocyte dedifferentiation by JJYMD-C. Hypertrophy, as a characteristic of chondrocyte ossification, was undetectable in the JJYMD-C groups. We used JJYMD-C at doses of 0.125, 0.25, and 0.5 µg/mL, and the strongest response was observed with 0.25 µg/mL. This study provides a basis for further studies on a novel agent in the treatment of articular cartilage defects.
Cell Death and Disease | 2017
Zhenhui Lu; Danqing Lei; Tongmeng Jiang; Lihui Yang; Li Zheng; Jinmin Zhao
Growth factors such as transforming growth factor beta1 (TGF-β1), have critical roles in the regulation of the chondrogenic differentiation of mesenchymal stem cells (MSCs), which promote cartilage repair. However, the clinical applications of the traditional growth factors are limited by their high cost, functional heterogeneity and unpredictable effects, such as cyst formation. It may be advantageous for cartilage regeneration to identify a low-cost substitute with greater chondral specificity and easy accessibility. As a neuropeptide, nerve growth factor (NGF) was involved in cartilage metabolism and NGF is hypothesized to mediate the chondrogenic differentiation of MSCs. We isolated NGF from Chinese cobra venom using a three-step procedure that we had improved upon from previous studies, and investigated the chondrogenic potential of NGF on bone marrow MSCs (BMSCs) both in vitro and in vivo. The results showed that NGF greatly upregulated the expression of cartilage-specific markers. When applied to cartilage repair for 4, 8 and 12 weeks, NGF-treated BMSCs have greater therapeutic effect than untreated BMSCs. Although inferior to TGF-β1 regarding its chondrogenic potential, NGF showed considerably lower expression of collagen type I, which is a fibrocartilage marker, and RUNX2, which is critical for terminal chondrocyte differentiation than TGF-β1, indicating its chondral specificity. Interestingly, NGF rarely induced BMSCs to differentiate into a neuronal phenotype, which may be due to the presence of other chondrogenic supplements. Furthermore, the underlying mechanism revealed that NGF-mediated chondrogenesis may be associated with the activation of PI3K/AKT and MAPK/ERK signaling pathways via the specific receptor of NGF, TrkA. In addition, NGF is easily accessed because of the abundance and low price of cobra venom, as well as the simplified methods for separation and purification. This study was the first to demonstrate the chondrogenic potential of NGF, which may provide a reference for cartilage regeneration in the clinic.
Bioorganic Chemistry | 2014
Zhenhui Lu; Shixiu Wei; Huayu Wu; Xiao Lin; Cuiwu Lin; Buming Liu; Li Zheng; Jinmin Zhao
Chondrocyte based therapy is promising to treat symptomatic chondral and osteochondral lesions. Growth factors to accelerate the proliferation and retain the phenotype of chondrocytes in vitro are imperative. However, the high cost and rapid degradation of growth factors limited their further application. Therefore, it is significant to find substitutes that can preserve chondrocytes phenotype and ensure sufficient cells for cytotherapy. Antioxidant and anti-inflammatory agents or their derivatives that have effect on arthritis may be an alternative. In this study, we synthesized sulfonamido-based gallate - LDQN-C and investigated its effect on rat articular chondrocytes through examination of the cell proliferation, morphology, viability, glycosaminoglycans (GAGs) synthesis and cartilage specific gene expression. Results showed that LDQN-C could enhance secretion and synthesis of cartilage extracellular matrix (ECM) by up-regulating expression levels of aggrecan, collagen II and Sox9 genes compared to the GA treated group and control group. Expression of collagen type II was effectively up-regulated while collagen I was down-regulated, which demonstrated that the inhibition of chondrocytes dedifferentiation by LDQN-C. Range of 1.36×10(-9)M to 1.36×10(-7)M is recommended dose of LDQN-C, among which the most profound response was observed with 1.36×10(-8)M. GA at concentration of 0.125μg/mL was compared. This study might provide a basis for the development of a novel agent for the treatment of articular cartilage defect.
PLOS ONE | 2015
Shixiu Wei; Zhenhui Lu; Yunfeng Zou; Xiao Lin; Cuiwu Lin; Buming Liu; Li Zheng; Jinmin Zhao
Gallic acid (GA) and its derivatives are anti-inflammatory agents reported to have an effect on osteoarthritis (OA). However, GA has much weaker anti-oxidant effects and inferior bioactivity compared with its derivatives. We modified GA with the introduction of sulfonamide to synthesize a novel compound named JEZ-C and analyzed its anti-arthritis and chondro-protective effects. Comparison of JEZ-C with its sources i.e. GA and Sulfamethoxazole (SMZ) was also performed. Results showed that JEZ-C could effectively inhibit the IL-1-mediated induction of MMP-1 and MMP-13 and could induce the expression of TIMP-1, which demonstrated its ability to reduce the progression of OA. JEZ-C can also exert chondro-protective effects by promoting cell proliferation and maintaining the phenotype of articular chondrocytes, as evidenced by improved cell growth, enhanced synthesis of cartilage specific markers such as aggrecan, collagen II and Sox9. Meanwhile, expression of the collagen I gene was effectively downregulated, revealing the inhibition of chondrocytes dedifferentiation by JEZ-C. Hypertrophy that may lead to chondrocyte ossification was also undetectable in JEZ-C groups. The recommended dose of JEZ-C ranges from 6.25×10-7 μg/ml to 6.25×10-5 μg/ml, among which the most profound response was observed with 6.25×10-6 μg/ml. In contrast, its source products of GA and SMZ have a weak effect not only in the inhibition of OA but also in the bioactivity of chondrocytes, which indicated the significance of this modification. This study revealed JEZ-C as a promising novel agent in the treatment of chondral and osteochondral lesions.
Cellular Physiology and Biochemistry | 2015
Zhenhui Lu; Liqin Wang; Hongmei Pan; Xiao Lin; Cuiwu Lin; Buming Liu; Li Zheng; Jinmin Zhao
Background: The phenotype of chondrocyte is easy to be lost when expanded in vitro by a process defined “dedifferentiation”. Traditional growth factors such as transforming growth factor (TGF-β1) are effective in preventing of dedifferentiation, but high costs and loss of activity limited their use. It is of significance to find substitutes which can reduce dedifferentiation and preserve chondrocytes phenotype to ensure sufficient differentiated cells for further study. Methods: We synthesized new type of sulfonamido-based gallates named ZXHA-C and investigated its effect on primary articular chondrocytes of rats. After preliminary screening by cytotoxicity test, ZXHA-C of 1.06 × 10-8, 1.06 × 10-7 and 1.06 × 10-6M were chosen for further studies. Cell proliferation, morphology, viability, GAG synthesis and cartilage specific gene expression were detected. Also the effects of ZXHA-C on Wnt/β-catenin signaling pathway were investigated. Results: ZXHA-C could significantly promote chondrocytes growth. And it could enhance ECM synthesis by up-regulating expression levels of cartilage specific markers like aggrecan, collagen II and Sox9. Expression of collagen I which marked chondrocytes dedifferentiation was also significantly down-regulated after treated by ZXHA-C. Further exploration of the molecular mechanism indicated that ZXHA-C activated the Wnt/β-catenin signal pathway in chondrocytes, as evidenced by up-regulated gene expression of β-catenin, Wnt-4, cyclin D1 and Frizzled-2 and decreased glycogen synthase kinase 3β (GSK-3β). Among the various concentrations, ZXHA-C of 1.06 × 10-7 M showed the best performance, which was close to positive control (group with TGF-β1). Conclusion: ZXHA-C might be potential a novel agent for the maintenances of chondrocytes phenotype.
Nano Research | 2018
Trever Todd; Zhenhui Lu; Jinmin Zhao; Benjamin Cline; Weizhong Zhang; Hongmin Chen; Anil Kumar; Wen Jiang; Franklin D. West; Samuel P. Franklin; Li Zheng; Jin Xie
Electrolytes can be taken orally or intravenously as supplements or therapeutics. However, their therapeutic window may exceed the serum toxicity threshold, making systemic delivery a poor option. Local injection is also not adequate due to rapid diffusion of electrolytes. Here, we solved this issue with a nanocapsule technology, comprising an electrolyte nanocrystal as the drug filling and a silica sheath to regulate drug release rates. In particular, we prepared LiF@SiO2 nanocapsules and investigated their potential as a delivery system for lithium, which was shown in recent studies to be an effective therapeutic agent for osteoarthritis (OA). We demonstrated that LiF@SiO2 can extend lithium release time from minutes to more than 60 h. After intraarticular (i.a.) injection into a rat OA model, the nanocapsules reduced the Osteoarthritis Research Society International (OARSI) score by 71% in 8 weeks while inducing no systemic toxicity. Our study opens new doors for improved delivery of electrolyte therapeutics, which have rarely been studied in the past.
ACS Chemical Biology | 2016
Zhenhui Lu; Huayu Wu; Xiao Lin; Buming Liu; Cuiwu Lin; Li Zheng; Jinmin Zhao
The effects of gallic acid (GA) on arthritis are limited by weak antioxidant effects and inferior biological properties of GA. We recently described a new series of synthesized GA derivatives by coupling with sulfonamides. Among these analogs, a novel compound synthesized from GA and sulfadimoxine (SDM) named ZXHA-TC exhibited the most robust anti-inflammatory potential. In this current study, the chondro-protective and antiarthritic effects of ZXHA-TC were investigated both in vitro and in vivo. In the in vitro study, ZXHA-TC exerted chondro-protective effects as evidenced by promoting cell proliferation and the maintaining of the phenotype of articular chondrocytes treated with interleukin-1-beta (IL-1β). The potential of ZXHA-TC to slow the progress of osteoarthritis (OA) was suggested by a reduction in matrix metalloproteinases (MMPs) and the up-regulation of the tissue inhibitor of metalloproteinase-1 (TIMP-1). In a rabbit anterior cruciate ligament transaction (ACLT) model of OA, ZXHA-TC exerted a protective effect on arthritis as assessed by macroscopic scores, histological, qRT-PCR, and immunohistochemical analyses. The effects of ZXHA-TC on inhibiting the production of inflammatory mediators in OA may be mediated partly by the suppression of the PI3K/AKT pathway or MAPK cascades, leading to NF-κB inactivation. Thus, this study indicates that ZXHA-TC may be developed as a potential therapeutic agent for OA.
Molecular Medicine Reports | 2018
Zhikang Miao; Zhenhui Lu; Shixing Luo; Danqing Lei; Yi He; Huayu Wu; Jinmin Zhao; Li Zheng
Mesenchymal stem cell (MSC)-based therapy has been commonly used in cartilage reconstruction, due to its self-renewing ability and multi-differentiation potential. Nerve growth factor (NGF) from cobra venom has been reported to regulate chondrogenesis of bone-derived MSCs (BMSCs) and chondrocyte metabolism. Therefore, the present study aimed to determine whether other sources of NGF behave in the same manner as NGF from natural venom. The present study compared the effects of NGF from two sources, the commercially purchased recombinant murine β-NGF (mNGF) and cobra venom-derived NGF (cvNGF), on chondrogenesis of BMSCs by performing hematoxylin and eosin and fluorescein diacetate/propidium iodide staining, DNA and glycosaminoglycan quantization and reverse transcription-quantitative polymerase chain reaction to investigate cell morphology, viability, proliferation, glycosaminoglycan synthesis and cartilage-specific gene expression. The results demonstrated that cvNGF significantly accelerated cell proliferation and upregulated the expression of cartilage-specific genes, including aggrecan, SRY-box 9 and collagen type II α1 chain. Conversely, cvNGF reduced the expression levels of collagen type I α1 chain (a fibrocartilage marker), runt-related transcription factor 2 and enolase 2 compared with in the mNGF and control groups. In addition, Chinese cobra venom, which is the main resource of cvNGF, is abundant and inexpensive, thus greatly decreasing the cost. In conclusion, the present study demonstrated that cvNGF may be considered a potential growth factor for inducing chondrogenic differentiation of BMSCs.