Network


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

Hotspot


Dive into the research topics where Zhenhua Feng is active.

Publication


Featured researches published by Zhenhua Feng.


International Immunopharmacology | 2017

Butein inhibits IL-1β-induced inflammatory response in human osteoarthritis chondrocytes and slows the progression of osteoarthritis in mice

Wenhao Zheng; Hui Zhang; Yonglong Jin; Quan Wang; Linwei Chen; Zhenhua Feng; Hua Chen; Yaosen Wu

Abstract Osteoarthritis (OA) is a progressive degenerative disease characterized by irreversible articular cartilage destruction. Butein, a polyphenolic compound isolated from the stem bark of cashews and Rhus verniciflua Stokes, has been reported to have anti‐inflammatory effects. This study aimed to assess the effect of butein on human OA chondrocytes and mice OA models induced by destabilization of the medial meniscus (DMM). In vitro, human OA chondrocytes were pretreated with butein at 10, 50 &mgr;M and subsequently stimulated with IL‐1&bgr; (10 ng/ml) for 24 h. Production of NO, PGE2, TNF‐&agr; and IL‐6 was evaluated by the Griess reaction and ELISAs. The mRNA expression of COX‐2, iNOS, TNF‐&agr;, IL‐6, MMP‐1, MMP‐3, MMP‐13, ADAMTS‐4, ADAMTS‐5, COL‐2 and SOX‐9 were measured by real‐time PCR. The protein expression of COX‐2, iNOS, MMP‐13, COL‐2, SOX‐9, p65 and I&kgr;B‐&agr; were detected by Western blot. P65 nuclear translocation was detected by immunofluorescence. In vivo, the severity of OA was determined by histological analysis. We found that butein significantly inhibited the IL‐1&bgr;‐induced production of NO and PGE2, expression of COX‐2, iNOS, TNF‐&agr;, IL‐6 and MMP‐13, degradation of COL‐2 and SOX‐9 at mRNA and protein levels as well as MMP‐1, MMP‐3, ADAMTS‐4 and ADAMTS‐5 gene expression. Furthermore, butein dramatically suppressed IL‐1&bgr;‐stimulated I&kgr;B‐&agr; degradation and NF‐kB p65 activation. In vivo, the cartilage in butein‐treated mice exhibited less Safranin O loss, cartilage erosion and lower OARSI scores. Butein also reduced subchondral bone plate thickness and alleviated synovitis. Taken together, these findings indicate that butein may be a potential agent in the treatment of OA. Graphical abstract Figure. No Caption available. HighlightsButein inhibited the IL‐1&bgr;‐induced inflammatory mediators in human osteoarthritis chondrocytes.Butein inhibited the IL‐1&bgr;‐induced degradation of type II collagen and SOX‐9 in human osteoarthritis chondrocytes.Butein inhibited the IL‐1&bgr;‐induced I&kgr;B‐&agr; degradation, NF‐kB p65 activation and NF‐&kgr;B p65 nuclear translocation in human osteoarthritis chondrocytes.Butein attenuated the arthritis in mice osteoarthritis models.


International Immunopharmacology | 2017

Fisetin inhibits IL-1β-induced inflammatory response in human osteoarthritis chondrocytes through activating SIRT1 and attenuates the progression of osteoarthritis in mice

Wenhao Zheng; Zhenhua Feng; Shengban You; Hui Zhang; Zhenyu Tao; Quan Wang; Hua Chen; Yaosen Wu

&NA; Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation and inflammation. Fisetin, a polyphenol extracted from fruits and vegetables, has been reported to have anti‐inflammatory effects. Our study aimed to investigate the effect of fisetin on OA both in vitro and in vivo. In vitro, chondrocytes were pretreated with fisetin alone or fisetin combined with sirtinol (an inhibitor of SIRT1) for 2 h before IL‐1&bgr; stimulation. Production of NO, PGE2, TNF‐&agr; and IL‐6 were evaluated by the Griess reaction and ELISAs. The mRNA (COX‐2, iNOS, MMP‐3, MMP‐13, ADAMTS‐5, Sox‐9, aggrecan and collagen‐II) and protein expression (COX‐2, iNOS, MMP‐3, MMP‐13, ADAMTS‐5 and SIRT1) were measured by qRT‐PCR and Western blot respectively. Immunofluorescence was used to assess the expression of collagen‐II and SIRT1. SIRT1 activity was quantified with SIRT1 fluorometric assay kit. The in vivo effect of fisetin was evaluated by gavage in mice OA models induced by destabilization of the medial meniscus (DMM). We found that fisetin inhibited IL‐1&bgr;‐induced expression of NO, PGE2, TNF‐&agr;, IL‐6, COX‐2, iNOS, MMP‐3, MMP‐13, ADAMTS‐5. Besides, fisetin remarkably decreased IL‐1&bgr;‐induced degradation of Sox‐9, aggrecan and collagen‐II. Furthermore, fisetin significantly inhibited IL‐1&bgr;‐induced SIRT1 decrease and inactivation. However, the inhibitory effect of fisetin was obvious abolished by sirtinol, suggesting that fisetin exerts anti‐inflammatory effects through activating SIRT1. In vivo, fisetin‐treated mice exhibited less cartilage destruction and lower OARSI scores. Moreover, fisetin reduced subchondral bone plate thickness and alleviated synovitis. Taken together, these findings indicate that fisetin may be a potential agent in the treatment of OA. Graphical abstract Figure. No caption available. HighlightsFisetin inhibited the IL‐1&bgr;‐induced inflammatory mediators in human osteoarthritis chondrocytes.Fisetin inhibited the IL‐1&bgr;‐induced degradation of Sox‐9, aggrecan and collagen‐II in human osteoarthritis chondrocytes.Fisetin inhibited the IL‐1&bgr;‐induced SIRT1 decrease and inactivation in human osteoarthritis chondrocytes.Sirtinol reversed the the inhibitory effect of fisetin on IL‐1&bgr;‐induced inflammatory response in human osteoarthritis chondrocytes.Fisetin alleviated the progression of osteoarthritis in mice models.


Cell Death and Disease | 2017

Trehalose ameliorates oxidative stress-mediated mitochondrial dysfunction and ER stress via selective autophagy stimulation and autophagic flux restoration in osteoarthritis development.

Qian Tang; Gang Zheng; Zhenhua Feng; Yu Chen; Yiting Lou; Chenggui Wang; Xiaolei Zhang; Yu Zhang; Hua-Zi Xu; Ping Shang; Haixiao Liu

Oxidative stress-related apoptosis and autophagy play crucial roles in the development of osteoarthritis (OA), a progressive cartilage degenerative disease with multifactorial etiologies. Here, we determined autophagic flux changes and apoptosis in human OA and tert-Butyl hydroperoxide (TBHP)-treated chondrocytes. In addition, we explored the potential protective effects of trehalose, a novel Mammalian Target of Rapamycin (mTOR)-independent autophagic inducer, in TBHP-treated mouse chondrocytes and a destabilized medial meniscus (DMM) mouse OA model. We found aberrant p62 accumulation and increased apoptosis in human OA cartilage and chondrocytes. Consistently, p62 and cleaved caspase-3 levels increased in mouse chondrocytes under oxidative stress. Furthermore, trehalose restored oxidative stress-induced autophagic flux disruption and targeted autophagy selectively by activating BCL2 interacting protein 3 (BNIP3) and Phosphoglycerate mutase family member 5 (PGAM5). Trehalose could ameliorate oxidative stress-mediated mitochondrial membrane potential collapse, ATP level decrease, dynamin-related protein 1 (drp-1) translocation into the mitochondria, and the upregulation of proteins involved in mitochondria and endoplasmic reticulum (ER) stress-related apoptosis pathway. In addition, trehalose suppressed the cleavage of caspase 3 and poly(ADP-ribose) polymerase (PARP) and prevented DNA damage under oxidative stress. However, the anti-apoptotic effects of trehalose in TBHP-treated chondrocytes were partially abolished by autophagic flux inhibitor chloroquine and BNIP3- siRNA. The protective effect of trehalose was also found in mouse OA model. Taken together, these results indicate that trehalose has anti-apoptotic effects through the suppression of oxidative stress-induced mitochondrial injury and ER stress which is dependent on the promotion of autophagic flux and the induction of selective autophagy. Thus, trehalose is a promising therapeutic agent for OA.


International Immunopharmacology | 2017

Cryptotanshinone protects against IL-1β-induced inflammation in human osteoarthritis chondrocytes and ameliorates the progression of osteoarthritis in mice.

Zhenhua Feng; Wenhao Zheng; Xiaobin Li; Jian Lin; Chenglong Xie; Hang Li; Liang Cheng; Aimin Wu; Wenfei Ni

Abstract Osteoarthritis (OA) is a common degenerative disease characterized by progressive erosion of articular cartilage, subchondral bone sclerosis and synovitis. Cryptotanshinone (CTS), an active component extracted from the root of Salvia miltiorrhiza Bunge, has been shown to have potent anti‐inflammatory effects. However, its effects on OA have not been clearly elucidated. This study aimed to assess the effect of CTS on human OA chondrocytes and mice OA models. Human OA chondrocytes were pretreated with CTS (5, 10 and 20 &mgr;M) for 2 h and subsequently stimulated with IL‐1&bgr; for 24 h. Production of NO, PGE2, IL‐6, TNF‐&agr; was evaluated by the Griess reaction and ELISA. The protein expression of COX‐2, iNOs, MMP‐3, MMP13, COX‐2, ADAMTS‐5, JNK, p‐JNK, ERK, p‐ERK, p38, p‐p38, p‐IKK&agr;/&bgr;, p65, p‐p65, I&kgr;B‐&agr;, and p‐I&kgr;B‐&agr; was tested by Western blot. In vivo, the severity of OA was determined by histological analysis. We found that CTS significantly inhibited the IL‐1&bgr;‐induced production of NO and PGE2; expression of COX‐2, iNOS, MMP‐3, MMP‐13, and ADAMTS‐5. Furthermore, CTS in dramatically suppressed IL‐1&bgr;‐stimulated NF‐&kgr;B and MAPK activation. Immunofluorescence staining demonstrated that CTS could suppress IL‐1&bgr;‐induced phosphorylation of p65 nuclear translocation. In vivo, treatment of CTS prevented the destruction of cartilage and the thickening of subchondral bone in mice OA models. These results indicate that the therapeutic effect of CTS on OA is accomplished through the inhibition of both NF‐&kgr;B and MAPK signaling pathways. Our findings provide the evidence to develop CTS as a potential therapeutic agent f or patients with OA. HighlightsCryptotanshinone inhibited the IL‐1&bgr;‐induced inflammatory mediators in human osteoarthritis chondrocytes.Cryptotanshinone inhibited IL‐1&bgr;‐induced inflammatory response through suppressing NF‐&kgr;B and MAPK activation.Cryptotanshinone alleviated the progression of osteoarthritis in mice models.


Oncotarget | 2017

Wogonoside inhibits IL-1β induced catabolism and hypertrophy in mouse chondrocyte and ameliorates murine osteoarthritis

Qian Tang; Gang Zheng; Zhenhua Feng; Min-Ji Tong; Jianxiang Xu; Zhiyan Hu; Ping Shang; Yu Chen; Chenggui Wang; Yiting Lou; Deheng Chen; Di Zhang; Majid Nisar; Xiaolei Zhang; Hua-Zi Xu; Haixiao Liu

The inflammatory environment is correlated with extracellular matrix (ECM) degradation and chondrocyte hypertrophy in the development of osteoarthritis (OA). Previous studies have reported the anti-inflammatory effects of wogonoside in several diseases. In the present study, we investigated the protective effects of wogonoside in relation to the development of OA and delineated the potential mechanism. In vitro, wogonoside decreased the production of pro-inflammatory cytokines like Nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6). It also inhibited the expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) both at gene and protein levels. Wogonoside also inhibited hypertrophy and the generation of vascular endothelial growth factor (VEGF) in interleukin-1β (IL-1β)-induced chondrocytes. Moreover, wogonoside promoted the expression of anabolic factors Sox-9, type two collagen and aggrecan while inhibiting the expression of catabolic factors such as matrix metalloproteinases (MMPs) and thrombospondin motifs 5 (ADAMTS-5) in mouse chondrocytes. Mechanistically, we found that wogonoside inhibited nuclear factor kappa B/ hypoxia-inducible factor two alpha (NF-κB/HIF-2α) activation via the phosphatidylinositol 3 kinase (PI3K) /AKT pathway. The protective effects of wogonoside were also observed in vivo and the pharmacokinetic results of wogonoside indicated that good systemic exposure was achievable after oral administration of wogonoside. In conclusion, our stduy demonstrates that wogonoside attenuates IL-1β-induced ECM degradation and hypertrophy in mouse chondrocytes via suppressing the activation of NF-κB/HIF-2α by the PI3K/AKT pathway. Moreover, wogonoside ameliorates OA progression in vivo, indicating that wogonoside may serve as a promising therapeutic agent for the treatment of OA.The inflammatory environment is correlated with extracellular matrix (ECM) degradation and chondrocyte hypertrophy in the development of osteoarthritis (OA). Previous studies have reported the anti-inflammatory effects of wogonoside in several diseases. In the present study, we investigated the protective effects of wogonoside in relation to the development of OA and delineated the potential mechanism. In vitro, wogonoside decreased the production of pro-inflammatory cytokines like Nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6). It also inhibited the expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) both at gene and protein levels. Wogonoside also inhibited hypertrophy and the generation of vascular endothelial growth factor (VEGF) in interleukin-1β (IL-1β)-induced chondrocytes. Moreover, wogonoside promoted the expression of anabolic factors Sox-9, type two collagen and aggrecan while inhibiting the expression of catabolic factors such as matrix metalloproteinases (MMPs) and thrombospondin motifs 5 (ADAMTS-5) in mouse chondrocytes. Mechanistically, we found that wogonoside inhibited nuclear factor kappa B/ hypoxia-inducible factor two alpha (NF-κB/HIF-2α) activation via the phosphatidylinositol 3 kinase (PI3K) /AKT pathway. The protective effects of wogonoside were also observed in vivo and the pharmacokinetic results of wogonoside indicated that good systemic exposure was achievable after oral administration of wogonoside. In conclusion, our stduy demonstrates that wogonoside attenuates IL-1β-induced ECM degradation and hypertrophy in mouse chondrocytes via suppressing the activation of NF-κB/HIF-2α by the PI3K/AKT pathway. Moreover, wogonoside ameliorates OA progression in vivo, indicating that wogonoside may serve as a promising therapeutic agent for the treatment of OA.


Oncotarget | 2017

Silibinin protects against osteoarthritis through inhibiting the inflammatory response and cartilage matrix degradation in vitro and in vivo

Wenhao Zheng; Zhenhua Feng; Yiting Lou; Chunhui Chen; Chuanxu Zhang; Zhenyu Tao; Hang Li; Liang Cheng; Xiaozhou Ying

Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation and inflammation. Silibinin, a polyphenolic flavonoid derived from fruits and seeds of Silybum marianum, has been reported to possess various potent beneficial biological effects, such as antioxidant, anti-cancer, hepatoprotective and anti-inflammatory activities. However, the anti-inflammatory effects of silibinin on OA have not been reported. This study aimed to assess the effects of silibinin on OA both in vitro and in vivo. In this study, we found that silibinin significantly inhibited the nterleukin-1β (IL-1β)-induced production of nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α) and IL-6, expression of cyclooxygenase2 (COX-2), inducible nitric oxide synthase (iNOS), matrix metalloproteinase-1 (MMP-1), MMP-3, MMP-13, a disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4) and ADAMTS-5, degradation of aggrecan and collagen-II in human OA chondrocytes. Furthermore, silibinin dramatically suppressed IL-1β-stimulated phosphatidylinositol 3 kinase/ protein kinase B (PI3K/Akt) phosphorylation and nuclear factor-kappa B (NF-kB) activation in human OA chondrocytes. In addition, treatment of silibinin not only prevented the destruction of cartilage and the thickening of subchondral bone but also relieved synovitis in mice OA models. Also, the immunohistochemistry results showed that silibinin significantly decreased the expression of MMP-13 and ADAMTS-5 and increased the expression of collagen-II and aggrecan in mice OA. Taken together, these results suggest that silibinin may be a potential agent in the treatment of OA.


Journal of Investigative Surgery | 2017

Comparison of the Conventional Surgery and the Surgery Assisted by 3d Printing Technology in the Treatment of Calcaneal Fractures

Wenhao Zheng; Zhenyu Tao; Yiting Lou; Zhenhua Feng; Hang Li; Liang Cheng; Hui Zhang; Jianshun Wang; Xiaoshan Guo; Hua Chen

ABSTRACT Purpose: This study was aimed to compare conventional surgery and surgery assisted by 3D printing technology in the treatment of calcaneal fractures. In addition, we also investigated the effect of 3D printing technology on the communication between doctors and patients. Methods: we enrolled 75 patients with calcaneal fracture from April 2014 to August 2016. They were divided randomly into two groups: 35 cases of 3D printing group, 40 cases of conventional group. The individual models were used to simulate the surgical procedures and carry out the surgery according to plan in 3D printing group. Operation duration, blood loss volume during the surgery, number of intraoperative fluoroscopy and fracture union time were recorded. The radiographic outcomes Böhler angle, Gissane angle, calcaneal width and calcaneal height and final functional outcomes including VAS and AOFAS score as well as the complications were also evaluated. Besides, we made a simple questionnaire to verify the effectiveness of the 3D-printed model for both doctors and patients. Results: The operation duration, blood loss volume and number of intraoperative fluoroscopy for 3D printing group was 71.4 ± 6.8 minutes, 226.1 ± 22.6 ml and 5.6 ± 1.9 times, and for conventional group was 91.3 ± 11.2 minutes, 288.7 ± 34.8 ml and 8.6 ± 2.7 times respectively. There was statistically significant difference between the conventional group and 3D printing group (p < 0.05). Additionally, 3D printing group achieved significantly better radiographic results than conventional group both postoperatively and at the final follow-up (p < 0.05). However, No significant difference was noted in the final functional outcomes between the two groups. As for complications, there was no significant difference between the two groups. Furthermore, the questionnaire showed that both doctors and patients exhibited high scores of overall satisfaction with the use of a 3D printing model. Conclusion: This study suggested the clinical feasibility of 3D printing technology in treatment of calcaneal fractures.


Journal of Cellular and Molecular Medicine | 2017

Salidroside attenuates neuroinflammation and improves functional recovery after spinal cord injury through microglia polarization regulation

Chenggui Wang; Qingqing Wang; Yiting Lou; Jianxiang Xu; Zhenhua Feng; Yu Chen; Qian Tang; Gang Zheng; Zengjie Zhang; Yaosen Wu; Nai-Feng Tian; Yifei Zhou; Hua-Zi Xu; Xiaolei Zhang

Spinal cord injury (SCI) is a severe neurological disease; however, few drugs have been proved to treat SCI effectively. Neuroinflammation is the major pathogenesis of SCI secondary injury and considered to be the therapeutic target of SCI. Salidroside (Sal) has been reported to exert anti‐inflammatory effects in airway, adipose and myocardial tissue; however, the role of Sal in SCI therapeutics has not been clarified. In this study, we showed that Sal could improve the functional recovery of spinal cord in rats as revealed by increased BBB locomotor rating scale, angle of incline, and decreased cavity of spinal cord injury and apoptosis of neurons in vivo. Immunofluorescence double staining of microglia marker and M1/M2 marker demonstrated that Sal could suppress M1 microglia polarization and activate M2 microglia polarization in vivo. To verify how Sal exerts its effects on microglia polarization and neuron protection, we performed the mechanism study in vitro in microglia cell line BV‐2 and neuron cell line PC12. The results showed that Sal prevents apoptosis of PC12 cells in coculture with LPS‐induced M1 BV‐2 microglia, also the inflammatory secretion phenotype of M1 BV‐2 microglia was suppressed by Sal, and further studies demonstrated that autophagic flux regulation through AMPK/mTOR pathway was involved in Sal regulated microglia polarization after SCI. Overall, our study illustrated that Sal could promote spinal cord injury functional recovery in rats, and the mechanism may relate to its microglia polarization modulation through AMPK‐/mTOR‐mediated autophagic flux stimulation.


The American Journal of Chinese Medicine | 2017

Endoplasmic Reticulum Stress and NF-κB Pathway in Salidroside Mediated Neuroprotection: Potential of Salidroside in Neurodegenerative Diseases

Chenggui Wang; Yiting Lou; Jianxiang Xu; Zhenhua Feng; Yu Chen; Qian Tang; Qingqing Wang; Haiming Jin; Yaosen Wu; Nai-Feng Tian; Yifei Zhou; Hua-Zi Xu; Xiaolei Zhang

Microglial activation leads to increased production of proinflammatory enzymes and cytokines, which is considered to play crucial role in neurodegenerative diseases, however there are only a few drugs that target microglia activation. Recent studies have indicated that the Traditional Chinese Medicine, salidroside (Sal), exerted anti-inflammatory effects. According to this evidence, our present study aims to explore the effect of the Sal (a phenylpropanoid glycoside compound which is isolated from rhodiola), on microglia activation in lipopolysaccharide (LPS)-stimulated BV-2 cells. Our results showed that Sal could significantly inhibit the excessive production of Nitric Oxide (NO) and Prostaglandin E2 (PGE2) in LPS-stimulated BV2 cells. Moreover, Sal treatment could suppress the mRNA and protein expressions of inflammatory enzymes, including Inducible Nitric Oxide Synthase (iNOS) and Cyclooxygenase-2 (COX-2). The mechanisms may be related to the inhibition of the activation of Nuclear Factor-kappaB (NF-[Formula: see text]B) and endoplasmic reticulum stress. Our study demonstrated that salidroside could inhibit lipopolysaccharide-induced microglia activation via the inhibition of the NF-[Formula: see text]B pathway and endoplasmic reticulum stress, which makes it a promising therapeutic agent for human neurodegenerative diseases.


BioMed Research International | 2017

The Outcomes of Minimally Invasive versus Open Posterior Approach Spinal Fusion in Treatment of Lumbar Spondylolisthesis: The Current Evidence from Prospective Comparative Studies

Ai-Min Wu; Chun-Hui Chen; Zhi-Hao Shen; Zhenhua Feng; Wan-Qing Weng; Shu-Min Li; Yong-Long Chi; Li-Hui Yin; Wen-Fei Ni

Purpose. To investigate the evidence of minimally invasive (MI) versus open (OP) posterior lumbar fusion in treatment of lumbar spondylolisthesis from current prospective literatures. Methods. The electronic literature database of Pubmed, Embase, and Cochrane library was searched at April 2016. The data of operative time, estimated blood loss and length of hospital stay, visual analog scale (VAS) of both lower back pain and leg pain, Oswestry disability index (ODI), SF-36 PCS (physical component scores) and SF-36 MCS (mental component scores), complications, fusion rate, and secondary surgery were extracted and analyzed by STATA 12.0 software. Results. Five nonrandom prospective comparative studies were included in this meta-analysis. The meta-analysis showed that the MI group had a significantly longer operative time than OP group, less blood loss, and shorter hospital stay. No significant difference was found in back pain, leg pain, ODI, SF-36 PCS, SF-36 MCS, complications, fusion rate, and secondary surgery between MI and OP groups. Conclusion. The prospective evidence suggested that MI posterior fusion for spondylolisthesis had less EBL and hospital stay than OP fusion; however it took more operative time. Both MI and OP fusion had similar results in pain and functional outcomes, complication, fusion rate, and secondary surgery.

Collaboration


Dive into the Zhenhua Feng's collaboration.

Top Co-Authors

Avatar

Wenhao Zheng

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Xiaobin Li

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Wen-Fei Ni

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Hui Zhang

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Ai-Min Wu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Qian Tang

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Yaosen Wu

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Yiting Lou

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Zhi-Chao Hu

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge